1
|
Chen J, Liu H, Fu Y, Chen X, Zhang S, Yang Y, Li S, Wang G, Lan T. Kaempferol attenuates macrophage M1 polarization and liver fibrosis by inhibiting mitogen-activated protein kinase/nuclear factor κB signaling pathway. J Pharmacol Exp Ther 2025; 392:103533. [PMID: 40139075 DOI: 10.1016/j.jpet.2025.103533] [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: 11/08/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Chronic liver inflammation is a major cause of death in patients with liver fibrosis and cirrhosis, which pose a serious health threat worldwide, and there is no effective anti-hepatic fibrosis drug. Kaempferol (KA), a flavonoid polyphenol extracted from many edible plants and traditional Chinese medicine, has been reported to possess anti-inflammatory, antioxidant, and antitumor activities and has an ameliorating effect on liver fibrosis or other fibroproliferative diseases. However, the specific regulatory mechanism of KA-reversed macrophage M1 polarization is still obscure. This study aimed to investigate the protective effects of KA on carbon tetrachloride (CCl4)-induced liver fibrosis in mice through M1 polarization. C57BL/6 mice were intraperitoneally injected with CCl4 twice weekly to induce liver fibrosis. Male mice were randomly divided into 4 groups (n = 5): the oil group, the CCl4 group, the low-dose KA-treatment CCl4 group (50 mg/kg/day KA), and the high-dose KA-treatment CCl4 group (100 mg/kg/day KA). An equal amount of solvent was given to each group by intraperitoneal injection. The results indicated that KA decreased liver pathologic changes, hepatic macrophage recruitment, and serum alanine aminotransferase levels. Notably, it reduced the activation of M1-type macrophages in the liver. The expression of proinflammatory cytokines and genes associated with M1 macrophages, such as tumor necrosis factor-α, interleukin-6, interleukin-1β, and inducible nitric oxide synthase, was also decreased. The core targets, signaling pathways, and possible mechanisms related to the M1 polarization of KA were analyzed by network pharmacology and molecular docking. Further analysis revealed that KA regulated mitogen-activated protein kinase (MAPK)/nuclear factor κB (NF-κB) signaling pathways. Finally, the results indicated that KA regulates M1 macrophage activation by modulating the MAPK/NF-κB signaling pathways. This study revealed that KA ameliorated liver injury, inflammation, and fibrosis by inhibiting macrophage M1 polarization through the MAPK/NF-κB signaling pathway, highlighting KA as a potential novel agent for the prevention and treatment of liver fibrosis. SIGNIFICANCE STATEMENT: Chronic liver inflammation is a leading cause of mortality in patients with liver fibrosis and cirrhosis, presenting a significant global health threat. Kaempferol, as a traditional Chinese medicine, effectively suppresses M1 polarization of macrophages through the mitogen-activated protein kinase/nuclear factor κB signaling pathway, thereby ameliorating liver injury, inflammation, and fibrosis. These findings underscore the potential of kaempferol as an innovative therapeutic agent for the prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Jiajia Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huanle Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanfang Fu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaolan Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shiqin Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongqi Yang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Shengwen Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guixiang Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Tian Lan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
2
|
Li Y, Shen Q, Feng L, Zhang C, Jiang X, Liu F, Pang B. A nanoscale natural drug delivery system for targeted drug delivery against ovarian cancer: action mechanism, application enlightenment and future potential. Front Immunol 2024; 15:1427573. [PMID: 39464892 PMCID: PMC11502327 DOI: 10.3389/fimmu.2024.1427573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/22/2024] [Indexed: 10/29/2024] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological malignancies in the world and is the leading cause of cancer-related death in women. The complexity and difficult-to-treat nature of OC pose a huge challenge to the treatment of the disease, Therefore, it is critical to find green and sustainable drug treatment options. Natural drugs have wide sources, many targets, and high safety, and are currently recognized as ideal drugs for tumor treatment, has previously been found to have a good effect on controlling tumor progression and reducing the burden of metastasis. However, its clinical transformation is often hindered by structural stability, bioavailability, and bioactivity. Emerging technologies for the treatment of OC, such as photodynamic therapy, immunotherapy, targeted therapy, gene therapy, molecular therapy, and nanotherapy, are developing rapidly, particularly, nanotechnology can play a bridging role between different therapies, synergistically drive the complementary role of differentiated treatment schemes, and has a wide range of clinical application prospects. In this review, nanoscale natural drug delivery systems (NNDDS) for targeted drug delivery against OC were extensively explored. We reviewed the mechanism of action of natural drugs against OC, reviewed the morphological composition and delivery potential of drug nanocarriers based on the application of nanotechnology in the treatment of OC, and discussed the limitations of current NNDDS research. After elucidating these problems, it will provide a theoretical basis for future exploration of novel NNDDS for anti-OC therapy.
Collapse
Affiliation(s)
- Yi Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Shen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Feng
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanlong Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Pang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Jover I, Ramos MC, Escámez MJ, Lozoya E, Tormo JR, de Prado-Verdún D, Mencía Á, Pont M, Puig C, Larraufie MH, Gutiérrez-Caballero C, Reyes F, Trincado JL, García-González V, Cerrato R, Andrés M, Crespo M, Vicente F, Godessart N, Genilloud O, Larcher F, Nueda A. Identification of novel small molecule-based strategies of COL7A1 upregulation and readthrough activity for the treatment of recessive dystrophic epidermolysis bullosa. Sci Rep 2024; 14:18969. [PMID: 39152155 PMCID: PMC11329504 DOI: 10.1038/s41598-024-67398-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/10/2024] [Indexed: 08/19/2024] Open
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is a rare genetic disease caused by loss of function mutations in the gene coding for collagen VII (C7) due to deficient or absent C7 expression. This disrupts structural and functional skin architecture, leading to blistering, chronic wounds, inflammation, important systemic symptoms affecting the mouth, gastrointestinal tract, cornea, and kidney function, and an increased skin cancer risk. RDEB patients have an extremely poor quality of life and often die at an early age. A frequent class of mutations in RDEB is premature termination codons (PTC), which appear in homozygosity or compound heterozygosity with other mutations. RDEB has no cure and current therapies are mostly palliative. Using patient-derived keratinocytes and a library of 8273 small molecules and 20,160 microbial extracts evaluated in a phenotypic screening interrogating C7 levels, we identified three active chemical series. Two of these series had PTC readthrough activity, and one upregulated C7 mRNA, showing synergistic activity when combined with the reference readthrough molecule gentamicin. These compounds represent novel potential small molecule-based systemic strategies that could complement topical-based treatments for RDEB.
Collapse
Affiliation(s)
- Irene Jover
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Maria C Ramos
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - María José Escámez
- Departamento de Bioingeniería E Ingeniería Aeroespacial (UC3M), División de Biomedicina Epitelial, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras, Universidad Carlos III de Madrid (UC3M), Madrid, Spain
- Unidad de Innovación Biomédica. Centro de Investigaciones Energéticas, U714-CIBER de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
- Instituto de Investigación Sanitaria, Fundación Jiménez Díaz (IISFJD), Madrid, Spain
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Estrella Lozoya
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - José R Tormo
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - Diana de Prado-Verdún
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Ángeles Mencía
- Departamento de Bioingeniería E Ingeniería Aeroespacial (UC3M), División de Biomedicina Epitelial, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras, Universidad Carlos III de Madrid (UC3M), Madrid, Spain
- Unidad de Innovación Biomédica. Centro de Investigaciones Energéticas, U714-CIBER de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
- Instituto de Investigación Sanitaria, Fundación Jiménez Díaz (IISFJD), Madrid, Spain
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Mercè Pont
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Carles Puig
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Marie-Helene Larraufie
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | | | - Fernando Reyes
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - Juan Luis Trincado
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Vicente García-González
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Rosario Cerrato
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Miriam Andrés
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Maribel Crespo
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Francisca Vicente
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - Nuria Godessart
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain
| | - Olga Genilloud
- Fundación MEDINA, Parque Tecnológico de La Salud, Av. Conocimiento 34, 18016, Granada, Spain
| | - Fernando Larcher
- Departamento de Bioingeniería E Ingeniería Aeroespacial (UC3M), División de Biomedicina Epitelial, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras, Universidad Carlos III de Madrid (UC3M), Madrid, Spain.
- Unidad de Innovación Biomédica. Centro de Investigaciones Energéticas, U714-CIBER de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain.
- Instituto de Investigación Sanitaria, Fundación Jiménez Díaz (IISFJD), Madrid, Spain.
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.
| | - Arsenio Nueda
- R&D Centre, Almirall S.A., Laureà Miró 408-410, 08980, Sant Feliu de Llobregat, Barcelona, Spain.
| |
Collapse
|
4
|
Qi J, Pan Z, Wang X, Zhang N, He G, Jiang X. Research advances of Zanthoxylum bungeanum Maxim. polyphenols in inflammatory diseases. Front Immunol 2024; 15:1305886. [PMID: 38343532 PMCID: PMC10853423 DOI: 10.3389/fimmu.2024.1305886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Zanthoxylum bungeanum Maxim., commonly known as Chinese prickly ash, is a well-known spice and traditional Chinese medicine ingredient with a rich history of use in treating inflammatory conditions. This review provides a comprehensive overview of the botanical classification, traditional applications, and anti-inflammatory effects of Z. bungeanum, with a specific focus on its polyphenolic components. These polyphenols have exhibited considerable promise, as evidenced by preclinical studies in animal models, suggesting their therapeutic potential in human inflammatory diseases such as ulcerative colitis, arthritis, asthma, chronic obstructive pulmonary disease, cardiovascular disease, and neurodegenerative conditions. This positions them as a promising class of natural compounds with the potential to enhance human well-being. However, further research is necessary to fully elucidate their mechanisms of action and develop safe and effective therapeutic applications.
Collapse
Affiliation(s)
- Jinxin Qi
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zhaoping Pan
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyun Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gu He
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Cheng C, Zhu R, Liu M, Yang H, Guo F, Du Q, Wang X, Li M, Song G, Qin R, Liu S. Kunxian capsule alleviates renal damage by inhibiting the JAK1/STAT1 pathway in lupus nephritis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116349. [PMID: 36924861 DOI: 10.1016/j.jep.2023.116349] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kunxian capsule (KXC) is a new traditional Chinese medicine drug included in "The key science and technology achievements" in the Ninth Five Year Plan of China. KXC has been clinically used for more than 10 years in the treatment of lupus nephritis (LN). However, the underlying role and molecular mechanism of KXC in LN remain unclear. AIM OF THE STUDY This study aimed to explore the efficacy and potential mechanisms of KXC through pharmacological network, in vitro and in vivo studies. MATERIALS AND METHODS Pharmacological network analysis of KXC treatment in LN was performed using data acquired from the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform (TCMSP, https://old.tcmsp-e.com/tcmsp.php) and NCBI Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo/database). HK-2 cells were chosen as an in vitro model of the tubular immune response by simulation with interferon γ (IFN-γ). MRL/lpr mice were used to explore the mechanism of KXC in vivo. Finally, the specific active molecules of KXC were further analyzed by molecular docking. RESULTS The pharmacological network analysis showed that STAT1 is a key factor in the effects of KXC. In vitro and in vivo experiments confirmed the therapeutic effect of KXC on LN renal function and tubular inflammation. The protective effect of KXC is mediated by STAT1 blockade, which further reduces T-cell infiltration and improves the renal microenvironment in LN. Two main components of KXC, Tripterygium hypoglaucum (H.Lév.) Hutch (Shanhaitang) and Epimedium brevicornu Maxim (Yinyanghuo) could block JAK1-STAT1 activation. Furthermore, we found 8 molecules that could bind to the ATP pocket of JAK1 with high affinities by performing docking analysis. CONCLUSIONS KXC inhibits renal damage and T-cell infiltration in LN by blocking the JAK1-STAT1 pathway.
