1
|
Qiu M, Ye C, Bao L, Wu K, Zhao Y, Zhao X, Tang R, Shang R, Shang S, Yuan C, Hu X, Zhang N, Fu Y, Wang J, Zhao C. Elevated muramyl dipeptide by sialic acid-facilitated postantibiotic pathobiont expansion contributes to gut dysbiosis-induced mastitis in mice. J Adv Res 2024:S2090-1232(24)00425-9. [PMID: 39374734 DOI: 10.1016/j.jare.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024] Open
Abstract
INTRODUCTION In responses to antibiotics exposure, gut dysbiosis is a risk factor not only for pathogen infection but also for facilitating pathobiont expansion, resulting in increased inflammatory responses in the gut and distant organs. However, how this process is regulated has not been fully elucidated. OBJECTIVES In this study, we investigated the role of sialic acid, a host-derived carbohydrate, in the pathogenesis of gut dysbiosis-derived inflammation in distant organs. METHODS Ampicillin (Amp)-induced gut dysbiotic mice were treated with N-glycolylneuraminic acid (Neu5Gc) and N-acetylneuraminic acid (Neu5Ac) for three weeks to assess the role of sialic acids in mastitis. The underlying mechanism by which sialic acids regulate mastitis was explored using 16S rRNA sequencing, transcriptomics and employed multiple molecular approaches. RESULTS Administration of Neu5Ac and Neu5Gc exacerbated gut dysbiosis-induced mastitis and systemic inflammation. The gut dysbiosis caused by Amp was also aggravated by sialic acid. Notably, increased Enterococcus expansion, which was positively correlated with inflammatory markers, was observed in both Neu5Ac- and Neu5Gc-treated gut dysbiotic mice. Treatment of mice with Enterococcus cecorum (E. cecorum) aggravated gut dysbiosis-induced mastitis. Mechanically, sialic acid-facilitated E. cecorum expansion promoted muramyl dipeptide (MDP) release, which induced inflammatory responses by activating the NOD2-RIP2-NF-κB axis. CONCLUSIONS Collectively, our data reveal a role of sialic acid-facilitated postantibiotic pathobiont expansion in gut dysbiosis-associated inflammation, highlighting a potential strategy for disease prevention by regulating the MDP-NOD2-RIP2 axis.
Collapse
Affiliation(s)
- Min Qiu
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China; Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Cong Ye
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Lijuan Bao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Keyi Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Yihong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Xiaotong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Ruibo Tang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Ruping Shang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Shan Shang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Chongshan Yuan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China.
| | - Jun Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, Jilin Province, China.
| | - Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, China.
| |
Collapse
|
2
|
Gouda NA, Alshammari SO, Abourehab MAS, Alshammari QA, Elkamhawy A. Therapeutic potential of natural products in inflammation: underlying molecular mechanisms, clinical outcomes, technological advances, and future perspectives. Inflammopharmacology 2023; 31:2857-2883. [PMID: 37950803 DOI: 10.1007/s10787-023-01366-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/06/2023] [Indexed: 11/13/2023]
Abstract
Chronic inflammation is a common underlying factor in many major diseases, including heart disease, diabetes, cancer, and autoimmune disorders, and is responsible for up to 60% of all deaths worldwide. Metformin, statins, and corticosteroids, and NSAIDs (non-steroidal anti-inflammatory drugs) are often given as anti-inflammatory pharmaceuticals, however, often have even more debilitating side effects than the illness itself. The natural product-based therapy of inflammation-related diseases has no adverse effects and good beneficial results compared to substitute conventional anti-inflammatory medications. In this review article, we provide a concise overview of present pharmacological treatments, the pathophysiology of inflammation, and the signaling pathways that underlie it. In addition, we focus on the most promising natural products identified as potential anti-inflammatory therapeutic agents. Moreover, preclinical studies and clinical trials evaluating the efficacy of natural products as anti-inflammatory therapeutic agents and their pragmatic applications with promising outcomes are reviewed. In addition, the safety, side effects and technical barriers of natural products are discussed. Furthermore, we also summarized the latest technological advances in the discovery and scientific development of natural products-based medicine.
Collapse
Affiliation(s)
- Noha A Gouda
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Ahmed Elkamhawy
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea.
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
3
|
Adepoju FO, Duru KC, Li E, Kovaleva EG, Tsurkan MV. Pharmacological Potential of Betulin as a Multitarget Compound. Biomolecules 2023; 13:1105. [PMID: 37509141 PMCID: PMC10377123 DOI: 10.3390/biom13071105] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Betulin is a natural triterpene, usually from birch bark, known for its potential wound-healing properties. Despite having a wide range of pharmacological targets, no studies have proposed betulin as a multitarget compound. Betulin has protective effects against cardiovascular and liver diseases, cancer, diabetes, oxidative stress, and inflammation. It reduces postprandial hyperglycemia by inhibiting α-amylase and α-glucosidase activity, combats tumor cells by inducing apoptosis and inhibiting metastatic proteins, and modulates chronic inflammation by blocking the expression of proinflammatory cytokines via modulation of the NFκB and MAPKs pathways. Given its potential to influence diverse biological networks with high target specificity, it can be hypothesized that betulin may eventually become a new lead for drug development because it can modify a variety of pharmacological targets. The summarized research revealed that the diverse beneficial effects of betulin in various diseases can be attributed, at least in part, to its multitarget anti-inflammatory activity. This review focuses on the natural sources, pharmacokinetics, pharmacological activity of betulin, and the multi-target effects of betulin on signaling pathways such as MAPK, NF-κB, and Nrf2, which are important regulators of the response to oxidative stress and inflammation in the body.
