1
|
Osorio R, Rodríguez-Lozano FJ, Toledano M, Toledano-Osorio M, García-Bernal D, Murcia L, López-García S. Mitigating lipopolysaccharide-induced impairment in human dental pulp stem cells with tideglusib-doped nanoparticles: Enhancing osteogenic differentiation and mineralization. Dent Mater 2024; 40:1591-1601. [PMID: 39068091 DOI: 10.1016/j.dental.2024.07.012] [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: 06/17/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVE Drug-loaded non-resorbable polymeric nanoparticles (NPs) are proposed as an adjunctive treatment for pulp regenerative strategies. The present in vitro investigation aimed to evaluate the effectiveness of tideglusib-doped nanoparticles (TDg-NPs) in mitigating the adverse effects of bacterial lipopolysaccharide endotoxin (LPS) on the viability, morphology, migration, differentiation and mineralization potential of human dental pulp stem cells (hDPSCs). METHODS Cell viability, proliferation, and differentiation were assessed using a MTT assay, cell migration evaluation, cell cytoskeleton staining analysis, Alizarin Red S staining and expression of the odontogenic related genes by a real-time quantitative polymerase chain reaction (RT-qPCR) were also performed. Cells were tested both with and without stimulation with LPS at various time points. One-way ANOVA and Tukey's test were employed for statistical analysis (p < 0.05). RESULTS Adequate cell viability was encountered in all groups and at every tested time point (24, 48, 72 and 168 h), without differences among the groups (p > 0.05). The analysis of cell cytoskeleton showed nuclear alteration in cultures with undoped NPs after LPS stimulation. These cells exhibited an in blue diffuse and multifocal appearance. Some nuclei looked fragmented and condensed. hDPSCs after LPS stimulation but in the presence of TDg-NPs exhibited less nuclei changes. LPS induced down-regulation of Alkaline phosphatase, Osteonectin and Collagen1 gene markers, after 21d. LPS half-reduced the cells production of calcium deposits in all groups (p < 0.05), except in the group with TDg-NPs (decrease about 10 %). SIGNIFICANCE LPS induced lower mineral deposition and cytoskeletal disorganization in hDPSCs. These effects were counteracted by TDg-NPs, enhancing osteogenic differentiation and mineralization.
Collapse
Affiliation(s)
- Raquel Osorio
- Faculty of Dentistry, University of Granada Colegio Máximo de Cartuja s/n, Granada 18071, Spain
| | - Francisco J Rodríguez-Lozano
- Department of Dermatology, Stomatology, Radiology and Physical Medicine, Morales Meseguer Hospital, Biomedical Research Institute (IMIB), Regional Campus of International Excellence "Campus Mare Nostrum", Faculty of Medicine, University of Murcia, Murcia 30008, Spain
| | - Manuel Toledano
- Faculty of Dentistry, University of Granada Colegio Máximo de Cartuja s/n, Granada 18071, Spain.
| | - Manuel Toledano-Osorio
- Postgraduate Program of Specialization in Periodontology, Faculty of Dentistry, University Complutense of Madrid, Madrid, Spain
| | - David García-Bernal
- Department of Biochemistry, Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Biomedical Research Institute (IMIB), Murcia 30120, Spain
| | - Laura Murcia
- Department of Health Sciences, Catholic University San Antonio of Murcia, Murcia 30107, Spain
| | - Sergio López-García
- Department of Dermatology, Stomatology, Radiology and Physical Medicine, Morales Meseguer Hospital, Biomedical Research Institute (IMIB), Regional Campus of International Excellence "Campus Mare Nostrum", Faculty of Medicine, University of Murcia, Murcia 30008, Spain
| |
Collapse
|
2
|
Uchendu AP, Omogbai EK, Obarisiagbon PA, Omogiade UG, Bafor EE. Chlorophyll Derivatives Exert Greater Potency Over Progesterone in the Prevention of Infection-Induced Preterm Birth in Murine Models. Am J Reprod Immunol 2024; 92:e70000. [PMID: 39422053 DOI: 10.1111/aji.70000] [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: 04/21/2024] [Revised: 09/12/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
PROBLEM Preterm birth (PTB) is a significant cause of maternal and neonatal morbidity and mortality worldwide. However, the effectiveness of progesterone (P4) which is clinically used for PTB management remains controversial and necessitates research into new therapeutic options METHOD OF STUDY: In the current study, we investigated the effectiveness of two chlorophyll derivatives, pheophorbide a (PBa) and pheophytin a (PTa), in counteracting PTB. Timed-pregnant mice (gestation day 17 ± 0.5) received lipopolysaccharide (LPS) (25 µg/mouse) or phosphate-buffered saline (PBS) intraperitoneally, with PBa, PTa, progesterone (P4), and co-administration of P4 and ibuprofen (IBP), administered orally 2 h prior. RESULTS The LPS group experienced PTB and 100% fetal mortality, whereas the PBa and PTa groups showed a delayed onset of LPS-induced PTB, with significantly decreased PTB rate and fetal mortality. In addition, PBa and PTa suppressed LPS-induced pro-inflammatory cytokines and NF-κB transcription factor while increasing anti-inflammatory cytokines in the placenta and uterus. CONCLUSIONS Our findings indicate that the chlorophyll derivatives, PBa and PTa increase fetal survival in infection-induced PTB and demonstrate greater efficacy than P4 in preventing PTB.
