1
|
Zhao Q, Chen L, Ma Y, Wang S. Scutellarin Attenuates Pro-Inflammatory Foam Cell Formation and Facilitates M2 Polarization in Microglia during Copper Homeostasis Imbalance via the MAPK Signaling Pathway. FRONT BIOSCI-LANDMRK 2025; 30:36255. [PMID: 40152387 DOI: 10.31083/fbl36255] [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: 12/09/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Clinical and experimental evidence indicates that copper has the ability to promote the progressive development of demyelinating diseases such as multiple sclerosis. Microglia-mediated neuroinflammation is believed to play a crucial role in this process. Scutellarin, a flavonoid compound, has anti-inflammatory, antioxidative, and neuroprotective effects. AIM We investigated the effect of scutellarin on copper-induced inflammatory foam cell formation in microglia. METHODS We exposed BV2 murine microglial cells to copper, then collected the conditioned medium and co-cultured it with MO3.13 human glial cells to mimic myelin damage in vitro. The Cell Counting kit-8 assay, quantitative (polymerase chain reaction) PCR, enzyme-linked immunosorbent assay, Luxol fast blue staining, and western blotting were used to detect the cell phenotype. To investigate whether exposure of BV2 cells to copper can cause neurotoxicity and indirect damage to myelin cells, we determined whether BV2 cells promote inflammation through foam cell formation by oil red O staining and detection of malondialdehyde (MDA) content. Finally, we treated cells with scutellarin to investigate its therapeutic effects. RESULTS Exposure to copper activated the pro-inflammatory phenotype of microglia, as assessed by measuring the transcription of M1/M2-related biomarkers. In addition, increased copper intake by microglia promoted intracellular lipid accumulation and oxidation, facilitating foam cell formation. Rescue experiments showed that copper chelator ammonium tetrathiomolybdate (ATTM) and the lipid oxidation inhibitor ferrostatin-1 (Fer-1) significantly inhibited copper-induced inflammation, reduced intracellular lipid accumulation and MDA levels, and decreased foam cell formation. Moreover, copper-induced phosphorylation of p38 mitogen-activated protein kinase (MAPK) in microglia led to a shift towards the M1 phenotype and foam cell transformation, which were effectively inhibited by ATTM, Fer-1, and the p38 MAPK inhibitor SB203580. Lastly, after treatment with scutellarin, copper-induced foam microglia exhibited inhibited p38 MAPK phosphorylation, increased production of neurotrophic factors, decreased expression of inflammatory mediators, reduced lipid accumulation, and induced polarization towards the M2 phenotype. CONCLUSIONS Here, we demonstrated that copper can induce microglia to damage myelinating cells, with the key mechanism involving the phosphorylation of p38 MAPK. Scutellarin partially reversed the positive effects of copper on promoting microglial M1 polarization, lipid deposition, and lipid oxidation by mediating the p38 MAPK signaling pathway. Taken together, these results suggest that scutellarin may be a promising drug for the treatment of demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Qiting Zhao
- Institute of Biomedical Engineering, Kunming Medical University, 650500 Kunming, Yunnan, China
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, 650051 Kunming, Yunnan, China
| | - Lingyi Chen
- Institute of Biomedical Engineering, Kunming Medical University, 650500 Kunming, Yunnan, China
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, 650051 Kunming, Yunnan, China
| | - Yantuanjin Ma
- Institute of Biomedical Engineering, Kunming Medical University, 650500 Kunming, Yunnan, China
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, 650051 Kunming, Yunnan, China
| | - Shufen Wang
- Institute of Biomedical Engineering, Kunming Medical University, 650500 Kunming, Yunnan, China
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, 650051 Kunming, Yunnan, China
| |
Collapse
|
2
|
Hernandez K, Jones N, Ortega SB. The efficacy of an allosteric modulator of the alpha 7 nicotinic acetylcholine receptor in a murine model of stroke. Front Neurosci 2025; 19:1525975. [PMID: 40012683 PMCID: PMC11860958 DOI: 10.3389/fnins.2025.1525975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025] Open
Abstract
Introduction Ischemic strokes contribute significantly to cardiovascular-related deaths in the U.S., with current interventions limited to thrombolytic agents. However, these agents present challenges such as a limited therapeutic window, incomplete reperfusion rates, risk of transformation, reperfusion-induced inflammation, and a lack of promoting neuroprotection. We investigated an additional strategy in which prior studies indicated a neuroprotective role. Using a murine transient middle cerebral artery occlusion (tMCAO) model, we sought to evaluate the neurotherapeutic efficacy of a positive allosteric modulator of the alpha7 nicotinic acetylcholine receptor (α7-nAChR), PNU-120596 (PNU), specifically examining whether PNU would modulate stroke-induced neurological dysfunction and neuropathology, with modulation of neuroinflammation as a possible mechanism. Methods Young male C57BL/6J mice received a subcutaneous injection of 20mg/kg of vehicle (DMSO) or PNU-120596 immediately after reperfusion, and infarct area and Bederson score were analyzed 24 hours post-stroke. In the 72-hour post-stroke study, the animals were injected with 20mg/kg of PNU or vehicle subcutaneously immediately after reperfusion, followed by two additional doses of 10mg/kg of PNU or vehicle at 24 and 48 hours post-tMCAO. Seventy-two hours later, behavior function and infarct area were assessed. Results In contrast to previous rat studies that demonstrated improvements in clinical outcomes, a single administration of PNU following stroke induction led to a reduction in acute neuropathology but did not produce a significant improvement in motor outcomes. Prolonged treatment showed no significant changes in acute neuropathology or sensorimotor function. Additionally, an assessment of neuroinflammation revealed no changes in CD4 T-cell cellularity or phenotype. Discussion These findings, alongside prior studies, suggest that the therapeutic efficacy of PNU may be contingent upon the timing of administration, dosage, and pharmacokinetics.
Collapse
Affiliation(s)
| | | | - Sterling B. Ortega
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
3
|
Nie S, Zhang S, Wu R, Zhao Y, Wang Y, Wang X, Zhu M, Huang P. Scutellarin: pharmacological effects and therapeutic mechanisms in chronic diseases. Front Pharmacol 2024; 15:1470879. [PMID: 39575387 PMCID: PMC11578714 DOI: 10.3389/fphar.2024.1470879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
Scutellarin (SCU), a flavonoid glucuronide derived from Scutellaria barbata and Erigeron breviscapus, exhibits broad pharmacological effects with promising therapeutic potential in treating various chronic diseases. It has demonstrated efficacy in modulating multiple biological pathways, including antioxidant, anti-inflammatory, anti-apoptotic, and vasodilatory mechanisms. These protective roles make SCU a valuable compound in treating chronic diseases such as cerebrovascular diseases, cardiovascular diseases, neurodegenerative disorders, and metabolic diseases. Despite its multi-targeted effects, SCU faces challenges such as low bioavailability and limited clinical data, which hinder its widespread therapeutic application. Current research supports its potential to prevent oxidative stress, reduce inflammatory responses, and enhance cell survival in cells and rats. However, more comprehensive studies are required to clarify its molecular mechanisms and to develop strategies that enhance its bioavailability for clinical use. SCU could emerge as a potent therapeutic agent for the treatment of chronic diseases with complex pathophysiological mechanisms. This review examines the current literature on Scutellarin to provide a comprehensive understanding of its pharmacological activity, mechanisms of action, and therapeutic potential in treating chronic diseases.
Collapse
Affiliation(s)
- Shanshan Nie
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shan Zhang
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Ruipeng Wu
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yuhang Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongxia Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xinlu Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Mingjun Zhu
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
4
|
Cheng Y, Chen X, Zhu G, Li N, Sun Y, Luo S, Liu Y, Lu X. Erigeron breviscapus: A Promising Medication for Protecting the Optic Nerve in Glaucoma. PLANTA MEDICA 2024; 90:992-1004. [PMID: 39303747 DOI: 10.1055/a-2409-2999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Glaucoma is a common eye condition characterized by the loss of retinal ganglion cells and their axons, optic nerve damage, and visual field defects, which seriously affect a patient's quality of life. The pathogenesis of glaucoma is still unclear at present. It presents as damage to retinal ganglion cells, and the main treatment is primarily to reduce intraocular pressure by surgery or taking medication. However, even with well-controlled intraocular pressure, retinal ganglion cells still undergo degeneration, progressive apoptosis, and axonal loss. Therefore, protecting the optic nerve and inhibiting the apoptosis of retinal ganglion cells are the current hot topic for prevention and treatment of glaucoma. Recently, Erigeron breviscapus, originating from Yunnan province in China, has been shown to be a promising herb with neuroprotective effects to treat glaucoma. Therefore, the traditional usage, botanical characteristics, and phytochemical composition of E. breviscapus were explored through a literature review. Furthermore, we have summarized the pharmacological mechanisms of E. breviscapus and its active components in inhibiting the apoptosis of retinal ganglion cells. These research findings can not only provide guidance and recommendations for the protection of retinal ganglion cells but also further explore the potential of E. breviscapus in the treatment of glaucoma.
