1
|
Zhang L, Li J, Feng M, Xu X, Tang W, Jiang Y, Xia Z, Liu H, Shen F, Li X, Jiang L. Tigecycline modulates LPS-induced inflammatory response in sepsis via NF-κB signalling pathways: Experimental insights into immune regulation. Int J Antimicrob Agents 2025; 66:107496. [PMID: 40139445 DOI: 10.1016/j.ijantimicag.2025.107496] [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: 12/10/2024] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Sepsis is associated with high morbidity and high mortality and has strongly motivated intense studies into its mechanisms. Antibiotics, aimed to eradicate bacteria, have some impact on the immune system due to anti-inflammatory properties. Tigecycline, an antibiotic of the glycylcycline class, is commonly used for severe infections. PURPOSE This study aimed to investigate tigecycline's mechanism on the inflammatory response of sepsis to find new targets for sepsis treatment. The objective included (i) to observe the changes in inflammatory factors in LPS (lipopolysaccharide) induced septic mice after tigecycline administration, (ii) to detect the effect of tigecycline on macrophages NF-κB (nuclear factor kappa B) signalling. METHODS For LPS-induced sepsis in mice and intervention with tigecycline, mice were first injected with tigecycline (6.5 mg/kg) via tail vein followed by LPS (15 mg/kg). Luminex analysis was performed on 16 mediators. NF-κB signalling pathway antibody chip detected the expression of target sites in macrophages of the LPS group and tigecycline + LPS group. RESULTS Tigecycline has inhibitory effects on LPS-induced inflammatory response in septic mice, decreasing the concentrations of IL (interleukin)-6, IL-27, TNF-α (tumour necrosis factor-α), TNF RII, IFN-γ (interferon-gamma), CCL5/RANTES (CC Motif Chemokine Ligand) while increasing IL-6Rα, IL-10, and TWEAK (TNF-related weak inducer of apoptosis). Tigecycline downregulated phosphorylation levels of key sites JNK (c-Jun N-terminal kinase)1/2/3, p-p65 (s468) and p-p105/p50 (s907) in NF-κB signalling. CONCLUSIONS Tigecycline may inhibit the excessive immune response induced by LPS in sepsis, which may cause a potential protective effect on the host through immune regulation.
Collapse
Affiliation(s)
- Lu Zhang
- Minhang Hospital, Fudan University, Shanghai, China
| | - Jun Li
- Department of Intensive Care Unit, Shanghai Deji Hospital, Shanghai, China
| | - Meiqing Feng
- School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaocheng Xu
- Minhang District of Shanghai Medical Emergency Center, Shanghai, China
| | - Weiyi Tang
- Minhang Hospital, Fudan University, Shanghai, China
| | | | - Zhuye Xia
- Minhang Hospital, Fudan University, Shanghai, China
| | - Hongjie Liu
- Minhang Hospital, Fudan University, Shanghai, China
| | - Feiyang Shen
- Minhang Hospital, Fudan University, Shanghai, China
| | - Xiang Li
- Minhang Hospital, Fudan University, Shanghai, China.
| | - Lijing Jiang
- Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Wei F, Hughes M, Omer M, Ngo C, Pugazhendhi AS, Kolanthai E, Aceto M, Ghattas Y, Razavi M, Kean TJ, Seal S, Coathup M. A Multifunctional Therapeutic Strategy Using P7C3 as A Countermeasure Against Bone Loss and Fragility in An Ovariectomized Rat Model of Postmenopausal Osteoporosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308698. [PMID: 38477537 PMCID: PMC11151083 DOI: 10.1002/advs.202308698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Indexed: 03/14/2024]
Abstract
By 2060, an estimated one in four Americans will be elderly. Consequently, the prevalence of osteoporosis and fragility fractures will also increase. Presently, no available intervention definitively prevents or manages osteoporosis. This study explores whether Pool 7 Compound 3 (P7C3) reduces progressive bone loss and fragility following the onset of ovariectomy (OVX)-induced osteoporosis. Results confirm OVX-induced weakened, osteoporotic bone together with a significant gain in adipogenic body weight. Treatment with P7C3 significantly reduced osteoclastic activity, bone marrow adiposity, whole-body weight gain, and preserved bone area, architecture, and mechanical strength. Analyses reveal significantly upregulated platelet derived growth factor-BB and leukemia inhibitory factor, with downregulation of interleukin-1 R6, and receptor activator of nuclear factor kappa-B (RANK). Together, proteomic data suggest the targeting of several key regulators of inflammation, bone, and adipose turnover, via transforming growth factor-beta/SMAD, and Wingless-related integration site/be-catenin signaling pathways. To the best of the knowledge, this is first evidence of an intervention that drives against bone loss via RANK. Metatranscriptomic analyses of the gut microbiota show P7C3 increased Porphyromonadaceae bacterium, Candidatus Melainabacteria, and Ruminococcaceae bacterium abundance, potentially contributing to the favorable inflammatory, and adipo-osteogenic metabolic regulation observed. The results reveal an undiscovered, and multifunctional therapeutic strategy to prevent the pathological progression of OVX-induced bone loss.
Collapse
Affiliation(s)
- Fei Wei
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
| | - Megan Hughes
- School of BiosciencesCardiff UniversityWalesCF10 3ATUK
| | - Mahmoud Omer
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
| | - Christopher Ngo
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | | | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC)University of Central FloridaOrlandoFL32826USA
| | - Matthew Aceto
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Yasmine Ghattas
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Mehdi Razavi
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Thomas J Kean
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Sudipta Seal
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC)University of Central FloridaOrlandoFL32826USA
| | - Melanie Coathup
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| |
Collapse
|
3
|
Jin J, Huang R, Chang Y, Yi X. Roles and mechanisms of optineurin in bone metabolism. Biomed Pharmacother 2024; 172:116258. [PMID: 38350370 DOI: 10.1016/j.biopha.2024.116258] [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: 12/03/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
Optineurin (OPTN) is a widely expressed multifunctional articulatory protein that participates in cellular or mitochondrial autophagy, vesicular transport, and endoplasmic reticulum (ER) stress via interactions with various proteins. Skeletal development is a complex biological process that requires the participation of various osteoblasts, such as bone marrow mesenchymal stem cells (BMSCs), and osteogenic, osteoclastic, and chondrogenic cells. OPTN was recently found to be involved in the regulation of osteoblast activity, which affects bone metabolism. OPTN inhibits osteoclastogenesis via signaling pathways, including NF-κB, IFN-β, and NRF2. OPTN can promote the differentiation of BMSCs toward osteogenesis and inhibit lipogenic differentiation by delaying BMSC senescence and autophagy. These effects are closely related to the development of bone metabolism disorders, such as Paget's disease of bone, rheumatoid arthritis, and osteoporosis. Therefore, this review aims to explore the role and mechanism of OPTN in the regulation of bone metabolism and related bone metabolic diseases. Our findings will provide new targets and strategies for the prevention and treatment of bone metabolic diseases.
Collapse
Affiliation(s)
- Junjie Jin
- School of Sports and Human Sciences, Shenyang Sport University, No. 36 Jinqiansong East Road, Sujiatun District, Shenyang, Liaoning 110115, China
| | - Ruiqi Huang
- School of Physical Education, Liaoning Normal University, Dalian 116029, China
| | - Yixing Chang
- Jilin University, No. 2699 Qianjin Street, Changchun, Jilin 130012, China
| | - Xuejie Yi
- Exercise and Health Research Center/Department of Kinesiology, Shenyang Sport University, No. 36 Jinqiansong East Road, Sujiatun District, Shenyang , Liaoning 110115, China.
| |
Collapse
|
4
|
Xu Y, Hu X, Cai J, Li Y, Zou Y, Wang Y, Xie C, Xu S, Wang Y, Zheng Y, Mahamat DA, Xu Y, Wang X, Li X, Liu A, Chen D, Zhu L, Guo J. Atractylenolide-III alleviates osteoarthritis and chondrocyte senescence by targeting NF-κB signaling. Phytother Res 2023; 37:4607-4620. [PMID: 37380363 DOI: 10.1002/ptr.7929] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/01/2023] [Accepted: 06/10/2023] [Indexed: 06/30/2023]
Abstract
Atractylenolide-III (AT-III) is well known as its role in antioxidant and anti-inflammatory. Present study was aimed to figure out its effects on osteoarthritis and potential mechanisms. Rat model, human osteoarthritis cartilage explants as well as rat/human chondrocyte cultures were prepared to test AT-III's effects on osteoarthritis progression and chondrocyte senescence. Potential targeted molecules of AT-III were predicted using network pharmacology and molecular docking, assessed by Western blotting and then verified with rescue experiments. AT-III treatment alleviated osteoarthritis severity (shown by OARSI grading score and micro-CT) and chondrocyte senescence (indexed by levels of SA-β-gal, P16, P53, MMP13, ROS and ratio of healthy/collapsed mitochondrial membrane potentials). Network pharmacology and molecular docking suggested that AT-III might play role through NF-κB pathway. Further experiments revealed that AT-III reduced phosphorylation of IKKα/β, IκBα and P65 in NF-κB pathway. As well as nuclear translocation of p65. Both in vivo and in vitro experiments indicated that AT-III's effects on osteoarthritis and anti-senescence were reversed by an NF-κB agonist. AT-III could alleviate osteoarthritis by inhibiting chondrocyte senescence through NF-κB pathway, which indicated that AT-III is a prospective drug for osteoarthritis treatment.
Collapse
Affiliation(s)
- Yizhou Xu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaofang Hu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiale Cai
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yunlun Li
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Zou
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yihan Wang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Changnan Xie
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuyi Xu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yanqing Wang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuli Zheng
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Djibril Adam Mahamat
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuantao Xu
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xianghai Wang
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xican Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aijun Liu
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dongfeng Chen
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixin Zhu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiasong Guo
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, National Demonstration Center for Experimental Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
5
|
Avelar-Rodríguez D, Peña-Vélez R, Popov J, Hill L, Ryan PM. Probiotics and non-alcoholic fatty liver disease in children and adolescents: a systematic review. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2023; 115:418-427. [PMID: 36412490 DOI: 10.17235/reed.2022.8796/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
BACKGROUND non-alcoholic fatty liver disease (NAFLD) in childhood is an increasing global public health issue with significant long-term consequences. NAFLD management mainly consists of lifestyle modifications, however, adjunct pharmacological therapies are currently lacking. Gut microbiota manipulation via probiotics may alter the course of pediatric NAFLD. The objective of this systematic review was to synthesize all the available literature on the use of probiotics in children and adolescents with NAFLD. METHODS PubMed, EBSCOhost, Scopus, Web of Science, and Cochrane Library were systematically searched for trials on the use of probiotics in pediatric NAFLD. A quantitative DerSimonian Laird random effects meta-analysis was performed when possible; otherwise, a narrative summary of the study outcomes was presented and discussed. A separate search was completed to include all the ongoing registered trials on probiotics use in pediatric NAFLD. RESULTS five randomized controlled trials met the inclusion criteria. Of these, four trials were included in the final quantitative analysis. Probiotic therapy significantly reduced the levels of alanine aminotransferase (ALT) (mean difference: -10.39 [-19.85, -0.93]), however significant heterogeneity between studies was identified (I2, 93 %). CONCLUSIONS there is insufficient evidence to support probiotics in the treatment of pediatric NAFLD given the substantial degree of discordance amongst the available trials. Lifestyle modifications focusing on maintaining a normal BMI and regular exercise continue to be the gold standard approach to treating NAFLD in children.