Collapse
Affiliation(s)
- Chen Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rongrong Zhu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mingjian Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China
| | - Hao Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fangfang Guo
- Center of Clinical Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Qunqun Du
- Baiyunshan Chenliji Pharmaceutical Co., Ltd, Guangzhou, 510288, China
| | - Xiaolan Wang
- Baiyunshan Chenliji Pharmaceutical Co., Ltd, Guangzhou, 510288, China
| | - Minmin Li
- Center of Clinical Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Gaopeng Song
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China.
| | - Renan Qin
- Baiyunshan Chenliji Pharmaceutical Co., Ltd, Guangzhou, 510288, China.
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
6
|
Kumar S, Mehan S, Narula AS. Therapeutic modulation of JAK-STAT, mTOR, and PPAR-γ signaling in neurological dysfunctions. J Mol Med (Berl) 2023; 101:9-49. [PMID: 36478124 DOI: 10.1007/s00109-022-02272-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade is a pleiotropic pathway that involves receptor subunit multimerization. The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine-threonine kinase that perceives and integrates a variety of intracellular and environmental stimuli to regulate essential activities such as cell development and metabolism. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a prototypical metabolic nuclear receptor involved in neural differentiation and axon polarity. The JAK-STAT, mTOR, and PPARγ signaling pathways serve as a highly conserved signaling hub that coordinates neuronal activity and brain development. Additionally, overactivation of JAK/STAT, mTOR, and inhibition of PPARγ signaling have been linked to various neurocomplications, including neuroinflammation, apoptosis, and oxidative stress. Emerging research suggests that even minor disruptions in these cellular and molecular processes can have significant consequences manifested as neurological and neuropsychiatric diseases. Of interest, target modulators have been proven to alleviate neuronal complications associated with acute and chronic neurological deficits. This research-based review explores the therapeutic role of JAK-STAT, mTOR, and PPARγ signaling modulators in preventing neuronal dysfunctions in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India.
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
7
|
Network Pharmacology Analysis and Experimental Validation of Kaempferol in the Treatment of Ischemic Stroke by Inhibiting Apoptosis and Regulating Neuroinflammation Involving Neutrophils. Int J Mol Sci 2022; 23:ijms232012694. [PMID: 36293548 PMCID: PMC9604352 DOI: 10.3390/ijms232012694] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Kaempferol, a natural plant flavonoid compound, has a neuroprotective effect on ischemic stroke, while the specific mechanism remains unclear. In the current study, we applied the comprehensive strategy that combines network pharmacology and experimental evaluation to explore the potential mechanism of kaempferol in the treatment of cerebral ischemia. First, network pharmacology analysis identified the biological process of kaempferol, suggesting that kaempferol may partly help in treating ischemic stroke by regulating apoptosis and inflammatory response. Then, we evaluated the efficacy of kaempferol in the acute stage of ischemic stroke and elucidated its effects and possible mechanisms on cell apoptosis and neuroinflammation involved by neutrophils. The results showed that kaempferol could significantly reduce the modified neurological severity score (mNSS), and reduce the volume of cerebral infarction and the degree of cerebral edema. In terms of anti-apoptosis, kaempferol could significantly reduce the number of TUNEL-positive cells, inhibit the expression of pro-apoptotic proteins and promote the expression of anti-apoptotic proteins. Kaempferol may play an anti-apoptotic role by up-regulating the expression level of the BDNF-TrkB-PI3K/AKT signaling pathway. In addition, we found that kaempferol inhibited neuron loss and the activation of glial cells, as well as the expression level of the inflammatory protein COX-2 and the classic pro-inflammatory signaling pathway TLR4/MyD88/NF-κB in the ischemic brain, reduced MPO activity and neutrophil counts in peripheral blood, and down-regulated neutrophil aggregation and infiltration in the ischemic brain. Western blot revealed that kaempferol down-regulated the activation of the JAK1/STAT3 signaling pathway in neutrophils and ischemic brains. Our study showed that kaempferol inhibited the activation and number of neutrophils in the rat peripheral blood and brain, which may be related to the down-regulation of the JAK1/STAT3 pathway.
Collapse
|
8
|
Shankar A, McAlees JW, Lewkowich IP. Modulation of IL-4/IL-13 cytokine signaling in the context of allergic disease. J Allergy Clin Immunol 2022; 150:266-276. [PMID: 35934680 PMCID: PMC9371363 DOI: 10.1016/j.jaci.2022.06.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/29/2022]
Abstract
Aberrant activation of CD4 TH2 cells and excessive production of TH2 cytokines such as IL-4 and IL-13 have been implicated in the pathogenesis of allergic diseases. Generally, IL-4 and IL-13 utilize Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathways for induction of inflammatory gene expression and the effector functions associated with disease pathology in many allergic diseases. However, it is increasingly clear that JAK/STAT pathways activated by IL-4/IL-13 can themselves be modulated in the presence of other intracellular signaling programs, thereby changing the overall tone and/or magnitude of IL-4/IL-13 signaling. Apart from direct activation of the canonic JAK/STAT pathways, IL-4 and IL-13 also induce proinflammatory gene expression and effector functions through activation of additional signaling cascades. These alternative signaling cascades contribute to several specific aspects of IL-4/IL-13-associated cellular and molecular responses. A more complete understanding of IL-4/IL-13 signaling pathways, including the precise conditions under which noncanonic signaling pathways are activated, and the impact of these pathways on cellular- and host-level responses, will better allow us to design agents that target specific pathologic outcomes or tailor therapies for the treatment of uncommon disease endotypes.
Collapse
|
9
|
Aloke C, Ohanenye IC, Aja PM, Ejike CECC. Phytochemicals from medicinal plants from African forests with potentials in rheumatoid arthritis management. J Pharm Pharmacol 2022; 74:1205-1219. [PMID: 35788356 DOI: 10.1093/jpp/rgac043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 06/04/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by inflammation, pain, and cartilage and bone damage. There is currently no cure for RA. It is however managed using nonsteroidal anti-inflammatory drugs, corticosteroids and disease-modifying anti-rheumatic drugs, often with severe side effects. Hidden within Africa's lush vegetation are plants with diverse medicinal properties including anti-RA potentials. This paper reviews the scientific literature for medicinal plants, growing in Africa, with reported anti-RA activities and identifies the most abundant phytochemicals deserving research attention. A search of relevant published scientific literature, using the major search engines, such as Pubmed/Medline, Scopus, Google Scholar, etc. was conducted to identify medicinal plants, growing in Africa, with anti-RA potentials. KEY FINDINGS Twenty plants belonging to 17 families were identified. The plants are rich in phytochemicals, predominantly quercetin, rutin, catechin, kaempferol, etc., known to affect some pathways relevant in RA initiation and progression, and therefore useful in its management. SUMMARY Targeted research is needed to unlock the potentials of medicinal plants by developing easy-to-use technologies for preparing medicines from them. Research attention should focus on how best to exploit the major phytochemicals identified in this review for the development of anti-RA 'green pharmaceuticals'.
Collapse
Affiliation(s)
- Chinyere Aloke
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ebonyi State, Nigeria.,Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein 2050, Johannesburg, South Africa
| | - Ikenna C Ohanenye
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa Ontario Canada
| | - Patrick M Aja
- Department of Biochemistry, Faculty of Science, Ebonyi State University Abakaliki, Ebonyi State, Nigeria
| | - Chukwunonso E C C Ejike
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ebonyi State, Nigeria
| |
Collapse
|
10
|
Song Y, Li X, Liu F, Zhu H, Shen Y. Isoalantolactone alleviates ovalbumin‑induced asthmatic inflammation by reducing alternatively activated macrophage and STAT6/PPAR‑γ/KLF4 signals. Mol Med Rep 2021; 24:701. [PMID: 34368878 DOI: 10.3892/mmr.2021.12340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/22/2021] [Indexed: 11/09/2022] Open
Abstract
Isoalantolactone (IAL), a sesquiterpene lactone, has anti‑inflammatory activity in lipopolysaccharide (LPS)‑induced sepsis. However, it remains to be elucidated whether IAL influences asthmatic inflammation. The present study found that IAL inhibited ovalbumin (OVA)‑induced asthmatic inflammation and attenuated OVA‑induced eosinophil infiltration, immunoglobulin E generation and the production of interleukin (IL)‑4, IL‑5, C‑C motif chemokine ligand (CCL)17 and CCL22. In addition, IAL treatment with IL‑4 reduced the expression of arginase‑1, Ym‑1, CCL17 and CCL22 in bone marrow‑derived macrophages in vitro. Furthermore, IAL inhibited IL‑4‑induced STAT6 phosphorylation and the expression of peroxisome proliferator‑activated receptor γ and Krüppel‑like factor 4. Collectively, the results suggested that IAL attenuated asthmatic inflammation and is a potential therapeutic agent for the treatment of asthma.
Collapse
Affiliation(s)
- Yunduan Song
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Xiaozong Li
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Fangfang Liu
- Department of Operation Management Office, Shanghai Pudong New Area Center for Disease Control and Prevention, Fudan University Pudong Institute of Preventive Medicine, Shanghai 200136, P.R. China
| | - Hongbo Zhu
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Yao Shen
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| |
Collapse
|
11
|
Jantrapirom S, Hirunsatitpron P, Potikanond S, Nimlamool W, Hanprasertpong N. Pharmacological Benefits of Triphala: A Perspective for Allergic Rhinitis. Front Pharmacol 2021; 12:628198. [PMID: 33995026 PMCID: PMC8120106 DOI: 10.3389/fphar.2021.628198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/16/2021] [Indexed: 12/24/2022] Open
Abstract
Allergic rhinitis (AR) is considered a major nasal condition impacting a large number of people around the world, and it is now becoming a global health problem. Because the underlying mechanisms of AR are complex, the development of single-drug treatment might not be enough to treat a wide spectrum of the disease. Although the standard guidelines classify and provide suitable diagnosis and treatment, the vast majority of people with AR are still without any means of controlling it. Moreover, the benefits of AR drugs are sometimes accompanied by undesirable side effects. Thus, it is becoming a significant challenge to find effective therapies with limited undesirable side effects for a majority of patients suffering from uncontrolled AR. Aller-7/NR-A2, a polyherbal formulation, has revealed promising results in patients by reducing nasal symptoms and eosinophil counts without serious adverse effects. Interestingly, three out of seven of the herbals in the Aller-7/NR-A2 formulation are also found in an Ayurvedic polyherbal formulation known as “Triphala,” which is a potential candidate for the treatment of AR. However, there are no current studies that have examined the effects of Triphala on the disease. This review aims to describe the complexity of AR pathophysiology, currently available treatments, and the effects of Triphala on AR in order to help develop it as a promising alternative treatment in the future.