Collapse
Affiliation(s)
- Yuxin Cheng
- Eye School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Ophthalmology, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM, Sichuan Province, Chengdu, China
| | - Xuanyi Chen
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guangyu Zhu
- Eye School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Ophthalmology, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM, Sichuan Province, Chengdu, China
| | - Na Li
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Sun
- Eye School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Ophthalmology, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM, Sichuan Province, Chengdu, China
| | - Shichun Luo
- Eye School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Ophthalmology, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM, Sichuan Province, Chengdu, China
| | - Yujie Liu
- Eye School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Ophthalmology, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM, Sichuan Province, Chengdu, China
| | - Xuejing Lu
- Eye School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Ophthalmology, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM, Sichuan Province, Chengdu, China
| |
Collapse
|
5
|
Kim JH, Dareowolabi BO, Thiruvengadam R, Moon EY. Application of Nanotechnology and Phytochemicals in Anticancer Therapy. Pharmaceutics 2024; 16:1169. [PMID: 39339205 PMCID: PMC11435124 DOI: 10.3390/pharmaceutics16091169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/22/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer is well recognized as a leading cause of mortality. Although surgery tends to be the primary treatment option for many solid cancers, cancer surgery is still a risk factor for metastatic diseases and recurrence. For this reason, a variety of medications has been adopted for the postsurgical care of patients with cancer. However, conventional medicines have shown major challenges such as drug resistance, a high level of drug toxicity, and different drug responses, due to tumor heterogeneity. Nanotechnology-based therapeutic formulations could effectively overcome the challenges faced by conventional treatment methods. In particular, the combined use of nanomedicine with natural phytochemicals can enhance tumor targeting and increase the efficacy of anticancer agents with better solubility and bioavailability and reduced side effects. However, there is limited evidence in relation to the application of phytochemicals in cancer treatment, particularly focusing on nanotechnology. Therefore, in this review, first, we introduce the drug carriers used in advanced nanotechnology and their strengths and limitations. Second, we provide an update on well-studied nanotechnology-based anticancer therapies related to the carcinogenesis process, including signaling pathways related to transforming growth factor-β (TGF-β), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3 kinase (PI3K), Wnt, poly(ADP-ribose) polymerase (PARP), Notch, and Hedgehog (HH). Third, we introduce approved nanomedicines currently available for anticancer therapy. Fourth, we discuss the potential roles of natural phytochemicals as anticancer drugs. Fifth, we also discuss the synergistic effect of nanocarriers and phytochemicals in anticancer therapy.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea; (B.O.D.); (E.-Y.M.)
| | - Boluwatife Olamide Dareowolabi
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea; (B.O.D.); (E.-Y.M.)
| | - Rekha Thiruvengadam
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha Medical College, Saveetha University, Chennai 600077, India;
| | - Eun-Yi Moon
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea; (B.O.D.); (E.-Y.M.)
| |
Collapse
|
6
|
Zhou Y, Gu C, Zhu Y, Zhu Y, Chen Y, Shi L, Yang Y, Lu X, Pang H. Pharmacological effects and the related mechanism of scutellarin on inflammation-related diseases: a review. Front Pharmacol 2024; 15:1463140. [PMID: 39188946 PMCID: PMC11345237 DOI: 10.3389/fphar.2024.1463140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/01/2024] [Indexed: 08/28/2024] Open
Abstract
Inflammation is a biological response of multicellular organisms caused by injuries, pathogens or irritants. An excessive inflammatory response can lead to tissue damage and various chronic diseases. Chronic inflammation is a common feature of many diseases, making the search for drugs to treat inflammation-related diseases urgent. Scutellarin, a natural flavonoid metabolite, is widely used in the treatment of various inflammation-related diseases for its anti-inflammatory, anti-oxidant and anti-cancer activities. Scutellarin can inhibit key inflammatory pathways (PI3K/Akt, MAPK, and NF-κB, etc.) and activate the anti-oxidant related pathways (Nrf2, ARE, ect.), thereby protecting tissues from inflammation and oxidative stress. Modern extraction technologies, such as microwave-assisted, ultrasound assisted, and supercritical fluid extraction, have been utilized to extract scutellarin from Scutellaria and Erigeron genera. These technologies improve efficiency and retain biological activity, making scutellarin suitable for large-scale production. Scutellarin has significant therapeutic effects in treating osteoarthritis, pulmonary fibrosis, kidney injury, and cardiovascular diseases. However, due to its low bioavailability and short half-life, its clinical application is limited. Researchers are exploring innovative formulations (β-cyclodextrin polymers, triglyceride mimetic active ingredients, and liposome precursors, etc.) to improve stability and absorption rates. Despite these challenges, the potential of scutellarin in anti-inflammatory, anti-oxidant, and anti-cancer applications remains enormous. By optimizing formulations, exploring combination therapies, and conducting in-depth mechanistic research, scutellarin can play an important role in treating various inflammatory diseases, providing patients with more and effective treatment options.
Collapse
Affiliation(s)
- Yang Zhou
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Chenlin Gu
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yan Zhu
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yuting Zhu
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yutong Chen
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Li Shi
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yang Yang
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Xin Lu
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Hanqing Pang
- School of Medicine, Institute of Translational Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| |
Collapse
|
7
|
Duan Z, Yang L, Xu D, Qi Z, Jia W, Wu C. Scutellarin Attenuates Microglia Activation in LPS-Induced BV-2 Microglia via miRNA-7036a/MAPT/PRKCG/ERK Axis. Adv Biol (Weinh) 2024; 8:e2400123. [PMID: 38684459 DOI: 10.1002/adbi.202400123] [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: 03/03/2024] [Revised: 04/08/2024] [Indexed: 05/02/2024]
Abstract
Scutellarin is an herbal agent which can exert anti-neuroinflammatory effects in activated microglia. However, it remains uncertain if it can inhibit microglia-mediated neuroinflammation by regulating miRNAs. This study sought to elucidate the upstream regulatory mechanisms by endogenous microRNAs and its target gene in activated microglia in lipopolysaccharide (LPS)-induced BV-2 microglia. Results show that scutellarin suppressed the expression of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and inducible nitric oxide synthase (iNOS) significantly in LPS-stimulated BV-2 microglia. As with the results of miRNAs function classification in vitro, the expression levels of mir-7036a-5p are upregulated in LPS-activated BV-2 microglia, but are downregulated by scutellarin. Rescue experiments indicated that mir-7036a-5p is a pro-inflammatory factor in activated BV-2 microglia. mir-7036a-5p agomir promoted the expression of phosphorylated tau proteins (p-tau), protein kinase C gamma type (PRKCG), extracellular regulated protein kinases (ERK1/2), but the is reversed by mir-7036a-5p antagomir in vitro. It is shown here that mir-7036a-5p is involved in microglia-mediated inflammation in LPS-induced BV-2 microglia. More important is the novel finding that scutellarin mitigated microglia inflammation by down-regulating the mir-7036a-5p/MAPT/PRKCG/ERK signaling pathway.