Collapse
Affiliation(s)
| | - Rubén Peña-Vélez
- Gastroenterología y Nutrición, Hospital General de Puebla "Dr. Eduardo Vázquez N", México
| | - Jelena Popov
- College of Medicine and Health. University College Cork
| | | | | |
Collapse
|
6
|
Li Y, Palmer A, Lupu L, Huber-Lang M. Inflammatory response to the ischaemia-reperfusion insult in the liver after major tissue trauma. Eur J Trauma Emerg Surg 2022; 48:4431-4444. [PMID: 35831749 DOI: 10.1007/s00068-022-02026-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Polytrauma is often accompanied by ischaemia-reperfusion injury to tissues and organs, and the resulting series of immune inflammatory reactions are a major cause of death in patients. The liver is one of the largest organs in the body, a characteristic that makes it the most vulnerable organ after multiple injuries. In addition, the liver is an important digestive organ that secretes a variety of inflammatory mediators involved in local as well as systemic immune inflammatory responses. Therefore, this review considers the main features of post-traumatic liver injury, focusing on the immuno-pathophysiological changes, the interactions between liver organs, and the principles of treatment deduced. METHODS We focus on the local as well as systemic immune response involving the liver after multiple injuries, with emphasis on the pathophysiological mechanisms. RESULTS An overview of the mechanisms underlying the pathophysiology of local as well as systemic immune responses involving the liver after multiple injuries, the latest research findings, and the current mainstream therapeutic approaches. CONCLUSION Cross-reactivity between various organs and cascade amplification effects are among the main causes of systemic immune inflammatory responses after multiple injuries. For the time being, the pathophysiological mechanisms underlying this interaction remain unclear. Future work will continue to focus on identifying potential signalling pathways as well as target genes and intervening at the right time points to prevent more severe immune inflammatory responses and promote better and faster recovery of the patient.
Collapse
Affiliation(s)
- Yang Li
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Annette Palmer
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Ludmila Lupu
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany.
| |
Collapse
|
7
|
Kaltschmidt B, Witte KE, Greiner JFW, Weissinger F, Kaltschmidt C. Targeting NF-κB Signaling in Cancer Stem Cells: A Narrative Review. Biomedicines 2022; 10:biomedicines10020261. [PMID: 35203471 PMCID: PMC8869483 DOI: 10.3390/biomedicines10020261] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/14/2022] Open
Abstract
Among the cell populations existing within a tumor, cancer stem cells are responsible for metastasis formation and chemotherapeutic resistance. In the present review, we focus on the transcription factor NF-κB, which is present in every cell type including cancer stem cells. NF-κB is involved in pro-tumor inflammation by its target gene interleukin 1 (IL1) and can be activated by a feed-forward loop in an IL1-dependent manner. Here, we summarize current strategies targeting NF-κB by chemicals and biologicals within an integrated cancer therapy. Specifically, we start with a tyrosine kinase inhibitor targeting epidermal growth factor (EGF)-receptor-mediated phosphorylation. Furthermore, we summarize current strategies of multiple myeloma treatment involving lenalidomide, bortezomib, and dexamethasone as potential NF-κB inhibitors. Finally, we discuss programmed death-ligand 1 (PD-L1) as an NF-κB target gene and its role in checkpoint therapy. We conclude, that NF-κB inhibition by specific inhibitors of IκB kinase was of no clinical use but inhibition of upstream and downstream targets with drugs or biologicals might be a fruitful way to treat cancer stem cells.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany;
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
| | - Kaya E. Witte
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Johannes F. W. Greiner
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Florian Weissinger
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
- Department of Hematology, Oncology, Internal Medicine, Bone Marrow and Stem Cell Transplantation, Palliative Medicine, and Tumor Center, Protestant Hospital of Bethel Foundation, University Hospital OWL of Bielefeld University, Schildescher Str. 99, 33611 Bielefeld, Germany
| | - Christian Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Correspondence: ; Tel.: +49-521-106-5625
| |
Collapse
|
8
|
Li Y, Lv X, Jiang M, Jin Z. Sitagliptin ameliorates hypoxia-induced damages in endometrial stromal cells: an implication in endometriosis. Bioengineered 2021; 13:800-809. [PMID: 34964708 PMCID: PMC8805946 DOI: 10.1080/21655979.2021.2012950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hypoxia-induced damage in endometrial stromal cells (ESCs) is an important event in the pathological progression of Endometriosis. It is reported that significant inflammation is induced by hypoxia in ESCs, mediated by serval inflammatory progressions, pathways, or factors. Sitagliptin, an important member of the dipeptidyl peptidase-4 (DPP-4) inhibitors family and has been widely used for the management of type 2 diabetes. It has been recently reported to exert significant anti-inflammatory effects. Here, we aim to assess whether Sitagliptin possesses a protective effect against hypoxia-induced damages in ESCs. Our findings indicate that exposure to hypoxia significantly increased oxidative stress in ESCs by increasing the production of reactive oxygen species (ROS) and decreasing the levels of reduced glutathione (GSH), which was ameliorated by Sitagliptin. Additionally, the excessively produced inflammatory mediators, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, monocyte chemoattractant protein-1 (MCP-1), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and high mobility group box (HMGB)-1, in hypoxia-treated HESCs were pronouncedly repressed by Sitagliptin. The activated p38 mitogen-activated protein kinases (MAPK) pathway was observed in hypoxia-stimulated HESCs, then greatly inhibited by the introduction of Sitagliptin. Lastly, hypoxia-induced phosphorylation and degradation of IκBα, as well as the upregulation of nuclear factor kappa-B (NF-κB) p65 and increased transcriptional activity of NF-κB, were dramatically abolished by Sitagliptin. Collectively, Sitagliptin ameliorated hypoxia-induced damages in ESCs by suppressing the inflammation.
Collapse
Affiliation(s)
- Ying Li
- Department of Outpatient, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China
| | - Xiaolin Lv
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China
| | - Mei Jiang
- Department of Outpatient, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China
| | - Zhili Jin
- Department of Rheumatology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China
| |
Collapse
|
9
|
Niu Y, Zhao Y, He J, Yun Y, Shen M, Gan Z, Zhang L, Wang T. Dietary dihydroartemisinin supplementation alleviates intestinal inflammatory injury through TLR4/NOD/NF-κB signaling pathway in weaned piglets with intrauterine growth retardation. ACTA ACUST UNITED AC 2021; 7:667-678. [PMID: 34430721 PMCID: PMC8361298 DOI: 10.1016/j.aninu.2020.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 01/10/2023]
Abstract
The aim of present study was to evaluate whether diets supplemented with dihydroartemisinin (DHA) could alleviate intestinal inflammatory injury in weaned piglets with intrauterine growth retardation (IUGR). Twelve normal birth weight (NBW) piglets and 12 piglets with IUGR were fed a basal diet (NBW-CON and IUCR-CON groups), and another 12 piglets with IUGR were fed the basal diet supplemented with DHA at 80 mg/kg (IUGR-DHA group) from 21 to 49 d of age. At 49 d of age, 8 piglets with similar body weight in each group were sacrificed. The jejunal and ileal samples were collected for further analysis. The results showed that IUGR impaired intestinal morphology, increased intestinal inflammatory response, raised enterocyte apoptosis and reduced enterocyte proliferation and activated transmembrane toll-like receptor 4 (TLR4)/nucleotide-binding and oligomerization domain (NOD)/nuclear factor-κB (NF-κB) signaling pathway. Dihydroartemisinin inclusion ameliorated intestinal morphology, indicated by increased villus height, villus height-to-crypt depth ratio, villus surface area and decreased villus width of piglets with IUGR (P < 0.05). Compared with NBW piglets, IUGR piglets supplemented with DHA exhibited higher apoptosis index and caspase-3 expression, and lower proliferation index and proliferating cell nuclear antigen expression in the intestine (P < 0.05). Dihydroartemisinin supplementation attenuated the intestinal inflammation of piglets with IUGR, indicated by increased concentrations of intestinal inflammatory cytokines and lipopolysaccharides (P < 0.05). In addition, DHA supplementation down-regulated the related mRNA expressions of TLR4/NOD/NF-κB signaling pathway and upregulated mRNA expressions of negative regulators of TLR4 and NOD signaling pathway in the intestine of piglets with IUGR (P < 0.05). Piglets in the IUGR-DHA group showed lower protein expressions of TLR4, phosphorylated NF-κB (pNF-κB) inhibitor α, nuclear pNF-κB, and higher protein expression of cytoplasmic pNF-κB in the intestine than those in the IUGR-CON group (P < 0.05). In conclusion, DHA supplementation could improve intestinal morphology, regulate enterocyte proliferation and apoptosis, and alleviate intestinal inflammation through TLR4/NOD/NF-κB signaling pathway in weaned piglets with IUGR.
Collapse
Affiliation(s)
- Yu Niu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yongwei Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jintian He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yang Yun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Mingming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhending Gan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
10
|
Kaltschmidt C, Greiner JFW, Kaltschmidt B. The Transcription Factor NF-κB in Stem Cells and Development. Cells 2021; 10:2042. [PMID: 34440811 PMCID: PMC8391683 DOI: 10.3390/cells10082042] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/26/2022] Open
Abstract
NF-κB (nuclear factor kappa B) belongs to a family of transcription factors known to regulate a broad range of processes such as immune cell function, proliferation and cancer, neuroprotection, and long-term memory. Upcoming fields of NF-κB research include its role in stem cells and developmental processes. In the present review, we discuss one role of NF-κB in development in Drosophila, Xenopus, mice, and humans in accordance with the concept of evo-devo (evolutionary developmental biology). REL domain-containing proteins of the NF-κB family are evolutionarily conserved among these species. In addition, we summarize cellular phenotypes such as defective B- and T-cell compartments related to genetic NF-κB defects detected among different species. While NF-κB proteins are present in nearly all differentiated cell types, mouse and human embryonic stem cells do not contain NF-κB proteins, potentially due to miRNA-dependent inhibition. However, the mesodermal and neuroectodermal differentiation of mouse and human embryonic stem cells is hampered upon the repression of NF-κB. We further discuss NF-κB as a crucial regulator of differentiation in adult stem cells such as neural crest-derived and mesenchymal stem cells. In particular, c-REL seems to be important for neuronal differentiation and the neuroprotection of human adult stem cells, while RELA plays a crucial role in osteogenic and mesodermal differentiation.
Collapse
Affiliation(s)
- Christian Kaltschmidt
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
| | - Johannes F. W. Greiner
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (C.K.); (J.F.W.G.)
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| |
Collapse
|
11
|
Folding and Stability of Ankyrin Repeats Control Biological Protein Function. Biomolecules 2021; 11:biom11060840. [PMID: 34198779 PMCID: PMC8229355 DOI: 10.3390/biom11060840] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 01/04/2023] Open
Abstract
Ankyrin repeat proteins are found in all three kingdoms of life. Fundamentally, these proteins are involved in protein-protein interaction in order to activate or suppress biological processes. The basic architecture of these proteins comprises repeating modules forming elongated structures. Due to the lack of long-range interactions, a graded stability among the repeats is the generic properties of this protein family determining both protein folding and biological function. Protein folding intermediates were frequently found to be key for the biological functions of repeat proteins. In this review, we discuss most recent findings addressing this close relation for ankyrin repeat proteins including DARPins, Notch receptor ankyrin repeat domain, IκBα inhibitor of NFκB, and CDK inhibitor p19INK4d. The role of local folding and unfolding and gradual stability of individual repeats will be discussed during protein folding, protein-protein interactions, and post-translational modifications. The conformational changes of these repeats function as molecular switches for biological regulation, a versatile property for modern drug discovery.