Collapse
Affiliation(s)
- Feyisayo O Adepoju
- Department of Technology for Organic Synthesis, Chemical Technology Institute, Ural Federal University, Mira 19, 620002 Yekaterinburg, Russia
| | - Kingsley C Duru
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854-8021, USA
| | - Erguang Li
- Medical School, Nanjing University, Nanjing, 22 Hankou Road, Nanjing 210093, China
| | - Elena G Kovaleva
- Department of Technology for Organic Synthesis, Chemical Technology Institute, Ural Federal University, Mira 19, 620002 Yekaterinburg, Russia
| | | |
Collapse
|
4
|
Zou H, Gong Y, Ye H, Yuan C, Li T, Zhang J, Ren L. Dietary regulation of peroxisome proliferator-activated receptors in metabolic syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154904. [PMID: 37267691 DOI: 10.1016/j.phymed.2023.154904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 05/15/2023] [Accepted: 05/28/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) are a class of ligand-activated nuclear transcription factors, members of the type nuclear receptor superfamily, with three subtypes, namely PPARα, PPARβ/δ, and PPARγ, which play a key role in the metabolic syndrome. In the past decades, a large number of studies have shown that natural products can act by regulating metabolic pathways mediated by PPARs. PURPOSE This work summarizes the physiological importance and clinical significance of PPARs and reviews the experimental evidence that natural products mediate metabolic syndrome via PPARs. METHODS This study reviews relevant literature on clinical trials, epidemiology, animals, and cell cultures published in NCBI PubMed, Scopus, Web of Science, Google Scholar, and other databases from 2001 to October 2022. Search keywords were "natural product" OR "botanical" OR "phytochemical" AND "PPAR" as well as free text words. RESULTS The modulatory involvement of PPARs in the metabolic syndrome has been supported by prior research. It has been observed that many natural products can treat metabolic syndrome by altering PPARs. The majority of currently described natural compounds are mild PPAR-selective agonists with therapeutic effects that are equivalent to synthetic medicines but less harmful adverse effects. CONCLUSION PPAR agonists can be combined with natural products to treat and prevent metabolic syndrome. Further human investigations are required because it is unknown how natural products cause harm and how they might have negative impacts.
Collapse
Affiliation(s)
- Haoyang Zou
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Yiyao Gong
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Haiqing Ye
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Cuiping Yuan
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun 130033, China
| | - Tiezhu Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun 130033, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
5
|
Wang XZ, Song XJ, Liu C, Xing C, Wu T, Zhang Y, Su J, Hao JY, Chen XY, Zhang ZY, Li YH, Liu YY. Active components and molecular mechanism of Syringa oblata Lindl. in the treatment of endometritis based on pharmacology network prediction. Front Vet Sci 2022; 9:885952. [PMID: 35937303 PMCID: PMC9355479 DOI: 10.3389/fvets.2022.885952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Antibiotic treatment of endometritis was limited by the inevitable antibiotic residues and risk of bacterial resistance. Therefore, the development of safe and effective strategies for endometritis treatment is urgently needed. Syringa oblata Lindl. (SOL) showed great pharmacological potential against endometritis. However, the active components and underlying mechanism of SOL for endometritis treatment remain indeterminate. In our study, the active components and possible molecular mechanism of SOL against endometritis were predicted through computer data mining and biological networks construction. It was predicted that the main active components of SOL were luteolin, kaempferol, oleanolic acid, and rutin, and their anti-endometritis effect was mainly attributed to the TLRs/NF-κB signaling pathway. Furthermore, a green and efficient deep eutectic solvent combined with ultrasound-assisted extraction (DES-UAE) was performed and optimized to obtain high contents of total flavonoid, rutin, and luteolin. The four predicted active components in the SOL extracts were qualitatively and quantitatively analyzed by LC/MS and HPLC. Finally, the pharmacological effects of SOL and active components have been verified by Staphylococcus aureus-endometritis models in mice. H&E staining and bacterial load in uterus tissues assays initially validated the pharmacodynamic effects of SOL, and quantitative real-time PCR (RT-qPCR) and ELISA results confirmed that SOL and four active components could ameliorate the uterus injury caused by Staphylococcus aureus, the mechanism of action is related to the TLRs/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiao-Zhen Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xue-Jiao Song
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chang Liu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chen Xing
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Tong Wu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yue Zhang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jing Su
- Heilongjiang Animal Disease Prevention and Control Center, Harbin, China
| | - Jing-You Hao
- Harbin Lvda Sheng Animal Medicine Manufacture Co., Ltd., Harbin, China
| | - Xue-Ying Chen
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhi-Yun Zhang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yan-Hua Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- *Correspondence: Yan-Yan Liu
| | - Yan-Yan Liu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Yan-Hua Li
| |
Collapse
|
6
|
Single-Chain Fragment Variables Targeting Leukocidin ED Can Alleviate the Inflammation of Staphylococcus aureus-Induced Mastitis in Mice. Int J Mol Sci 2021; 23:ijms23010334. [PMID: 35008761 PMCID: PMC8745144 DOI: 10.3390/ijms23010334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus aureus is a vital bovine mastitis pathogen causing huge economic losses to the dairy industry worldwide. In our previous studies, leukotoxin ED (LukED) was detected in most S. aureus strains isolated from bovine mastitis. Here, four single-chain fragment variables (scFvs) (ZL8 and ZL42 targeting LukE, ZL22 and ZL23 targeting LukD) were obtained using purified LukE and LukD proteins as the antigens after five rounds of bio-panning. The complementarity-determining region 3 (CDR3) of the VH domain of these scFvs exhibited significant diversities. In vitro, the scFvs significantly decreased LukED-induced cell killing by inhibiting the binding of LukED to chemokine receptors (CCR5 and CXCR2) and reduced the death rates of bovine neutrophils and MAC-T cells caused by LukED and S. aureus (p < 0.05). In an S. aureus-induced mouse mastitis model, histopathology and MPO results revealed that scFvs ameliorated the histopathological damages and reduced the infiltration of inflammatory cells (p < 0.05). The ELISA and qPCR assays showed that scFvs reduced the transcription and expression levels of Tumor Necrosis Factor-alpha (TNF-α), interleukin-1β (IL-1β), IL-6, IL-8 and IL-18 (p < 0.05). The overall results demonstrated the protective anti-inflammatory effect of scFvs in vitro and in vivo, enlightening the potential role of scFvs in the prevention and treatment of S. aureus-induced mastitis.