Collapse
Affiliation(s)
- Zhaoda Duan
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P. R. China
| | - Li Yang
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P. R. China
| | - Dongyao Xu
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P. R. China
| | - Zhi Qi
- Department of Neurology, No.2 Affiliated Hospital, Kunming Medical University, 374 Dianmian Road, Kunming, 650101, P. R. China
| | - Wenji Jia
- Department of Neurology, No.2 Affiliated Hospital, Kunming Medical University, 374 Dianmian Road, Kunming, 650101, P. R. China
| | - Chunyun Wu
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, 650500, P. R. China
| |
Collapse
|
8
|
Lee CT, Lin KD, Hsieh CF, Wang JY. SGLT2 Inhibitor Canagliflozin Alleviates High Glucose-Induced Inflammatory Toxicity in BV-2 Microglia. Biomedicines 2023; 12:36. [PMID: 38255143 PMCID: PMC10813070 DOI: 10.3390/biomedicines12010036] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Patients with diabetes mellitus can experience hyperglycemia, which affects brain function and produces cognitive impairment or neurodegeneration. Neuroinflammation is an important cause of cognitive dysfunction. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are antihyperglycemic agents that reportedly possess anti-inflammatory properties and may produce beneficial cognitive effects. We hypothesized that SGLT2 inhibitors alleviate hyperglycemia-related inflammation in brain immune cells. Cultured BV-2 microglia were exposed to high glucose (HG) in the absence or presence of SGLT2 inhibitors including canagliflozin (Cana), dapagliflozin (Dapa), empagliflozin (Empa), and ertugliflozin (Ertu). Afterward, we evaluated the cytotoxic and inflammatory responses by specific biochemical assays. Treatments with non-toxic Cana or Dapa, but not Empa or Ertu, inhibited proliferation without cell death. Only Cana rescued BV-2 microglia from HG-induced cytotoxicity, including apoptosis or autophagic degradation. None of SGLT2 inhibitors affected the HG-stimulated induction of stress proteins HO-1 and HSP70. Also, compared to the other three SGLT2 inhibitors, Cana was better at inhibiting HG-induced oxidative/inflammatory stress, as evidenced by its ability to repress proinflammatory factors (e.g., oxygen free radicals, iNOS, NLRP3, IL-1β, and TNF-α) other than COX-2. Cana's action to alleviate HG insults was mediated not by altering SGLT2 protein expression, but by reducing HG-stimulated signaling activities of NFκB, JNK, p38, and PI3K/Akt pathways. Particularly, Cana imitated the effects of NFκB inhibitor on HG-induced iNOS and COX-2. Of the four SGLT2 inhibitors, Cana provided BV-2 microglia with the best protection against HG-induced inflammatory toxicity. Thus, Cana may help to reduce innate neuroimmune damage caused by hyperglycemia.
Collapse
Affiliation(s)
- Ching-Tien Lee
- Department of Medical and Healthcare Business, Hsin-Sheng College of Medical Care and Management, Taoyuan 32544, Taiwan;
| | | | - Cheng-Fang Hsieh
- Division of Geriatrics and Gerontology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Jiz-Yuh Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| |
Collapse
|
9
|
Ma C, Sheng N, Li Y, Zheng H, Wang Z, Zhang J. A comprehensive perspective on the disposition, metabolism, and pharmacokinetics of representative multi-components of Dengzhan Shengmai in rats with chronic cerebral hypoperfusion after oral administration. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116212. [PMID: 36739927 DOI: 10.1016/j.jep.2023.116212] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dengzhan Shengmai capsule (DZSM), an evidence-based Chinese medicine comprising Erigeron breviscapus (Vaniot) Hand. -Mazz., Panax ginseng C.A.Mey., Ophiopogon japonicus (Thunb.) Ker Gawl., and Schisandra chinensis (Turcz.) Baill., exhibits an excellent efficacy in treating cardio- and cerebrovascular diseases. It contains caffeoyl compounds, flavonoids, saponins, and lignans as primary active components. However, so far, the characteristics of disposition, metabolism, and pharmacokinetics of its active components remain mostly unclear. AIM OF STUDY To elucidate disposition, metabolism, and pharmacokinetics of representative components of DZSM in rats with chronic cerebral hypoperfusion (CCH) by integrating ex vivo and in situ approaches. MATERIALS AND METHODS Exposure and distribution of absorbed prototypes and their metabolites were comprehensively investigated using sensitive LC-MS/MS and high-resolution LC-Q-TOF/MS. Pharmacokinetics of representative 16 components (12 prototypes and 4 metabolites) with different chemical categories, relatively high in vivo levels, wide tissue distribution, and reported neuroprotective activities were profiled. The ex vivo everted gut sac and in situ linked-rat models were adopted. RESULTS Representative 12 prototypes including 6 caffeoyl compounds (CA, 5-CQA, 3-CQA, 4-CQA, 1,3-CQA, and 3,4-CQA), 1 flavonoid (Scu), 2 saponins (Rd and Rg2), and 3 lignans (SchA, SchB, and SolA) presented characteristic absorption, disposition, and pharmacokinetics profiles in CCH rats. The caffeoyl compounds and flavonoid were well absorbed, exhibited wide distribution, and underwent extensive intestinal metabolism, such as methylation, isomerization, and sulfoconjugation. For CA, 5-CQA, Scu, and 4 related metabolites, the enterohepatic circulation was observed and resulted in bimodal or multimodal pharmacokinetic profiles. Saponins showed relatively low systemic exposure and limited distribution. The PPD-type ginsenoside Rd exhibited longer elimination half-life and systemic circulation than the PPT-type ginsenoside Rg2. No enterohepatic circulation was observed regarding saponins, suggesting that the multimodal pharmacokinetic profile of Rd could be due to its multi-site intestinal absorption. Lignans presented a low in vivo exposure and broad distribution. They were mainly transformed into hydroxylated metabolites. Corresponding to its bimodal pharmacokinetic profile, one metabolite of lignans completed the enterohepatic cycle. CONCLUSION The disposition, metabolism, and pharmacokinetic profiles of representative active components of DZSM were comprehensively characterized and elucidated.
Collapse
Affiliation(s)
- Congyu Ma
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| | - Ning Sheng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| | - Yuanyuan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| | - Hao Zheng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| | - Zhe Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| | - Jinlan Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
10
|
Liu H, Zhang X, Shi P, Yuan J, Jia Q, Pi C, Chen T, Xiong L, Chen J, Tang J, Yue R, Liu Z, Shen H, Zuo Y, Wei Y, Zhao L. α7 Nicotinic acetylcholine receptor: a key receptor in the cholinergic anti-inflammatory pathway exerting an antidepressant effect. J Neuroinflammation 2023; 20:84. [PMID: 36973813 PMCID: PMC10041767 DOI: 10.1186/s12974-023-02768-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/17/2023] [Indexed: 03/28/2023] Open
Abstract
Depression is a common mental illness, which is related to monoamine neurotransmitters and the dysfunction of the cholinergic, immune, glutamatergic, and neuroendocrine systems. The hypothesis of monoamine neurotransmitters is one of the commonly recognized pathogenic mechanisms of depression; however, the drugs designed based on this hypothesis have not achieved good clinical results. A recent study demonstrated that depression and inflammation were strongly correlated, and the activation of alpha7 nicotinic acetylcholine receptor (α7 nAChR)-mediated cholinergic anti-inflammatory pathway (CAP) in the cholinergic system exhibited good therapeutic effects against depression. Therefore, anti-inflammation might be a potential direction for the treatment of depression. Moreover, it is also necessary to further reveal the key role of inflammation and α7 nAChR in the pathogenesis of depression. This review focused on the correlations between inflammation and depression as well-discussed the crucial role of α7 nAChR in the CAP.
Collapse
Affiliation(s)
- Huiyang Liu
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Xiaomei Zhang
- grid.469520.c0000 0004 1757 8917Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, Institute of Medicinal Chemistry of Chinese Medicine, Chongqing Academy of Chinese Materia Medica, Chongqing, 400065 People’s Republic of China
| | - Peng Shi
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jiyuan Yuan
- grid.488387.8Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Qiang Jia
- grid.488387.8Ethics Committee Office, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Chao Pi
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
| | - Tao Chen
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Linjin Xiong
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jinglin Chen
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jia Tang
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ruxu Yue
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Zerong Liu
- Central Nervous System Drug Key Laboratory of Sichuan Province, Sichuan Credit Pharmaceutical CO., Ltd., Luzhou, 646000 Sichuan China
- grid.190737.b0000 0001 0154 0904Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Hongping Shen
- grid.488387.8Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ying Zuo
- grid.488387.8Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan China
| | - Yumeng Wei
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ling Zhao
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| |
Collapse
|
11
|
Liu F, Gao X, Li Z, Zhang X, Fan H, Yu G, Bello BK, Feng X, Li D, Teng D, Chen Y, Zhao P, Fu M, Dong J. Protective Effects of Scutellarin on Acute Alcohol Intestinal Injury. Chem Biodivers 2022; 19:e202100856. [PMID: 35263019 DOI: 10.1002/cbdv.202100856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/09/2022] [Indexed: 11/09/2022]
Abstract
The present study aims to investigate the roles of scutellarin (SCU) on acute alcohol intestinal injury. Mice were divided into six groups: alcohol, three administration, negative control and positive drug bifendate control. The administration group mice were intraperitoneally injected with SCU for 3 consecutive days followed by alcohol gavage at an interval of 1 h. After the mice were sacrificed, colon tissue damage was evaluated by histopathological examination; the activities of inducible nitric oxide synthase (iNOS) and catalase (CAT), as well as the content of malondialdehyde (MDA) were detected using biochemical kits; the levels of inflammatory cytokines mRNA were determined by real-time fluorescence quantitative PCR; the protein expression levels of hemeoxygenase-1 (HO-1) and phosphorylated nuclear factor-ĸB p65 were measured via western blotting. The results showed that alcohol induced severe colon morphological degradation, epithelia atrophy, and more inflammatory cells infiltration in the submucosa. SCU treatment prevented this process, especially in the middle and high dose groups. Alcohol treatment caused excessive lipid peroxidation product accumulation of MDA, restrained the activity of antioxidant enzyme CAT, induced HO-1 expression in the colon, whereas low dose SCU treatment significantly down-regulated the MDA level, enhanced the CAT level, and accelerated HO-1 signals. SCU prevented alcohol stimulation triggered inflammatory response in colon tissues through significantly downregulating the iNOS activity, transcript levels of Tnf-α, Il-1β and Il-6, and phosphorylation levels of NF-κB p65. These findings suggest that SCU protects the colon via antioxidant and anti-inflammatory mechanisms, making it a promising drug against alcohol-induced colon damage.