Collapse
|
12
|
Tan EE, Hopkins RA, Lim CK, Jamuar SS, Ong C, Thoon KC, Koh MJ, Shin EM, Lian DW, Weerasooriya M, Lee CZ, Soetedjo AAP, Lim CS, Au VB, Chua E, Lee HY, Jones LA, James SS, Kaliaperumal N, Kwok J, Tan ES, Thomas B, Wu LX, Ho L, Fairhurst AM, Ginhoux F, Teo AK, Zhang YL, Ong KH, Yu W, Venkatesh B, Tergaonkar V, Reversade B, Chin KC, Tan AM, Liew WK, Connolly JE. Dominant-negative NFKBIA mutation promotes IL-1β production causing hepatic disease with severe immunodeficiency. J Clin Invest 2021; 130:5817-5832. [PMID: 32750042 DOI: 10.1172/jci98882] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Although IKK-β has previously been shown as a negative regulator of IL-1β secretion in mice, this role has not been proven in humans. Genetic studies of NF-κB signaling in humans with inherited diseases of the immune system have not demonstrated the relevance of the NF-κB pathway in suppressing IL-1β expression. Here, we report an infant with a clinical pathology comprising neutrophil-mediated autoinflammation and recurrent bacterial infections. Whole-exome sequencing revealed a de novo heterozygous missense mutation of NFKBIA, resulting in a L34P IκBα variant that severely repressed NF-κB activation and downstream cytokine production. Paradoxically, IL-1β secretion was elevated in the patient's stimulated leukocytes, in her induced pluripotent stem cell-derived macrophages, and in murine bone marrow-derived macrophages containing the L34P mutation. The patient's hypersecretion of IL-1β correlated with activated neutrophilia and liver fibrosis with neutrophil accumulation. Hematopoietic stem cell transplantation reversed neutrophilia, restored a resting state in neutrophils, and normalized IL-1β release from stimulated leukocytes. Additional therapeutic blockade of IL-1 ameliorated liver damage, while decreasing neutrophil activation and associated IL-1β secretion. Our studies reveal a previously unrecognized role of human IκBα as an essential regulator of canonical NF-κB signaling in the prevention of neutrophil-dependent autoinflammatory diseases. These findings also highlight the therapeutic potential of IL-1 inhibitors in treating complications arising from systemic NF-κB inhibition.
Collapse
Affiliation(s)
- Enrica Ek Tan
- Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - Richard A Hopkins
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Chrissie K Lim
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Saumya S Jamuar
- Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - Christina Ong
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - Koh C Thoon
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - Mark Ja Koh
- Duke-NUS Medical School, Singapore.,Dermatology Service, KK Women's and Children's Hospital, Singapore
| | - Eun Mong Shin
- Institute of Molecular and Cell Biology, A*STAR, Singapore.,Cancer Science Institute of Singapore, Singapore.,National University of Singapore, Singapore
| | - Derrick Wq Lian
- Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore.,Duke-NUS Medical School, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Madhushanee Weerasooriya
- Department of Microbiology and Immunology and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | | | | | | | - Veonice B Au
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Edmond Chua
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Hui Yin Lee
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Leigh Ann Jones
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Sharmy S James
- Department of Microbiology and Immunology and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Nivashini Kaliaperumal
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Jeffery Kwok
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Ee Shien Tan
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - Biju Thomas
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - Lynn Xue Wu
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Lena Ho
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | | | | | - Adrian Kk Teo
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Yong Liang Zhang
- Department of Microbiology and Immunology and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Kok Huar Ong
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Weimiao Yu
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | | | - Vinay Tergaonkar
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology, A*STAR, Singapore.,Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia.,Faculty of Health Sciences, University of Macau, Macau, China
| | - Bruno Reversade
- Institute of Molecular and Cell Biology, A*STAR, Singapore.,Department of Medical Genetics, School of Medicine, Koç University, Istanbul, Turkey.,Department of Paediatrics, National University of Singapore, Singapore.,Institute of Medical Biology, A*STAR, Singapore
| | - Keh Chuang Chin
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore.,Department of Physiology and
| | | | - Woei Kang Liew
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - John E Connolly
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore.,Department of Paediatrics and.,Department of Microbiology and Immunity, National University of Singapore, Singapore.,Institute of Biomedical Studies, Baylor University Medical Center, Waco, Texas, USA
| |
Collapse
|
13
|
Qin T, Jia Y, Liu Y, Dai R, Zhou L, Okada S, Tsumura M, Ohnishi H, Kato Z, Kanegane H, Sun X, Zhao X. A Novel Homozygous Mutation Destabilizes IKKβ and Leads to Human Combined Immunodeficiency. Front Immunol 2021; 11:517544. [PMID: 33658989 PMCID: PMC7917045 DOI: 10.3389/fimmu.2020.517544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
Mutations in the IKBKB gene cause severe immunodeficiency, characterized clinically by persistent respiratory or gastrointestinal infections. Targeted gene panel sequencing revealed a novel homozygous missense mutation in the IKBKB gene of a patient with immune dysregulation and combined T and B cell functional defects. PBMCs from the patient, Ikbkb Y397H mice, and transfected cells were used to elucidate how the Y395H mutation triggers IKKβ deficiency and impairs immune function. Here, we found that cells from both the patient and Ikbkb Y397H mice lacked or showed decreased levels of IKKβ protein, along with impaired lymphocyte function. IKKα and IKKγ protein expression by human PBMCs harboring the Y395H mutation was normal, but degradation of IKKβ protein was accelerated. Binding of human NF-κB to DNA in patient PBMCs fell upon stimulation with TNF-α or LPS. Additionally, a structural model of Y395H revealed loss of the hydrogen bond with D389. These data suggest that IKBKB deficiency induces abnormal IKKβ protein degradation, leading to impaired NF-κB signaling and immune function. We postulate that the Y395H variant in the IKKβ protein lost the hydrogen bond with D389, thereby affecting interaction between Y395 and D389 and increasing protein instability.
Collapse
Affiliation(s)
- Tao Qin
- Department of Infection, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanjun Jia
- National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Department of Rheumatism and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuhang Liu
- National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Rongxin Dai
- National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Department of Rheumatism and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lina Zhou
- National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Department of Rheumatism and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Satoshi Okada
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Miyuki Tsumura
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Hidenori Ohnishi
- Department of Pediatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Zenichiro Kato
- Department of Pediatrics, Gifu University Graduate School of Medicine, Gifu, Japan.,Structural Medicine, United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Xiulian Sun
- Department of Brain Research Institute, Qilu Hospital of Shandong University, Shandong, China
| | - Xiaodong Zhao
- National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Department of Rheumatism and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
14
|
Takenaka M, Takahashi Y, Takakura Y. Intercellular delivery of NF-κB inhibitor peptide utilizing small extracellular vesicles for the application of anti-inflammatory therapy. J Control Release 2020; 328:435-443. [DOI: 10.1016/j.jconrel.2020.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/11/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022]
|
15
|
Chibber P, Kumar C, Singh A, Assim Haq S, Ahmed I, Kumar A, Singh S, Vishwakarma R, Singh G. Anti-inflammatory and analgesic potential of OA-DHZ; a novel semisynthetic derivative of dehydrozingerone. Int Immunopharmacol 2020; 83:106469. [PMID: 32251963 DOI: 10.1016/j.intimp.2020.106469] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022]
Abstract
Despite various advances in the arena of the current system of medicine, there are numerous side effects associated with the therapeutics which essentially demand research on the development of safer therapeutics. One way is to explore the bioactive plant secondary metabolites and their semisynthetic derivatives. In context to this, we analyzed OA-DHZ, a dehydrozingerone derivative as the later has been reported to show anti-inflammatory and analgesic properties. OA-DHZ was found to be having promising anti-inflammatory and analgesic potential. OA-DHZ was found to inhibit the carrageenan-induced edema and leukocyte migration, acetic acid-induced increase in vascular permeability and lipopolysaccharide-induced pro-inflammatory cytokines like TNF-α, IL-6, and IL-1β. Meanwhile, it was also found to potentially inhibit thermally as well as chemically induced pain signifying its analgesic/nociceptive potential. Further, safety pharmacology studies using in vivo animal models for the central nervous system, gastrointestinal tract, the cardio-respiratory system suggest that optimum functioning of vital organ systems does not get altered after single oral administration. Also, the acute toxicity study revealed its nontoxic nature up to 2000 mg/kg. This study paves the way for future exploration and development of OA-DHZ based on its potent activity and nontoxic nature.
Collapse
Affiliation(s)
- Pankaj Chibber
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; PK-PD Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India.
| | - Chetan Kumar
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India
| | - Amarinder Singh
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; PK-PD Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India; Discovery Biology, Syngene International Pvt. Ltd., Bangalore, India
| | - Syed Assim Haq
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; PK-PD Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India
| | - Irfan Ahmed
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; PK-PD Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India
| | - Anil Kumar
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; Inflammation Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India
| | - Surjeet Singh
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; PK-PD Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India
| | - Ram Vishwakarma
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; PK-PD Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India
| | - Gurdarshan Singh
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India; PK-PD Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, Jammu and Kashmir 180001, India.
| |
Collapse
|
16
|
Cheng FS, Pan D, Chang B, Jiang M, Sang LX. Probiotic mixture VSL#3: An overview of basic and clinical studies in chronic diseases. World J Clin Cases 2020; 8:1361-1384. [PMID: 32368530 PMCID: PMC7190945 DOI: 10.12998/wjcc.v8.i8.1361] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/27/2020] [Accepted: 04/08/2020] [Indexed: 02/05/2023] Open
Abstract
Probiotics are known as “live microorganisms” and have been proven to have a health effect on hosts at the proper dose. Recently, a kind of probiotic mixture including eight live bacterial strains, VSL#3, has attracted considerable attention for its combined effect. VSL#3 is the only probiotic considered as a kind of medical food; it mainly participates in the regulation of the intestinal barrier function, including improving tight junction protein function, balancing intestinal microbial composition, regulating immune-related cytokine expression and so on. The objective of this review is to discuss the treatment action and mechanism for the administration of VSL#3 in chronic diseases of animals and humans (including children). We found that VSL#3 has a therapeutic or preventive effect in various systemic diseases per a large number of studies, including digestive systemic diseases (gastrointestinal diseases and hepatic diseases), obesity and diabetes, allergic diseases, nervous systemic diseases, atherosclerosis, bone diseases, and female reproductive systemic diseases.
Collapse
Affiliation(s)
- Fang-Shu Cheng
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
- Class 85 of 101k, China Medical University, Shenyang 110004, Liaoning Province, China
| | - Dan Pan
- Department of Geriatrics, the First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Bing Chang
- Department of Gastroenterology, the First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Min Jiang
- Department of Gastroenterology, the First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Li-Xuan Sang
- Department of Geriatrics, the First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
17
|
Li X, Zhu J, Tian L, Ma X, Fan X, Luo L, Yu J, Sun Y, Yang X, Tang W, Ma W, Yan J, Xu X, Liang H. Agmatine Protects Against the Progression of Sepsis Through the Imidazoline I2 Receptor-Ribosomal S6 Kinase 2-Nuclear Factor-κB Signaling Pathway. Crit Care Med 2020; 48:e40-e47. [PMID: 31634234 DOI: 10.1097/ccm.0000000000004065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The knowledge that agmatine is found in the human body has existed for several years; however, its role in sepsis has not yet been studied. In the present study, we investigate the role of agmatine in the progression and treatment of sepsis. DESIGN Clinical/laboratory investigations. SETTING Medical centers/University-based research laboratory. SUBJECTS Elective ICU patients with severe sepsis and healthy volunteers; C57BL/6 mice weighing 18-22 g. INTERVENTIONS Serum agmatine level and its associations with inflammatory markers were assessed in patients with sepsis. Agmatine was administered intraperitoneally to mice before a lipopolysaccharide challenge. Human peripheral blood mononuclear cells and murine macrophages were pretreated with agmatine followed by lipopolysaccharide stimulation. MEASUREMENTS AND MAIN RESULTS Serum agmatine levels were significantly decreased in patients with sepsis and lipopolysaccharide-induced mice, and correlated with Acute Physiology and Chronic Health Evaluation II score, procalcitonin, tumor necrosis factor-α, and interleukin-6 levels. In a therapeutic experiment, exogenous agmatine attenuated the cytokine production of peripheral blood mononuclear cells from patients with sepsis and healthy controls. Agmatine also exerted a significant beneficial effect in the inflammatory response and organ damage and reduced the death rate in lipopolysaccharide-induced mice. Imidazoline I2 receptor agonist 2-benzofuran-2-yl blocked the pharmacological action of agmatine; whereas, other imidazoline receptor ligands did not. Furthermore, agmatine significantly impaired the inflammatory response by inactivating nuclear factor-κB, but not protein 38 mitogen-activated protein kinase, c-Jun N-terminal kinase, extracellular signal-regulated kinase, and inducible nitric oxide synthase signaling in macrophages. Activation of imidazoline I2 receptor or knockdown of ribosomal S6 kinase 2 counteracted the effects of agmatine on phosphorylation and degradation of inhibitor of nuclear factor-κBα. CONCLUSIONS Endogenous agmatine metabolism correlated with the progression of sepsis. Supplemental exogenous agmatine could ameliorate the lipopolysaccharide-induced systemic inflammatory responses and multiple organ injuries through the imidazoline I2 receptor-ribosomal S6 kinase 2-nuclear factor-κB pathway. Agmatine could be used as both a clinical biomarker and a promising pharmaconutrient in patients with severe sepsis.