Collapse
Affiliation(s)
- Adaeze P Uchendu
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Eric K Omogbai
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Philip A Obarisiagbon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Uyi G Omogiade
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Enitome E Bafor
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, USA
| |
Collapse
|
3
|
Mehrani R, Mondal J, Ghazanfari D, Goetz DJ, McCall KD, Bergmeier SC, Sharma S. Capturing the Effects of Single Atom Substitutions on the Inhibition Efficiency of Glycogen Synthase Kinase-3β Inhibitors via Markov State Modeling and Experiments. J Chem Theory Comput 2024; 20:6278-6286. [PMID: 38975986 DOI: 10.1021/acs.jctc.4c00311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Small modifications in the chemical structure of ligands are known to dramatically change their ability to inhibit the activity of a protein. Unraveling the mechanisms that govern these dramatic changes requires scrutinizing the dynamics of protein-ligand binding and unbinding at the atomic level. As an exemplary case, we have studied Glycogen Synthase Kinase-3β (GSK-3β), a multifunctional kinase that has been implicated in a host of pathological processes. As such, there is a keen interest in identifying ligands that inhibit GSK-3β activity. One family of compounds that are highly selective and potent inhibitors of GSK-3β is exemplified by a molecule termed COB-187. COB-187 consists of a five-member heterocyclic ring with a thione at C2, a pyridine substituted methyl at N3, and a hydroxyl and phenyl at C4. We have studied the inhibition of GSK-3β by COB-187-related ligands that differ in a single heavy atom from each other (either in the location of nitrogen in their pyridine ring, or with the pyridine ring replaced by a phenyl ring), or in the length of the alkyl group joining the pyridine and the N3. The inhibition experiments show a large range of half-maximal inhibitory concentration (IC50) values from 10 nM to 10 μM, implying that these ligands exhibit vastly different propensities to inhibit GSK-3β. To explain these differences, we perform Markov State Modeling (MSM) using fully atomistic simulations. Our MSM results are in excellent agreement with the experiments in that they accurately capture differences in the binding propensities of the ligands. The simulations show that the binding propensities are related to the ligands' ability to attain a compact conformation where their two aromatic rings are spatially close. We rationalize this result by sampling numerous binding and unbinding events via funnel metadynamics simulations, which show that indeed while approaching the bound state, the ligands prefer to be in their compact conformation. We find that the presence of nitrogen in the aromatic ring increases the probability of attaining the compact conformation. Protein-ligand binding is understood to be dictated by the energetics of interactions and entropic factors, like the release of bound water from the binding pockets. This work shows that changes in the conformational distribution of ligands due to atom-level modifications in the structure play an important role in protein-ligand binding.
Collapse
Affiliation(s)
- Ramin Mehrani
- Department of Mechanical Engineering, Ohio University, Athens, Ohio 45701, United States
| | - Jagannath Mondal
- Center for Interdisciplinary Sciences, Tata Institute of Fundamental Research, Hyderabad 500046, India
| | - Davoud Ghazanfari
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio 45701, United States
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio 45701, United States
- Biomedical Engineering Program, Ohio University, Athens, Ohio 45701, United States
| | - Kelly D McCall
- Biomedical Engineering Program, Ohio University, Athens, Ohio 45701, United States
- Department of Specialty Medicine, Ohio University, Athens, Ohio 45701, United States
- The Diabetes Institute, Ohio University, Athens, Ohio 45701, United States
- Molecular and Cellular Biology Program, Ohio University, Athens, Ohio 45701, United States
- Translational Biomedical Sciences Program, Ohio University, Athens, Ohio 45701, United States
| | - Stephen C Bergmeier
- Biomedical Engineering Program, Ohio University, Athens, Ohio 45701, United States
- Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio 45701, United States
| | - Sumit Sharma
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio 45701, United States
| |
Collapse
|
4
|
Ghazanfari D, Courreges MC, Belinski LE, Hogrell MJ, Lloyd J, C Bergmeier S, McCall KD, Goetz DJ. Mechanistic insights into SARS-CoV-2 spike protein induction of the chemokine CXCL10. Sci Rep 2024; 14:11179. [PMID: 38750069 PMCID: PMC11096305 DOI: 10.1038/s41598-024-61906-6] [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: 01/20/2023] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
During a SARS-CoV-2 infection, macrophages recognize viral components resulting in cytokine production. While this response fuels virus elimination, overexpression of cytokines can lead to severe COVID-19. Previous studies suggest that the spike protein (S) of SARS-CoV-2 can elicit cytokine production via the transcription factor NF-κB and the toll-like receptors (TLRs). In this study, we found that: (i) S and the S2 subunit induce CXCL10, a chemokine implicated in severe COVID-19, gene expression by human macrophage cells (THP-1); (ii) a glycogen synthase kinase-3 inhibitor attenuates this induction; (iii) S and S2 do not activate NF-κB but do activate the transcription factor IRF; (iv) S and S2 do not require TLR2 to elicit CXCL10 production or activate IRF; and (v) S and S2 elicit CXCL10 production by peripheral blood mononuclear cells (PBMCs). We also discovered that the cellular response, or lack thereof, to S and S2 is a function of the recombinant S and S2 used. While such a finding raises the possibility of confounding LPS contamination, we offer evidence that potential contaminating LPS does not underly induced increases in CXCL10. Combined, these results provide insights into the complex immune response to SARS-CoV-2 and suggest possible therapeutic targets for severe COVID-19.