Collapse
Affiliation(s)
- Feixue Liu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xuzhu Gao
- Department of Central Laboratory, The Second People's Hospital of Lianyungang City, Lianyungang, 222000, China
| | - Zhixing Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xiao Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Hui Fan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Guili Yu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Babatunde Kazeem Bello
- State Key Laboratory of Rice Biology, Lianyungang Academy of Agricultural Sciences, Lianyungang, 222000, China
| | - Xin Feng
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Debang Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Daoyang Teng
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yi Chen
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - PanPan Zhao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Mian Fu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, 222005, China
| |
Collapse
|
12
|
Ha S, Vetrivel P, Kim S, Bhosale P, Kim H, Pak J, Heo J, Kim Y, Kim G. Inhibitory effect of membrane‑free stem cell components derived from adipose tissues on skin inflammation in keratinocytes. Mol Med Rep 2022; 25:125. [DOI: 10.3892/mmr.2022.12641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/24/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Sang Ha
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam‑do 52828, Republic of Korea
| | - Preethi Vetrivel
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam‑do 52828, Republic of Korea
| | - Seong Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam‑do 52828, Republic of Korea
| | - Pritam Bhosale
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam‑do 52828, Republic of Korea
| | - Hun Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam‑do 52828, Republic of Korea
| | - Jung Pak
- T‑Stem Co., Ltd., Changwon, Gyeongsangnam‑do 51573, Republic of Korea
| | - Jeong Heo
- Biological Resources Research Group, Bioenvironmental Science and Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), Jinju, Gyeongsangnam‑do 52834, Republic of Korea
| | - Young Kim
- T‑Stem Co., Ltd., Changwon, Gyeongsangnam‑do 51573, Republic of Korea
| | - Gon Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsangnam‑do 52828, Republic of Korea
| |
Collapse
|
13
|
Lim HS, Sohn E, Kim YJ, Kim BY, Kim JH, Jeong SJ. Ethanol Extract of Elaeagnus glabra f. oxyphylla Branches Alleviates the Inflammatory Response Through Suppression of Cyclin D3/Cyclin-Dependent Kinase 11p58 Coupled to Lipopolysaccharide-Activated BV-2 Microglia. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221075079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Neuroinflammation plays a pivotal role in the pathogenesis of neurodegenerative diseases and is characterized by microglial dysregulation. Here, we explored the beneficial effects of a leaf extract of Elaeagnus glabra f. oxyphylla (EGFO), a native medicinal plant to Korea, South China, Japan, and Taiwan, on neuroinflammation using lipopolysaccharide (LPS)-stimulated BV-2 microglia. Levels of the inflammatory mediators were determined by enzyme-linked immunosorbent assays and reverse transcription–polymerase chain reaction. The phospho levels of mitogen-activated protein kinases, which are key kinase molecules in the inflammatory signaling pathway in microglia, were analyzed by Western blotting. Treatment with EGFO significantly suppressed the LPS-mediated induction of nitric oxide and prostaglandin E2. Consistently, EGFO treatment in LPS-stimulated BV-2 cells markedly reduced the inflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) levels. The best concentration of EGFO that could reduce TNF-α and IL-6 was 100 μg/mL. EGFO relatively reduced the messenger RNA expression of TNF-α and IL-6 by 0.36 and 0.32-fold ratio, respectively, compared to LPS treatment. Moreover, EGFO markedly reduced the phospho levels of p38 and the c-jun N-terminal kinase. Furthermore, antibody microarray and immunoblotting data revealed that the pharmacological mechanisms driving the antineuroinflammatory action of EGFO involve prevention of the cyclin D3/cyclin-dependent kinase 11p58 (CDK11p58) interaction. In conclusion, our results demonstrate that EGFO alleviates the inflammatory response through the suppression of cyclin D3/CDK11p58 coupling in LPS-activated BV-2 microglia. We propose the potential of EGFO as a novel drug candidate for neurodegenerative diseases by targeting neuroinflammation.
Collapse
Affiliation(s)
- Hye-Sun Lim
- Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Eunjin Sohn
- Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Yu Jin Kim
- Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Bu-Yeo Kim
- Korea Institute of Oriental Medicine, Daejeon, South Korea
| | | | - Soo-Jin Jeong
- Korea Institute of Oriental Medicine, Daejeon, South Korea
| |
Collapse
|
14
|
Liu X, Li Y, Bai N, Yu C, Xiao Y, Li C, Liu Z. Updated evidence of Dengzhan Shengmai capsule against ischemic stroke: A systematic review and meta-analysis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114675. [PMID: 34571078 DOI: 10.1016/j.jep.2021.114675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ischemic stroke is the most common type of stroke, with high mortality, disability and recurrence rate, which brings a heavy burden to individuals, families and the medical system. Therefore, the intervention and treatment of ischemic stroke are of great significance. Chinese herbal medicine is widely used in treating stroke, for example, Dengzhan shengmai (DZSM) capsule. The current systematic review aims to comprehensively evaluate the efficacy and safety of the DZSM capsule in treating ischemic stroke. MATERIALS AND METHODS Eligible randomized controlled trials (RCTs) were included to evaluate the efficacy and safety of Chinese herbal medicine DZSM capsule in treating ischemic stroke. Eight electronic databases were searched up to January 27, 2021. The risk ratio (RR), standardized mean difference (SMD), or weighted mean difference (WMD) with 95% confidence interval (CI) were used to assess DZSM capsule treatment outcomes. RESULTS A total of 28 RCTs involving 6683 participants were included in the systematic review and meta-analysis. Compared with conventional therapy group, DZSM capsule plus conventional therapy improved Barthel Index scores (WMD: 8.97, 95%CI: 5.88-12.05) and reduced modified Rankin Scale (WMD: -0.75, 95%CI: -1.02∼ -0.48), reduced neurological functional deficit scores (WMD: -2.81, 95%CI: -4.17∼ -1.44), recurrence rate (RR: 0.57, 95%CI: 0.44-0.73) and mortality (RR: 0.54, 95%CI: 0.31-0.95), improved clinical effect (RR: 1.18, 95%CI: 1.12-1.24) and quality of life (WMD: 21.67, 95%CI: 6.74-36.61), exhibited a beneficial effect on hemorheology such as elevated levels of APTT (SMD: 1.17, 95%CI: 0.87-1.47) and INR (SMD: 1.12, 95%CI: 0.82-1.42), and on lipid metabolism such as levels of TC (SMD: -0.62, 95%CI: -1.04 ∼ -0.20), TG (SMD: -0.72, 95%CI: -1.18∼ -0.26), LDL (SMD: -1.14, 95%CI: -1.57∼ -0.71) and HDL (SMD: 0.93, 95%CI: 0.36-1.50). No trials reported severe adverse events. CONCLUSION DZSM capsule appears to be safe and effective in clinical applications for ischemic stroke. Based on conventional therapy, adding the DZSM capsule could reduce the mortality, recurrence rate, and neurological functional deficit scores, improve clinical effect and quality of life. In addition, compared with conventional therapy, the addition of the DZSM capsule played a beneficial role in hemorheology and lipid metabolism, which may attribute to the potential mechanism.