Collapse
Affiliation(s)
- Xuanfei Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Junyu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Lixing Tian
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Xiaoyuan Ma
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Xia Fan
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Li Luo
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Jing Yu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Yu Sun
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Xue Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Wanqi Tang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Wei Ma
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Jun Yan
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Xiang Xu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| |
Collapse
|
18
|
Neotuberostemonine inhibits osteoclastogenesis via blockade of NF-κB pathway. Biochimie 2018; 157:81-91. [PMID: 30439408 DOI: 10.1016/j.biochi.2018.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/08/2018] [Indexed: 01/27/2023]
Abstract
Osteoporosis has been attributed to low bone mass arising from cellular communications between bone formation and bone resorption. Osteoclastogenesis is induced by M-CSF and RANKL in hematopoietic lineage cells. Once RANK/RANKL complex is formed, TRAF6 is recruited and triggers the activation of NF-κB pathway and the expression of osteoclast-related genes including NFATc1. Neotuberostemonine (NTS) is an active compound isolated from Stemona tuberosa Lour. Pharmacologically, NTS has been known to possess antitussive, anti-fibrotic and anti-inflammatory activities through regulation of macrophage. However, the influence of NTS to osteoclastogenesis has not been reported. The purpose of this study is to investigate whether NTS can modulate the osteoclastogenesis induced by RANKL or cancer cells. We found that NTS inhibits RANKL- or cancer cell-mediated osteoclastogenesis via blockade of TRAF6 and NF-κB activation. NTS also impairs the formation of F-actin ring structure, an important feature of osteoclast differentiation and function. These results indicate that NTS can be a preventive and therapeutic candidate for bone-related disease and that NTS provides insights underlying molecular mechanisms that influence osteoclastogenesis.
Collapse
|
19
|
Zou Y, Chen Z, Li J, Gong W, Zhang L, Xu F, Chen L, Liu P, Huang H. Progestin and AdipoQ Receptor 3 Upregulates Fibronectin and Intercellular Adhesion Molecule-1 in Glomerular Mesangial Cells via Activating NF-κB Signaling Pathway Under High Glucose Conditions. Front Endocrinol (Lausanne) 2018; 9:275. [PMID: 29930535 PMCID: PMC5999916 DOI: 10.3389/fendo.2018.00275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 05/09/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Progestin and adipoQ receptor 3 (PAQR3), is a Golgi-anchored membrane protein containing seven transmembrane helices. It has been demonstrated that PAQR3 mediates insulin resistance, glucose and lipid metabolism, and inflammation. In addition, kidney inflammatory fibrosis is an important pathological feature of diabetic nephropathy (DN). Therefore, we aimed to investigate the role of PAQR3 in diabetic kidney fibrosis as well as inflammation in DN. OBJECT The effect of PAQR3 on NF-κB signaling pathway, expressions of fibronectin (FN) and intercellular adhesion molecule-1 (ICAM-1) in glomerular mesangial cells (GMCs) cultured by high glucose (HG) were examined. METHOD Diabetic mouse and rat models were induced by streptozotocin (STZ). GMCs were treated with HG and transfected with PAQR3 plasmids or small-interfering RNA targeting PAQR3 or NF-κB. The protein levels of FN and ICAM-1 were examined by Western blotting, and the transcriptional activity and DNA binding activity of NF-κB were measured by dual luciferase reporter assay and electrophoretic mobility shift assay (EMSA). The interaction between PAQR3 and IKKβ (inhibitor of nuclear factor κB kinase β) was analyzed by co-immunoprecipitation. RESULTS PAQR3 was increased in both STZ-induced diabetic models and HG-treated GMCs. PAQR3 overexpression further increased HG-induced FN and ICAM-1 upregulation. In contrast, silencing of PAQR3 suppressed the expressions of FN and ICAM-1. PAQR3 overexpression promoted the nuclear accumulation, DNA binding activity, and transcriptional activity of NF-κB. Mechanically, PAQR3 directly interacted with IKKβ. The upregulation effect of PAQR3 overexpression on the expressions of FN and ICAM-1 was abolished by the treatment of NF-κB siRNA or PDTC (ammonium pyrrolidinedithiocarbamate) in HG-treated GMCs. CONCLUSION PAQR3 promotes the expressions of FN and ICAM-1 via activating NF-κB signaling pathway. Mechanistically, PAQR3 activates NF-κB signaling pathway to mediate kidney inflammatory fibrosis through direct interaction with IKKβ in DN.
Collapse
Affiliation(s)
- Yezi Zou
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhiquan Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jie Li
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Wenyan Gong
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Futian Xu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lihao Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peiqing Liu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Heqing Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
20
|
Pimenta TS, Chaves NF, Rodrigues APD, Diniz CWP, DaMatta RA, Diniz Junior JAP. Granulocyte macrophage colony-stimulating factor alone reduces Toxoplasma gondii replication in microglial culture by superoxide and nitric oxide, without IFN-γ production: a preliminary report. Microbes Infect 2018; 20:385-390. [DOI: 10.1016/j.micinf.2018.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/03/2018] [Accepted: 05/30/2018] [Indexed: 12/25/2022]
|
21
|
Ushio A, Eto K. RBM3 expression is upregulated by NF‐κB p65 activity, protecting cells from apoptosis, during mild hypothermia. J Cell Biochem 2018; 119:5734-5749. [DOI: 10.1002/jcb.26757] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/25/2018] [Indexed: 01/19/2023]
Affiliation(s)
- Ayako Ushio
- Department of Biological SciencesGraduate School of Science and TechnologyKumamoto UniversityKumamotoJapan
| | - Ko Eto
- Department of Biological SciencesGraduate School of Science and TechnologyKumamoto UniversityKumamotoJapan
| |
Collapse
|
22
|
Nicorandil modulated macrophages activation and polarization via NF-κb signaling pathway. Mol Immunol 2017; 88:69-78. [PMID: 28609713 DOI: 10.1016/j.molimm.2017.06.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/26/2017] [Accepted: 06/03/2017] [Indexed: 12/25/2022]
Abstract
Nicorandil, a drug with both nitrate-like and ATP-sensitive potassium (KATP) channel-activating properties, has been well demonstrated in various aspects of myocardial infarction (MI), especially in inhibiting cell apoptosis and increasing coronary flow. However, the role of nicorandil in regulating inflammation and angiogenesis following myocardial infarction is still unrevealed. In the present study, we explored the effect of nicorandil on macrophage phenotype transition and inflammation regulation and the potential underlying mechanisms. For the phenotype transition and phagocytosis ability of macrophages detection, flow cytometry analysis was used. The inflammation factors were measured with ELISA and qRT-PCR. Western blot was used to assess the levels of NF-κb and its target genes and VEGF expression. The tube formation ability of endothelial cells was examined on matrigel. We discovered that nicorandil can obviously inhibit the differentiation of monocytes into mature macrophages and decrease M1 phenotype transition both in peritoneal macrophages and cultured macrophage cell line in normal or hypoxia and serum deprivation (H/SD) conditions. Meanwhile, nicorandil can induce an anti-inflammatory M2 phenotype. Thereby, nicorandil regulated macrophages switching to M1/M2 status. Our data further showed that NF-κb and the expression of its target genes were pivotal players in the regulation of macrophages phenotype. Besides, we also showed that nicorandil can promote the tube formation and VEGF expression in endothelial cells. We concluded that nicorandil may serve as an effective modulator of NF-κb signaling pathway during the pathogenesis of MI via regulating M1/M2 status and promoting angiogenesis.
Collapse
|
23
|
The Nutraceutical Dehydrozingerone and Its Dimer Counteract Inflammation- and Oxidative Stress-Induced Dysfunction of In Vitro Cultured Human Endothelial Cells: A Novel Perspective for the Prevention and Therapy of Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1246485. [PMID: 28050226 PMCID: PMC5165227 DOI: 10.1155/2016/1246485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/19/2016] [Indexed: 12/18/2022]
Abstract
Atherosclerosis is characterized by endothelial dysfunction, mainly induced by inflammation and oxidative stress. Increased reactive oxygen species (ROS) production together with increased adhesion molecules and thrombogenic tissue factor (TF) expression on endothelial cells has a key role in proatherogenic mechanisms. Therefore downmodulation of these molecules could be useful for reducing the severity of inflammation and atherosclerosis progression. Dehydrozingerone (DHZ) is a nutraceutical compound with anti-inflammatory and antioxidant activities. In this study we evaluated the ability of DHZ and its symmetric dimer to modulate hydrogen peroxide- (H2O2-) induced ROS production in human umbilical vein endothelial cells (HUVEC). We also evaluated intercellular adhesion molecule- (ICAM-) 1, vascular cell adhesion molecule- (VCAM-) 1, and TF expression in HUVEC activated by tumor necrosis factor- (TNF-) α. HUVEC pretreatment with DHZ and DHZ dimer reduced H2O2-induced ROS production and inhibited adhesion molecule expression and secretion. Of note, only DHZ dimer was able to reduce TF expression. DHZ effects were in part mediated by the inhibition of the nuclear factor- (NF-) κB activation. Overall, our findings demonstrate that the DHZ dimer exerts a potent anti-inflammatory, antioxidant, and antithrombotic activity on endothelial cells and suggest potential usefulness of this compound to contrast the pathogenic mechanisms involved in atherosclerosis progression.
Collapse
|
24
|
Pancreatic Acinar Cells Employ miRNAs as Mediators of Intercellular Communication to Participate in the Regulation of Pancreatitis-Associated Macrophage Activation. Mediators Inflamm 2016; 2016:6340457. [PMID: 27546996 PMCID: PMC4980583 DOI: 10.1155/2016/6340457] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/22/2016] [Indexed: 12/27/2022] Open
Abstract
Macrophage activation plays an important role in the inflammatory response in acute pancreatitis. In the present study, the activation of AR42J pancreatic acinar cells was induced by taurolithocholate treatment. The results showed that the culture medium from the activated AR42J cells significantly enhanced NFκB activation in the macrophages compared to that without taurolithocholate treatment. Additionally, the precipitates obtained from ultracentrifugation of the culture media that were rich in exosomes were markedly more potent in activating macrophages compared with the supernatant fraction lacking exosomes. The results indicated that the mediators carried by the exosomes played important roles in macrophage activation. Exosomal miRNAs were extracted and examined using microarrays. A total of 115 differentially expressed miRNAs were identified, and 30 showed upregulated expression, while 85 displayed downregulated expression. Target genes of the differentially expressed miRNAs were predicted using TargetScan, MiRanda, and PicTar software programs. The putative target genes were subjected to KEGG functional analysis. The functions of the target genes were primarily enriched in MAPK pathways. Specifically, the target genes regulated macrophage activation through the TRAF6-TAB2-TAK1-NIK/IKK-NFκB pathway. As the mediators of signal transduction, miRNAs and their predicted target mRNAs regulate every step in the MAPK pathway.