Collapse
Affiliation(s)
- Salinee Jantrapirom
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Drosophila Center for Human Diseases and Drug Discovery (DHD), Faculty of Medicine, Chiang Mai, University, Chiang Mai, Thailand
| | - Pannaphak Hirunsatitpron
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Graduate School, Chiang Mai University, Chiang Mai, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nutthiya Hanprasertpong
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
12
|
The Active Compounds and Therapeutic Mechanisms of Pentaherbs Formula for Oral and Topical Treatment of Atopic Dermatitis Based on Network Pharmacology. PLANTS 2020; 9:plants9091166. [PMID: 32916837 PMCID: PMC7569866 DOI: 10.3390/plants9091166] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023]
Abstract
To examine the molecular targets and therapeutic mechanism of a clinically proven Chinese medicinal pentaherbs formula (PHF) in atopic dermatitis (AD), we analyzed the active compounds and core targets, performed network and molecular docking analysis, and investigated interacting pathways. Information on compounds in PHF was obtained from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, and target prediction was performed using the Drugbank database. AD-related genes were gathered using the GeneCards and Online Mendelian Inheritance in Man (OMIM) databases. Network analysis was performed by Cytoscape software and protein-protein interaction was analyzed by the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). The Database for Annotation, Visualization and Integrated Discovery (DAVID) Bioinformatics Resources were applied for the enrichment analysis of the potential biological process and pathways associated with the intersection targets between PHF and AD. Autodock software was used to perform protein compound docking analysis. We identified 43 active compounds in PHF associated with 117 targets, and 57 active compounds associated with 107 targets that form the main pathways linked to oral and topical treatment of AD, respectively. Among them, quercetin, luteolin, and kaempferol are key chemicals targeting the core genes involved in the oral use of PHF against AD, while apigenin, ursolic acid, and rosmarinic acid could be used in topical treatment of PHF against AD. The compound–target–disease network constructed in the current study reveals close interactions between multiple components and multiple targets. Enrichment analysis further supports the biological processes and signaling pathways identified, indicating the involvement of IL-17 and tumor necrosis factor signaling pathways in the action of PHF on AD. Our data demonstrated the main compounds and potential pharmacological mechanisms of oral and topical application of PHF in AD.
Collapse
|
13
|
Beken B, Serttas R, Yazicioglu M, Turkekul K, Erdogan S. Quercetin Improves Inflammation, Oxidative Stress, and Impaired Wound Healing in Atopic Dermatitis Model of Human Keratinocytes. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2020; 33:69-79. [PMID: 34678092 DOI: 10.1089/ped.2019.1137] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Atopic dermatitis (AD) is a common inflammatory skin disease with complex pathogenesis. Natural flavonoids exhibit strong anti-inflammatory and antioxidant properties in many human diseases. In this study, the potential bioactive effect of quercetin, a polyphenolic plant-derived flavonoid, on the AD model of human keratinocytes was evaluated. Methods: Immortalized human HaCaT keratinocytes were treated with interleukin (IL) -4, -13, and tumor necrosis factor-α to mimic AD features in vitro. Then effects of quercetin on inflammation, oxidative stress, and wound healing were assessed. Results: Pretreatment of the cells with 1.5 μM of quercetin significantly reduced the expression of AD-induced IL-1β, IL-6, IL-8, and thymic stromal lymphopoietin, while it strongly enhanced the expression of superoxide dismutase-1 (SOD1), SOD2, catalase, glutathione peroxidase, and IL-10. Quercetin promoted wound healing by inducing epithelial-mesenchymal transition, which was supported by the upregulation of Twist and Snail mRNA expression. Unexpectedly, quercetin pretreatment of AD-induced cells upregulated the mRNA expression of occludin and E-cadherin, while downregulating matrix metalloproteinase 1 (MMP1), MMP2, and MMP9 expression. The pretreatment inhibited AD-induced phosphorylation of extracellular signal-regulated kinase 1/2/mitogen-activated protein kinase (ERK1/2 MAPK) and the expression of nuclear factor-kappa B (NF-κB), but it did not alter signal transducer and activator of transcription 6 (STAT6) phosphorylation. Conclusion: Quercetin may serve as a potential bioactive substance for atopic dermatitis-related symptoms through anti-inflammatory and antioxidant activities along with its acceleration of wound healing via ERK1/2 MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Burcin Beken
- Department of Pediatric Allergy and Immunology, School of Medicine, Trakya University, Edirne, Turkey
| | - Riza Serttas
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| | - Mehtap Yazicioglu
- Department of Pediatric Allergy and Immunology, School of Medicine, Trakya University, Edirne, Turkey
| | - Kader Turkekul
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| | - Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
14
|
HuoXueTongFu Formula Alleviates Intraperitoneal Adhesion by Regulating Macrophage Polarization and the SOCS/JAK2/STAT/PPAR- γ Signalling Pathway. Mediators Inflamm 2019; 2019:1769374. [PMID: 31772499 PMCID: PMC6854253 DOI: 10.1155/2019/1769374] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/08/2019] [Indexed: 12/17/2022] Open
Abstract
Intraperitoneal adhesion is a common complication after abdominal surgery, which seriously affects the quality of life of patients. HuoXueTongFu Formula (HXTF) plays an important role in the prevention and treatment of intraperitoneal adhesions. However, the molecular-related mechanisms are still not fully known. In this study, the model of Intrapetitoneal adhesion was established by cecum abrasion and treated with HXTF for one week. RAW264.7 cells were given LPS, IFN-γ, IL-4, HXTF-medicated serum, and PPAR-γ agonist/antagonist, respectively. Histopathology, flow cytometry, ELISA, real-time PCR, and Western blotting were used to further detect the related protein, M1/M2 polarization tendency, and PPAR-γ nuclear translocation. The deposition of collagen fibres reduced in the local area of rats after the operation with HXTF treatment. Similar to IL-4, HXTF induced a tendency for macrophages to polarize toward M2 and promoted peroxisome proliferator-activated receptor-gamma (PPAR-γ) nuclear translocation. Furthermore, the use of HXTF and PPAR-γ agonists downregulated macrophage M1 polarization-related factors IL-1, IL-6, and TNF-alpha and upregulated M2 polarization-related factors IL-4, IL-10, and TGF-beta 1. Meanwhile, the use of HXTF and PPAR-γ agonists downregulated the SOCS3/JAK2/STAT1 pathway and activated the SOCS1/STAT6/PPAR-γ pathway. These results show that HXTF may reduce intraperitoneal adhesion by inducing macrophage M2 polarization and regulating the SOCS/JAK2/STAT/PPAR-γ pathway.
Collapse
|
15
|
Champ CE, Kundu-Champ A. Maximizing Polyphenol Content to Uncork the Relationship Between Wine and Cancer. Front Nutr 2019; 6:44. [PMID: 31114789 PMCID: PMC6502998 DOI: 10.3389/fnut.2019.00044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 03/27/2019] [Indexed: 12/12/2022] Open
Abstract
Studies have revealed conflicting results regarding the risk of cancer from alcohol consumption. Furthermore, some studies have suggested that wine may have benefits that separate it from other alcoholic beverages. As wine contains a significant amount of chemicals, specifically polyphenols like anthocyanins and proanthocyanidins (PA), that can affect cellular function and promote health, this hypothesis is reasonably supported by recent research. Polyphenols promote several anticancer cellular pathways, including xenobiotic metabolism, support of innate antioxidant production, and stimulation of phase I and II detoxification of carcinogens. However, the multitude of growing and production conditions of grapes, including temperature, water availability, soil type, maceration, and aging can result in a remarkably varying final product based on the available literature. Thus, we hypothesize that wines produced from grapes cultivated between steady daily temperatures at 15–25°C with moderate sun exposure from flowering to harvest, lower vine-water status, resulting either from lower precipitation, and irrigation practices or more permeable soil types, limitation of fertilizers, extended maceration, and aging in oak will impact the concentration of anthocyanins and PA in the finished wine and may have a differential impact on cancer. This higher concentration of polyphenols would, in theory, create a healthier wine, thus explaining the conflicting reports on the benefits or harms of wine.
Collapse
Affiliation(s)
- Colin E. Champ
- Cancer Prevention Project, Pittsburgh, PA, United States
- Department of Radiation Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- *Correspondence: Colin E. Champ
| | | |
Collapse
|
16
|
Rojas DA, Iturra PA, Méndez A, Ponce CA, Bustamante R, Gallo M, Bórquez P, Vargas SL. Increase in secreted airway mucins and partial Muc5b STAT6/FoxA2 regulation during Pneumocystis primary infection. Sci Rep 2019; 9:2078. [PMID: 30765827 PMCID: PMC6376022 DOI: 10.1038/s41598-019-39079-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/19/2018] [Indexed: 12/19/2022] Open
Abstract
Airway mucus responses to subclinical infections may explain variations in progression of chronic lung diseases and differences in clinical expression of respiratory infections across individuals. Pneumocystis associates to more severe Chronic Obstructive Pulmonary Disease (COPD), asthma, respiratory distress of premature newborns, and is a consistent subclinical infection between 2 and 5 months of age when hospitalizations for respiratory cause and infant mortality are higher. This atypical fungus associates to increased mucin 5AC (MUC5AC), a central effector of Th2-type allergic inflammation, in infant lungs. However, mucus progression, expression of MUC5B essential for airway defense, and potential for pharmacologic modulation of mucus during Pneumocystis infection remain unknown. We measured MUC5B and Pneumocystis in infant lungs, and progression of mucin levels and effect of inhibition of the STAT6/FoxA2 mucus pathway using Kaempferol, a JAK/STAT6 inhibitor, in immunocompetent rats during Pneumocystis primary infection. Pneumocystis associated to increased MUC5B in infant lungs. Muc5b increased earlier and more abundantly than Muc5ac during experimental primary infection suggesting an acute defensive response against Pneumocystis as described against bacteria, while increased Muc5ac levels supports an ongoing allergic, Th2 lymphocyte-type response during primary Pneumocystis infection. Kaempferol partly reversed Muc5b stimulation suggesting limited potential for pharmacological modulation via the STAT6-FoxA2 pathway.
Collapse
Affiliation(s)
- Diego A Rojas
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Pablo A Iturra
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Andrea Méndez
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Carolina A Ponce
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Rebeca Bustamante
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Miriam Gallo
- Servicio Médico Legal de Santiago, Av. La Paz 1012, Independencia, Santiago, 8380454, Chile
| | - Pamela Bórquez
- Servicio Médico Legal de Santiago, Av. La Paz 1012, Independencia, Santiago, 8380454, Chile
| | - Sergio L Vargas
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile.
| |
Collapse
|
17
|
Potential Beneficial Effects of Wine Flavonoids on Allergic Diseases. Diseases 2019; 7:diseases7010008. [PMID: 30650667 PMCID: PMC6473930 DOI: 10.3390/diseases7010008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
Wine, a widely consumed beverage, comprises several biophenols that promote health. Flavonoids, majorly present in red wine, have been shown to have antioxidant, anti-inflammatory, anticancer, and immunomodulatory activities. Regular consumption of red wine (100 mL/day) is estimated to provide an average of 88 mg of flavonoids, whereas recent epidemiological studies indicate that wine is one of the major sources of flavonoid intake amongst wine lovers in European countries (providing an average intake of 291⁻374 mg/day of flavonoids). In addition to being antioxidants, in vitro studies suggest that flavonoids also have anti-allergic activities that inhibit IgE synthesis, activation of mast cells and basophils or other inflammatory cells, and production of inflammatory mediators, including cytokines. Furthermore, they affect the differentiation of naïve CD4+ T cells into effector T cell subsets. Moreover, several studies have reported the benefits of flavonoids in allergic models such as atopic dermatitis, asthma, anaphylaxis, and food allergy; however, evidence in humans is limited to allergic rhinitis and respiratory allergy. Although further evaluation is required, it is expected that an appropriate intake of flavonoids may be beneficial in preventing, and eventually managing, allergic diseases.