Collapse
Affiliation(s)
- Xueyan Liu
- Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Yiqi Li
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350108, China
| | - Ningning Bai
- Guangzhou Chinese Medicinal Materials Magazine Co., Ltd, Guangzhou, Guangdong, 510000, China
| | - Chuanyu Yu
- Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Yun Xiao
- Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Chi Li
- World Federation of Chinese Medicine Societies, Beijing, 100101, China.
| | - Zhiqiang Liu
- Fujian Medical University, Fuzhou, Fujian, 350122, China.
| |
Collapse
|
15
|
Lu L, Yang LK, Yue J, Wang XS, Qi JY, Yang F, Feng B, Liu SB. Scutellarin alleviates depression-like behaviors induced by LPS in mice partially through inhibition of astrocyte-mediated neuroinflammation. Neurosci Lett 2021; 765:136284. [PMID: 34624394 DOI: 10.1016/j.neulet.2021.136284] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/23/2022]
Abstract
Depression is a kind of common mental disorder associated with neuroinflammation, and astrocytes play a vital role in regulating and mediating neuroinflammation in central nervous system. Scutellarin has significant anti-inflammatory and neuroprotective effects. However, whether scutellarin exerts antidepressant effect remains unknown. In present study, it was found that scutellarin suppressed LPS-induced neuroinflammation in the hippocampus and alleviated depression-like behaviors in mice. In addition, scutellarin inhibited LPS-induced elevation of TNFα, IL-1β, IL-6 and iNOS, and reversed the downregulation of IL-4 and BDNF in astrocytes in vitro. Furthermore, the activated TLR4/NF-κB pathway in LPS-treated astrocytes was suppressed by scutellarin. Collectively, these results suggest that scutellarin ameliorates depression-like behaviors induced by neuroinflammation partially through inhibiting the TLR4/NF-κB pathway in astrocytes.
Collapse
Affiliation(s)
- Liang Lu
- Department of Pharmacology and Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Liu-Kun Yang
- Department of Pharmacology and Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Jiao Yue
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Pharmacy, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xin-Shang Wang
- Department of Pharmacology and Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Jing-Yu Qi
- Department of Pharmacology and Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Fan Yang
- Department of Pharmacology and Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ban Feng
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Pharmacy, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Shui-Bing Liu
- Department of Pharmacology and Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
16
|
Dejani NN, Elshabrawy HA, Bezerra Filho CDSM, de Sousa DP. Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives. Biomolecules 2021; 11:biom11081254. [PMID: 34439920 PMCID: PMC8394099 DOI: 10.3390/biom11081254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/14/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023] Open
Abstract
In 2019, COVID-19 emerged as a severe respiratory disease that is caused by the novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). The disease has been associated with high mortality rate, especially in patients with comorbidities such as diabetes, cardiovascular and kidney diseases. This could be attributed to dysregulated immune responses and severe systemic inflammation in COVID-19 patients. The use of effective antiviral drugs against SARS-CoV-2 and modulation of the immune responses could be a potential therapeutic strategy for COVID-19. Studies have shown that natural phenolic compounds have several pharmacological properties, including anticoronavirus and immunomodulatory activities. Therefore, this review discusses the dual action of these natural products from the perspective of applicability at COVID-19.
Collapse
Affiliation(s)
- Naiara Naiana Dejani
- Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA;
| | - Carlos da Silva Maia Bezerra Filho
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
| | - Damião Pergentino de Sousa
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
- Correspondence: ; Tel.: +55-83-3216-7347
| |
Collapse
|
17
|
Zhou J, Lin H, Xu P, Yao L, Xie Q, Mao L, Wang Y. Matcha green tea prevents obesity-induced hypothalamic inflammation via suppressing the JAK2/STAT3 signaling pathway. Food Funct 2021; 11:8987-8995. [PMID: 33001081 DOI: 10.1039/d0fo01500h] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Obesity is an increasingly severe global health problem, leading to chronic inflammation and metabolic disorders in both peripheral tissues and the central nervous system. Matcha is a powdered green tea, and it is very popular in recent years as a beverage and food additive. Matcha green tea has been reported to have outstanding potential in regulating obesity-related metabolic syndrome. However, there are few studies on the regulation mechanism of matcha green tea on the central nervous system. In this study, we established a high-fat diet-induced obese mouse model. The results showed that dietary supplementation with matcha could effectively inhibit the weight gain, fat accumulation, glycemia and lipidemia increase, and excessive activation of microglia in the arcuate nucleus of the hypothalamus. Furthermore, we used different concentrations (100%, 80%, 60%, 40%, and 20%, v/v) of ethanol solution to prepare matcha ethanol extracts, and investigated their effects on palmitic acid-induced inflammation of microglial BV-2 cells. The results showed that matcha ethanol extracts could significantly reduce the release of inflammatory cytokines and the expression and phosphorylation of JAK2 and STAT3.
Collapse
Affiliation(s)
- Jihong Zhou
- Tea Research Institute, Zhejiang University, Zijingang Campus, Hangzhou 310058, P. R. China.
| | - Haiyu Lin
- Tea Research Institute, Zhejiang University, Zijingang Campus, Hangzhou 310058, P. R. China.
| | - Ping Xu
- Tea Research Institute, Zhejiang University, Zijingang Campus, Hangzhou 310058, P. R. China.
| | - Liyun Yao
- Tea Research Institute, Zhejiang University, Zijingang Campus, Hangzhou 310058, P. R. China.
| | - Qingyi Xie
- Tea Research Institute, Zhejiang University, Zijingang Campus, Hangzhou 310058, P. R. China.
| | - Limin Mao
- Tea Research Institute, Zhejiang University, Zijingang Campus, Hangzhou 310058, P. R. China. and Zhejiang Tea Group Co., Ltd, Hangzhou 310058, P. R. China
| | - Yuefei Wang
- Tea Research Institute, Zhejiang University, Zijingang Campus, Hangzhou 310058, P. R. China.
| |
Collapse
|
18
|
Piovesana R, Salazar Intriago MS, Dini L, Tata AM. Cholinergic Modulation of Neuroinflammation: Focus on α7 Nicotinic Receptor. Int J Mol Sci 2021; 22:ijms22094912. [PMID: 34066354 PMCID: PMC8125157 DOI: 10.3390/ijms22094912] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
All nervous system pathologies (e.g., neurodegenerative/demyelinating diseases and brain tumours) develop neuroinflammation, a beneficial process during pathological events, aimed at removing damaged cells, toxic agents, and/or pathogens. Unfortunately, excessive inflammation frequently occurs during nervous system disorders, becoming a detrimental event capable of enhancing neurons and myelinating glial cell impairment, rather than improving their survival and activity. Consequently, targeting the neuroinflammation could be relevant for reducing brain injury and rescuing neuronal and glial cell functions. Several studies have highlighted the role of acetylcholine and its receptors in the regulation of central and peripheral inflammation. In particular, α7 nicotinic receptor has been described as one of the main regulators of the “brain cholinergic anti-inflammatory pathway”. Its expression in astrocytes and microglial cells and the ability to modulate anti-inflammatory cytokines make this receptor a new interesting therapeutic target for neuroinflammation regulation. In this review, we summarize the distribution and physiological functions of the α7 nicotinic receptor in glial cells (astrocytes and microglia) and its role in the modulation of neuroinflammation. Moreover, we explore how its altered expression and function contribute to the development of different neurological pathologies and exacerbate neuroinflammatory processes.
Collapse
Affiliation(s)
- Roberta Piovesana
- Département de Neurosciences, Université de Montréal, Montréal, QC H3C 3J7, Canada;
- Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | | | - Luciana Dini
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza, University of Rome, 00185 Rome, Italy; (M.S.S.I.); (L.D.)
| | - Ada Maria Tata
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza, University of Rome, 00185 Rome, Italy; (M.S.S.I.); (L.D.)