Collapse
|
7
|
Anti- Staphylococcus aureus Single-Chain Fragment Variables Play a Protective Anti-Inflammatory Role In Vitro and In Vivo. Vaccines (Basel) 2021; 9:vaccines9111300. [PMID: 34835231 PMCID: PMC8618225 DOI: 10.3390/vaccines9111300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is a causative agent of bovine mastitis, capable of causing significant economic losses to the dairy industry worldwide. This study focuses on obtaining single-chain fragment variables (scFvs) against the virulence factors of S. aureus and evaluates the protective effect of scFvs on bovine mammary epithelial (MAC-T) cells and mice mammary gland tissues infected by S. aureus. After five rounds of bio-panning, four scFvs targeting four virulence factors of S. aureus were obtained. The complementarity-determining regions (CDRs) of these scFvs exhibited significant diversities, especially CDR3 of the VH domain. In vitro, each of scFvs was capable of inhibiting S. aureus growth and reducing the damage of MAC-T cells infected by S. aureus. Preincubation of MAC-T cells with scFvs could significantly attenuate the effect of apoptosis and necrosis compared with the negative control group. In vivo, the qPCR and ELISA results demonstrated that scFvs reduced the transcription and expression of Tumor Necrosis Factor alpha (TNF-α), interleukin-1β (IL-1β), IL-6, IL-8, and IL-18. Histopathology and myeloperoxidase (MPO) results showed that scFvs ameliorated the histopathological damages and reduced the inflammatory cells infiltration. The overall results demonstrated the positive anti-inflammatory effect of scFvs, revealing the potential role of scFvs in the prevention and treatment of S. aureus infections.
Collapse
|
8
|
Ma X, Song M, Yan Y, Ren G, Hou J, Qin G, Wang W, Li Z. Albiflorin alleviates cognitive dysfunction in STZ-induced rats. Aging (Albany NY) 2021; 13:18287-18297. [PMID: 34319254 PMCID: PMC8351685 DOI: 10.18632/aging.203274] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/14/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND To explore the effect of albiflorin (AL) on streptozotocin (STZ)-induced Alzheimer's disease (AD) in rats. METHODS A mouse model of diabetic encephalopathy was established by intraperitoneal injection of 1%STZ. Step down test and water maze test were used to test the cognitive function of rats. Congo Red Staining was used to detect the distribution of Aβ plaques in the hippocampus of rats. Cytokine levels in serum and hippocampus were measured using ELISA. Serum insulin, oral glucose tolerance (OGTT), serum superoxide dismutase (SOD) activity and malondialdehyde (MDA) content were measured by commercial kits. And the content of Nrf-2/HO-1/HMGB1/NF-kB in the hippocampus of diabetic rats were detected by western blot. RESULTS AND CONCLUSION Compared with the STZ model group, the average escape latency of rats in the AL group in the Morris water maze test was significantly shortened, and the average number of platform crossings and the ratio of distance/total swimming distance in the target quadrant were increased significantly. Staining of tissue sections and ELISA showed a decrease in Aβ plaque density in the hippocampus of rats in the AL group. And serum insulin levels of rats in the ALgroup were significantly reduced and OGTT was improved. In addition, AL could also regulate the Nrf-2/HO-1/HMGB1/NF-kB signal pathway in the hippocampus. Therefore, AL may ameliorate STZ-induced cognitive impairment in rats by regulating oxidative stress and inflammation in the brain.