Collapse
Affiliation(s)
- Davoud Ghazanfari
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | | | - Lydia E Belinski
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Michael J Hogrell
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Jacob Lloyd
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Stephen C Bergmeier
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, USA
| | - Kelly D McCall
- Department of Specialty Medicine, Ohio University, Athens, OH, 45701, USA
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- The Diabetes Institute, Ohio University, Athens, OH, 45701, USA
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, 45701, USA
- Department of Biological Sciences, Ohio University College of Arts & Sciences, Athens, OH, 45701, USA
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, 45701, USA
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA.
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
5
|
Grayck MR, McCarthy WC, Solar M, Golden E, Balasubramaniyan N, Zheng L, Sherlock LG, Wright CJ. GSK3β/NF-κB -dependent transcriptional regulation of homeostatic hepatocyte Tnf production. Am J Physiol Gastrointest Liver Physiol 2024; 326:G374-G384. [PMID: 38193163 PMCID: PMC11211040 DOI: 10.1152/ajpgi.00229.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/18/2023] [Accepted: 12/23/2023] [Indexed: 01/10/2024]
Abstract
Maintenance of hepatocyte homeostasis plays an important role in mediating the pathogenesis of many diseases. A growing body of literature has established a critical role played by tumor necrosis factor-α (TNFα) in maintaining hepatocyte homeostasis; however, the transcriptional mechanisms underlying constitutive Tnf expression are unknown. Whole liver fractions and primary hepatocytes from adult control C57BL/6 mice and the murine hepatocyte cell line AML12 were assessed for constitutive Tnf expression. Impacts of glycogen synthase kinase-3 β (GSK3β) and nuclear factor κB (NF-κB) inhibition on constitutive Tnf expression were assessed in AML12 cells. Finally, AML12 cell proliferation following GSK3β and NF-κB inhibition was evaluated. Constitutive Tnf gene expression is present in whole liver, primary hepatocytes, and cultured AML12 hepatocytes. Cytokine-induced Tnf gene expression is regulated by NF-κB activation. Pharmacological inhibition of GSK3β resulted in a time- and dose-dependent inhibition of Tnf gene expression. GSK3β inhibition decreased nuclear levels of the NF-κB subunits p65 and p50. We determined that NF-κB transcription factor subunit p65 binds to consensus sequence elements present in the murine TNFα promoter and inhibition of GSK3β decreases binding and subsequent Tnf expression. Finally, AML12 cell growth was significantly reduced following GSK3β and NF-κB inhibition. These results demonstrate that GSK3β and NF-κB are essential for mediating Tnf expression and constitutive hepatocyte cell growth. These findings add to a growing body of literature on TNFα mediated hepatocyte homeostasis and identify novel molecular mechanisms involved in mediating response to various disease states in the liver.NEW & NOTEWORTHY Maintenance of hepatocyte homeostasis plays an important role in controlling the pathogenesis of many diseases. Our findings add to a growing body of literature on tumor necrosis factor-α (TNFα)-mediated hepatocyte homeostasis and identify novel molecular mechanisms involved in regulating this response.
Collapse
Affiliation(s)
- Maya R Grayck
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - William C McCarthy
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Mack Solar
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Emma Golden
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Natarajan Balasubramaniyan
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Lijun Zheng
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Laura G Sherlock
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| |
Collapse
|
6
|
Chen H, Zhang GX, Zhou XY. Identification of hub genes associated with Helicobacter pylori infection and type 2 diabetes mellitus: A pilot bioinformatics study. World J Diabetes 2024; 15:170-185. [PMID: 38464370 PMCID: PMC10921168 DOI: 10.4239/wjd.v15.i2.170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/21/2023] [Accepted: 12/27/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection is related to various extragastric diseases including type 2 diabetes mellitus (T2DM). However, the possible mechanisms connecting H. pylori infection and T2DM remain unknown. AIM To explore potential molecular connections between H. pylori infection and T2DM. METHODS We extracted gene expression arrays from three online datasets (GSE60427, GSE27411 and GSE115601). Differentially expressed genes (DEGs) commonly present in patients with H. pylori infection and T2DM were identified. Hub genes were validated using human gastric biopsy samples. Correlations between hub genes and immune cell infiltration, miRNAs, and transcription factors (TFs) were further analyzed. RESULTS A total of 67 DEGs were commonly presented in patients with H. pylori infection and T2DM. Five significantly upregulated hub genes, including TLR4, ITGAM, C5AR1, FCER1G, and FCGR2A, were finally identified, all of which are closely related to immune cell infiltration. The gene-miRNA analysis detected 13 miRNAs with at least two gene cross-links. TF-gene interaction networks showed that TLR4 was coregulated by 26 TFs, the largest number of TFs among the 5 hub genes. CONCLUSION We identified five hub genes that may have molecular connections between H. pylori infection and T2DM. This study provides new insights into the pathogenesis of H. pylori-induced onset of T2DM.