- Research Centre of Neurobiology “Daniel Bovet”, Sapienza, University of Rome, 00185 Rome, Italy
- Correspondence: ; Tel.: +39-06-4991-2822
| |
Collapse
|
19
|
Ye J, Zeng B, Zhong M, Li H, Xu L, Shu J, Wang Y, Yang F, Zhong C, Ye X, He X, Ouyang D. Scutellarin inhibits caspase-11 activation and pyroptosis in macrophages via regulating PKA signaling. Acta Pharm Sin B 2021; 11:112-126. [PMID: 33532184 PMCID: PMC7838020 DOI: 10.1016/j.apsb.2020.07.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/21/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory caspase-11 senses and is activated by intracellular lipopolysaccharide (LPS) leading to pyroptosis that has critical role in defensing against bacterial infection, whereas its excess activation under pathogenic circumstances may cause various inflammatory diseases. However, there are few known drugs that can control caspase-11 activation. We report here that scutellarin, a flavonoid from Erigeron breviscapus, acted as an inhibitor for caspase-11 activation in macrophages. Scutellarin dose-dependently inhibited intracellular LPS-induced release of caspase-11p26 (indicative of caspase-11 activation) and generation of N-terminal fragment of gasdermin D (GSDMD-NT), leading to reduced pyroptosis. It also suppressed the activation of non-canonical nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome as evidenced by reduced apoptosis-associated speck-like protein containing a CARD (ASC) speck formation and decreased interleukin-1 beta (IL-1β) and caspase-1p10 secretion, whereas the NLRP3-specific inhibitor MCC950 only inhibited IL-1β and caspase-1p10 release and ASC speck formation but not pyroptosis. Scutellarin also suppressed LPS-induced caspase-11 activation and pyroptosis in RAW 264.7 cells lacking ASC expression. Moreover, scutellarin treatment increased Ser/Thr phosphorylation of caspase-11 at protein kinase A (PKA)-specific sites, and its inhibitory action on caspase-11 activation was largely abrogated by PKA inhibitor H89 or by adenylyl cyclase inhibitor MDL12330A. Collectively, our data indicate that scutellarin inhibited caspase-11 activation and pyroptosis in macrophages at least partly via regulating the PKA signaling pathway.
Collapse
Affiliation(s)
- Jiezhou Ye
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Bo Zeng
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Meiyan Zhong
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hongchun Li
- Wuzhongpei Memorial Hospital of Shunde, Foshan 528300, China
| | - Lihui Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Junxiang Shu
- Wuzhongpei Memorial Hospital of Shunde, Foshan 528300, China
| | - Yaofeng Wang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Fan Yang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Chunsu Zhong
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xunjia Ye
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xianhui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Dongyun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
20
|
Xiao J, Wang Y, Yang Y, Liu J, Chen G, Lin B, Hou Y, Li N. Natural potential neuroinflammatory inhibitors from Stephania epigaea H.S. Lo. Bioorg Chem 2020; 107:104597. [PMID: 33450546 DOI: 10.1016/j.bioorg.2020.104597] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 12/23/2022]
Abstract
Stephania epigaea H. S. Lo is a folk medicine widely distributed in the south of China, especially in Yunnan and Guangxi province. An in vitro anti-neuroinflammatory study showed that total alkaloids of it can potently inhibit LPS-induced NO releasing of BV2 cells with an IC50 value of 10.05 ± 2.03 μg/mL (minocycline as the positive drug, IC50 15.49 ± 2.14 μM). The phytochemical investigation of the total alkaloids afforded three new phenanthrene (1-3), two lactams (4a, 4b), and nine aporphine derivatives (5-13). The final structure of 1 was identified by computer-assisted structure elucidation (ACD/Structure Elucidator software and the 13C NMR calculation with GIAO method) due to many possibilities of the substituent pattern. All isolates were evaluated for their anti-neuroinflammatory effects, and as a result, 5, 8, 10, and 11 exhibited stronger inhibitory activities than the minocycline. The results suggested S. epigaea could provide potential therapeutic agents for neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiao Xiao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, People's Republic of China
| | - Yingjie Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, People's Republic of China
| | - Yanqiu Yang
- College of Life and Health Sciences, Northeastern University, Shenyang 110004, People's Republic of China
| | - Jingyu Liu
- College of Life and Health Sciences, Northeastern University, Shenyang 110004, People's Republic of China
| | - Gang Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, People's Republic of China
| | - Bin Lin
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yue Hou
- College of Life and Health Sciences, Northeastern University, Shenyang 110004, People's Republic of China.
| | - Ning Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, People's Republic of China.
| |
Collapse
|
21
|
Peng L, Wen L, Shi QF, Gao F, Huang B, Meng J, Hu CP, Wang CM. Scutellarin ameliorates pulmonary fibrosis through inhibiting NF-κB/NLRP3-mediated epithelial-mesenchymal transition and inflammation. Cell Death Dis 2020; 11:978. [PMID: 33188176 PMCID: PMC7666141 DOI: 10.1038/s41419-020-03178-2] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is featured with inflammation and extensive lung remodeling caused by overloaded deposition of extracellular matrix. Scutellarin is the major effective ingredient of breviscapine and its anti-inflammation efficacy has been reported before. Nevertheless, the impact of scutellarin on IPF and the downstream molecular mechanism remain unclear. In this study, scutellarin suppressed BLM-induced inflammation via NF-κB/NLRP3 pathway both in vivo and in vitro. BLM significantly elevated p-p65/p65 ratio, IκBα degradation, and levels of NLRP3, caspase-1, caspase-11, ASC, GSDMDNterm, IL-1β, and IL-18, while scutellarin reversed the above alterations except for that of caspase-11. Scutellarin inhibited BLM-induced epithelial-mesenchymal transition (EMT) process in vivo and in vitro. The expression levels of EMT-related markers, including fibronectin, vimentin, N-cadherin, matrix metalloproteinase 2 (MMP-2) and MMP-9, were increased in BLM group, and suppressed by scutellarin. The expression level of E-cadherin showed the opposite changes. However, overexpression of NLRP3 eliminated the anti-inflammation and anti-EMT functions of scutellarin in vitro. In conclusion, scutellarin suppressed inflammation and EMT in BLM-induced pulmonary fibrosis through NF-κB/NLRP3 signaling.
Collapse
Affiliation(s)
- Ling Peng
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of the National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| | - Li Wen
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China
| | - Qing-Feng Shi
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China
| | - Feng Gao
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China
| | - Bin Huang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China
| | - Jie Meng
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of the National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| | - Cheng-Ping Hu
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of the National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China.
| | - Chang-Ming Wang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541002, P.R. China.
| |
Collapse
|
22
|
Gamage R, Wagnon I, Rossetti I, Childs R, Niedermayer G, Chesworth R, Gyengesi E. Cholinergic Modulation of Glial Function During Aging and Chronic Neuroinflammation. Front Cell Neurosci 2020; 14:577912. [PMID: 33192323 PMCID: PMC7594524 DOI: 10.3389/fncel.2020.577912] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex biological process that increases the risk of age-related cognitive degenerative diseases such as dementia, including Alzheimer’s disease (AD), Lewy Body Dementia (LBD), and mild cognitive impairment (MCI). Even non-pathological aging of the brain can involve chronic oxidative and inflammatory stress, which disrupts the communication and balance between the brain and the immune system. There has been an increasingly strong connection found between chronic neuroinflammation and impaired memory, especially in AD. While microglia and astrocytes, the resident immune cells of the central nervous system (CNS), exerting beneficial effects during the acute inflammatory phase, during chronic neuroinflammation they can become more detrimental. Central cholinergic circuits are involved in maintaining normal cognitive function and regulating signaling within the entire cerebral cortex. While neuronal-glial cholinergic signaling is anti-inflammatory and anti-oxidative, central cholinergic neuronal degeneration is implicated in impaired learning, memory sleep regulation, and attention. Although there is evidence of cholinergic involvement in memory, fewer studies have linked the cholinergic anti-inflammatory and anti-oxidant pathways to memory processes during development, normal aging, and disease states. This review will summarize the current knowledge of cholinergic effects on microglia and astroglia, and their role in both anti-inflammatory and anti-oxidant mechanisms, concerning normal aging and chronic neuroinflammation. We provided details on how stimulation of α7 nicotinic acetylcholine (α7nACh) receptors can be neuroprotective by increasing amyloid-β phagocytosis, decreasing inflammation and reducing oxidative stress by promoting the nuclear factor erythroid 2-related factor 2 (Nrf2) pathways and decreasing the release of pro-inflammatory cytokines. There is also evidence for astroglial α7nACh receptor stimulation mediating anti-inflammatory and antioxidant effects by inhibiting the nuclear factor-κB (NF-κB) pathway and activating the Nrf2 pathway respectively. We conclude that targeting cholinergic glial interactions between neurons and glial cells via α7nACh receptors could regulate neuroinflammation and oxidative stress, relevant to the treatment of several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rashmi Gamage
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ingrid Wagnon
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ilaria Rossetti
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ryan Childs
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Garry Niedermayer
- School of Science, Western Sydney University, Penrith, NSW, Australia
| | - Rose Chesworth
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Erika Gyengesi
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
23
|
Chumsakul O, Wakayama K, Tsuhako A, Baba Y, Takai Y, Kurose T, Honma Y, Watanabe S. Apigenin Regulates Activation of Microglia and Counteracts Retinal Degeneration. J Ocul Pharmacol Ther 2020; 36:311-319. [PMID: 32379991 DOI: 10.1089/jop.2019.0163] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Purpose: Photoreceptor degeneration is a major cause of blindness. Microglia are known to play key roles in the pathogenesis and progression of neural degeneration. We examined the possible use of apigenin, which is a naturally occurring flavonoid, for the treatment of photoreceptor degeneration through regulation of microglial activities. Methods: As in vitro analyses, BV2 and MG5 mouse microglia cell lines were stimulated in the presence or absence of apigenin, and their activation profile was examined. In vivo study was done using rd1 photoreceptor degeneration model, and apigenin was administered by intravitreal injection, and pathological feature was examined. Results: Cell survival was not affected by apigenin in either BV2 and MG5. Apigenin suppressed lipopolysaccharide (LPS)-induced chemokine production in both BV2 and MG5 cells, but phagocytosis was suppressed in MG5 cells but not in BV2 cells. Apigenin inhibited LPS-induced M1 activation but could not drive microglia toward the M2 phenotype. Apigenin suppressed the expression of miR-155 in a dose-dependent manner. Furthermore, the Ets protein level was suppressed by treatment of BV2 cells with apigenin. When rd1 mice were treated with apigenin by intravitreal injection, the expression of inflammatory chemokines in the retina was reduced, and activation of microglia and Müller glia was suppressed. Furthermore, the thickness of the outer nuclear layer of the retina of rd1 mice was thicker in apigenin-treated retinas. Conclusions: Taken together, local administration of apigenin to the retina is a potential therapeutic treatment for photoreceptor degeneration, which involves downregulation of microglia in the retina when photoreceptors are damaged.