Collapse
|
18
|
Choi W, Yang AX, Waltenburg MA, Choe S, Steiner M, Radwan A, Lin J, Maddox CW, Stern AW, Fredrickson RL, Lau GW. FOXA2 depletion leads to mucus hypersecretion in canine airways with respiratory diseases. Cell Microbiol 2018; 21:e12957. [DOI: 10.1111/cmi.12957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Woosuk Choi
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Alina X. Yang
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Michelle A. Waltenburg
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Shawn Choe
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Madeline Steiner
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Ahmed Radwan
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Jingjun Lin
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Carrol W. Maddox
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
- Veterinary Diagnostic Laboratory, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Adam W. Stern
- Veterinary Diagnostic Laboratory, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
- Department of Veterinary Clinical Medicine, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Richard L. Fredrickson
- Veterinary Diagnostic Laboratory, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
- Department of Veterinary Clinical Medicine, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Gee W. Lau
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| |
Collapse
|
19
|
Kapinova A, Kubatka P, Golubnitschaja O, Kello M, Zubor P, Solar P, Pec M. Dietary phytochemicals in breast cancer research: anticancer effects and potential utility for effective chemoprevention. Environ Health Prev Med 2018; 23:36. [PMID: 30092754 PMCID: PMC6085646 DOI: 10.1186/s12199-018-0724-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022] Open
Abstract
Cancerous tissue transformation developing usually over years or even decades of life is a highly complex process involving strong stressors damaging DNA, chronic inflammation, comprehensive interaction between relevant molecular pathways, and cellular cross-talk within the neighboring tissues. Only the minor part of all cancer cases are caused by inborn predisposition; the absolute majority carry a sporadic character based on modifiable risk factors which play a central role in cancer prevention. Amongst most promising candidates for dietary supplements are bioactive phytochemicals demonstrating strong anticancer effects. Abundant evidence has been collected for beneficial effects of flavonoids, carotenoids, phenolic acids, and organosulfur compounds affecting a number of cancer-related pathways. Phytochemicals may positively affect processes of cell signaling, cell cycle regulation, oxidative stress response, and inflammation. They can modulate non-coding RNAs, upregulate tumor suppressive miRNAs, and downregulate oncogenic miRNAs that synergically inhibits cancer cell growth and cancer stem cell self-renewal. Potential clinical utility of the phytochemicals is discussed providing examples for chemoprevention against and therapy for human breast cancer. Expert recommendations are provided in the context of preventive medicine.
Collapse
Affiliation(s)
- A. Kapinova
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Malá Hora 4C, 036 01 Martin, Slovak Republic
| | - P. Kubatka
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Malá Hora 4C, 036 01 Martin, Slovak Republic
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Malá Hora 4, 036 01 Martin, Slovak Republic
| | - O. Golubnitschaja
- Radiological Clinic, Breast Cancer Research Center, Center for Integrated Oncology, Cologne-Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Str 25, 53105 Bonn, Germany
| | - M. Kello
- Faculty of Medicine, Department of Pharmacology, University of Pavol Jozef Šafárik, Trieda SNP 1, 040 11, Košice, Slovak Republic
| | - P. Zubor
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Malá Hora 4C, 036 01 Martin, Slovak Republic
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollárova 2, 03601 Martin, Slovak Republic
| | - P. Solar
- Faculty of Medicine, Department of Medical Biology, University of Pavol Jozef Šafárik, Trieda SNP 1, 040 11 Košice, Slovak Republic
| | - M. Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Malá Hora 4, 036 01 Martin, Slovak Republic
| |
Collapse
|
20
|
Pandey S, Cabot PJ, Shaw PN, Hewavitharana AK. Anti-inflammatory and immunomodulatory properties of Carica papaya. J Immunotoxicol 2016; 13:590-602. [PMID: 27416522 DOI: 10.3109/1547691x.2016.1149528] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation is linked with the generation and progression of various diseases such as cancer, diabetes and atherosclerosis, and anti-inflammatory drugs therefore have the potential to assist in the treatment of these conditions. Carica papaya is a tropical plant that is traditionally used in the treatment of various ailments including inflammatory conditions. A literature search was conducted by using the keywords "papaya", "anti-inflammatory and inflammation" and "immunomodulation and immune" along with cross-referencing. Both in vitro and in vivo investigation studies were included. This is a review of all studies published since 2000 on the anti-inflammatory activity of papaya extracts and their effects on various immune-inflammatory mediators. Studies on the anti-inflammatory activities of recognized phytochemicals present in papaya are also included. Although in vitro and in vivo studies have shown that papaya extracts and papaya-associated phytochemicals possess anti-inflammatory and immunomodulatory properties, clinical studies are lacking.
Collapse
Affiliation(s)
- Saurabh Pandey
- a School of Pharmacy , The University of Queensland , Brisbane , Australia
| | - Peter J Cabot
- a School of Pharmacy , The University of Queensland , Brisbane , Australia
| | - P Nicholas Shaw
- a School of Pharmacy , The University of Queensland , Brisbane , Australia
| | | |
Collapse
|
21
|
Chung MY, Shin HS, Choi DW, Shon DH. Citrus Tachibana
Leaf Extract Mitigates Symptoms of Food Allergy by Inhibiting Th2-Associated Responses. J Food Sci 2016; 81:H1537-45. [DOI: 10.1111/1750-3841.13315] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 02/25/2016] [Accepted: 03/24/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Min-Yu Chung
- Div. of Creative Food Science for Health; Korea Food Research Inst; Seongnam 463-746 Republic of Korea
| | - Hee Soon Shin
- Food Biotechnology Program; Korea Univ. of Science and Technology; Daejeon 305-350 Republic of Korea
| | - Dae Woon Choi
- Div. of Creative Food Science for Health; Korea Food Research Inst; Seongnam 463-746 Republic of Korea
| | - Dong-Hwa Shon
- Food Biotechnology Program; Korea Univ. of Science and Technology; Daejeon 305-350 Republic of Korea
| |
Collapse
|
22
|
Beekmann K, de Haan LHJ, Actis-Goretta L, van Bladeren PJ, Rietjens IMCM. Effect of Glucuronidation on the Potential of Kaempferol to Inhibit Serine/Threonine Protein Kinases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:1256-1263. [PMID: 26808477 DOI: 10.1021/acs.jafc.5b05456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
To study the effect of metabolic conjugation of flavonoids on the potential to inhibit protein kinase activity, the inhibitory effects of the dietary flavonol kaempferol and its major plasma conjugate kaempferol-3-O-glucuronide on protein kinases were studied. To this end, the inhibition of the phosphorylation activity of recombinant protein kinase A (PKA) and of cell lysate from the hepatocellular carcinoma cell line HepG2 on 141 putative serine/threonine phosphorylation sites derived from human proteins was assessed. Glucuronidation reduced the inhibitory potency of kaempferol on the phosphorylation activity of PKA and HepG2 lysate on average about 16 and 3.5 times, respectively, but did not appear to affect the target selectivity for kinases present in the lysate. The data demonstrate that, upon glucuronidation, kaempferol retains part of its intrinsic kinase inhibition potential, which implies that K3G does not necessarily need to be deconjugated to the aglycone for a potential inhibitory effect on protein kinases.
Collapse
Affiliation(s)
- Karsten Beekmann
- Division of Toxicology, Wageningen University , Postbus 8000, 6700EA, Wageningen, The Netherlands
| | - Laura H J de Haan
- Division of Toxicology, Wageningen University , Postbus 8000, 6700EA, Wageningen, The Netherlands
| | - Lucas Actis-Goretta
- Nestlé Research Center, Nestec Ltd., Vers-chez-les-Blanc, Case Postale 44, 1000 Lausanne 26, Switzerland
| | - Peter J van Bladeren
- Division of Toxicology, Wageningen University , Postbus 8000, 6700EA, Wageningen, The Netherlands
- Nestlé Research Center, Nestec Ltd., Vers-chez-les-Blanc, Case Postale 44, 1000 Lausanne 26, Switzerland
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University , Postbus 8000, 6700EA, Wageningen, The Netherlands
| |
Collapse
|
23
|
Wang Z, Dabrosin C, Yin X, Fuster MM, Arreola A, Rathmell WK, Generali D, Nagaraju GP, El-Rayes B, Ribatti D, Chen YC, Honoki K, Fujii H, Georgakilas AG, Nowsheen S, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich B, Yang X, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Halicka D, Mohammed SI, Azmi AS, Bilsland A, Keith WN, Jensen LD. Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol 2015; 35 Suppl:S224-S243. [PMID: 25600295 PMCID: PMC4737670 DOI: 10.1016/j.semcancer.2015.01.001] [Citation(s) in RCA: 336] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 12/25/2014] [Accepted: 01/08/2015] [Indexed: 12/20/2022]
Abstract
Deregulation of angiogenesis--the growth of new blood vessels from an existing vasculature--is a main driving force in many severe human diseases including cancer. As such, tumor angiogenesis is important for delivering oxygen and nutrients to growing tumors, and therefore considered an essential pathologic feature of cancer, while also playing a key role in enabling other aspects of tumor pathology such as metabolic deregulation and tumor dissemination/metastasis. Recently, inhibition of tumor angiogenesis has become a clinical anti-cancer strategy in line with chemotherapy, radiotherapy and surgery, which underscore the critical importance of the angiogenic switch during early tumor development. Unfortunately the clinically approved anti-angiogenic drugs in use today are only effective in a subset of the patients, and many who initially respond develop resistance over time. Also, some of the anti-angiogenic drugs are toxic and it would be of great importance to identify alternative compounds, which could overcome these drawbacks and limitations of the currently available therapy. Finding "the most important target" may, however, prove a very challenging approach as the tumor environment is highly diverse, consisting of many different cell types, all of which may contribute to tumor angiogenesis. Furthermore, the tumor cells themselves are genetically unstable, leading to a progressive increase in the number of different angiogenic factors produced as the cancer progresses to advanced stages. As an alternative approach to targeted therapy, options to broadly interfere with angiogenic signals by a mixture of non-toxic natural compound with pleiotropic actions were viewed by this team as an opportunity to develop a complementary anti-angiogenesis treatment option. As a part of the "Halifax Project" within the "Getting to know cancer" framework, we have here, based on a thorough review of the literature, identified 10 important aspects of tumor angiogenesis and the pathological tumor vasculature which would be well suited as targets for anti-angiogenic therapy: (1) endothelial cell migration/tip cell formation, (2) structural abnormalities of tumor vessels, (3) hypoxia, (4) lymphangiogenesis, (5) elevated interstitial fluid pressure, (6) poor perfusion, (7) disrupted circadian rhythms, (8) tumor promoting inflammation, (9) tumor promoting fibroblasts and (10) tumor cell metabolism/acidosis. Following this analysis, we scrutinized the available literature on broadly acting anti-angiogenic natural products, with a focus on finding qualitative information on phytochemicals which could inhibit these targets and came up with 10 prototypical phytochemical compounds: (1) oleanolic acid, (2) tripterine, (3) silibinin, (4) curcumin, (5) epigallocatechin-gallate, (6) kaempferol, (7) melatonin, (8) enterolactone, (9) withaferin A and (10) resveratrol. We suggest that these plant-derived compounds could be combined to constitute a broader acting and more effective inhibitory cocktail at doses that would not be likely to cause excessive toxicity. All the targets and phytochemical approaches were further cross-validated against their effects on other essential tumorigenic pathways (based on the "hallmarks" of cancer) in order to discover possible synergies or potentially harmful interactions, and were found to generally also have positive involvement in/effects on these other aspects of tumor biology. The aim is that this discussion could lead to the selection of combinations of such anti-angiogenic compounds which could be used in potent anti-tumor cocktails, for enhanced therapeutic efficacy, reduced toxicity and circumvention of single-agent anti-angiogenic resistance, as well as for possible use in primary or secondary cancer prevention strategies.