Collapse
Affiliation(s)
- Han Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210003, Jiangsu Province, China
| | - Guo-Xin Zhang
- Department of Gastroenterology, Jiangsu Province Hospital, Nanjing 210029, Jiangsu Province, China
| | - Xiao-Ying Zhou
- Department of Gastroenterology, Jiangsu Province Hospital, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
7
|
Chan M, Kang Y, Osborne S, Zager M, Gujral TS. A kinase to cytokine explorer to identify molecular regulators and potential therapeutic opportunities. eLife 2024; 12:RP91472. [PMID: 38305363 PMCID: PMC10945549 DOI: 10.7554/elife.91472] [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] [Indexed: 02/03/2024] Open
Abstract
Cytokines and chemokines are secreted proteins that regulate various biological processes, such as inflammation, immune response, and cell differentiation. Therefore, disruption of signaling pathways involving these proteins has been linked to a range of diseases, including cancer. However, targeting individual cytokines, chemokines, or their receptors is challenging due to their regulatory redundancy and incomplete understanding of their signaling networks. To transform these difficult-to-drug targets into a pharmacologically manageable class, we developed a web-based platform called KinCytE. This platform was designed to link the effects of kinase inhibitors, a well-established class of drugs, with cytokine and chemokine release and signaling networks. The resulting KinCytE platform enables users to investigate protein kinases that regulate specific cytokines or chemokines, generate a ranked list of FDA-approved kinase inhibitors that affect cytokine/chemokine activity, and explore and visualize cytokine signaling network thus facilitating drugging this challenging target class. KinCytE is freely accessible via https://atlas.fredhutch.org/kincyte.
Collapse
Affiliation(s)
- Marina Chan
- Human Biology Division, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Yuqi Kang
- Human Biology Division, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Shannon Osborne
- Data Visualization Core, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Michael Zager
- Data Visualization Core, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Taranjit S Gujral
- Human Biology Division, Fred Hutchinson Cancer CenterSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| |
Collapse
|
8
|
Yu J, Hu G, Guo X, Cao H, Zhang C. Quercetin Alleviates Inflammation and Energy Deficiency Induced by Lipopolysaccharide in Chicken Embryos. Animals (Basel) 2023; 13:2051. [PMID: 37443849 DOI: 10.3390/ani13132051] [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: 04/05/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
Energy deficiency causes multiple organ dysfunctions after LPS induction. Quercetin is a phenolic compound found in herbal medicines. However, the effects of quercetin in alleviating LPS-induced energy deficiency remain unclear. In the present study, an in vivo LPS-induced inflammation model was established in chicken embryos. Specific pathogen-free chicken embryos (n = 120) were allocated to control, PBS with or without ethanol, quercetin (10, 20, or 40 nmol, respectively), and LPS (125 ng/egg) with or without quercetin groups. Fifteen day old embryonated eggs were injected with the abovementioned solutions via the allantoic cavity. On embryonic day 19, the tissues of the embryos were collected for histopathological examination using frozen oil red O staining, RNA extraction, real-time quantitative polymerase chain reaction, and immunohistochemical investigations. The glycogen and lipid contents in the liver increased after LPS stimulation as compared with the PBS group, whereas quercetin decreased the accumulation as compared with the LPS group. The mRNA expressions of AMPKα1 and AMPKα2 in the duodena, ceca, and livers were upregulated after LPS induction as compared with the PBS group, while quercetin could downregulate these expressions as compared with the LPS group. The immunopositivity of AMPKα2 in the villus, crypt, lamina propria, tunica muscularis, and myenteric plexus in the duodena and in the cytoplasms of hepatocytes significantly increased after LPS induction when compared with the PBS group (p < 0.01), whereas the immunopositivity to AMPKα2 in the quercetin treatment group significantly decreased when compared with the LPS group (p < 0.01 or p < 0.05). The LPS-induced high expressions of transcription factor PPARα and glucose transporter (SGLT1) were blocked by quercetin in the duodena, ceca, and livers. Quercetin treatment improved the LPS-induced decrease in APOA4 in the duodena, ceca, and livers. The mRNA expression of PEPT1 in the duodena and ceca increased after LPS challenge, whereas quercetin could downregulate PEPT1 gene expression. These data demonstrate that quercetin improved the energy deficiency induced by LPS in chicken embryos. The LPS-induced inflammation model was established to avoid the effect of LPS exposure from the environment and intestinal flora. The results form the basis the administration of quercetin pretreatment (in ovo infection) to improve the energy state of chicken embryos and improve the inflammation response.