Collapse
Affiliation(s)
- Xiaojun Ma
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Min Song
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yushan Yan
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Gaofei Ren
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jingwen Hou
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Guijun Qin
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Zhizhen Li
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
9
|
Akhtar M, Shaukat A, Zahoor A, Chen Y, Wang Y, Yang M, Umar T, Guo M, Deng G. Hederacoside-C Inhibition of Staphylococcus aureus-Induced Mastitis via TLR2 & TLR4 and Their Downstream Signaling NF-κB and MAPKs Pathways In Vivo and In Vitro. Inflammation 2021; 43:579-594. [PMID: 31845052 DOI: 10.1007/s10753-019-01139-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Hederacoside-C (HDC) is a biological active ingredient, extracted from the leaves of Hedera helix. It has been reported to have anti-inflammatory properties. However, the effects of HDC on Staphylococcus aureus (S. aureus)-induced mastitis have not been reported yet. Here, we evaluated the anti-inflammatory effects of HDC on S. aureus-induced mastitis both in vivo on mammary gland tissues and in vitro on RAW 264.7 cells. The ascertained histopathological changes and MPO activity revealed that HDC defended mammary glands from tissue destruction and inflammatory cell infiltration induced by S. aureus. The results of ELISA, western blot, and qRT-PCR indicated that HDC significantly inhibited the expressions IL-6, IL-1β, and TNF-α and enhanced the IL-10 by downregulating and upregulating their relevant genes, respectively. Furthermore, HDC markedly suppressed the TLR2 and TLR4 expressions by attenuating the MAPKs (p38, ERK, JNK) and NF-κB (p65 and IκBα) pathways followed by decreasing the phosphorylation of p38, ERK, JNK, p65, and IκBα. The above parameters enhanced the mammary gland defense and reduced inflammation. These findings suggested that HDC may have the potential to be an effective anti-inflammatory drug for the S. aureus-induced mice mastitis and in RAW 264.7 cells.
Collapse
Affiliation(s)
- Muhammad Akhtar
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Arshad Zahoor
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ying Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Mei Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Talha Umar
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Mengyao Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
10
|
Zahoor A, Yang Y, Yang C, Akhtar M, Guo Y, Shaukat A, Guo MY, Deng G. Gas6 negatively regulates the Staphylococcus aureus-induced inflammatory response via TLR signaling in the mouse mammary gland. J Cell Physiol 2020; 235:7081-7093. [PMID: 32052456 DOI: 10.1002/jcp.29604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Staphylococcus aureus (S. aureus)-induced mastitis is the most frequent, pathogenic, and prevalent infection of the mammary gland. The ligand growth arrest-specific 6 (Gas6) is a secretory protein that binds to and activates Tyro3, Axl, and MerTK receptors. This study explored the role of Gas6 in S. aureus-induced mastitis. Our results revealed that TLR receptors initiate the innate immune response in mammary gland tissues and epithelial cells and that introducing S. aureus activates TLR2 and TLR6 to drive multiple intracellular mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) pathways. Moreover, S. aureus also induces Gas6, which then activates the TAM receptor kinase pathway, which is related to the inhibition of TLR2- and TLR6-mediated inflammatory pathways through SOCS1 and SOCS3 induction. Gas6 absence alone was found to be involved in the downregulation of TAM receptor-mediated anti-inflammatory effects by inducing significantly prominent expression of TRAF6 and low protein and messenger RNA expression of SOCS1 and SOCS3. S. aureus-induced MAPK and NF-ĸB p65 phosphorylation were also dependent on Gas6, which negatively regulated the production of Pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) in S. aureus-treated mammary tissues and mammary epithelial cells. Our in vivo and in vitro study uncovered the Gas6-mediated negative feedback mechanism, which inhibits TLR2- and TLR6-mediated MAPK and NF-ĸB signaling by activating TAM receptor kinase (MerTK, Axl, and Tyro3) through the induction of SOCS1/SOCS3 proteins.
Collapse
Affiliation(s)
- Arshad Zahoor
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Chao Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Muhammad Akhtar
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Yingfang Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Meng-Yao Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
11
|
Li J, Jiang B, Chen C, Fan B, Huang H, Chen G. Biotransformation of betulin by Mucor subtilissimus to discover anti-inflammatory derivatives. PHYTOCHEMISTRY 2019; 166:112076. [PMID: 31351331 DOI: 10.1016/j.phytochem.2019.112076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 06/10/2023]
Abstract
Biotransformation of lupane-type triterpenoid betulin was carried out with Mucor subtilissimus CGMCC 3.2456. Yielded nine previously undescribed hydroxylated compounds. M. subtilissimus biotransformation provided C-7, C-11, C-15 and C-24 hydroxylated compounds along with C-7 oxidized and C-28 acetylated derivatives. The structures of the metabolites were established based on extensive NMR and HR-ESI-MS data analyses. Furthermore, we found that most of the metabolites exhibited pronounced inhibitory activities on lipopolysaccharides-induced NO production in RAW264.7 cells.