Collapse
Affiliation(s)
- Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Charlotta Dabrosin
- Department of Oncology, Linköping University, Linköping, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Xin Yin
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, San Diego, CA, USA
| | - Mark M Fuster
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, San Diego, CA, USA
| | - Alexandra Arreola
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - W Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Daniele Generali
- Molecular Therapy and Pharmacogenomics Unit, AO Isituti Ospitalieri di Cremona, Cremona, Italy
| | - Ganji P Nagaraju
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - Yi Charlie Chen
- Department of Biology, Alderson Broaddus University, Philippi, WV, USA
| | - Kanya Honoki
- Department of Orthopedic Surgery, Arthroplasty and Regenerative Medicine, Nara Medical University, Nara, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Arthroplasty and Regenerative Medicine, Nara Medical University, Nara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirate University, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirate University, United Arab Emirates
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust Laboratory, Guilford, Surrey, UK
| | | | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Asfar S Azmi
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Lasse D Jensen
- Department of Medical, and Health Sciences, Linköping University, Linköping, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Tan L, Akahane K, McNally R, Reyskens KMSE, Ficarro SB, Liu S, Herter-Sprie GS, Koyama S, Pattison MJ, Labella K, Johannessen L, Akbay EA, Wong KK, Frank DA, Marto JA, Look TA, Arthur JSC, Eck MJ, Gray NS. Development of Selective Covalent Janus Kinase 3 Inhibitors. J Med Chem 2015; 58:6589-606. [PMID: 26258521 DOI: 10.1021/acs.jmedchem.5b00710] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Janus kinases (JAKs) and their downstream effectors, signal transducer and activator of transcription proteins (STATs), form a critical immune cell signaling circuit, which is of fundamental importance in innate immunity, inflammation, and hematopoiesis, and dysregulation is frequently observed in immune disease and cancer. The high degree of structural conservation of the JAK ATP binding pockets has posed a considerable challenge to medicinal chemists seeking to develop highly selective inhibitors as pharmacological probes and as clinical drugs. Here we report the discovery and optimization of 2,4-substituted pyrimidines as covalent JAK3 inhibitors that exploit a unique cysteine (Cys909) residue in JAK3. Investigation of structure-activity relationship (SAR) utilizing biochemical and transformed Ba/F3 cellular assays resulted in identification of potent and selective inhibitors such as compounds 9 and 45. A 2.9 Å cocrystal structure of JAK3 in complex with 9 confirms the covalent interaction. Compound 9 exhibited decent pharmacokinetic properties and is suitable for use in vivo. These inhibitors provide a set of useful tools to pharmacologically interrogate JAK3-dependent biology.
Collapse
Affiliation(s)
- Li Tan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | - Randall McNally
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Kathleen M S E Reyskens
- Division of Cell Signaling and Immunology, College of Life Sciences, University of Dundee , Dundee DD1 5EH. U.K
| | - Scott B Ficarro
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | | | | | - Michael J Pattison
- Division of Cell Signaling and Immunology, College of Life Sciences, University of Dundee , Dundee DD1 5EH. U.K
| | | | - Liv Johannessen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | | | | | - Jarrod A Marto
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | - J Simon C Arthur
- Division of Cell Signaling and Immunology, College of Life Sciences, University of Dundee , Dundee DD1 5EH. U.K
| | - Michael J Eck
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| |
Collapse
|
25
|
Lin Y, Chen Z, Kato S. Receptor-selective IL-4 mutein modulates inflammatory vascular cell phenotypes and attenuates atherogenesis in apolipoprotein E-knockout mice. Exp Mol Pathol 2015; 99:116-27. [DOI: 10.1016/j.yexmp.2015.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/15/2015] [Indexed: 01/25/2023]
|
26
|
Kaempferol pretreatment modulates systemic inflammation and oxidative stress following hemorrhagic shock in mice. Chin Med 2015; 10:6. [PMID: 25798187 PMCID: PMC4369346 DOI: 10.1186/s13020-015-0035-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 03/06/2015] [Indexed: 11/16/2022] Open
Abstract
Background Kaempferol has been reported as beneficial for both acute and chronic inflammatory diseases. This study aims to investigate whether kaempferol affects systemic inflammation and oxidative stress in the heart, lung, and liver after hemorrhagic shock in mice. Methods Male C57/BL6 mice underwent hemorrhagic shock (mean arterial pressure of 35 mmHg for 90 min) and were arbitrarily divided into Sham, hemorrhagic shock (HS), and Kae groups (n = 10 in each group). Mice in the Kae groups received a kaempferol (10-mg/kg body weight) injection 12 h prior to (Group Kae PT) or 90 min after (Group Kae T) the initiation of hemorrhagic shock. Plasma proinflammatory cytokines (TNF-α and IL-6), organ myeloperoxidase (MPO) and superoxide dismutase (SOD) activities, and organ malondialdehyde (MDA) concentrations and heme oxygenase-1 (HO-1) expression levels were assessed by enzyme-linked immunosorbent assay (ELISA) or western blot assay. Results Compared with the HS group and the Kae T group, pretreatment with kaempferol significantly decreased proinflammatory cytokines TNF-α (P = 0.012 and 0.015, respectively) and IL-6 (P = 0.023 and 0.014, respectively) following hemorrhagic shock. Kae pretreatment reverted MPO, SOD, and MDA to basal levels in the heart, lung, and liver (Ps < 0.05), while the Kae T group showed no significant differences in these biomarkers compared with the HS group (Ps > 0.05). HO-1 expression was significantly increased in the Kae PT group compared with the other groups (P = 0.011 vs. HS group and P = 0.02 vs. Kae T group). Conclusions Pretreatment of hemorrhagic shock mice with kaempferol significantly decreased plasma levels of TNF-α and IL-6; reverted MPO, SOD, and MDA in the heart, lung, and liver; and increased expression of HO-1 in the same organs.
Collapse
|
27
|
Mycoplasma pneumoniae modulates STAT3-STAT6/EGFR-FOXA2 signaling to induce overexpression of airway mucins. Infect Immun 2014; 82:5246-55. [PMID: 25287927 DOI: 10.1128/iai.01989-14] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aberrant mucin secretion and accumulation in the airway lumen are clinical hallmarks associated with various lung diseases such as asthma, chronic obstructive pulmonary disease, and cystic fibrosis. Mycoplasma pneumoniae, long appreciated as one of the triggers of acute exacerbations of chronic pulmonary diseases, has recently been reported to promote excessive mucus secretion. However, the mechanism of mucin overproduction induced by M. pneumoniae remains unclear. This study aimed to determine the mechanism by which M. pneumoniae induces mucus hypersecretion by using M. pneumoniae infection of mouse lungs, human primary bronchial epithelial (NHBE) cells cultured at the air-liquid interface, and the conventionally cultured airway epithelial NCI-H292 cell line. We demonstrated that M. pneumoniae induced the expression of mucins MUC5AC and MUC5B by activating the STAT6-STAT3 and epidermal growth factor receptor (EGFR) signal pathways, which in turn downregulated FOXA2, a transcriptional repressor of mucin biosynthesis. The upstream stimuli of these pathways, including interleukin-4 (IL-4), IL-6, and IL-13, increased dramatically upon exposure to M. pneumoniae. Inhibition of the STAT6, STAT3, and EGFR signaling pathways significantly restored the expression of FOXA2 and attenuated the expression of airway mucins MUC5AC and MUC5B. Collectively, these studies demonstrated that M. pneumoniae induces airway mucus hypersecretion by modulating the STAT/EGFR-FOXA2 signaling pathways.
Collapse
|
28
|
So JS, Kim GC, Song M, Lee CG, Park E, Kim HJ, Kim YS, Jun CD, Im SH. 6-Methoxyflavone inhibits NFAT translocation into the nucleus and suppresses T cell activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:2772-2783. [PMID: 25114106 DOI: 10.4049/jimmunol.1400285] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
NFAT plays a crucial role in the immune system by regulating the transcription of inducible genes during immune responses. In T cells, NFAT proteins govern various cellular events related to T cell development, activation, tolerance induction, and differentiation. We previously reported the NFAT1-dependent enhancer activity of conserved noncoding sequence (CNS)-9, a distal cis-acting element, in the regulation of IL-10 transcription in T cells. In this study, we developed a T cell-based reporter system to identify compounds that modulate the regulatory activity of CNS-9. Among the identified candidates, 6-methoxyflavone (6-MF) significantly inhibited the enhancer activity of CNS-9, thereby reducing IL-10 expression in T cells without affecting cell viability. 6-MF also downregulated the transcription of NFAT1 target genes such as IL-4, IL-13, and IFN-γ. Treatment of 6-MF inhibited the translocation of NFAT1 into the nucleus, which consequently interrupted NFAT1 binding to the target loci, without affecting the expression or dephosphorylation of NFAT1. Treatment of 6-MF to CD4(+) T cells or B cells isolated from mice with atopic dermatitis significantly reduced disease-associated cytokine production, as well as the levels of IgE. In addition, oral administration of 6-MF to atopic dermatitis mice ameliorated disease symptoms by reducing serum IgE levels and infiltrating lymphocytes. Conclusively, our results suggest that 6-MF can be a potential candidate for the development of an effective immunomodulator via the suppression of NFAT-mediated T cell activation.
Collapse
Affiliation(s)
- Jae-Seon So
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10021
| | - Gi-Cheon Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea; Academy of Immunology and Microbiology, Institute for Basic Science, Pohang 790-784, Republic of Korea
| | - Minkyung Song
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY 10021
| | - Choong-Gu Lee
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang 790-784, Republic of Korea
| | - Eunbee Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea; Academy of Immunology and Microbiology, Institute for Basic Science, Pohang 790-784, Republic of Korea
| | - Ho Jin Kim
- National Cancer Center, Korea, Goyang 410-769, Republic of Korea
| | - Young Sup Kim
- Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, Daejeon 305-600, Republic of Korea; and
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea
| | - Sin-Hyeog Im
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang 790-784, Republic of Korea; Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| |
Collapse
|
29
|
Kaempferol, a potential cytostatic and cure for inflammatory disorders. Eur J Med Chem 2014; 86:103-12. [PMID: 25147152 DOI: 10.1016/j.ejmech.2014.08.011] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 08/04/2014] [Accepted: 08/04/2014] [Indexed: 12/30/2022]
Abstract
Kaempferol (3,5,7-trihydroxy-2-(4-hydroxyphenyl)-4H-1-benzopyran-4-one) is a flavonoid found in many edible plants (e.g., tea, broccoli, cabbage, kale, beans, endive, leek, tomato, strawberries, and grapes) and in plants or botanical products commonly used in traditional medicine (e.g., Ginkgo biloba, Tilia spp, Equisetum spp, Moringa oleifera, Sophora japonica and propolis). Its anti-oxidant/anti-inflammatory effects have been demonstrated in various disease models, including those for encephalomyelitis, diabetes, asthma, and carcinogenesis. Moreover, kaempferol act as a scavenger of free radicals and superoxide radicals as well as preserve the activity of various anti-oxidant enzymes such as catalase, glutathione peroxidase, and glutathione-S-transferase. The anticancer effect of this flavonoid is mediated through different modes of action, including anti-proliferation, apoptosis induction, cell-cycle arrest, generation of reactive oxygen species (ROS), and anti-metastasis/anti-angiogenesis activities. In addition, kaempferol was found to exhibit its anticancer activity through the modulation of multiple molecular targets including p53 and STAT3, through the activation of caspases, and through the generation of ROS. The anti-tumor effects of kaempferol have also been investigated in tumor-bearing mice. The combination of kaempferol and conventional chemotherapeutic drugs produces a greater therapeutic effect than the latter, as well as reduces the toxicity of the latter. In this review, we summarize the anti-oxidant/anti-inflammatory and anticancer effects of kaempferol with a focus on its molecular targets and the possible use of this flavonoid for the treatment of inflammatory diseases and cancer.