Collapse
Affiliation(s)
- Onuma Chumsakul
- Pharmacological Research Group, Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Kizugawa, Kyoto, Japan
| | - Kanaho Wakayama
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Asano Tsuhako
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yukihiro Baba
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yoshihiro Takai
- Pharmacological Research Group, Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Kizugawa, Kyoto, Japan
| | - Takahiro Kurose
- Pharmacological Research Group, Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Kizugawa, Kyoto, Japan
| | - Yoichi Honma
- Pharmacological Research Group, Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Kizugawa, Kyoto, Japan
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
24
|
A Potential Anti-cancer Compound Separated from the Chloroform Extract of the Chinese Medicine Formula Shenqi San. Curr Med Sci 2020; 40:138-144. [PMID: 32166676 DOI: 10.1007/s11596-020-2157-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/27/2019] [Indexed: 12/28/2022]
Abstract
This study examined anti-cancer compounds present in the chloroform extract of the Chinese medicine formula Shenqi San (CE-SS). Silica gel column chromatography, Sephadex LH-20, octadecylsilyl (ODS) column chromatography, and high performance liquid chromatography (HPLC) were used to separate the compounds from CE-SS. The structural formulas of the separated compounds were determined using 1D 1H and 13C experiments as well as high resolution electrospray ionization mass spectroscopy (HRESIMS). The corresponding results were compared with the reported literature data. A total of six compounds were separated and their structures were identified on the basis of corresponding spectroscopic and physico-chemical properties. They were Saikogenin F (I), Prosaikogenin D (II), Prosaikogenin F (III), β-sitosterol (IV), 3β,16β,23-trihydroxy-13,28-epoxyurs-11-ene-3-O-β-D-glucopyranoside (V), and methyl ursolic acid (VI). The separated compounds were evaluated in vitro for their inhibitory ability against the proliferation of A549 cells via MTT assay. Apoptosis was investigated using Annexin V-FITC/propidium iodide (PI) by flow cytometry. Apoptosis-associated proteins were examined by Western blotting. All the compounds were observed to have inhibitory activities against the proliferation of A549 cells to different degrees. Flow cytometry showed that compound V increased the proportion of apoptotic A549 cells in a dose-dependent manner. Western blotting showed that compound V increased the expression of Bax, cleaved-caspase-3, cleaved-caspase-9 and cleaved-poly ADP-ribose polymerase (PARP), and decreased the expression of Bcl-2. These results indicated that compound V featured a significant inhibitory effect on A549 cells when compared with other compounds, and it may be considered a potential drug against cancers.
Collapse
|
25
|
Liu F, Li L, Lu W, Ding Z, Huang W, Li YT, Cheng C, Shan WS, Xu J, He W, Zhanghui, Yin Z. Scutellarin ameliorates cartilage degeneration in osteoarthritis by inhibiting the Wnt/β-catenin and MAPK signaling pathways. Int Immunopharmacol 2020; 78:105954. [DOI: 10.1016/j.intimp.2019.105954] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 10/25/2022]
|
26
|
Wang W, Li J, Li F, Peng J, Xu M, Shangguan Y, Li Y, Zhao Y, Qiu C, Qu R, Li W, Zhang C, Zhang T. Scutellarin suppresses cartilage destruction in osteoarthritis mouse model by inhibiting the NF-κB and PI3K/AKT signaling pathways. Int Immunopharmacol 2019; 77:105928. [PMID: 31711940 DOI: 10.1016/j.intimp.2019.105928] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/20/2019] [Accepted: 09/20/2019] [Indexed: 12/22/2022]
Abstract
Osteoarthritis (OA), a common and severe disease, is predominantly characterized by cartilage destruction, which results in the degeneration of joint surfaces. Nowadays, it is accepted that TNFα plays a critical role in OA. Scutellarin, the main bioactive flavonoid glycoside extracted form Erigeron breviscapus, has been reported to exert positive effects on anti-inflammatory reactions. However, the effect of scutellarin in OA is still unknown. In this study, we isolated and cultured primary murine chondrocytes, stimulating TNF-α, in the presence or absence of scutellarin treatment. We found that the inflammatory response stimulated by TNF-α was significantly inhibited by the addition of scutellarin. Moreover, we established OA mouse models induced by surgery. In this mouse model, both inflammatory reaction and cartilage degeneration were markedly inhibited by oral administration of scutellarin. Furthermore, the cellular mechanism underlying the protective effect of scutellarin in OA was clearly associated with the NF-κB and PI3K/AKT signaling pathways. Collectively, this study proposes scutellarin as a potential therapeutic to treat joint degenerative diseases, including OA.
Collapse
Affiliation(s)
- Wenhan Wang
- Department of Pathology, The School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, PR China; Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Jiayi Li
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Feng Li
- Department of Medical Imaging, First People's Hospital of Jinan, Jinan, Shandong 250012, PR China
| | - Jiangfan Peng
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Mingyang Xu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Yangtao Shangguan
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Yuanming Li
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Yunpeng Zhao
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Cheng Qiu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Ruize Qu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Weiwei Li
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Cuijuan Zhang
- Department of Pathology, The School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, PR China; Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Tingguo Zhang
- Department of Pathology, The School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
27
|
Galangin Suppresses Renal Inflammation via the Inhibition of NF- κB, PI3K/AKT and NLRP3 in Uric Acid Treated NRK-52E Tubular Epithelial Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3018357. [PMID: 31240210 PMCID: PMC6556363 DOI: 10.1155/2019/3018357] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/22/2019] [Accepted: 04/16/2019] [Indexed: 01/10/2023]
Abstract
Renal inflammation can result in renal injury. Uric acid (UA) is the final product of purine metabolism in humans and because of the lack of urate oxidase, UA may accumulate in tissues, including kidney, causing inflammation. Galangin was isolated from a traditional Chinese medicine plant and possesses several beneficial effects, working as an anti-oxidant, anti-mutagenic, anti-tumor, anti-inflammatory, anti-microbial, and anti-viral agent. Therefore, this study aimed at investigating the molecular mechanism of galangin in the attenuation of UA induced renal inflammation in normal rat kidney epithelial cells NRK-52E. Our findings suggested that galangin treatment efficiently protected NRK-52E cells against UA induced renal inflammation by decreasing tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-18, prostaglandin E2 (PGE2), and nitric oxide (NO) release, and it inhibited nitric oxide synthase (iNOS), prostaglandin endoperoxide synthase 2 (PTGS2), TNF-α, IL-1β, and IL-18 mRNA expression. In addition, galangin was not exerting any cytotoxicity at the concentrations that were effective against inflammation as assessed by CCK8 assay. Moreover, western blotting showed that galangin treatment effectively inhibited nuclear factor-kappa B (NF-κB), phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT) and nucleotide-binding domain- (NOD-) like receptor protein 3 (NLRP3) signaling pathway activation. Taken together, these findings suggested that galangin plays a pivotal role in renal inflammation by suppressing inflammatory responses, which might be closely associated with the inhibition of NLRP3 inflammasome, NF-κB and PI3K/AKT signaling pathway activation.