Collapse
Affiliation(s)
- Jinhai Yu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
9
|
Patients with Infections of The Central Nervous System Have Lowered Gut Microbiota Alpha Diversity. Curr Issues Mol Biol 2022; 44:2903-2914. [PMID: 35877424 PMCID: PMC9318043 DOI: 10.3390/cimb44070200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
There are multiple lines of evidence for the existence of communication between the central nervous system (CNS), gut, and intestinal microbiome. Despite extensive analysis conducted on various neurological disorders, the gut microbiome was not yet analyzed in neuroinfections. In the current study, we analyzed the gut microbiome in 47 consecutive patients hospitalized with neuroinfection (26 patients had viral encephalitis/meningitis; 8 patients had bacterial meningitis) and in 20 matched for age and gender health controls. Using the QIIME pipeline, 16S rRNA sequencing and classification into operational taxonomic units (OTUs) were performed on the earliest stool sample available. Bacterial taxa such as Clostridium, Anaerostipes, Lachnobacterium, Lachnospira, and Roseburia were decreased in patients with neuroinfection when compared to controls. Alpha diversity metrics showed lower within-sample diversity in patients with neuroinfections, though there were no differences in beta diversity. Furthermore, there was no significant change by short-term (1-3 days) antibiotic treatment on the gut microbiota, although alpha diversity metrics, such as Chao1 and Shannon's index, were close to being statistically significant. The cause of differences between patients with neuroinfections and controls is unclear and could be due to inflammation accompanying the disease; however, the effect of diet modification and/or hospitalization cannot be excluded.
Collapse
|
10
|
Anti-inflammatory effect of essential oil from Amomum Tsaoko Crevost et Lemarie. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
11
|
Ghazanfari D, Courreges MC, Belinski L, Bergmeier SC, McCall KD, Goetz DJ. Evidence for investigating GSK-3 inhibitors as potential therapeutics for severe COVID-19. Biochem Biophys Res Commun 2022; 605:171-176. [PMID: 35367865 PMCID: PMC8924054 DOI: 10.1016/j.bbrc.2022.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/07/2022] [Indexed: 11/18/2022]
Abstract
A key component of severe COVID-19 is a "cytokine storm" i.e., the excessive expression of unneeded cytokines. Previous studies suggest that SARS-CoV-2 proteins can induce macrophages to secrete pro-inflammatory cytokines; a process that may involve Toll-like receptors (TLRs). Glycogen synthase kinase-3 (GSK-3) has been implicated in TLR signal transduction and a selective GSK-3 inhibitor, termed COB-187, dramatically attenuates cytokine expression induced by the TLR ligand lipopolysaccharide (LPS). In the present study, we provide evidence that the SARS-CoV-2 spike protein (S) and the S2 subunit (S2) induce production of CXCL10 (a chemokine elevated in severe COVID-19) by a human macrophage cell line. Further, we report that two clinically relevant GSK-3 inhibitors and COB-187 attenuate S and S2 protein-induced CXCL10 production. Combined, our observations provide impetus for investigating GSK-3 inhibitors as potential therapeutics for severe COVID-19.
Collapse
Affiliation(s)
- Davoud Ghazanfari
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, United States
| | | | - Lydia Belinski
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, United States
| | - Stephen C Bergmeier
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, United States; Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, United States
| | - Kelly D McCall
- Department of Specialty Medicine, Ohio University, Athens, OH, 45701, United States; Biomedical Engineering Program, Ohio University, Athens, OH, 45701, United States; The Diabetes Institute, Ohio University, Athens, OH, 45701, United States
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, United States; Biomedical Engineering Program, Ohio University, Athens, OH, 45701, United States.
| |
Collapse
|
12
|
Dhupar R, Jones KE, Powers AA, Eisenberg SH, Ding K, Chen F, Nasarre C, Cen Z, Gong YN, LaRue AC, Yeh ES, Luketich JD, Lee AV, Oesterreich S, Lotze MT, Gemmill RM, Soloff AC. Isoforms of Neuropilin-2 Denote Unique Tumor-Associated Macrophages in Breast Cancer. Front Immunol 2022; 13:830169. [PMID: 35651620 PMCID: PMC9149656 DOI: 10.3389/fimmu.2022.830169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) exert profound influence over breast cancer progression, promoting immunosuppression, angiogenesis, and metastasis. Neuropilin-2 (NRP2), consisting of the NRP2a and NRP2b isoforms, is a co-receptor for heparin-binding growth factors including VEGF-C and Class 3 Semaphorins. Selective upregulation in response to environmental stimuli and independent signaling pathways endow the NRP2 isoforms with unique functionality, with NRP2b promoting increased Akt signaling via receptor tyrosine kinases including VEGFRs, MET, and PDGFR. Although NRP2 has been shown to regulate macrophage/TAM biology, the role of the individual NRP2a/NRP2b isoforms in TAMs has yet to be evaluated. Using transcriptional profiling and spectral flow cytometry, we show that NRP2 isoform expression was significantly higher in TAMs from murine mammary tumors. NRP2a/NRP2b levels in human breast cancer metastasis were dependent upon the anatomic location of the tumor and significantly correlated with TAM infiltration in both primary and metastatic breast cancers. We define distinct phenotypes of NRP2 isoform-expressing TAMs in mouse models of breast cancer and within malignant pleural effusions from breast cancer patients which were exclusive of neuropilin-1 expression. Genetic depletion of either NRP2 isoform in macrophages resulted in a dramatic reduction of LPS-induced IL-10 production, defects in phagosomal processing of apoptotic breast cancer cells, and increase in cancer cell migration following co-culture. By contrast, depletion of NRP2b, but not NRP2a, inhibited production of IL-6. These results suggest that NRP2 isoforms regulate both shared and unique functionality in macrophages and are associated with distinct TAM subsets in breast cancer.