Collapse
|
30
|
Kaempferol Inhibits IL-1β-Stimulated, RANKL-mediated Osteoclastogenesis via Downregulation of MAPKs, c-Fos, and NFATc1. Inflammation 2014; 37:1221-30. [DOI: 10.1007/s10753-014-9849-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
31
|
Lin CW, Chen PN, Chen MK, Yang WE, Tang CH, Yang SF, Hsieh YS. Kaempferol reduces matrix metalloproteinase-2 expression by down-regulating ERK1/2 and the activator protein-1 signaling pathways in oral cancer cells. PLoS One 2013; 8:e80883. [PMID: 24278338 PMCID: PMC3835430 DOI: 10.1371/journal.pone.0080883] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 10/18/2013] [Indexed: 12/13/2022] Open
Abstract
Background Kaempferol has been proposed as a potential drug for cancer chemoprevention and treatment because it is a natural polyphenol contained in plant-based foods. Recent studies have demonstrated that kaempferol protects against cardiovascular disease and cancer. Based on this finding, we investigated the mechanisms by which kaempferol produces the anti-metastatic effect in human tongue squamous cell carcinoma SCC4 cells. Methodology/Principal Findings In this study, we provided molecular evidence associated with the anti-metastatic effect of kaempferol by demonstrating a substantial suppression of SCC4 cell migration and invasion. This effect was associated with reduced expressions of MMP-2 and TIMP-2 mRNA and protein levels. Analysis of the transcriptional regulation indicated that kaempferol inhibited MMP-2 transcription by suppressing c-Jun activity. Kaempferol also produced an inhibitory effect on the phosphorylation of ERK1/2. Conclusions These findings provide new insights into the molecular mechanisms involved in the anti-metastatic effect of kaempferol, and are valuable in the prevention of oral cancer metastasis.
Collapse
Affiliation(s)
- Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail: (Y-SH); (S-FY)
| | - Yih-Shou Hsieh
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail: (Y-SH); (S-FY)
| |
Collapse
|
32
|
STAT6 siRNA matrix-loaded gelatin nanocarriers: formulation, characterization, and ex vivo proof of concept using adenocarcinoma cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:858946. [PMID: 24191252 PMCID: PMC3806510 DOI: 10.1155/2013/858946] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/13/2013] [Indexed: 12/22/2022]
Abstract
The clinical utility of siRNA therapy has been hampered due to poor cell penetration, nonspecific effects, rapid degradation, and short half-life. We herewith proposed the formulation development of STAT6 siRNA (S6S) nanotherapeutic agent by encapsulating them within gelatin nanocarriers (GNC). The prepared nanoformulation was characterized for size, charge, loading efficiency, release kinetics, stability, cytotoxicity, and gene silencing assay. The stability of S6S-GNC was also assessed under conditions of varying pH, serum level, and using electrophoretic assays. In vitro cytotoxicity performance was evaluated in human adenocarcinoma A549 cells following MTT assay. The developed formulation resulted in an average particle size, surface charge, and encapsulation efficiency as 70 ± 6.5 nm, +10 ± 1.5 mV, and 85 ± 4.0%, respectively. S6S-GNC showed an insignificant (P < 0.05) change in the size and charge in the presence of buffer solutions (pH 6.4 to 8.4) and FBS (10% v/v). A549 cells were treated with native S6S, S6S-lipofectamine, placebo-GNC, and S6S-GNC using untreated cells as a control. It was observed that cell viability was decreased significantly with S6S-GNC by 55 ± 4.1% (P < 0.001) compared to native S6S (2.0 ± 0.55%) and S6S-lipofectamine complex (40 ± 3.1%). This investigation infers that gelatin polymer-based nanocarriers are a robust, stable, and biocompatible strategy for the delivery of siRNA.
Collapse
|
33
|
Sanson M, Distel E, Fisher EA. HDL induces the expression of the M2 macrophage markers arginase 1 and Fizz-1 in a STAT6-dependent process. PLoS One 2013; 8:e74676. [PMID: 23991225 PMCID: PMC3749183 DOI: 10.1371/journal.pone.0074676] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 08/03/2013] [Indexed: 12/22/2022] Open
Abstract
Our lab has previously shown in a mouse model that normalization of a low HDL level achieves atherosclerotic plaque regression. This included the shift from a pro ("M1") to an anti-inflammatory ("M2") phenotypic state of plaque macrophages. Whether HDL can directly cause this phenotypic change and, if so, what the signaling mechanism is, were explored in the present studies. Murine primary macrophages treated with HDL showed increased gene expression for the M2 markers Arginase-1 (Arg-1) and Fizz-1, which are classically induced by IL-4. HDL was able to potentiate the IL-4-induced changes in Arg-1, and tended to do the same for Fizz-1, while suppressing the expression of inflammatory genes in response to IFNγ. The effects of either IL-4 or HDL were suppressed when macrophages were from STAT6(-/-) mice, but inhibitor studies suggested differential utilization of JAK isoforms by IL-4 and HDL to activate STAT6 by phosphorylation. Overall, our results describe a new function of HDL, namely its ability to directly enrich macrophages in markers of the M2, anti-inflammatory, state in a process requiring STAT6.
Collapse
Affiliation(s)
- Marie Sanson
- The Leon H. Charney Division of Cardiology and Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Emilie Distel
- The Leon H. Charney Division of Cardiology and Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Edward A. Fisher
- The Leon H. Charney Division of Cardiology and Marc and Ruti Bell Program in Vascular Biology, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
34
|
Klementiev B, Enevoldsen MN, Li S, Carlsson R, Liu Y, Issazadeh-Navikas S, Bock E, Berezin V. Antiinflammatory properties of a peptide derived from interleukin-4. Cytokine 2013; 64:112-21. [PMID: 23972727 DOI: 10.1016/j.cyto.2013.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 06/19/2013] [Accepted: 07/20/2013] [Indexed: 12/23/2022]
Abstract
Interleukin-4 (IL-4) is a potent antiinflammatory cytokine. However its use in the clinic is hampered by side effects. We here describe the identification of a novel synthetic peptide, termed Ph8, derived from α-helix C of IL-4, which interacts with IL-4 receptor α (IL-4Rα). Employing various cultured genetically engineered cell lines and primary lymphocytes, surface plasmon resonance, qPCR, ELISA and immunoblotting techniques we found that Ph8 bound IL-4Rα and mimicked the anti-inflammatory effects of IL-4 by inhibiting TNF-α production by macrophages in vitro. It induced phosphorylation of STAT6 65kD but inhibited phosphorylation of STAT6 110 kD induced by IL-4 in a B-cell line that expressed the type I receptor. It also inhibited the IL-4-stimulated expression of a STAT6-inducible reporter gene in cells that expressed the type II receptor. Ph8 inhibited the proliferation of Th1/2 cells and downregulated the production of IFN-γ in stimulated Th1 cells. Moreover, Ph8 did not induce any shift in Th1/Th2 profile. This is a favorable effect and it is indicating that Ph8 could block general T cell activation and inflammatory responses without further inducing the side effects generally associated with IL-4 signaling. These data collectively show that Ph8 is only a partial agonist of IL-4 mimicking its desirable properties. In agreement, Ph8 treatment of rats with collagen-induced arthritis, a Th1- and antibody- mediated disease of joint, delayed the manifestation of chronic inflammation and reduced acute inflammation in carrageenan-induced edema. Our findings indicate that Ph8 is a promising potential drug candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Boris Klementiev
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
35
|
YOON HAYONG, LEE EUNGYEONG, LEE HYUN, CHO INJIN, CHOI YUNJUNG, SUNG MYUNGSOON, YOO HANGYUL, YOO WANHEE. Kaempferol inhibits IL-1β-induced proliferation of rheumatoid arthritis synovial fibroblasts and the production of COX-2, PGE2 and MMPs. Int J Mol Med 2013; 32:971-7. [DOI: 10.3892/ijmm.2013.1468] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 08/01/2013] [Indexed: 11/06/2022] Open
|
36
|
Flavonoids and asthma. Nutrients 2013; 5:2128-43. [PMID: 23752494 PMCID: PMC3725497 DOI: 10.3390/nu5062128] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 05/03/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
Asthma is a chronic disease, characterized by airway inflammation, airflow limitation, hyper-reactivity and airway remodeling. It is believed that asthma is caused by the interaction between genetic and environmental factors. The prevalence of allergic diseases, including asthma, has increased worldwide during the past two decades. Although the precise reasons that have caused this increase remain unknown, dietary change is thought to be one of the environmental factors. Flavonoids, which are polyphenolic plant secondary metabolites ubiquitously present in vegetables, fruits and beverages, possess antioxidant and anti-allergic traits, as well as immune-modulating activities. Flavonoids are powerful antioxidants and anti-allergic nutrients that inhibit the release of chemical mediators, synthesis of Th2 type cytokines, such as interleukin (IL)-4 and IL-13, and CD40 ligand expression by high-affinity immunoglobulin E (IgE) receptor-expressing cells, such as mast cells and basophils. They also inhibit IL-4-induced signal transduction and affect the differentiation of naïve CD4+ T cells into effector T-cells through their inhibitory effect on the activation of the aryl hydrocarbon receptor. Various studies of flavonoids in asthmatic animal models have demonstrated their beneficial effects. The results of several epidemiological studies suggest that an increase in flavonoid intake is beneficial for asthma. Moreover, clinical trials of flavonoids have shown their ameliorative effects on symptoms related to asthma. However, these human studies are currently limited; further validation is required to clarify whether an appropriate intake of flavonoids may constitute dietary treatment and for part of a preventive strategy for asthma.
Collapse
|
37
|
Chen AY, Chen YC. A review of the dietary flavonoid, kaempferol on human health and cancer chemoprevention. Food Chem 2012; 138:2099-107. [PMID: 23497863 DOI: 10.1016/j.foodchem.2012.11.139] [Citation(s) in RCA: 597] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Revised: 11/03/2012] [Accepted: 11/29/2012] [Indexed: 01/08/2023]
Abstract
Kaempferol is a polyphenol antioxidant found in fruits and vegetables. Many studies have described the beneficial effects of dietary kaempferol in reducing the risk of chronic diseases, especially cancer. Epidemiological studies have shown an inverse relationship between kaempferol intake and cancer. Kaempferol may help by augmenting the body's antioxidant defence against free radicals, which promote the development of cancer. At the molecular level, kaempferol has been reported to modulate a number of key elements in cellular signal transduction pathways linked to apoptosis, angiogenesis, inflammation, and metastasis. Significantly, kaempferol inhibits cancer cell growth and angiogenesis and induces cancer cell apoptosis, but on the other hand, kaempferol appears to preserve normal cell viability, in some cases exerting a protective effect. The aim of this review is to synthesize information concerning the extraction of kaempferol, as well as to provide insights into the molecular basis of its potential chemo-preventative activities, with an emphasis on its ability to control intracellular signaling cascades that regulate the aforementioned processes. Chemoprevention using nanotechnology to improve the bioavailability of kaempferol is also discussed.