Collapse
|
25
|
Srivastava AK, Mishra S, Ali W, Shukla Y. Protective effects of lupeol against mancozeb-induced genotoxicity in cultured human lymphocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:714-724. [PMID: 27235710 DOI: 10.1016/j.phymed.2016.03.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 03/17/2016] [Accepted: 03/19/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND Lup-20(29)-en-3H-ol (Lupeol), a dietary pentacyclic triterpenoid has been shown to possess multiple medicinal activities including anti-inflammatory, anti-oxidant and anti-carcinogenic effects. Mancozeb is a widely used broad-spectrum fungicide with well-known carcinogenic hazards in rodents. PURPOSE The present study has been designed to investigate the protective effects of lupeol against mancozeb-induced genotoxicity and apoptosis in cultured human lymphocytes (CHLs). METHODS The genotoxic effect of mancozeb was evaluated by chromosomal aberration and micronucleus assays. The cell cycle kinetics and intracellular reactive oxygen species (ROS) generation was measured by flow cytometry. The levels of anti-oxidant enzymes and lipid peroxidation (LPO) were estimated by enzymatic assays. The localization of p65NF-κB was measured by immunocytochemical analysis. The differential expression of genes associated with genotoxicity was measured by qRT-PCR. RESULTS Mancozeb exposure (5µg/ml) for 24h caused significant induction of chromosomal aberrations (CAs) and micronuclei (MN) formation in CHLs. Pre-and post-treatment (25 and 50µg/ml) of lupeol for 24h significantly (p<0.05) reduced the frequency of CAs and MN induction, in a dose-dependent manner in mancozeb treated CHLs. Concomitantly, lupeol pre-treatment for 24h significantly increased the levels of anti-oxidant enzymes, superoxide dismutase (SOD) and catalase and decreased ROS generation and LPO. Additionally, lupeol pre-treatment significantly reduced mancozeb-induced apoptosis as shown by Sub-G1 peak analysis and annexin V-PI assay, in a dose dependent manner. Moreover, pre-treatment with lupeol attenuated mancozeb-induced NF-κB activation in CHLs. Furthermore, the results of qRT-PCR showed that lupeol pre-treatment significantly (p<0.05) decreased mancozeb-induced expression of DNA damage (p53, MDM2, COX-2, GADD45α and p21) and increased expression of DNA repair responsive genes (hOGG1 and XRCC1) in CHLs. CONCLUSION Taken together, our findings suggest that lupeol could attenuate mancozeb-induced oxidative stress, which in turn could inhibit NF-κB activation and thus provide protection against mancozeb-induced genotoxicity and apoptosis. So, lupeol could be used as a potent anti-oxidant regimen against pesticide induced genotoxicity in agricultural farm workers.
Collapse
Affiliation(s)
- Amit Kumar Srivastava
- Proteomics & Environmental Carcinogenesis Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India
| | - Sanjay Mishra
- Proteomics & Environmental Carcinogenesis Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India
| | - Wahid Ali
- Department of Pathology, Chatrapati Shahuji Maharaj Medical University, Lucknow Chowk, Lucknow U.P. India- 226003
| | - Yogeshwer Shukla
- Proteomics & Environmental Carcinogenesis Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India.
| |
Collapse
|
26
|
Inhibition of Acute Lung Injury by TNFR-Fc through Regulation of an Inflammation-Oxidative Stress Pathway. PLoS One 2016; 11:e0151672. [PMID: 26990441 PMCID: PMC4798551 DOI: 10.1371/journal.pone.0151672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/02/2016] [Indexed: 01/11/2023] Open
Abstract
Background Acute lung injury (ALI), characterized by disruption of the lung alveolar-capillary membrane barrier and resultant pulmonary edema, and associated with a proteinaceous alveolar exudate, is a leading cause of morbidity and mortality. Currently, inflammation-oxidative stress interaction between TNF-α and NF-κB was identified as a key pathway of ALI. We hypothesized that a TNFR-Fc fusion protein would have beneficial effects in experimental ALI, and sought to test this idea in mice by blocking TNF-α. Methods and Results Intratracheal instillation of lipopolysaccharide (LPS) into the lungs of ALI mice led to histiocyte apoptosis, and detection of serum and bronchoalveolar lavage fluid (BALF) cytokines, feedback between NF-κB and TNF-α, lung albumin leakage, lung damage, IκB kinase (IKK) and NF-κB activation, I-κB degradation, and oxidative injury. LPS administration raised pulmonary inflammation as reflected by increased inflammatory cytokines, alveoli protein concentration, and ALI scores. IKK is phosphorylated following LPS challenge, leading to I-κB degradation and NF-κB p65 phosphorylation. Furthermore, NF-κB is translocated into the nucleus and up-regulates TNF-α gene transcription. Infusion of TNFR-Fc 24h before LPS challenge significantly abrogated the increase of inflammatory cytokines, especially serum TNF-α concentration, as well as pulmonary alveoli protein levels, and diminished IKK and NF-κB activation and I-κB degradation. The nuclear translocation of NF-κB was inhibited, following by down-regulation of TNF-α gene transcription. In addition, LPS intratracheal instillation induced marked oxidative damage, such as a decrease in total anti-oxidation products and an increase in malondialdehyde (MDA), as well as up-regulation of oxidation enzymes. Histologic analysis and apoptosis scores revealed that the extent of tissue lesions was significantly reduced, but not abrogated, by TNF-α blockade. Conclusion Treatment with LPS alone increased inflammation and oxidative stress in ALI mice, while administration of TNFR-Fc 24h before LPS challenge broke the feedback between NF-κB and TNF-α, resulting in decreased pulmonary inflammation/oxidative damage and tissue destruction. These results suggest a potential role for TNF-α therapy to treat clinical ALI.
Collapse
|
27
|
Yuan S, Jin J, Shi J, Hou Y. Inhibitor of growth-4 is a potential target for cancer therapy. Tumour Biol 2016; 37:4275-9. [PMID: 26803518 DOI: 10.1007/s13277-016-4842-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/12/2016] [Indexed: 12/11/2022] Open
Abstract
The inhibitor of growth-4 (ING-4) belongs to the inhibitor of growth (ING) family that is a type II tumor suppressor gene including five members (ING1-5). As a tumor suppressor, ING4 inhibits tumor growth, invasion, and metastasis by multiple signaling pathways. In addition to that, ING4 can facilitate cancer cell sensitivity to chemotherapy and radiotherapy. Although ING4 loss is observed for many types of cancers, increasing evidences show that ING4 can be used for gene therapy. In this review, the recent progress of ING4 regulating tumorigenesis is discussed.
Collapse
Affiliation(s)
- Shuping Yuan
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China.,Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Jianhua Jin
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China
| | - Juanjuan Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| | - Yongzhong Hou
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China. .,Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| |
Collapse
|
28
|
Lee J, Noh ALSM, Zheng T, Kang JH, Yim M. Eriodicyol inhibits osteoclast differentiation and ovariectomy-induced bone loss in vivo. Exp Cell Res 2015; 339:380-8. [DOI: 10.1016/j.yexcr.2015.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/30/2015] [Accepted: 10/02/2015] [Indexed: 01/02/2023]
|
29
|
Inhibitory Effect of a French Maritime Pine Bark Extract-Based Nutritional Supplement on TNF-α-Induced Inflammation and Oxidative Stress in Human Coronary Artery Endothelial Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:260530. [PMID: 26664450 PMCID: PMC4664804 DOI: 10.1155/2015/260530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 11/17/2022]
Abstract
Oxidative stress and inflammation, leading to endothelial dysfunction, contribute to the pathogenesis of atherosclerosis. The popularity of natural product supplements has increased in recent years, especially those with purported anti-inflammatory and/or antioxidant effects. The efficacy and mechanism of many of these products are not yet well understood. In this study, we tested the antioxidant and anti-inflammatory effects of a supplement, HIPER Health Supplement (HIPER), on cytokine-induced inflammation and oxidative stress in human coronary artery endothelial cells (HCAECs). HIPER is a mixture of French maritime pine bark extract (PBE), honey, aloe vera, and papaya extract. Treatment for 24 hours with HIPER reduced TNF-α-induced reactive oxygen species (ROS) generation that was associated with decreased NADPH oxidase 4 and increased superoxide dismutase-1 expression. HIPER inhibited TNF-α induced monocyte adhesion to HCAECs that was in keeping with decreased expression of vascular cell adhesion molecule-1 and intercellular cell adhesion molecule-1 and decreased nuclear factor-kappa B (NF-κB) activation. Further investigation of mechanism showed HIPER reduced TNF-α induced IκBα and p38 and MEK1/2 MAP kinases phosphorylation. Our findings show that HIPER has potent inhibitory effects on HCAECs inflammatory and oxidative stress responses that may protect against endothelial dysfunction that underlies early atherosclerotic lesion formation.
Collapse
|
30
|
Abstract
Myocardial infarction is defined as sudden ischemic death of myocardial tissue. In the clinical context, myocardial infarction is usually due to thrombotic occlusion of a coronary vessel caused by rupture of a vulnerable plaque. Ischemia induces profound metabolic and ionic perturbations in the affected myocardium and causes rapid depression of systolic function. Prolonged myocardial ischemia activates a "wavefront" of cardiomyocyte death that extends from the subendocardium to the subepicardium. Mitochondrial alterations are prominently involved in apoptosis and necrosis of cardiomyocytes in the infarcted heart. The adult mammalian heart has negligible regenerative capacity, thus the infarcted myocardium heals through formation of a scar. Infarct healing is dependent on an inflammatory cascade, triggered by alarmins released by dying cells. Clearance of dead cells and matrix debris by infiltrating phagocytes activates anti-inflammatory pathways leading to suppression of cytokine and chemokine signaling. Activation of the renin-angiotensin-aldosterone system and release of transforming growth factor-β induce conversion of fibroblasts into myofibroblasts, promoting deposition of extracellular matrix proteins. Infarct healing is intertwined with geometric remodeling of the chamber, characterized by dilation, hypertrophy of viable segments, and progressive dysfunction. This review manuscript describes the molecular signals and cellular effectors implicated in injury, repair, and remodeling of the infarcted heart, the mechanistic basis of the most common complications associated with myocardial infarction, and the pathophysiologic effects of established treatment strategies. Moreover, we discuss the implications of pathophysiological insights in design and implementation of new promising therapeutic approaches for patients with myocardial infarction.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
31
|
Fan C, Xie Y, Dong Y, Su Y, Upton Z. Investigating the potential of Shikonin as a novel hypertrophic scar treatment. J Biomed Sci 2015; 22:70. [PMID: 26275605 PMCID: PMC4537585 DOI: 10.1186/s12929-015-0172-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 07/24/2015] [Indexed: 11/29/2022] Open
Abstract
Background Hypertrophic scarring is a highly prevalent condition clinically and results from a decreased number of apoptotic fibroblasts and over-abundant production of collagen during scar formation following wound healing. Our previous studies indicated that Shikonin, an active component extracted from Radix Arnebiae, induces apoptosis and reduces collagen production in hypertrophic scar-derived fibroblasts. In the study reported here, we further evaluate the potential use of Shikonin as a novel scar remediation therapy by examining the effects of Shikonin on both keratinocytes and fibroblasts using Transwell® co-culture techniques. The underlying mechanisms were also revealed. In addition, effects of Shikonin on the expression of cytokines in Transwell co-culture “conditioned” medium were investigated. Results Our results indicate that Shikonin preferentially inhibits cell proliferation and induces apoptosis in fibroblasts without affecting keratinocyte function. In addition, we found that the proliferation-inhibiting and apoptosis-inducing abilities of SHI might be triggered via MAPK and Bcl-2/Caspase 3 signalling pathways. Furthermore, SHI has been found to attenuate the expression of TGF-β1 in Transwell co-cultured “conditioned” medium. Conclusions The data generated from this study provides further evidence that supports the potential use of Shikonin as a novel scar remediation therapy.