Collapse
|
28
|
TEEG Induced A549 Cell Autophagy by Regulating the PI3K/AKT/mTOR Signaling Pathway. Anal Cell Pathol (Amst) 2019; 2019:7697610. [PMID: 31183317 PMCID: PMC6515120 DOI: 10.1155/2019/7697610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/20/2019] [Accepted: 04/07/2019] [Indexed: 12/14/2022] Open
Abstract
TEEG (3β,16β,23-trihydroxy-13,28-epoxyurs-11-ene-3-O-β-D-glucopyranoside) is derived from the chloroform extract of the Chinese medicine formula Shenqi San (CE-SS). In the present study, we aimed to elucidate the anticancer effect and possible molecular mechanism underlying the action of TEEG against the human non-small cell lung cancer (NSCLC) cell line A549 in vitro. A549 cells were incubated with different concentrations of TEEG. Cell proliferation was assessed by MTT assay. Autophagy was evaluated by immunofluorescence staining. Autophagy-associated proteins were examined by Western blot analysis. TEEG markedly inhibited A549 cell proliferation in a concentration-dependent manner. Immunofluorescence staining showed that TEEG induced autophagy in A549 cells. The LC3-II : LC3-I conversion ratio and the expression of Beclin-1, Atg5, Atg7, and Atg12 increased with the concentration of TEEG. In addition, increased TEEG concentration enhanced the expression of Class III p-PI3K and reduced the expression of Class I p-PI3K, p-AKT, p-mTOR, and p-P70S6K. These results indicate that TEEG induces autophagy of A549 cells through regulation of the PI3K/AKT/mTOR signaling pathway.
Collapse
|
29
|
Sun CY, Nie J, Zheng ZL, Zhao J, Wu LM, Zhu Y, Su ZQ, Zheng GJ, Feng B. Renoprotective effect of scutellarin on cisplatin-induced renal injury in mice: Impact on inflammation, apoptosis, and autophagy. Biomed Pharmacother 2019; 112:108647. [PMID: 30797149 DOI: 10.1016/j.biopha.2019.108647] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/16/2022] Open
Abstract
Cisplatin remains the standard first-line chemotherapeutic agent in the treatment of many types of cancers, but its clinical application is hindered by its severe nephrotoxicity. Previous studies reported that scutellarin enhanced the anti-cancer activity of cisplatin in lung cancer cells, with no confirmation on cisplatin-induced renal damage. Here, we investigated the nephroprotective effect of scutellarin on cisplatin-induced renal injury and its underlying mechanisms. Renal function, histological change, inflammation, apoptosis, autophagy and involved pathways were investigated. Pretreatment with scutellarin prevented cisplatin-induced decline of renal function including BUN, CRE, and histological damage. Scutellarin also reduced renal inflammation by suppressing the levels of pro-inflammatory cytokine, TNF-α and IL-6. Similarly, scutellarin administration inhibited apoptosis triggered by cisplatin through reducing the expressions of Cleaved caspase-3, Cleaved PARP, p53, and the ratio of Bax/Bcl-2. Moreover, scutellarin prevented cisplatin-induced inhibition of autophagy via enhancing LC3-II/LC3-I and Atg7, and inhibition of p62. Of note, the activations of JNK, ERK, p38 and stat3 induced by cisplatin were strikingly attenuated in scutellarin-treated mice. Thus, these results provide compelling evidence that scutellarin is a novel nephroprotectant against cisplatin-induced renal toxicity.
Collapse
Affiliation(s)
- Chao-Yue Sun
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Juan Nie
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zuo-Liang Zheng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Jie Zhao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Liu-Mei Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, no 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, China
| | - Ying Zhu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Zu-Qing Su
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Guang-Juan Zheng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Bing Feng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
30
|
Xu K, Guo L, Bu H, Wang H. Daphnetin inhibits high glucose-induced extracellular matrix accumulation, oxidative stress and inflammation in human glomerular mesangial cells. J Pharmacol Sci 2018; 139:91-97. [PMID: 30595336 DOI: 10.1016/j.jphs.2018.11.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/11/2018] [Accepted: 11/26/2018] [Indexed: 01/22/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the most common causes of end-stage renal disease (ESRD). Oxidative stress and inflammation have been documented to play important roles in the pathogenesis of DN. Daphnetin, a natural coumarin compound, possesses antioxidant and anti-inflammatory activities. However, the role of daphnetin in DN has not yet been investigated. The aim of the present study was to explore the function of daphnetin in DN and the underlying mechanism in vitro. Our results demonstrated that daphnetin alleviated cell proliferation induced by high glucose (HG) in human mesangial cells (MCs). Daphnetin strikingly reduced reactive oxygen species (ROS) and malonaldehyde (MDA) levels, and induced the superoxide dismutase (SOD) activity in HG-stimulated MCs. Besides, the production of TNF-α, IL-1β, IL-6, fibronectin (FN) and collagen IV (Col IV) was also inhibited by daphnetin in HG-stimulated MCs. In addition, daphnetin enhanced the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2) and inhibited the levels of p-Akt and p-p65 in HG-stimulated MCs. The results indicated that daphnetin inhibited HG-induced oxidative stress, inflammatory response, and ECM accumulation in human MCs. The effect is partially mediated by Nrf2/keap1 and Akt/NF-κB pathways. The findings suggested that daphnetin might be a therapeutic or preventive agent for DN.
Collapse
Affiliation(s)
- Ke Xu
- Department of Nephrology, Xinxiang Central Hospital, Xinxiang 453000, Henan, China.
| | - Liqin Guo
- Department of Nephrology, Xinxiang Central Hospital, Xinxiang 453000, Henan, China
| | - Haixia Bu
- Department of Nephrology, Xinxiang Central Hospital, Xinxiang 453000, Henan, China
| | - Huan Wang
- Department of Nephrology, Xinxiang Central Hospital, Xinxiang 453000, Henan, China
| |
Collapse
|
31
|
Shang L, Wang T, Tong D, Kang W, Liang Q, Ge S. Prolyl hydroxylases positively regulated LPS-induced inflammation in human gingival fibroblasts via TLR4/MyD88-mediated AKT/NF-κB and MAPK pathways. Cell Prolif 2018; 51:e12516. [PMID: 30091492 PMCID: PMC6528886 DOI: 10.1111/cpr.12516] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Prolyl hydroxylases (PHDs) play essential roles in oxygen-sensing system, whereas the effects of PHDs on inflammation have not been totally uncovered. Our study aimed to investigate the role of PHDs in lipopolysaccharide (LPS)-induced inflammation of human gingival fibroblasts (HGFs) and clarify the potential mechanisms. MATERIALS AND METHODS A pan hydroxylase inhibitor, dimethyloxallyl glycine (DMOG), and RNA interference were used to explore the role of PHDs in inflammation. Cytotoxic effect of DMOG was determined by cell-counting kit-8 and flow cytometry respectively. The secretion levels of IL-6 and IL-8 were assessed by ELISA. The mRNA levels of inflammatory cytokines, Toll-like receptor (TLR) 4 and MyD88 were evaluated by quantitative real-time PCR. The activation of NF-κB, mitogen-activated protein kinase (MAPK) and PI3K/AKT pathways were detected by western blot and the nuclear translocation of NF-κB p65 was examined by immunofluorescence. Downregulation of PHD1 and PHD2 was performed with siRNA transfection. RESULTS Dimethyloxallyl glycine inhibited LPS-induced inflammatory cytokine, TLR4 and MyD88 expression in gene level and the elevated secretion of IL-6 and IL-8 was also downregulated. Additionally, LPS-induced activation of NF-κB, MAPK and AKT pathways was abolished by DMOG treatment. Importantly, LPS-induced inflammatory cytokine expression was merely suppressed by PHD2 knockdown. CONCLUSIONS Prolyl hydroxylases acted as a positive regulator in LPS-induced inflammation of HGFs via TLR4/MyD88-mediated NF-κB, MAPK and AKT signalling pathways and PHD2 among three isoforms was principally responsible for the effects.
Collapse
Affiliation(s)
- Lingling Shang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Ting Wang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Dongdong Tong
- Department of Oral maxillofacial SurgerySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Wenyan Kang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Qianyu Liang
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| | - Shaohua Ge
- Shandong Provincial Key Laboratory of Oral Tissue RegenerationSchool of Stomatology, Shandong UniversityShandong, JinanChina
- Department of PeriodontologySchool of Stomatology, Shandong UniversityShandong, JinanChina
| |
Collapse
|