Collapse
Affiliation(s)
- Allen Y Chen
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
| | | |
Collapse
|
38
|
Luo H, Jiang B, Li B, Li Z, Jiang BH, Chen YC. Kaempferol nanoparticles achieve strong and selective inhibition of ovarian cancer cell viability. Int J Nanomedicine 2012; 7:3951-9. [PMID: 22866004 PMCID: PMC3410694 DOI: 10.2147/ijn.s33670] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ovarian cancer is one of the leading causes of cancer death for women throughout the Western world. Kaempferol, a natural flavonoid, has shown promise in the chemoprevention of ovarian cancer. A common concern about using dietary supplements for chemoprevention is their bioavailability. Nanoparticles have shown promise in increasing the bioavailability of some chemicals. Here we developed five different types of nanoparticles incorporating kaempferol and tested their efficacy in the inhibition of viability of cancerous and normal ovarian cells. We found that positively charged nanoparticle formulations did not lead to a significant reduction in cancer cell viability, whereas nonionic polymeric nanoparticles resulted in enhanced reduction of cancer cell viability. Among the nonionic polymeric nanoparticles, poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) (PEO-PPO-PEO) nanoparticles incorporating kaempferol led to significant reduction in cell viability of both cancerous and normal cells. Poly(DL-lactic acid-co-glycolic acid) (PLGA) nanoparticles incorporating kaempferol resulted in enhanced reduction of cancer cell viability together with no significant reduction in cell viability of normal cells compared with kaempferol alone. Therefore, both PEO-PPO-PEO and PLGA nanoparticle formulations were effective in reducing cancer cell viability, while PLGA nanoparticles incorporating kaempferol had selective toxicity against cancer cells and normal cells. A PLGA nanoparticle formulation could be advantageous in the prevention and treatment of ovarian cancers. On the other hand, PEO-PPO-PEO nanoparticles incorporating kaempferol were more effective inhibitors of cancer cells, but they also significantly reduced the viability of normal cells. PEO-PPO-PEO nanoparticles incorporating kaempferol may be suitable as a cancer-targeting strategy, which could limit the effects of the nanoparticles on normal cells while retaining their potency against cancer cells. We have identified two nanoparticle formulations incorporating kaempferol that may lead to breakthroughs in cancer treatment. Both PEO-PPO-PEO and PLGA nanoparticle formulations had superior effects compared with kaempferol alone in reducing cancer cell viability.
Collapse
Affiliation(s)
- Haitao Luo
- Department of Biology, Natural Science Division, Alderson-Broaddus College, Philippi, WV 26416, USA
| | | | | | | | | | | |
Collapse
|
39
|
Zhu LY, Pan PP, Fang W, Shao JZ, Xiang LX. Essential role of IL-4 and IL-4Rα interaction in adaptive immunity of zebrafish: insight into the origin of Th2-like regulatory mechanism in ancient vertebrates. THE JOURNAL OF IMMUNOLOGY 2012; 188:5571-84. [PMID: 22547699 DOI: 10.4049/jimmunol.1102259] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The roles of IL-4 and IL-4Rα in Th2-mediated immunity have been well characterized in humans and other mammals. In contrast, few reports have been documented in ancient vertebrates. Several putative IL-4- and IL-4Rα-like molecules were identified recently from a few fish species, providing preliminary insight into the occurrence of Th2-type immunity in teleosts. However, functional determination still is required to address this hypothesis. To this end, these two molecules were characterized functionally in zebrafish (Danio rerio). Besides the identification of a full-length IL-4Rα molecule and an isoform lacking most of the cytoplasmic region as predicted previously, two novel alternatively spliced soluble variants with the extracellular domain only also were identified. Zebrafish IL-4Rα (DrIL-4Rα) shared overall conserved structural features of the IL-4Rα family. Immunofluorescence staining showed that DrIL-4Rα distributed on B cells. In vitro binding assays demonstrated that zebrafish IL-4 (DrIL-4) can bind specifically to DrIL-4Rα. In vivo administration of DrIL-4 significantly upregulated B cell proliferation and Ab production. These DrIL-4-elicited immune responses were downregulated by the administration of zebrafish soluble IL-4Rα or by DrIL-4Rα blockade using anti-DrIL-4Rα Abs. In addition, Th2-related cytokines or transcription factors were upregulated by DrIL-4. The DrIL-4-DrIL-4Rα interaction promoted CD40 expression on B cells and enhanced the CD154-CD40 costimulatory response, both of which are crucial for the initiation of Th2-type immunity. To our knowledge, this is the first report showing that a possible Th2-mediated regulatory mechanism may have appeared before the divergence of teleosts and mammals. These results add greater insight into the evolutionary history of adaptive immunity.
Collapse
Affiliation(s)
- Lv-yun Zhu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | | | | | | | | |
Collapse
|
40
|
Giron-Michel J, Azzi S, Khawam K, Mortier E, Caignard A, Devocelle A, Ferrini S, Croce M, François H, Lecru L, Charpentier B, Chouaib S, Azzarone B, Eid P. Interleukin-15 plays a central role in human kidney physiology and cancer through the γc signaling pathway. PLoS One 2012; 7:e31624. [PMID: 22363690 PMCID: PMC3283658 DOI: 10.1371/journal.pone.0031624] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 01/16/2012] [Indexed: 12/16/2022] Open
Abstract
The ability of Interleukin-15 (IL-15) to activate many immune antitumor mechanisms renders the cytokine a good candidate for the therapy of solid tumors, particularly renal cell carcinoma. Although IL-15 is being currently used in clinical trials, the function of the cytokine on kidney's components has not been extensively studied; we thus investigated the role of IL-15 on normal and tumor renal epithelial cells. Herein, we analyzed the expression and the biological functions of IL-15 in normal renal proximal tubuli (RPTEC) and in their neoplastic counterparts, the renal clear cell carcinomas (RCC). This study shows that RPTEC express a functional heterotrimeric IL-15Rαβγc complex whose stimulation with physiologic concentrations of rhIL-15 is sufficient to inhibit epithelial mesenchymal transition (EMT) commitment preserving E-cadherin expression. Indeed, IL-15 is not only a survival factor for epithelial cells, but it can also preserve the renal epithelial phenotype through the γc-signaling pathway, demonstrating that the cytokine possess a wide range of action in epithelial homeostasis. In contrast, in RCC in vitro and in vivo studies reveal a defect in the expression of γc-receptor and JAK3 associated kinase, which strongly impacts IL-15 signaling. Indeed, in the absence of the γc/JAK3 couple we demonstrate the assembly of an unprecedented functional high affinity IL-15Rαβ heterodimer, that in response to physiologic concentrations of IL-15, triggers an unbalanced signal causing the down-regulation of the tumor suppressor gene E-cadherin, favoring RCC EMT process. Remarkably, the rescue of IL-15/γc-dependent signaling (STAT5), by co-transfecting γc and JAK3 in RCC, inhibits EMT reversion. In conclusion, these data highlight the central role of IL-15 and γc-receptor signaling in renal homeostasis through the control of E-cadherin expression and preservation of epithelial phenotype both in RPTEC (up-regulation) and RCC (down-regulation).
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Sandy Azzi
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Krystel Khawam
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Erwan Mortier
- INSERM UMRS 892, Institut de Recherche Thérapeutique de l'Université de Nantes (IRT UN), Nantes, France
| | - Anne Caignard
- Institut Cochin, Université Paris Descartes, INSERM U1016, Paris, France
| | - Aurore Devocelle
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Silvano Ferrini
- Laboratory of Immunotherapy, Instituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Michela Croce
- Laboratory of Immunotherapy, Instituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Hélène François
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Lola Lecru
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Bernard Charpentier
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Salem Chouaib
- INSERM UMR 753, Université de Paris-Sud, Institut Gustave Roussy (IGR), Villejuif, France
| | - Bruno Azzarone
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
- * E-mail: (BA); (PE)
| | - Pierre Eid
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
- * E-mail: (BA); (PE)
| |
Collapse
|
41
|
Hao Y, Kuang Z, Walling BE, Bhatia S, Sivaguru M, Chen Y, Gaskins HR, Lau GW. Pseudomonas aeruginosa pyocyanin causes airway goblet cell hyperplasia and metaplasia and mucus hypersecretion by inactivating the transcriptional factor FoxA2. Cell Microbiol 2011; 14:401-15. [PMID: 22103442 DOI: 10.1111/j.1462-5822.2011.01727.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The redox-active exotoxin pyocyanin (PCN) can be recovered in 100 µM concentrations in the sputa of bronchiectasis patients chronically infected with Pseudomonas aeruginosa (PA). However, the importance of PCN within bronchiectatic airways colonized by PA remains unrecognized. Recently, we have shown that PCN is required for chronic PA lung infection in mice, and that chronic instillation of PCN induces goblet cell hyperplasia (GCH), pulmonary fibrosis, emphysema and influx of immune cells in mouse airways. Many of these pathological features are strikingly similar to the mouse airways devoid of functional FoxA2, a transcriptional repressor of GCH and mucus biosynthesis. In this study, we postulate that PCN causes and exacerbates GCH and mucus hypersecretion in bronchiectatic airways chronically infected by PA by inactivating FoxA2. We demonstrate that PCN represses the expression of FoxA2 in mouse airways and in bronchial epithelial cells cultured at an air-liquid interface or conventionally, resulting in GCH, increased MUC5B mucin gene expression and mucus hypersecretion. Immunohistochemical and inhibitor studies indicate that PCN upregulates the expression of Stat6 and EGFR, both of which in turn repress the expression of FoxA2. These studies demonstrate that PCN induces GCH and mucus hypersecretion by inactivating FoxA2.
Collapse
Affiliation(s)
- Yonghua Hao
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Fang C, Corrigan CJ, Ying S. Identifying and testing potential new anti-asthma agents. Expert Opin Drug Discov 2011; 6:1027-44. [PMID: 22646862 DOI: 10.1517/17460441.2011.608659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Inhaled corticosteroids alone or with long-acting beta-2 agonists (LABA) are the basic treatment for stable asthma. While the majority of patients are controllable, some patients retain chronic severe disease and develop permanent alterations in airway function. For patients such as these it is important to better understand the mechanisms of asthma so that alternative approaches can be developed. AREA COVERED Based on data from in vitro cell culture, animal models and clinical trials, this review discusses potential agents targeting either key effector cells, mediators and their receptors in asthma pathogenesis or their signaling cascade molecules. EXPERT OPINION As targeting single Th2 cytokines and their receptors has been shown to have limited clinical benefit, it is important to identify and test potential new therapeutic agents. Recent studies suggest that blockade of IgE synthesis, its interaction with its receptors and downstream signaling, identification of molecular targets in innate immune and airways structural cells, and fresh anti-neutrophil strategies should be prominent among these. Further studies are required to clarify the relationship between airways remodeling and asthma severity so that appropriate patients may be targeted.