Collapse
Affiliation(s)
- Jianlin Li
- School of Pharmacy, Nantong University, Nantong, China
| | | | - Chen Chen
- School of Pharmacy, Nantong University, Nantong, China
| | - Boyi Fan
- School of Pharmacy, Nantong University, Nantong, China
| | - Huilian Huang
- Key Laboratory of Modern Preparation of TCM, Jiangxi University of Traditional Chinese Medicine, Ministry of Education, Nanchang, China
| | | |
Collapse
|
12
|
Tsai CW, Tsai RT, Liu SP, Chen CS, Tsai MC, Chien SH, Hung HS, Lin SZ, Shyu WC, Fu RH. Neuroprotective Effects of Betulin in Pharmacological and Transgenic Caenorhabditis elegans Models of Parkinson's Disease. Cell Transplant 2018; 26:1903-1918. [PMID: 29390878 PMCID: PMC5802634 DOI: 10.1177/0963689717738785] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common degenerative disorder of the central nervous system in the elderly. It is characterized by progressive loss of dopaminergic neurons in the substantia nigra pars compacta, as well as by motor dysfunction. Although the causes of PD are not well understood, aggregation of α-synuclein (α-syn) in neurons contributes to this disease. Current therapeutics for PD provides satisfactory symptom relief but not a cure. Treatment strategies include attempts to identify new drugs that will prevent or arrest the progressive course of PD by correcting disease-specific pathogenic process. Betulin is derived from the bark of birch trees and possesses anticancer, antimicrobial, and anti-inflammatory properties. The aim of the present study was to evaluate the potential for betulin to ameliorate PD features in Caenorhabditis elegans (C. elegans) models. We demonstrated that betulin diminished α-syn accumulation in the transgenic C. elegans model. Betulin also reduced 6-hydroxydopamine-induced dopaminergic neuron degeneration, reduced food-sensing behavioral abnormalities, and reversed life-span decreases in a pharmacological C. elegans model. Moreover, we found that the enhancement of proteasomes activity by promoting rpn1 expression and downregulation of the apoptosis pathway gene, egl-1, may be the molecular mechanism for betulin-mediated protection against PD pathology. Together, these findings support betulin as a possible treatment for PD and encourage further investigations of betulin as an antineurodegenerative agent.
Collapse
Affiliation(s)
- Chia-Wen Tsai
- 1 Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Rong-Tzong Tsai
- 2 Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Shih-Ping Liu
- 3 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,4 Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Chang-Shi Chen
- 5 Department of Biochemical and Molecular Biology, National Cheng Kung University, Tainan, Taiwan
| | - Min-Chen Tsai
- 3 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shao-Hsuan Chien
- 3 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Huey-Shan Hung
- 3 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,4 Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- 6 Bioinnovation Center, Tzu Chi foundation, Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan
| | - Woei-Cherng Shyu
- 3 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,4 Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Ru-Huei Fu
- 3 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,4 Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan.,7 Department of Psychology, Asia University, Taichung, Taiwan
| |
Collapse
|
13
|
Betulin inhibits lipopolysaccharide/D-galactosamine-induced acute liver injury in mice through activating PPAR-γ. Biomed Pharmacother 2018; 106:941-945. [DOI: 10.1016/j.biopha.2018.07.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/27/2018] [Accepted: 07/01/2018] [Indexed: 12/11/2022] Open
|
14
|
Luteoloside Protects the Uterus from Staphylococcus aureus-Induced Inflammation, Apoptosis, and Injury. Inflammation 2018; 41:1702-1716. [DOI: 10.1007/s10753-018-0814-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
15
|
Zhang W, Jiang H, Jin M, Wang Q, Sun Q, Du Y, Cao L, Xu H. UHPLC-Q-TOF-MS/MS based screening and identification of the metabolites in vivo after oral administration of betulin. Fitoterapia 2018; 127:29-41. [DOI: 10.1016/j.fitote.2018.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/09/2018] [Accepted: 04/12/2018] [Indexed: 11/25/2022]
|
16
|
Luteolin reduces inflammation in Staphylococcus aureus-induced mastitis by inhibiting NF-kB activation and MMPs expression. Oncotarget 2018; 8:28481-28493. [PMID: 28415707 PMCID: PMC5438666 DOI: 10.18632/oncotarget.16092] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/02/2017] [Indexed: 11/29/2022] Open
Abstract
Mastitis is a serious and prevalent disease caused by infection by pathogens such as Staphylococcus aureus. We evaluated the anti-inflammatory effects and mechanism of luteolin, a natural flavonoid with a wide range of pharmacological activities, in a mouse model of S. aureus mastitis. We also treated cultured mouse mammary epithelial cells (mMECs) with S. aureus and luteolin. Histopathological changes were examined by H&E staining and the levels of inflammatory cytokine proteins were analyzed using ELISAs. We determined mRNA levels with qPCR and the level of NF-κB and matrix metalloproteinase (MMP) proteins by Western blotting. The observed histopathological changes showed that luteolin protected mammary glands with S. aureus infection from tissue destruction and inflammatory cell infiltration. Luteolin inhibited the expression of TNF-α, IL-1β, and IL-6, all of which were increased with S. aureus infection of mammary tissues and mMECs. S. aureus-induced TLR2 and TLR4 was suppressed by luteolin, as were levels of IκBα and NF-κB p65 phosphorylation and expression of MMP-2 and MMP-9. Levels of tissue inhibitor of metalloproteinases (TIMP)-1 and TIMP-2 were enhanced. These findings suggest luteolin is a potentially effective new treatment to reduce tissue damage and inflammation from S. aureus-induced mastitis.