Collapse
Affiliation(s)
- Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Surgical Services Division, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - Katherine E Jones
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Amy A Powers
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Seth H Eisenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kai Ding
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee Women's Research Institute, Pittsburgh, PA, United States
| | - Fangyuan Chen
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee Women's Research Institute, Pittsburgh, PA, United States
| | - Cecile Nasarre
- Division of Hematology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Division of Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Zhanpeng Cen
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- School of Medicine, Tsinghua University, Beijing, China
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yi-Nan Gong
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Amanda C LaRue
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
- Research Service, Ralph H. Johnson VA Health Care System, Charleston, SC, United States
| | - Elizabeth S Yeh
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Simon Cancer Center, Indianapolis, IN, United States
| | - James D Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee Women's Research Institute, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee Women's Research Institute, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael T Lotze
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Robert M Gemmill
- Division of Hematology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Division of Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Adam C Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
- Research Service, Ralph H. Johnson VA Health Care System, Charleston, SC, United States
| |
Collapse
|
13
|
Sang N, Jiang L, Wang Z, Zhu Y, Lin G, Li R, Zhang J. Bacteria-targeting liposomes for enhanced delivery of cinnamaldehyde and infection management. Int J Pharm 2022; 612:121356. [PMID: 34919996 DOI: 10.1016/j.ijpharm.2021.121356] [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: 06/08/2021] [Revised: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Drug-resistant gram-negative bacteria have emerged as a global crisis. Therefore, novel antibiotics and novel anti-infection strategies are urgently needed. Current antibiotics remain unsatisfactory due to poor targeting efficiency and poor drug penetration through the bacterial cell wall. Thus, targeted delivery of antibiotics into gram-negative bacteria should be a promising approach. Moreover, gram-negative bacteria can release lipopolysaccharide (LPS) to induce inflammatory response and septic shock, further increasing the disease burden. Hence, it is also promising to neutralize LPS while delivering antibiotics. This study aims to develop a multifunctional bacteria-targeting liposome that could enhance the delivery of antibiotics and adsorb LPS. METHODS A polymyxin B (PMB)-modified liposomal system (P-Lipo) was developed as novel carrier of cinnamaldehyde (CA) by using a thin-film evaporation method. Liposome morphology, size, zeta potential, stability, entrapment efficiency, and in vitro release were systematically evaluated. The bacteria-targeting effect and LPS-neutralizing capacity of P-Lipo were evaluated both in vitro and in vivo. The antibacterial effect of CA-loaded P-Lipo was assessed in Escherichia coli (E. coli) O157:H7 and Pseudomonas aeruginosa (P. aeruginosa). Ultimately, the therapeutic effect of P-CA-Lipo was investigated in E. coli O157:H7-infected mice. RESULTS P-Lipo was successfully synthesized and encapsulated with CA, which was well characterized. Both in vivo and in vitro experiments demonstrated that P-Lipo could efficiently target the E. coli after modification with PMB. Compared with free CA, CA-Lipo, and P-Lipo, P-CA-Lipo exhibited a significantly enhanced inhibitory effect on E. coli and P. aeruginosa. Further analysis demonstrated that P-CA-Lipo improved the bacterial uptake of CA and enhanced its antibacterial effect. It was also confirmed that P-Lipo could neutralize the LPS to avoid the inflammatory responses and inhibit the release of proinflammatory cytokines in both macrophages and mice. Finally, P-CA-Lipo inhibited E. coli-induced skin damage and death in mice and showed good biocompatibility. CONCLUSION The P-Lipo could target E. coli by binding with LPS and enhancing the delivery and internalization of CA. In addition, P-Lipo could adsorb free LPS synergistically, thus promoting the infection management. We believe that this strategy can provide innovative insights into antibacterial agent delivery for the treatment of persistent and severe bacterial infections.
Collapse
Affiliation(s)
- Nina Sang
- Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lixian Jiang
- Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zefeng Wang
- Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuying Zhu
- Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guoqiang Lin
- Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, China; Shanghai Frontiers Science Center for Traditional Chinese Medicine Chemical Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, China; Shanghai Frontiers Science Center for Traditional Chinese Medicine Chemical Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jiange Zhang
- Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai, China; Shanghai Frontiers Science Center for Traditional Chinese Medicine Chemical Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
14
|
Peng LY, Li BB, Deng KB, Wang WG. MicroRNA-214-3p facilitates M2 macrophage polarization by targeting GSK3B. Kaohsiung J Med Sci 2022; 38:347-356. [PMID: 35005835 DOI: 10.1002/kjm2.12487] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/25/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Allergic rhinitis (AR) is a chronic inflammatory disease of the nasal mucosa. M2 macrophage polarization can reduce inflammation and repair tissue injury during AR development. Studies have substantiated the involvement of miRNAs in AR pathogenesis. Herein, the molecular mechanism of miR-214-3p in AR development was explored. To mimic the AR environment, ovalbumin (OVA) was used to treat macrophages. MiR-214-3p and glycogen synthase kinase 3 beta (GSK3B) expression in nasal mucus tissues and macrophages was assessed by RT-qPCR. The M2 phenotypic signature of CD206 in macrophages was assessed by flow cytometry. The protein expression of GSK3B and M2 macrophage markers (ARG-1 and IL-10) was evaluated by western blotting. The correlation between miR-214-3p and GSK3B was validated by a luciferase reporter assay. We found that miR-214-3p was overexpressed in macrophages and nasal mucus tissues from AR patients. MiR-214-3p facilitated M2 polarization of macrophages upon OVA stimulation. Mechanistically, miR-214-3p targeted the GSK3B 3' untranslated region in macrophages. In addition, GSK3B was downregulated in macrophages and nasal mucus tissues from AR patients. In rescue assays, GSK3B downregulation reversed the inhibitory effects of miR-214-3p silencing on M2 polarization of macrophages treated with OVA. Overall, miR-214-3p facilitates M2 macrophage polarization by targeting GSK3B.