Collapse
Affiliation(s)
- Cailong Fang
- Guy's Hospital, King's College London, MRC and Asthma UK Centre in Allergic Mechanisms of Asthma , Department of Asthma , Allergy and Respiratory Science, 5th Floor, Tower Wing, London SE1 9RT , UK +44 207 188 3392 ;
| | | | | |
Collapse
|
43
|
González R, Ballester I, López-Posadas R, Suárez MD, Zarzuelo A, Martínez-Augustin O, Sánchez de Medina F. Effects of flavonoids and other polyphenols on inflammation. Crit Rev Food Sci Nutr 2011; 51:331-62. [PMID: 21432698 DOI: 10.1080/10408390903584094] [Citation(s) in RCA: 366] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Flavonoids are a family of polyphenolic compounds which are widespread in nature (vegetables) and are consumed as part of the human diet in significant amounts. There are other types of polyphenols, including, for example, tannins and resveratrol. Flavonoids and related polyphenolic compounds have significant antiinflammatory activity, among others. This short review summarizes the current knowledge on the effects of flavonoids and related polyphenolic compounds on inflammation, with a focus on structural requirements, the mechanisms involved, and pharmacokinetic considerations. Different molecular (cyclooxygenase, lipoxygenase) and cellular targets (macrophages, lymphocytes, epithelial cells, endothelium) have been identified. In addition, many flavonoids display significant antioxidant/radical scavenging properties. There is substantial structural variation in these compounds, which is bound to have an impact on their biological profile, and specifically on their effects on inflammatory conditions. However, in general terms there is substantial consistency in the effects of these compounds despite considerable structural variations. The mechanisms have been studied mainly in myeloid cells, where the predominant effect is an inhibition of NF-κB signaling and the downregulation of the expression of proinflammatory markers. At present there is a gap in knowledge of in vitro and in vivo effects, although the pharmacokinetics of flavonoids has advanced considerably in the last decade. Many flavonoids have been studied for their intestinal antiinflammatory activity which is only logical, since the gastrointestinal tract is naturally exposed to them. However, their potential therapeutic application in inflammation is not restricted to this organ and extends to other sites and conditions, including arthritis, asthma, encephalomyelitis, and atherosclerosis, among others.
Collapse
Affiliation(s)
- R González
- Department of Pharmacology, CIBERehd, School of Pharmacy, University of Granada, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
44
|
Henkels KM, Frondorf K, Gonzalez-Mejia ME, Doseff AL, Gomez-Cambronero J. IL-8-induced neutrophil chemotaxis is mediated by Janus kinase 3 (JAK3). FEBS Lett 2010; 585:159-66. [PMID: 21095188 DOI: 10.1016/j.febslet.2010.11.031] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 11/15/2010] [Accepted: 11/16/2010] [Indexed: 12/11/2022]
Abstract
Janus kinase 3 (JAK3) is a non-receptor tyrosine kinase vital to the regulation of T-cells. We report that JAK3 is a mediator of interleukin-8 (IL-8) stimulation of a different class of hematopoietic relevant cells: human neutrophils. IL-8 induced a time- and concentration-dependent activation of JAK3 activity in neutrophils and differentiated HL-60 leukemic cells. JAK3 was more robustly activated by IL-8 than other kinases: p70S6K, mTOR, MAPK or PKC. JAK3 silencing severely inhibited IL-8-mediated chemotaxis. Thus, IL-8 stimulates chemotaxis through a mechanism mediated by JAK3. Further, JAK3 activity and chemotaxis were inhibited by the flavonoid apigenin (4',5,7-trihydroxyflavone) at ∼5nM IC(50). These new findings lay the basis for understanding the molecular mechanism of cell migration as it relates to neutrophil-mediated chronic inflammatory processes.
Collapse
Affiliation(s)
- Karen M Henkels
- Department of Biochemistry and Molecular Biology, Wright State University School Medicine, Dayton, OH 45435, USA
| | | | | | | | | |
Collapse
|
45
|
Huang CH, Jan RL, Kuo CH, Chu YT, Wang WL, Lee MS, Chen HN, Hung CH. Natural Flavone Kaempferol Suppresses Chemokines Expression in Human Monocyte THP-1 Cells through MAPK Pathways. J Food Sci 2010; 75:H254-9. [DOI: 10.1111/j.1750-3841.2010.01812.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Chi X, Tai HH. Interleukin-4 up-regulates 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in human lung cancer cells. Exp Cell Res 2010; 316:2251-9. [PMID: 20632471 DOI: 10.1016/j.yexcr.2010.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
IL-4, an anti-inflammatory cytokine, was found to stimulate 15-PGDH activity in A549 and other lung cancer cells. Increase in 15-PGDH activity was due to increased transcription and decreased protein turnover of 15-PGDH. MMP-9 was shown to result in decreased levels of 15-PGDH in A549 cells. IL-4 induced down-regulation of MMP-9 mRNA and up-regulation of TIMP-1, an inhibitor of MMP-9. These data suggest that IL-4-induced down-regulation of MMP-9 activity may contribute to up-regulation of 15-PGDH in A549 cells. The role of JAK-STAT6 in IL-4-induced 15-PGDH expression was examined by using inhibitors. Inhibitors of JAKs, kaempferol and JAK inhibitor I, attenuated IL-4-stimulated STAT6 phosphorylation and 15-PGDH activity in a comparable concentration-dependent manner. Furthermore, JAK inhibitor I blocked IL-4-induced down-regulation of MMP-9 mRNA and up-regulation of TIMP-1 but not IL-4-stimulated up-regulation of 15-PGDH mRNA. These results indicate that JAK-STAT6 participated in IL-4-induced up-regulation of 15-PGDH through inhibition of MMP-9-mediated degradation. The roles of other signaling kinases in IL-4-induced 15-PGDH expression were also examined by using various inhibitors. Inhibitors of various MAPKs, PI-3K and PKC attenuated significantly IL-4-stimulated 15-PGDH activity indicating that MAPKs, PI-3K/Akt and PKC pathways participated in IL-4-induced up-regulation of 15-PGDH. Our results indicate that IL-4 up-regulates 15-PGDH by increased gene transcription and decreased protein turnover and the up-regulation can be mediated by JAK-STAT6 as well as MAPKs, PI-3K/Akt and PKC pathways.
Collapse
Affiliation(s)
- Xiuling Chi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0082, USA
| | | |
Collapse
|
47
|
Cocoa polyphenols suppress TNF-α-induced vascular endothelial growth factor expression by inhibiting phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase kinase-1 (MEK1) activities in mouse epidermal cells. Br J Nutr 2010; 104:957-64. [PMID: 20550744 DOI: 10.1017/s0007114510001704] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cocoa polyphenols have antioxidant and anti-inflammatory effects. TNF-α is a pro-inflammatory cytokine that has a vital role in the pathogenesis of inflammatory diseases such as cancer and psoriasis. Vascular endothelial growth factor (VEGF) expression is associated with tumorigenesis, CVD, rheumatoid arthritis and psoriasis. We tested whether cocoa polyphenol extract (CPE) inhibited TNF-α-induced VEGF expression in promotion-sensitive JB6 mouse epidermal cells. CPE significantly inhibited TNF-α-induced up-regulation of VEGF via reducing TNF-α-induced activation of the nuclear transcription factors activator protein-1 (AP-1) and NF-κB, which are key regulators of VEGF expression. CPE also inhibited TNF-α-induced phosphorylation of protein kinase B (Akt) and extracellular signal-regulated kinase. CPE blocked activation of their downstream kinases, p70 kDa ribosomal protein S6 kinase and p90 kDa ribosomal protein S6 kinase. CPE suppressed phosphoinositide 3-kinase (PI3K) activity via binding PI3K directly. CPE did not affect TNF-α-induced phosphorylation of mitogen-activated protein kinase kinase-1 (MEK1) but suppressed TNF-α-induced MEK1 activity. Collectively, these results indicate that CPE reduced TNF-α-induced up-regulation of VEGF by directly inhibiting PI3K and MEK1 activities, which may contribute to its chemopreventive potential.
Collapse
|
48
|
Malaviya R, Laskin DL, Malaviya R. Janus kinase-3 dependent inflammatory responses in allergic asthma. Int Immunopharmacol 2010; 10:829-36. [PMID: 20430118 DOI: 10.1016/j.intimp.2010.04.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 03/25/2010] [Accepted: 04/20/2010] [Indexed: 12/14/2022]
Abstract
Allergic asthma is a chronic inflammatory condition of the lung characterized by reversible airway obstruction, high serum immunoglobulin (Ig) E levels, and chronic airway inflammation. A number of cells including mast cells, T cells, macrophages and dendritic cells play a role in the pathogenesis of the disease. Janus kinase (JAK)-3, a non-receptor protein tyrosine kinase, traditionally known to mediate cytokine signaling, also regulates functional responses of these cells. In this review the role of JAK-3 in regulating various pathogenic processes in allergic asthma is discussed. We propose that targeting JAK-3 is a rationale approach to control the inflammatory responses of multiple cell types responsible for the pathogenesis of allergic asthma.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| | | | | |
Collapse
|
49
|
Kim BH, Jee JG, Yin CH, Sandoval C, Jayabose S, Kitamura D, Bach EA, Baeg GH. NSC114792, a novel small molecule identified through structure-based computational database screening, selectively inhibits JAK3. Mol Cancer 2010; 9:36. [PMID: 20149240 PMCID: PMC2830973 DOI: 10.1186/1476-4598-9-36] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 02/11/2010] [Indexed: 01/10/2023] Open
Abstract
Background Human or animals lacking either JAK3 or the common gamma chain (γc) expression display severe combined immunodeficiency disease, indicating the crucial role of JAK3 in T-cell development and the homeostasis of the immune system. JAK3 has also been suggested to contribute to the pathogenesis of tumorigenesis. Recent studies identified activating JAK3 mutations in patients with various hematopoietic malignancies, including acute megakaryoblastic leukemia. Importantly, functional analyses of some of those JAK3 mutations have been shown to cause lethal hematopoietic malignancies in animal models. These observations make JAK3 an ideal therapeutic target for the treatment of various human diseases. To identify novel small molecule inhibitors of JAK3, we performed structure-based virtual screen using the 3D structure of JAK3 kinase domain and the NCI diversity set of compounds. Results We identified NSC114792 as a lead compound. This compound directly blocked the catalytic activity of JAK3 but not that of other JAK family members in vitro. In addition, treatment of 32D/IL-2Rβ cells with the compound led to a block in IL-2-dependent activation of JAK3/STAT5 but not IL-3-dependent activation of JAK2/STAT5. Consistent with the specificity of NSC114792 for JAK3, it selectively inhibited persistently-activated JAK3, but failed to affect the activity of other JAK family members and other oncogenic kinases in various cancer cell lines. Finally, we showed that NSC114792 decreases cell viability by inducing apoptosis through down-regulating anti-apoptotic gene expression only in cancer cells harboring persistently-active JAK3. Conclusions NSC114792 is a lead compound that selectively inhibits JAK3 activity. Therefore, our study suggests that this small molecule inhibitor of JAK3 can be used as a starting point to develop a new class of drugs targeting JAK3 activity, and may have therapeutic potential in various diseases that are caused by aberrant JAK3 activity.
Collapse
Affiliation(s)
- Byung-Hak Kim
- Department of Pediatrics, Division of Hematology/Oncology, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Flavones suppress type I IL-4 receptor signaling by down-regulating the expression of common gamma chain. FEBS Lett 2009; 584:775-9. [PMID: 20040389 DOI: 10.1016/j.febslet.2009.12.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 12/21/2009] [Accepted: 12/21/2009] [Indexed: 11/22/2022]
Abstract
Immunoglobulin E (IgE) production is induced by interleukin (IL)-4 signaling mediated by type I IL-4 receptor (IL-4R) in B cells. We found that flavones inhibited IL-4-induced epsilon germline transcription which is essential for IgE class switching, and the phosphorylation of signal transducer and activator of transcription 6, janus kinase 3, and IL-4Ralpha, whereas IL-4 signaling mediated through type II IL-4R was unaffected by flavones. Furthermore, flavones reduced the expression of common gamma chain, a characteristic constituent subunit of type I IL-4R, suggesting that flavones suppress type I IL-4R signaling.
Collapse
|