Collapse
Affiliation(s)
- Chen Fan
- Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4059, Australia.
| | - Yan Xie
- Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4059, Australia. .,Tissue Organ Bank & Tissue Engineering Centre, General Hospital of Ningxia Medical University, Yinchuan, China.
| | - Ying Dong
- Cancer Research Program, Translational Research Institute, Queensland University of Technology, Brisbane, Australia.
| | - Yonghua Su
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai, China.
| | - Zee Upton
- Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4059, Australia.
| |
Collapse
|
32
|
Han Y, Zhou M, Xing L, Jiang M, Bai G, Luo G. Identification of NF-κB inhibitors in Qishenyiqi dropping pills for myocardial infarction treatment based on bioactivity-integrated UPLC-Q/TOF MS. Biomed Chromatogr 2015; 29:1612-8. [DOI: 10.1002/bmc.3468] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/21/2015] [Accepted: 02/28/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Yanqi Han
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin 300071 People's Republic of China
- Department of Traditional Chinese Medicine; Tianjin Institute of Pharmaceutical Research; Tianjin 300193 People's Republic of China
| | - Mengge Zhou
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin 300071 People's Republic of China
| | - Lu Xing
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin 300071 People's Republic of China
| | - Min Jiang
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin 300071 People's Republic of China
| | - Gang Bai
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin 300071 People's Republic of China
| | - Guoan Luo
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin 300071 People's Republic of China
- Department of Chemistry; Tsinghua University; Beijing 100084 People's Republic of China
| |
Collapse
|
33
|
CPEB regulation of TAK1 synthesis mediates cytokine production and the inflammatory immune response. Mol Cell Biol 2014; 35:610-8. [PMID: 25452303 DOI: 10.1128/mcb.00800-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The cytoplasmic-element-binding (CPEB) protein is a sequence-specific RNA-binding protein that regulates cytoplasmic polyadenylation-induced translation. In mouse embryo fibroblasts (MEFs) lacking CPEB, many mRNAs encoding proteins involved in inflammation are misregulated. Correlated with this aberrant translation in MEFs, a macrophage cell line depleted of CPEB and treated with lipopolysaccharide (LPS) to stimulate the inflammatory immune response expresses high levels of interleukin-6 (IL-6), which is due to prolonged nuclear retention of NF-κB. Two proteins involved in NF-κB nuclear localization and IL-6 expression, IκBα and transforming growth factor beta-activated kinase 1 (TAK1), are present at excessively low and high steady-state levels, respectively, in LPS-treated CPEB-depleted macrophages. However, only TAK1 has an altered synthesis rate that is CPEB dependent and CPEB/TAK1 double depletion alleviates high IL-6 production. Peritoneal macrophages isolated from CPEB knockout (KO) mice treated with LPS in vitro also have prolonged NF-κB nuclear retention and produce high IL-6 levels. LPS-injected CPEB KO mice secrete prodigious amounts of IL-6 and other proinflammatory cytokines and exhibit hypersensitivity to endotoxic shock; these effects are mitigated when the animals are also injected with (5Z)-7-oxozeaenol, a potent and specific inhibitor of TAK1. These data show that CPEB control of TAK1 mRNA translation mediates the inflammatory immune response.
Collapse
|
34
|
Jiang C, Li Z, Quan H, Xiao L, Zhao J, Jiang C, Wang Y, Liu J, Gou Y, An S, Huang Y, Yu W, Zhang Y, He W, Yi Y, Chen Y, Wang J. Osteoimmunology in orthodontic tooth movement. Oral Dis 2014; 21:694-704. [PMID: 25040955 DOI: 10.1111/odi.12273] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 06/18/2014] [Accepted: 06/26/2014] [Indexed: 02/05/2023]
Abstract
The skeletal and immune systems share a multitude of regulatory molecules, including cytokines, receptors, signaling molecules, and signaling transducers, thereby mutually influencing each other. In recent years, several novel insights have been attained that have enhanced our current understanding of the detailed mechanisms of osteoimmunology. In orthodontic tooth movement, immune responses mediated by periodontal tissue under mechanical force induce the generation of inflammatory responses with consequent alveolar bone resorption, and many regulators are involved in this process. In this review, we take a closer look at the cellular/molecular mechanisms and signaling involved in osteoimmunology and at relevant research progress in the context of the field of orthodontic tooth movement.
Collapse
Affiliation(s)
- C Jiang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Z Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - H Quan
- Qingdao First Sanatorium of Jinan Military Distract of PLA, Qingdao, Shandong, China
| | - L Xiao
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Zhao
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - C Jiang
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Y Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Liu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Y Gou
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - S An
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Huang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - W Yu
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Zhang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - W He
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Yi
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Chen
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Huang TT, Wu SP, Chong KY, Ojcius DM, Ko YF, Wu YH, Wu CY, Lu CC, Martel J, Young JD, Lai HC. The medicinal fungus Antrodia cinnamomea suppresses inflammation by inhibiting the NLRP3 inflammasome. JOURNAL OF ETHNOPHARMACOLOGY 2014; 155:154-164. [PMID: 24858059 DOI: 10.1016/j.jep.2014.04.053] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/09/2014] [Accepted: 04/29/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Antrodia cinnamomea--a medicinal fungus that is indigenous to Taiwan--has been used as a health tonic by aboriginal tribes and the Asian population. Recent studies indicate that Antrodia cinnamomea extracts exhibit hepato-protective, anti-hypertensive, anti-oxidative, anti-inflammatory, immuno-modulatory, and anti-cancer effects on cultured cells and laboratory animals. This study aims to explore the anti-inflammatory activity of an Antrodia cinnamomea ethanol extract (ACEE) and elucidate its underlying mechanisms of action using lipopolysaccharide (LPS)-primed, ATP-stimulated human THP-1 macrophages. MATERIALS AND METHODS The effects of ACEE on cell viability were studied using the MTT assay. The expressions of genes, proteins, and pro-inflammatory cytokines were measured by quantitative real-time RT-PCR, Western blotting and ELISA, respectively. The ACEE was further investigated for its effects on reactive oxygen species (ROS) production using ROS detection kit. RESULTS Our results showed that ACEE significantly inhibits ATP-induced secretion of interleukin-1β (IL-1β), interleukin-18 (IL-18) and tumor necrosis factor-α (TNF-α) by LPS-primed macrophages. ACEE also suppresses the transcription and activation of caspase-1, which is responsible for the cleavage and activation of IL-1β and IL-18. Of note, ACEE not only reduces expression of the inflammasome component NLRP3 and the purinergic receptor P2X7R but also inhibits ATP-induced ROS production and caspase-1 activation. Furthermore, the anti-inflammatory properties of ACEE correlate with reduced activation of the MAPK and NF-κB pathways. CONCLUSION The results of the present study indicate that Antrodia cinnamomea suppresses the secretion of IL-1β and IL-18 associated with inhibition of the NLRP3 inflammasome in macrophages. These findings suggest that ACEE may have therapeutic potential for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Tsung-Teng Huang
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 333, Taiwan; Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Laboratory of Nanomaterials, Chang Gung University, Taoyuan 333, Taiwan; Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan 333, Taiwan
| | - Sian-Pu Wu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 333, Taiwan; Department of Molecular Cell Biology and Health Sciences Research Institute, University of California, Merced, Merced, CA 95343, USA
| | - Yun-Fei Ko
- Biochemical Engineering Research Center, Ming Chi University of Technology, Taipei 24301, Taiwan; Chang Gung Biotechnology Corporation, Taipei 10508, Taiwan
| | - Yi-Hui Wu
- Cancer Research Center, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Cheng-Yeu Wu
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 333, Taiwan; Laboratory of Nanomaterials, Chang Gung University, Taoyuan 333, Taiwan; Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan 333, Taiwan
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, Taipei 24205, Taiwan
| | - Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 333, Taiwan; Laboratory of Nanomaterials, Chang Gung University, Taoyuan 333, Taiwan
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 333, Taiwan; Laboratory of Nanomaterials, Chang Gung University, Taoyuan 333, Taiwan; Biochemical Engineering Research Center, Ming Chi University of Technology, Taipei 24301, Taiwan; Chang Gung Biotechnology Corporation, Taipei 10508, Taiwan; Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, NY 10021, USA.
| | - Hsin-Chih Lai
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 333, Taiwan; Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
36
|
Mantle cell lymphoma: taking therapeutic advantage of new insights into the biology. Curr Hematol Malig Rep 2014; 9:254-61. [PMID: 25023397 DOI: 10.1007/s11899-014-0221-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Mantle cell lymphoma (MCL) is an uncommon, incurable B-cell non-Hodgkin's lymphoma that afflicts the elderly. There is no standard course of treatment, with options varying from observation in asymptomatic patients to aggressive induction/consolidation regimens in younger patients with rapidly progressive disease. Emerging data regarding the role of the ubiquitin-proteasome system, B-cell receptor and mTOR signaling pathways, cell cycle regulation, and epigenetic and immune-modulation in the pathogenesis of MCL have resulted in the development of novel therapies, with a shift away from conventional cytotoxic chemotherapy to relatively less toxic, more targeted treatment. The challenge now is to determine the optimal sequence and combination of the various available and emerging therapies for use in patients with MCL.
Collapse
|
37
|
Rim HK, Yun CH, Shin JS, Cho YW, Jang DS, Ryu JH, Park H, Lee KT. 5,6,7-Trimethoxyflavone suppresses pro-inflammatory mediators in lipopolysaccharide-induced RAW 264.7 macrophages and protects mice from lethal endotoxin shock. Food Chem Toxicol 2013; 62:847-55. [DOI: 10.1016/j.fct.2013.10.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/14/2013] [Accepted: 10/16/2013] [Indexed: 01/03/2023]
|
38
|
Huang TT, Lai HC, Chen YB, Chen LG, Wu YH, Ko YF, Lu CC, Chang CJ, Wu CY, Martel J, Ojcius DM, Chong KY, Young JD. cis-Resveratrol produces anti-inflammatory effects by inhibiting canonical and non-canonical inflammasomes in macrophages. Innate Immun 2013; 20:735-50. [DOI: 10.1177/1753425913507096] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Resveratrol, a natural phenolic compound found in red grapes and wine, exists as cis and trans isomers. Recent studies have shown that trans-resveratrol possesses anti-inflammatory, anti-oxidant, anti-carcinogenic, anti-tumor and immunomodulatory properties. However, it remains unclear whether cis-resveratrol may exhibit similar activities. The objective of the present study was to examine the effects of cis- and trans-resveratrol on the production of pro-inflammatory cytokines and mediators in human macrophages. We examined the possibility that cis- and trans-resveratrol may affect cytokine secretion by modulating inflammasomes, intracellular multi-protein complexes, the assembly of which leads to caspase-1 activation and secretion of active IL-1β by macrophages. Our results show that pre-treatment of macrophages with cis-resveratrol not only reduces pro-IL-1β production and IL-1β secretion, but also suppresses ATP-induced transcription and activation of caspase-1 and caspase-4. Notably, cis-resveratrol inhibits the expression of the purinergic receptor, P2X7R, and the endoplasmic reticulum stress marker, Glc-regulated protein 78, but also reduces reactive oxygen species production. Moreover, cis-resveratrol attenuates cyclooxygenase-2 expression and prostaglandin E2 production. cis-Resveratrol also decreases the phosphorylation of p38 MAPK and expression of the c-Jun protein. These results indicate that cis-resveratrol produces anti-inflammatory effects by inhibiting both the canonical and non-canonical inflammasomes, and associated pathways in human macrophages.