Collapse
|
17
|
Pozharitskaya ON, Karlina MV, Shikov AN, Kosman VM, Makarov VG, Casals E, Rosenholm JM. Pharmacokinetics and Tissue Disposition of Nanosystem-Entrapped Betulin After Endotracheal Administration to Rats. Eur J Drug Metab Pharmacokinet 2017; 42:327-332. [PMID: 27155877 DOI: 10.1007/s13318-016-0340-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Betulin is a triterpene extracted from the cork layer of the outer bark of Betula spp. It has a wide spectrum of pharmacological activities, including being lung protective; however, its bioavailability is low. To increase its bioavailability, betulin was entrapped in a nanosystem (BN). In this study, we investigated the pharmacokinetics and tissue distribution of nanosystem-entrapped betulin after single dose endotracheal administration to rats. METHOD Betulin was nanosystem-entrapped using a solvent exchange technique. The surface morphology and size of the nanosystem were characterized by transmission electron microscopy and dynamic light scattering. The plasma and tissue concentrations of betulin were determined using a validated high-performance liquid chromatography method. RESULTS The highest concentration of betulin was found in lungs and liver, and the lowest in the heart. Betulin did not penetrate highly vascularized tissues or tissue with an average degree of vascularization, nor did it cross the blood-brain barrier. Tissue availability in the lungs was 1.3 times higher for BN than for free betulin. Betulin was detected in the bloodstream at 15 min after administration of BN compared with only at 1 h after administration of free betulin. Penetration of betulin in the liver tissue was characterized by a high degree of intensity both for BN and free betulin. Betulin in the heart tissue was detected in much smaller quantities than in the liver. CONCLUSION Entrapment of betulin in nanosystem form shows promise as a novel strategy in the treatment of pulmonary diseases.
Collapse
Affiliation(s)
- Olga N Pozharitskaya
- Saint Petersburg Institute of Pharmacy, Leningrad Region, Vsevolozhsky District, 188663, Kuzmolovo P 245, Russia
| | - Marina V Karlina
- Saint Petersburg Institute of Pharmacy, Leningrad Region, Vsevolozhsky District, 188663, Kuzmolovo P 245, Russia
| | - Alexander N Shikov
- Saint Petersburg Institute of Pharmacy, Leningrad Region, Vsevolozhsky District, 188663, Kuzmolovo P 245, Russia.
| | - Vera M Kosman
- Saint Petersburg Institute of Pharmacy, Leningrad Region, Vsevolozhsky District, 188663, Kuzmolovo P 245, Russia
| | - Valery G Makarov
- Saint Petersburg Institute of Pharmacy, Leningrad Region, Vsevolozhsky District, 188663, Kuzmolovo P 245, Russia
| | - Eudald Casals
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity (3rd Floor), Tykistökatu 6A, 20520, Turku, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity (3rd Floor), Tykistökatu 6A, 20520, Turku, Finland
| |
Collapse
|
18
|
Ci X, Zhou J, Lv H, Yu Q, Peng L, Hua S. Betulin exhibits anti-inflammatory activity in LPS-stimulated macrophages and endotoxin-shocked mice through an AMPK/AKT/Nrf2-dependent mechanism. Cell Death Dis 2017; 8:e2798. [PMID: 28518138 PMCID: PMC5520743 DOI: 10.1038/cddis.2017.39] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 12/14/2022]
Abstract
Continued oxidative stress can lead to chronic inflammation, which in turn could mediate most chronic diseases including cancer. Nuclear factor erythroid 2-related factor (Nrf2), a critical transcriptional activator for antioxidative responses, has envolved to be an attractive drug target for the treatment or prevention of human diseases. In the present study, we investigated the effects and mechanisms of betulin on Nrf2 activation and its involvement in the lipopolysaccharide (LPS)-triggered inflammatory system. In macrophages, betulin activated the Nrf2 signaling pathway and increased Nrf2-targeted antioxidant and detoxifying enzymes, including NADPH, quinine oxidoreductase 1 (NQO1), heme oxygenase-1 (HO-1), γ-glutamyl cysteine synthetase catalytic subunit (GCLC) and modifier subunit (GCLM) in a dose and time dependent manner. Importantly, we found betulin-induced activation of Nrf2 is AMPK/AKT/GSK3β dependent, as pharmacologically inactivating AMPK blocked the activating effect of betulin on AKT, GSK3β and Nrf2. Furthermore, betulin attenuated LPS-induced inflammatory mediators (iNOS and COX-2) and MAPK inflammatory signaling pathway. The effect of betulin on HO-1 and NQO1 upregulation, iNOS and COX-2 the downregulation, and survival time extension was largely weakened when Nrf2 was depleted in vitro and in vivo. Our results demonstrate that the AMPK/AKT/Nrf2 pathways are essential for the anti-inflammatory effects of betulin in LPS-stimulated macrophages and endotoxin-shocked mice.