Collapse
Affiliation(s)
- Ling-Yan Peng
- Department of Otorhinolaryngology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Bi-Bao Li
- Department of Rehabilitation Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Ke-Bin Deng
- Department of Otorhinolaryngology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Wen-Guang Wang
- Department of Pediatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
15
|
Xia Y, Yan M, Wang P, Hamada K, Yan N, Hao H, Gonzalez FJ, Yan T. Withaferin A in the treatment of liver diseases: progress and pharmacokinetic insights. Drug Metab Dispos 2021; 50:685-693. [PMID: 34903587 DOI: 10.1124/dmd.121.000455] [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: 03/09/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022] Open
Abstract
Withaferin A (WA) is a natural steroidal compound used in Ayurvedic medicine in India and elsewhere. While WA was used as an anti-cancer reagent for decades, its role in the treatment of liver diseases has only recently been experimentally explored. Here, the effects of WA in the treatment of liver injury, systematic inflammation, and liver cancer are reviewed, and the toxicity and metabolism of WA as well as pharmacological potentials of other extracts from W. somnifera discussed. The pharmacokinetic behaviors of WA are summarized and pharmacokinetic insights into current progress and future opportunities are highlighted. Significance Statement This review outlines the current experimental progress of WA hepatoprotective activities and highlights gaps in the field. This work also discusses the pharmacokinetics of WA that can be used to guide future studies for the possible treatment of liver diseases with this compound.
Collapse
Affiliation(s)
- Yangliu Xia
- School of Life Science and Medicine, Dalian University of Technology, China
| | - Mingrui Yan
- School of Life Science and Medicine, Dalian University of Technology, China
| | - Ping Wang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, China
| | - Keisuke Hamada
- Laboratory of Metabolism, National Cancer Institute, United States
| | - Nana Yan
- Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, China
| | - Haiping Hao
- State Key laboratory of Natural Medicines, China Pharmaceutical University, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, United States
| | | |
Collapse
|
16
|
A novel GSK-3 inhibitor binds to GSK-3β via a reversible, time and Cys-199-dependent mechanism. Bioorg Med Chem 2021; 40:116179. [PMID: 33991821 DOI: 10.1016/j.bmc.2021.116179] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 11/21/2022]
Abstract
Glycogen synthase kinase-3 (GSK-3) has been implicated in numerous pathologies making GSK-3 an attractive therapeutic target. Our group has identified a compound termed COB-187 that is a potent and selective inhibitor of GSK-3. In this study, we probed the mechanism by which COB-187 inhibits GSK-3β. Progress curves, generated via real-time monitoring of kinase activity, indicated that COB-187 inhibition of GSK-3β is time-dependent and subsequent jump dilution assays revealed that COB-187 binding to GSK-3β is reversible. Further, a plot of the kinetic constant (kobs) versus COB-187 concentration suggested that, within the range of concentrations studied, COB-187 binds to GSK-3β via an induced-fit mechanism. There is a critical cysteine residue at the entry to the active site of GSK-3β (Cys-199). We generated a mutant version of GSK-3β wherein Cys-199 was substituted with an alanine. This mutation caused a dramatic decrease in the activity of COB-187; specifically, an IC50 in the nM range for wild type versus >100 µM for the mutant. A screen of COB-187 against 34 kinases that contain a conserved cysteine in their active site revealed that COB-187 is highly selective for GSK-3 indicating that COB-187's inhibition of GSK-3β via Cys-199 is specific. Combined, these findings suggest that COB-187 inhibits GSK-3β via a specific, reversible, time and Cys-199-dependent mechanism.