Collapse
Affiliation(s)
- Tsung-Teng Huang
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Laboratory of Nanomaterials, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Hsin-Chih Lai
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Young-Bin Chen
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Lih-Geeng Chen
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi, Taiwan, Republic of China
| | - Yi-Hui Wu
- Cancer Research Center, National Cheng Kung University Hospital, Tainan, Taiwan, Republic of China
| | - Yun-Fei Ko
- Biochemical Engineering Research Center, Ming Chi University of Technology, Taipei, Taiwan, Republic of China
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, Taipei, Taiwan, Republic of China
| | - Chih-Jung Chang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Cheng-Yeu Wu
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Laboratory of Nanomaterials, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Laboratory of Nanomaterials, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Molecular Cell Biology, Health Sciences Research Institute, University of California Merced, Merced, CA, USA
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Laboratory of Nanomaterials, Chang Gung University, Taoyuan, Taiwan, Republic of China
- Biochemical Engineering Research Center, Ming Chi University of Technology, Taipei, Taiwan, Republic of China
- Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, NY, USA
| |
Collapse
|
39
|
Abstract
The transcription factor NF-κB is a family of proteins involved in signaling pathways essential for normal cellular functions and development. Deletion of various components of this pathway resulted with abnormal skeletal development. Research in the last decade has established that NF-κB signaling mediates RANK ligand-induced osteoclastogenesis. Consistently, it was shown that inhibition of NF-κB was an effective approach to inhibit osteoclast formation and bone resorptive activity. Identification of the molecular machinery underlying NF-κB activation permitted osteoclast-specific deletion of the major components of this pathway. As a result, it was clear that deletion of members of the proximal IKK kinase complex and the distal NF-κB subunits and downstream regulators affected skeletal development. These studies provided several targets of therapeutic intervention in osteolytic diseases. NF-κB activity has been also described as the centerpiece of inflammatory responses and is considered a potent mediator of inflammatory osteolysis. Indeed, inflammatory insults exacerbate physiologic RANKL-induced NF-κB signals leading to exaggerated responses and to inflammatory osteolysis. These superimposed NF-κB activities appear to underlie several bone pathologies. This review will describe the individual roles of NF-κB molecules in bone resorption and inflammatory osteolysis.
Collapse
Affiliation(s)
- Y Abu-Amer
- Department of Orthopedic Surgery, Department of Cell Biology & Physiology, Washington University School of Medicine, 660S. Euclid Avenue, Saint Louis, MO 63110, USA.
| |
Collapse
|
40
|
Chen Q, Liu K, Robinson AR, Clauson CL, Blair HC, Robbins PD, Niedernhofer LJ, Ouyang H. DNA damage drives accelerated bone aging via an NF-κB-dependent mechanism. J Bone Miner Res 2013; 28:1214-28. [PMID: 23281008 PMCID: PMC3662975 DOI: 10.1002/jbmr.1851] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 11/15/2012] [Accepted: 11/26/2012] [Indexed: 12/21/2022]
Abstract
Advanced age is one of the most important risk factors for osteoporosis. Accumulation of oxidative DNA damage has been proposed to contribute to age-related deregulation of osteoblastic and osteoclastic cells. Excision repair cross complementary group 1-xeroderma pigmentosum group F (ERCC1-XPF) is an evolutionarily conserved structure-specific endonuclease that is required for multiple DNA repair pathways. Inherited mutations affecting expression of ERCC1-XPF cause a severe progeroid syndrome in humans, including early onset of osteopenia and osteoporosis, or anomalies in skeletal development. Herein, we used progeroid ERCC1-XPF-deficient mice, including Ercc1-null (Ercc1(-/-)) and hypomorphic (Ercc1(-/Δ)) mice, to investigate the mechanism by which DNA damage leads to accelerated bone aging. Compared to their wild-type littermates, both Ercc1(-/-) and Ercc1(-/Δ) mice display severe, progressive osteoporosis caused by reduced bone formation and enhanced osteoclastogenesis. ERCC1 deficiency leads to atrophy of osteoblastic progenitors in the bone marrow stromal cell (BMSC) population. There is increased cellular senescence of BMSCs and osteoblastic cells, as characterized by reduced proliferation, accumulation of DNA damage, and a senescence-associated secretory phenotype (SASP). This leads to enhanced secretion of inflammatory cytokines known to drive osteoclastogenesis, such as interleukin-6 (IL-6), tumor necrosis factor α (TNFα), and receptor activator of NF-κB ligand (RANKL), and thereby induces an inflammatory bone microenvironment favoring osteoclastogenesis. Furthermore, we found that the transcription factor NF-κB is activated in osteoblastic and osteoclastic cells of the Ercc1 mutant mice. Importantly, we demonstrated that haploinsufficiency of the p65 NF-κB subunit partially rescued the osteoporosis phenotype of Ercc1(-/Δ) mice. Finally, pharmacological inhibition of the NF-κB signaling via an I-κB kinase (IKK) inhibitor reversed cellular senescence and SASP in Ercc1(-/Δ) BMSCs. These results demonstrate that DNA damage drives osteoporosis through an NF-κB-dependent mechanism. Therefore, the NF-κB pathway represents a novel therapeutic target to treat aging-related bone disease.
Collapse
Affiliation(s)
- Qian Chen
- Department of Medicine, University of Pittsburgh School of Medicine, 1218 Scaife Hall 3550 Terrace Street, Pittsburgh, PA 15261
| | - Kai Liu
- Department of Restorative Dentistry and Comprehensive Care, University of Pittsburgh School of Dental Medicine, 3023 Salk Annex, 3501 Terrace Street, Pittsburgh, PA 15261 USA
| | - Andria R. Robinson
- Department of Human Genetics, University of Pittsburgh School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15261 USA
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, 2.6, Pittsburgh, PA 15213 USA
| | - Cheryl L. Clauson
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, 2.6, Pittsburgh, PA 15213 USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 427 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Harry C. Blair
- Pittsburgh Veteran’s Affairs Medical Center, Laboratory Service-646, 7180 Highland Drive, Pittsburgh, PA 15206 USA
- Department of Pathology, University of Pittsburgh School of Medicine, Scaife Hall S705, 3501 Terrace Street, Pittsburgh, PA 15261 USA
| | - Paul D. Robbins
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, 2.6, Pittsburgh, PA 15213 USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 427 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Laura J. Niedernhofer
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Hillman Cancer Center, 2.6, Pittsburgh, PA 15213 USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 427 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Hongjiao Ouyang
- Department of Restorative Dentistry and Comprehensive Care, University of Pittsburgh School of Dental Medicine, 3023 Salk Annex, 3501 Terrace Street, Pittsburgh, PA 15261 USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 427 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA 15219 USA
- McGowan Institute for Regenerative Medicine, 450 Technology Drive Suite 300 Pittsburgh, PA 15219 USA
| |
Collapse
|
41
|
Hu P, Huang P, Chen MW. Curcumin attenuates cyclooxygenase-2 expression via inhibition of the NF-κB pathway in lipopolysaccharide-stimulated human gingival fibroblasts. Cell Biol Int 2013; 37:443-8. [PMID: 23494805 DOI: 10.1002/cbin.10050] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 01/11/2013] [Indexed: 01/22/2023]
Abstract
Porphyromonas gingivalis lipopolysaccharide (LPS) induces the expression of the cyclooxygenase-2 (COX-2), which contributes to the process of periodontitis. Curcumin, a constituent of turmeric, exhibits anti-inflammatory properties. We have investigated the anti-inflammatory effect of curcumin in human gingival fibroblasts (HGFs) stimulated by P. gingivalis LPS and its mechanism of action. HGFs pretreated with curcumin were stimulated by P. gingivalis LPS. COX-2 mRNA and protein expressions were analysed by real-time PCR and Western blot analysis. Activation of nuclear factor kappa B (NF-κB) was analysed by the NF-κB-dependent luciferase activity and electrophoretic mobility-shift assay (EMSA). Curcumin inhibited COX-2 mRNA and protein synthesis in LPS-stimulated HGFs in a dose-dependent manner. P. gingivalis LPS activated NF-κB-dependent transcription in HGFs, which were also downregulated by pretreatment with curcumin. Therefore, curcumin can inhibit P. gingivalis LPS-induced COX-2 expression, which may be due to the inhibition of the NF-κB pathway.
Collapse
Affiliation(s)
- Ping Hu
- Center of Stomatology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, No. 1095, Jiefang Road, Wuhan 430030, People's Republic of China
| | | | | |
Collapse
|
42
|
Roundy LM, Jia W, Zhang J, Ye X, Prestwich GD, OottamasathienQ S. LL-37 induced cystitis and the receptor for advanced glycation end-products (RAGE) pathway. ACTA ACUST UNITED AC 2013; 4:1-8. [DOI: 10.4236/abb.2013.48a2001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
Shi DD, Shi H, Lu D, Li R, Zhang Y, Zhang J. NDR1/STK38 potentiates NF-κB activation by its kinase activity. Cell Biochem Funct 2012; 30:664-70. [DOI: 10.1002/cbf.2846] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 04/13/2012] [Accepted: 05/10/2012] [Indexed: 11/10/2022]
Affiliation(s)
- Dan-Dan Shi
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology (Ministry of Health); Peking University Health Science Center; Beijing; China
| | - Hu Shi
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology (Ministry of Health); Peking University Health Science Center; Beijing; China
| | - Dan Lu
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology (Ministry of Health); Peking University Health Science Center; Beijing; China
| | - Rui Li
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology (Ministry of Health); Peking University Health Science Center; Beijing; China
| | - Yu Zhang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology (Ministry of Health); Peking University Health Science Center; Beijing; China
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology (Ministry of Health); Peking University Health Science Center; Beijing; China
| |
Collapse
|
44
|
Chung JW, Choi RJ, Seo EK, Nam JW, Dong MS, Shin EM, Guo LY, Kim YS. Anti-inflammatory effects of (Z)-ligustilide through suppression of mitogen-activated protein kinases and nuclear factor-κB activation pathways. Arch Pharm Res 2012; 35:723-32. [PMID: 22553066 DOI: 10.1007/s12272-012-0417-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 09/29/2011] [Accepted: 10/23/2011] [Indexed: 11/28/2022]
Abstract
The roots of Angelica tenuissima have been commonly used for the treatment of cardiovascular diseases and menstrual discomfort in Asian countries, such as China and Korea. The primary volatile flavor components are essential oil ingredients, phthalide lactones. In this study, (Z)-ligustilide was tested for its anti-inflammatory activities in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. We found that (Z)-ligustilide strongly inhibitis the induction of LPS-induced inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) at both the mRNA and protein levels in a dose-dependent manner. The transcriptional activity of nuclear factor kappa B (NF-B) was also down-regulated in a concentration-dependent manner. Further study revealed that (Z)-ligustilide inhibited the phosphorylation and subsequent degradation of IBα, an inhibitor protein of NF-B. In addition, (Z)-ligustilide inhibited the phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK), extracellular signal-regulated kinase (ERK) and c-Jun NH(2)-terminal kinase (JNK) in a dose-dependent manner. Taken together, these data suggest that (Z)-ligustilide can exert its antiinflammatory effects by regulating the NF-B and MAPK signal pathways.