Collapse
Affiliation(s)
- Xinxin Ci
- Institute of Translational Medicine, Department of Respiratory Medicine, The First Hospital, Jilin University, Changchun 130001, China
| | - Junfeng Zhou
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Hongming Lv
- Institute of Translational Medicine, Department of Respiratory Medicine, The First Hospital, Jilin University, Changchun 130001, China
| | - Qinlei Yu
- Institute of Translational Medicine, Department of Respiratory Medicine, The First Hospital, Jilin University, Changchun 130001, China
| | - Liping Peng
- Institute of Translational Medicine, Department of Respiratory Medicine, The First Hospital, Jilin University, Changchun 130001, China
| | - Shucheng Hua
- Institute of Translational Medicine, Department of Respiratory Medicine, The First Hospital, Jilin University, Changchun 130001, China
| |
Collapse
|
19
|
Jiang KF, Zhao G, Deng GZ, Wu HC, Yin NN, Chen XY, Qiu CW, Peng XL. Polydatin ameliorates Staphylococcus aureus-induced mastitis in mice via inhibiting TLR2-mediated activation of the p38 MAPK/NF-κB pathway. Acta Pharmacol Sin 2017; 38:211-222. [PMID: 27890916 DOI: 10.1038/aps.2016.123] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/18/2016] [Indexed: 01/01/2023]
Abstract
Recent studies show that Polydatin (PD) extracted from the roots of Polygonum cuspidatum Sieb, a widely used traditional Chinese remedies, possesses anti-inflammatory activity in several experimental models. In this study, we investigated the anti-inflammatory effects of PD on Staphylococcus aureus-induced mastitis in mice and elucidated the potential mechanisms. In mice with S aureus-induced mastitis, administration of PD (15, 30, 45 mg/kg, ip) or dexamethasone (Dex, 5 mg/kg, ip) significantly suppressed the infiltration of inflammatory cells, ameliorated the mammary structural damage, and inhibited the activity of myeloperoxidase, a biomarker of neutrophils accumulation. Furthermore, PD treatment dose-dependently decreased the levels of TNF-α, IL-1β, IL-6 and IL-8 in the mammary gland tissues. PD treatment also dose-dependently decreased the expression of TLR2, MyD88, IRAK1, IRAK4 and TRAF6 as well as the phosphorylation of TAK1, MKK3/6, p38 MAPK, IκB-α and NF-κB in the mammary gland tissues. In mouse mammary epithelial cells (mMECs) infected by S aureus in vitro, pretreatment with PD dose-dependently suppressed the upregulated pro-inflammatory cytokines and signaling proteins, and the nuclear translocation of NF-κB p65 and AP-1. A TLR2-neutralizing antibody mimicked PD in its suppression on S aureus-induced upregulation of MyD88, p-p38 and p-p65 levels in mMECs. PD (50, 100 μg/mL) affected neither the growth of S aureus in vitro, nor the viability of mMECs. In conclusion, PD does not exhibit antibacterial activity against S aureus, its therapeutic effects in mouse S aureus-induced mastitis depend on its ability to down-regulate pro-inflammatory cytokine levels via inhibiting TLR2-mediated activation of the p38 MAPK/NF-κB signaling pathway.
Collapse
|
20
|
Ra HJ, Lee HJ, Jo HS, Nam DC, Lee YB, Kang BH, Moon DK, Kim DH, Lee CJ, Hwang SC. Betulin suppressed interleukin-1β-induced gene expression, secretion and proteolytic activity of matrix metalloproteinase in cultured articular chondrocytes and production of matrix metalloproteinase in the knee joint of rat. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 21:19-26. [PMID: 28066137 PMCID: PMC5214907 DOI: 10.4196/kjpp.2017.21.1.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/31/2022]
Abstract
We investigated whether betulin affects the gene expression, secretion and proteolytic activity of matrix metalloproteinase-3 (MMP-3) in primary cultured rabbit articular chondrocytes, as well as in vivo production of MMP-3 in the rat knee joint to evaluate the potential chondroprotective effect of betulin. Rabbit articular chondrocytes were cultured and reverse transcription-polymerase chain reaction (RT-PCR) was used to measure interleukin-1β (IL-1β)-induced gene expression of MMP-3, MMP-1, MMP-13, a disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS-4), ADAMTS-5 and type II collagen. Effect of betulin on IL-1β-induced secretion and proteolytic activity of MMP-3 was investigated using western blot analysis and casein zymography, respectively. Effect of betulin on MMP-3 protein production was also examined in vivo. The results were as follows: (1) betulin inhibited the gene expression of MMP-3, MMP-1, MMP-13, ADAMTS-4, and ADAMTS-5, but increased the gene expression of type II collagen; (2) betulin inhibited the secretion and proteolytic activity of MMP-3; (3) betulin suppressed the production of MMP-3 protein in vivo. These results suggest that betulin can regulate the gene expression, secretion, and proteolytic activity of MMP-3, by directly acting on articular chondrocytes.
Collapse
Affiliation(s)
- Ho Jong Ra
- Department of Orthopedic Surgery, Gangneung Asan Hospital, College of Medicine, University of Ulsan, Gangneung 25440, Korea
| | - Hyun Jae Lee
- Department of Health Management, Sahmyook University, Seoul 01795, Korea
| | - Ho Seung Jo
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju 52727, Korea
| | - Dae Cheol Nam
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju 52727, Korea
| | - Young Bok Lee
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju 52727, Korea
| | - Byeong Hun Kang
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju 52727, Korea
| | - Dong Kyu Moon
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju 52727, Korea
| | - Dong Hee Kim
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju 52727, Korea
| | - Choong Jae Lee
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Sun-Chul Hwang
- Department of Orthopedic Surgery and Institute of Health Sciences, School of Medicine and Hospital, Gyeongsang National University, Jinju 52727, Korea
| |
Collapse
|
21
|
Ma C, Long H. Protective effect of betulin on cognitive decline in streptozotocin (STZ)-induced diabetic rats. Neurotoxicology 2016; 57:104-111. [PMID: 27640960 DOI: 10.1016/j.neuro.2016.09.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/13/2016] [Accepted: 09/14/2016] [Indexed: 12/14/2022]
Abstract
Betulin is extracted from birch tree bark and exerts diverse pharmacological activities. The present study was designed to investigate the protective effect of betulin (BE) on cognitive decline in streptozotocin (STZ)-induced diabetic rats. The diabetic model was built by streptozotocin (STZ) (30mg/kg, ip). After 4 weeks, the diabetic rats were treated with vehicle or BE (20mg/kg, 40mg/kg) for 4 weeks. The oral glucose tolerance (OGTT) and serum insulin were detected. Three days later, Morris water maze (MWM) test was used to evaluate memory function. Superoxide dismutase (SOD) activity and malondialdehyde (MDA) content in hippocampus were examined. Inflammatory cytokines including interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) in serum and hippocampus were measured. The protein expressions of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1) and NF-κB pathways-related molecules in hippocampus were examined. As a results, BE could improve glucose intolerance and modify basal learning performance. Treatment with BE significantly restored SOD activity and decreased MDA content in hippocampus. BE also markedly reduced the contents of inflammatory cytokines in serum and hippocampus. Furthermore, administration of BE effectively upregulated the expressions of Nrf2, HO-1 and blocked the phosphorylations of IκB, NF-κB. In summary, BE might exhibit protective effect on cognitive decline in STZ-induced diabetic rats through HO-1/Nrf-2/NF-κB pathway.