Collapse
|
17
|
Li R, Chinnathambi A, Alharbi SA, Shair OHM, Veeraraghavan VP, Surapaneni KM, Rengarajan T. Anti-inflammatory effects of rhaponticin on LPS-induced human endothelial cells through inhibition of MAPK/NF-κβ signaling pathways. J Biochem Mol Toxicol 2021; 35:e22733. [PMID: 33605003 DOI: 10.1002/jbt.22733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/08/2020] [Accepted: 01/19/2021] [Indexed: 01/11/2023]
Abstract
The untreated systemic chronic inflammation leads to autoimmune diseases, hyperglycemia, cardiovascular diseases, type 2 diabetes, hypertension, osteoporosis, and so on. Phytochemicals effectively inhibit the inflammation, and numerous studies have proved that the phytocomponents possess anti-inflammatory property via inhibiting the cyclooxygenase and lipoxygenase signaling pathways. Rhaponticin is one such phytochemical obtained from the perennial plant Rheum rhaponticum L. belonging to Polygonaceae family. We assessed the anti-inflammatory potency of rhaponticin in endothelial cells induced with lipopolysaccharides (LPS). Four different endothelial cells induced with LPS were treated with rhaponticin and assessed for the nitric oxide generation. The cytotoxic potency of rhaponticin was evaluated in endothelial cells using the 3-(4,5-dimethylthizaol-2yl)-2,5-diphenyl tetrazolium bromide assay. The tumor necrosis factor-α (TNF-α) synthesis was quantified using the commercially available assay kit. The inflammatory signaling protein gene expression of TNF-α, inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX2), and interleukin-1β (IL-1β) was analyzed with quantitative polymerase chain reaction (PCR) analysis. The gene expression of NADPH oxidase (NOX) cytoplasmic catalytic subunits gp91phox , p47phox , and p22phox was assessed with real-time PCR analysis. Finally, to confirm the anti-inflammatory potency of rhaponticin, the nuclear factor kappa B (NFκB) and mitogen-activated protein kinase (MAPK) signaling protein expression was analyzed with immunoblotting analysis. Rhaponticin treatment significantly decreased the levels of nitric oxide and TNF-α synthesis in LPS-induced endothelial cells. It significantly decreased the gene expression of inflammatory proteins and NOX signaling protein. The protein expression of NFκB and MAPK signaling proteins was drastically decreased in rhaponticin-treated endothelial cells induced with LPS. Overall, our results confirm that rhaponticin effectively inhibited the inflammation triggered by LPS in endothelial cells via downregulating iNOS, COX2, and NFκB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Rougang Li
- Department of General Surgery, The First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Omar H M Shair
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Krishna Mohan Surapaneni
- Departments of Biochemistry, Clinical Skills & Simulation, and Research, Panimalar Medical College Hospital & Research Institute, Chennai, Tamil Nadu, India
| | | |
Collapse
|
18
|
Kauffman MR, Nazemidashtarjandi S, Ghazanfari D, Allen AE, Reynolds NM, Faik A, Burdick MM, McCall KD, Goetz DJ. Evidence that knock down of GSK-3β in Chronic Myelogenous Leukemia cells augments IFN-γ-induced apoptosis. Leuk Res 2020; 99:106464. [PMID: 33130330 PMCID: PMC7740760 DOI: 10.1016/j.leukres.2020.106464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 12/25/2022]
Abstract
The role of interferon-gamma (IFN-γ) in Chronic Myelogenous/Myeloid Leukemia (CML) and in the treatment of CML remains unclear; specifically, the effect of IFN-γ on apoptosis. There is reported interplay between IFN-γ and glycogen synthase kinase-3 (GSK-3), a kinase which has been implicated in both cell death and, conversely, cell survival. Thus, we utilized the CML-derived HAP1 cell line and a mutant HAP1 GSK-3β knocked-down cell line (GSK-3β 31bp) to investigate whether GSK-3 modulates IFN-γ's action on CML cells. Significantly less GSK-3β 31bp cells, relative to HAP1 cells, were present after 48 h treatment with IFN-γ. IFN-γ treatment significantly decreased GSK-3β 31bp substrate adhesiveness (relative to HAP1 cells); an observation often correlated with cell death. Fluorescence microscopy revealed that IFN-γ induces a modest level of apoptosis in the HAP1 cells and that IFN-γ induced apoptosis is significantly enhanced in GSK-3β 31bp cells. Utilizing a complementary GSK-3β knocked-down cell line (8bp) we found, via flow cytometric analysis, that IFN-γ induced apoptosis is significantly enhanced in GSK-3β 8bp cells relative to HAP1 cells. Combined, our findings suggest that IFN-γ induces apoptosis of CML cells and that loss of GSK-3β significantly augments IFN-γ-induced apoptosis.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- CRISPR-Cas Systems
- Cell Adhesion/drug effects
- Cell Line, Tumor/drug effects
- Codon, Nonsense
- Drug Interactions
- Flow Cytometry
- Frameshift Mutation
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Gene Knockdown Techniques
- Glycogen Synthase Kinase 3 beta/antagonists & inhibitors
- Glycogen Synthase Kinase 3 beta/genetics
- Humans
- Interferon-gamma/pharmacology
- Interferon-gamma/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Neoplasm Proteins/antagonists & inhibitors
- Protein Kinase Inhibitors/pharmacology
- Spectrometry, Fluorescence
- RNA, Guide, CRISPR-Cas Systems
Collapse
Affiliation(s)
- Melissa R Kauffman
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA
| | | | - Davoud Ghazanfari
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA
| | - Abigail E Allen
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA
| | - Nathan M Reynolds
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| | - Ahmed Faik
- Environmental and Plant Biology Department, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
| | - Monica M Burdick
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| | - Kelly D McCall
- Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA; Department of Specialty Medicine, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH 45701, USA; Biomedical Engineering Program, Ohio University, Athens, OH 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA; Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|