Collapse
Affiliation(s)
- Ji Won Chung
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 151-742, Korea
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Impellizzeri D, Mazzon E, Di Paola R, Galuppo M, Bramanti P, Zhang J, Bobb K, Monie D, Meshulam J, Sliskovic D, Cuzzocrea S. PBS-1086, a Rel Inhibitor of NF-κB, Ameliorates Collagen-Induced Arthritis in Mice. EUR J INFLAMM 2012. [DOI: 10.1177/1721727x1201000106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The family of nuclear factor-kappaB (NF-κB) transcription factors is intimately involved in the regulation of expression of numerous genes in the setting of the inflammatory response. Inflammation, cartilage degradation, cell proliferation, angiogenesis and pannus formation are hallmarks of the pathogenesis of both collagen-induced arthritis (CIA) in rodents and rheumatoid arthritis (RA) in humans. The aim of this study is to investigate the effect of PBS-1086, a ReI inhibitor of NF-κB, on the modulation of the inflammatory response in mice subjected to CIA in comparison to the effect of etanercept. CIA was induced in mice by an intradermal injection of bovine type II collagen (CII) emulsion and complete Freund's adjuvant (CFA) at the base of the tail. On day 21, a second injection of CII in CFA was administered. Mice developed erosive hind paw arthritis when immunised with CII in CFA. Macroscopic clinical evidence of CIA first appeared as peri-articular erythema and oedema in the hind paws. The incidence of CIA was 100% by day 28 in the CII challenged mice and the severity of CIA progressed over a 35-day period with a resorption of bone. The histopathology of CIA included erosion of the cartilage at the joint. Treatment with PBS-1086 starting at the onset of arthritis (day 21) ameliorated the clinical signs at days 21–35 and improved histological status in the joint and paw. In addition, it also reduced the neutrophil infiltration which is a key mediator of RA. In this study, we demonstrate that PBS-1086 exerts an anti-inflammatory effect during chronic inflammation and ameliorates the tissue damage associated with CIA. The anti-inflammatory activities of PBS-1086 are comparable to those of etanercept treatment.
Collapse
Affiliation(s)
- D. Impellizzeri
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | - E. Mazzon
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy
| | - R. Di Paola
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | - M. Galuppo
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | - P. Bramanti
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | - J. Zhang
- Profectus BioSciences, Inc., Baltimore, MD, USA
| | - K. Bobb
- Profectus BioSciences, Inc., Baltimore, MD, USA
| | - D. Monie
- Profectus BioSciences, Inc., Baltimore, MD, USA
| | - J. Meshulam
- Profectus BioSciences, Inc., Baltimore, MD, USA
| | | | - S. Cuzzocrea
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| |
Collapse
|
46
|
Atochina-Vasserman EN. S-nitrosylation of surfactant protein D as a modulator of pulmonary inflammation. Biochim Biophys Acta Gen Subj 2011; 1820:763-9. [PMID: 22183030 DOI: 10.1016/j.bbagen.2011.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 10/13/2011] [Accepted: 12/04/2011] [Indexed: 01/26/2023]
Abstract
BACKGROUND Surfactant protein D (SP-D) is a member of the family of proteins termed collagen-like lectins or "collectins" that play a role in non-antibody-mediated innate immune responses [1]. The primary function of SP-D is the modulation of host defense and inflammation [2]. SCOPE OF REVIEW This review will discuss recent findings on the physiological importance of SP-D S-nitrosylation in biological systems and potential mechanisms that govern SP-D mediated signaling. MAJOR CONCLUSIONS SP-D appears to have both pro- and anti-inflammatory signaling functions. SP-D multimerization is a critical feature of its function and plays an important role in efficient innate host defense. Under baseline conditions, SP-D forms a multimer in which the N-termini are hidden in the center and the C-termini are on the surface. This multimeric form of SP-D is limited in its ability to activate inflammation. However, NO can modify key cysteine residues in the hydrophobic tail domain of SP-D resulting in a dissociation of SP-D multimers into trimers, exposing the S-nitrosylated N-termini. The exposed S-nitrosylated tail domain binds to the calreticulin/CD91 receptor complex and initiates a pro-inflammatory response through phosphorylation of p38 and NF-κB activation [3,4]. In addition, the disassembled SP-D loses its ability to block TLR4, which also results in activation of NF-κB. GENERAL SIGNIFICANCE Recent studies have highlighted the capability of NO to modify SP-D through S-nitrosylation, causing the activation of a pro-inflammatory role for SP-D [3]. This represents a novel mechanism both for the regulation of SP-D function and NO's role in innate immunity, but also demonstrates that the S-nitrosylation can control protein function by regulating quaternary structure. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation.
Collapse
Affiliation(s)
- Elena N Atochina-Vasserman
- Pulmonary, Allergy and Critical Care Division, Department of Medicine University of Pennsylvania, Vernon and Shirley Hill Pavilion, #H410C, 380 S. University Ave., Philadelphia, PA 19104-4539, USA.
| |
Collapse
|
47
|
Zhong F, Chen H, Han L, Jin Y, Wang W. Curcumin attenuates lipopolysaccharide-induced renal inflammation. Biol Pharm Bull 2011; 34:226-32. [PMID: 21415532 DOI: 10.1248/bpb.34.226] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Renal inflammation is the main pathological change in many acute and chronic kidney diseases. Curcumin, a yellow pigment present in the rhizome of turmeric (Curcuma longa L. Zingiberaceae), was found to be a potential anti-inflammatory agent. The present study aimed to investigate the effects of curcumin on the inflammation of mice kidney and cultured renal tubular epithelial cells (HK-2 cells) induced by lipopolysaccharide (LPS) and to explore the mechanism. Curcumin was injected intraperitoneally before LPS administration. Renal inflammation was assessed by evaluating monocyte chemoattractant protein-1 (MCP-1) expression and macrophage infiltration in renal tissue using immunohistochemical methods, and also by measuring renal MCP-1 mRNA level using Real-Time polymerase chain reaction (PCR). HK-2 cells were cultured to investigate the in vitro effect of curcumin against LPS-induced renal inflammation. The expression of MCP-1 and interleukin-8 (IL-8) mRNA was measured by Real-Time PCR. The expression of MCP-1 and IL-8 protein in supernatant was detected by enzyme-linked immunosorbent assay (ELISA). The activity of nuclear factor (NF)-κB was detected by electrophoretic mobility shift assay (EMSA). The results demonstrated that curcumin could inhibit LPS-induced renal MCP-1 mRNA expression. Curcumin also significantly inhibited the expression of MCP-1 and IL-2 mRNA in HK-2 cells, and partially inhibited the secretion of MCP-1 and IL-8. Furthermore, curcumin was found to inhibit the DNA-binding activity of NF-κB. The present study demonstrated that curcumin has a protective effect on LPS-induced experimental renal inflammation, and this effect might be attributed to its inhibitory effects on MCP-1 mRNA expression and DNA-binding activity of NF-κB. Hence, curcumin might be potentially useful in some kidney diseases by preventing renal inflammation.
Collapse
Affiliation(s)
- Fang Zhong
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, PR China
| | | | | | | | | |
Collapse
|
48
|
Cuesta S, Kireev R, García C, Forman K, Vara E, Tresguerres JA. Effect of Growth Hormone Treatment on Pancreatic Inflammation, Oxidative Stress, and Apoptosis Related to Aging in SAMP8 Mice. Rejuvenation Res 2011; 14:501-12. [DOI: 10.1089/rej.2011.1166] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Sara Cuesta
- Department Physiology, Medical School, University Complutense of Madrid Medical School, University Complutense of Madrid, Madrid, Spain
| | - Roman Kireev
- Department Physiology, Medical School, University Complutense of Madrid Medical School, University Complutense of Madrid, Madrid, Spain
| | - Cruz García
- Department Biochemistry and Molecular Biology, Medical School, University Complutense of Madrid Medical School, University Complutense of Madrid, Madrid, Spain
| | - Katherine Forman
- Department Physiology, Medical School, University Complutense of Madrid Medical School, University Complutense of Madrid, Madrid, Spain
| | - Elena Vara
- Department Biochemistry and Molecular Biology, Medical School, University Complutense of Madrid Medical School, University Complutense of Madrid, Madrid, Spain
| | - Jesús A.F. Tresguerres
- Department Physiology, Medical School, University Complutense of Madrid Medical School, University Complutense of Madrid, Madrid, Spain
| |
Collapse
|
49
|
Buhrmann C, Mobasheri A, Busch F, Aldinger C, Stahlmann R, Montaseri A, Shakibaei M. Curcumin modulates nuclear factor kappaB (NF-kappaB)-mediated inflammation in human tenocytes in vitro: role of the phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem 2011; 286:28556-66. [PMID: 21669872 PMCID: PMC3151097 DOI: 10.1074/jbc.m111.256180] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Inflammatory processes play essential roles in the pathogenesis of tendinitis and tendinopathy. These events are accompanied by catabolic processes initiated by pro-inflammatory cytokines such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). Pharmacological treatments for tendinitis are restricted to the use of non-steroidal anti-inflammatory drugs. Recent studies in various cell models have demonstrated that curcumin targets the NF-κB signaling pathway. However, its potential for the treatment of tendinitis has not been explored. Herein, we used an in vitro model of human tenocytes to study the mechanism of curcumin action on IL-1β-mediated inflammatory signaling. Curcumin at concentrations of 5-20 μm inhibited IL-1β-induced inflammation and apoptosis in cultures of human tenocytes. The anti-inflammatory effects of curcumin included down-regulation of gene products that mediate matrix degradation (matrix metalloproteinase-1, -9, and -13), prostanoid production (cyclooxygenase-2), apoptosis (Bax and activated caspase-3), and stimulation of cell survival (Bcl-2), all known to be regulated by NF-κB. Furthermore, curcumin suppressed IL-1β-induced NF-κB activation via inhibition of phosphorylation and degradation of inhibitor of κBα, inhibition of inhibitor of κB-kinase activity, and inhibition of nuclear translocation of NF-κB. Furthermore, the effects of IL-1β were abrogated by wortmannin, suggesting a role for the phosphatidylinositol 3-kinase (PI-3K) pathway in IL-1β signaling. Curcumin suppressed IL-1β-induced PI-3K p85/Akt activation and its association with IKK. These results demonstrate, for the first time, a potential role for curcumin in treating tendon inflammation through modulation of NF-κB signaling, which involves PI-3K/Akt and the tendon-specific transcription factor scleraxis in tenocytes.
Collapse
Affiliation(s)
- Constanze Buhrmann
- Institute of Anatomy, Ludwig-Maximilian-University Munich, 80336 Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
50
|
Wu S, Morrison A, Sun H, De Luca F. Nuclear factor-kappaB (NF-kappaB) p65 interacts with Stat5b in growth plate chondrocytes and mediates the effects of growth hormone on chondrogenesis and on the expression of insulin-like growth factor-1 and bone morphogenetic protein-2. J Biol Chem 2011; 286:24726-34. [PMID: 21592969 DOI: 10.1074/jbc.m110.175364] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Growth hormone (GH) stimulates growth plate chondrogenesis and longitudinal bone growth with its stimulatory effects primarily mediated by insulin-like growth factor-1 (IGF-1) both systemically and locally in the growth plate. It has been shown that the transcription factor Stat5b mediates the GH promoting effect on IGF-1 expression and on chondrogenesis, yet it is not known whether other signaling molecules are activated by GH in growth plate chondrocytes. We have previously demonstrated that nuclear factor-κB p65 is a transcription factor expressed in growth plate chondrocytes where it facilitates chondrogenesis. We have also shown that fibroblasts isolated from a patient with growth failure and a heterozygous mutation of inhibitor-κBα (IκB; component of the nuclear factor-κB (NF-κB) signaling pathway) exhibit GH insensitivity. In this study, we cultured rat metatarsal bones in the presence of GH and/or pyrrolidine dithiocarbamate (PDTC), a known NF-κB inhibitor. The GH-mediated stimulation of metatarsal longitudinal growth and growth plate chondrogenesis was neutralized by PDTC. In cultured chondrocytes isolated from rat metatarsal growth plates, GH induced NF-κB-DNA binding and chondrocyte proliferation and differentiation and prevented chondrocyte apoptosis. The inhibition of NF-κB p65 expression and activity (by NF-κB p65 siRNA and PDTC, respectively) in chondrocytes reversed the GH-mediated effects on chondrocyte proliferation, differentiation, and apoptosis. Lastly, the inhibition of Stat5b expression in chondrocytes prevented the GH promoting effects on NF-κB-DNA binding, whereas the inhibition of NF-κB p65 expression or activity prevented the GH-dependent activation of IGF-1 and bone morphogenetic protein-2 expression.
Collapse
Affiliation(s)
- Shufang Wu
- Section of Endocrinology and Diabetes, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania 19134, USA
| | | | | | | |
Collapse
|