Collapse
Affiliation(s)
- Chunhua Ma
- Central Laboratory, Nanjing Municipal Hospital of T.C.M., The Third Affiliated Hospital of Nanjing University of T.C.M., Nanjing 210001, China
| | - Hongyan Long
- Central Laboratory, Nanjing Municipal Hospital of T.C.M., The Third Affiliated Hospital of Nanjing University of T.C.M., Nanjing 210001, China.
| |
Collapse
|
22
|
Chunhua M, Long H, Zhu W, Liu Z, Jie R, Zhang Y, Wang Y. Betulin inhibited cigarette smoke-induced COPD in mice. Biomed Pharmacother 2016; 85:679-686. [PMID: 27899253 DOI: 10.1016/j.biopha.2016.11.079] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/03/2023] Open
Abstract
The purpose of the present study was to evaluate the protective effect of betulin (BE) on CS (cigarette smoke)-induced COPD in mice and explore its underlying mechanisms. 60 male ICR mice were randomly assigned to five groups: control group, model group, dexamethasone (2mg/kg) group, BE (20mg/kg) group and BE (40mg/kg) group. The COPD mice were induced by cigarette smoke exposure for 8 weeks. The result of H&E staining demonstrated that BE inhibited CS-induced pathological injury in lung tissue. Besides, BE could restore the activities of superoxide dismutase (SOD) in serum and in lung, catalase (CAT) in serum and reduce the content of malondialdehyde (MDA) in serum and in lung. BE also inhibited the overproductions of pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β). Furthermore, the administration of BE significantly inhibited the protein expression of ROCK/NF-κB pathway in CS-induced mice. Our findings suggested that BE might effectively ameliorate the progression of COPD via ROCK/NF-κB pathway in mice.
Collapse
Affiliation(s)
- Ma Chunhua
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China
| | - Hongyan Long
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China.
| | - Weina Zhu
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China
| | - Zheng Liu
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China
| | - Ruan Jie
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China
| | - Yajie Zhang
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China
| | - Yarui Wang
- Central Laboratory, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China
| |
Collapse
|
23
|
IFN-τ inhibits S. aureus-induced inflammation by suppressing the activation of NF-κB and MAPKs in RAW 264.7 cells and mice with pneumonia. Int Immunopharmacol 2016; 35:332-340. [DOI: 10.1016/j.intimp.2016.02.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 01/09/2023]
|
24
|
Zhao G, Wu H, Jiang K, Chen X, Wang X, Qiu C, Guo M, Deng G. The Anti-Inflammatory Effects of Interferon Tau by Suppressing NF-κB/MMP9 in Macrophages Stimulated with Staphylococcus aureus. J Interferon Cytokine Res 2016; 36:516-24. [PMID: 27142785 DOI: 10.1089/jir.2015.0170] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Previous studies have reported that interferon tau (IFNT) significantly mitigates tissue inflammation. However, this effect and its regulating pathways have not been reported for Staphylococcus aureus-induced inflammation. In this study, RAW 264.7 cells stimulated with S. aureus were used to identify the anti-inflammatory effects and mechanism of IFNT. First, IFNT was found to be noncytotoxic to macrophages treated with the high dose of 200 ng/mL IFNT. ELISA and qPCR revealed that IFNT decreased the expression of proinflammatory cytokines such as TNF-α, IL-1β, and IL-6. TLR2, which is involved in the immune response during S. aureus infection, directly affected NF-κB pathway activation and was also downregulated by IFNT. Subsequent Western blotting showed that the phosphorylation of IκBα and NF-κB p65 was inhibited by IFNT. Therefore, although the MMP9 levels were significantly downregulated in a dose-dependent manner by IFNT, little change in MMP2 was observed in S. aureus-stimulated RAW 264.7 cells. Furthermore, PDTC, an inhibitor of NF-κB, also significantly decreased MMP9 levels by inhibiting NF-κB p65 activation. All of these findings strongly suggested that IFNT suppresses the NF-κB/MMP9 signal transduction pathway and subsequently exerts its anti-inflammatory effects in S. aureus-stimulated RAW 264.7 cells.
Collapse
Affiliation(s)
- Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan, People's Republic of China
| | - Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan, People's Republic of China
| | - Xiuying Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan, People's Republic of China
| | - Xiaoyan Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan, People's Republic of China
| | - Changwei Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan, People's Republic of China
| | - Mengyao Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan, People's Republic of China
| |
Collapse
|