1
|
Sun J, Lu L, Lian Y, Xu S, Zhu Y, Wu Y, Lin Q, Hou J, Li Y, Yu Z. Sodium butyrate attenuates microglia-mediated neuroinflammation by modulating the TLR4/MyD88/NF-κB pathway and microbiome-gut-brain axis in cardiac arrest mice. Mol Brain 2025; 18:13. [PMID: 39962509 PMCID: PMC11834616 DOI: 10.1186/s13041-025-01179-w] [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: 12/02/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Cardiac arrest (CA) is one of the most common illnesses worldwide. Post-CA brain injury (PCABI) is a major cause of death and poor recovery in CA patients and the current CA treatments are not very effective. The microbiome-gut-brain axis has been found to significantly affect brain ischemia injury. Furthermore, in ischemic stroke patients, short-chain fatty acids (SCFA), especially sodium butyrate (SB), have been observed to promote neuroprotective effects by modulating inflammatory response and microglial polarization in the cortex. However, the precise mechanism of SB on CA-induced injury remains elusive. Therefore, this research study established an oxygen-glucose deprivation and reoxygenation (OGD/R) model using BV-2 microglial and HT22 cells to simulate cerebral ischemia/reperfusion injury in vitro and a potassium chloride-induced CA mouse model to mimic CA in vivo. The data revealed that SB markedly improved neurological scores and reduced neuronal death and apoptosis. Moreover, it reduced M1 microglia and neuroinflammation in CA mice. In addition, SB increased intestinal integrity and alleviated systemic inflammation. The 16S rDNA sequencing analysis indicated that SB intervention mitigated CA-induced gut microbiota dysbiosis and SCFA depletion. It was also observed that CA mice's brain and OGD/R-exposed BV2 cells had substantially increased levels of MyD88, phosphorylated NF-κB p65, and TLR4 proteins, which were reduced after SB treatment. In summary, this study revealed that SB can protect against cerebral ischemia-reperfusion injury by controlling microglia polarization and microbiome-gut-brain axis to inhibit brain inflammation via the TLR4/MyD88/NF-κB pathway.
Collapse
Affiliation(s)
- Jianfei Sun
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liping Lu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
| | - Yingtao Lian
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
| | - Ying Zhu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yanping Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianhui Lin
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Hou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yinping Li
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430060, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China.
| |
Collapse
|
2
|
Yu H, Ren K, Jin Y, Zhang L, Liu H, Huang Z, Zhang Z, Chen X, Yang Y, Wei Z. Mitochondrial DAMPs: Key mediators in neuroinflammation and neurodegenerative disease pathogenesis. Neuropharmacology 2025; 264:110217. [PMID: 39557152 DOI: 10.1016/j.neuropharm.2024.110217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are increasingly linked to mitochondrial dysfunction and neuroinflammation. Central to this link are mitochondrial damage-associated molecular patterns (mtDAMPs), including mitochondrial DNA, ATP, and reactive oxygen species, released during mitochondrial stress or damage. These mtDAMPs activate inflammatory pathways, such as the NLRP3 inflammasome and cGAS-STING, contributing to the progression of neurodegenerative diseases. This review delves into the mechanisms by which mtDAMPs drive neuroinflammation and discusses potential therapeutic strategies targeting these pathways to mitigate neurodegeneration. Additionally, it explores the cross-talk between mitochondria and the immune system, highlighting the complex interplay that exacerbates neuronal damage. Understanding the role of mtDAMPs could pave the way for novel treatments aimed at modulating neuroinflammation and slowing disease progression, ultimately improving patient outcome.
Collapse
Affiliation(s)
- Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Zhen Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Ziheng Zhang
- College of Life Sciences, Xinjiang University, Urumqi, Xinjiang, 830046, PR China
| | - Xing Chen
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
3
|
Kim HS, Ahn JW, Park JY, Joo SS. Identification, characterization, and anti-inflammatory activity of a lipocalin-like protein cloned from Oenanthe javanica. Food Sci Biotechnol 2025; 34:721-731. [PMID: 39958172 PMCID: PMC11822164 DOI: 10.1007/s10068-024-01700-2] [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: 03/26/2024] [Revised: 08/08/2024] [Accepted: 08/23/2024] [Indexed: 02/18/2025] Open
Abstract
This study investigated the anti-inflammatory effects of OJlipo1, a lipocalin-like protein derived from Oenanthe javanica. Through cloning and expressing OJlipo1 in E. coli, and subsequent rigorous characterization including amino acid analysis and mass spectrometry, its potential against inflammation was evaluated. Studies on lipopolysaccharide-stimulated RAW 264.7 cells highlighted its capability to suppress nitric oxide synthase and cyclooxygenase-2 expression, as well as its interference with nuclear factor kappa B and mitogen-activated protein kinase pathways, which are essential for toll-like receptor 4 (TLR4) signaling. Utilizing TAK242, a TLR4 pathway inhibitor, reinforced OJlipo1's specific targeting mechanism. These findings underscore OJlipo1's significant anti-inflammatory potential, aligning with the traditional uses of O. javanica, and suggest new therapeutic avenues, especially for diseases associated with TLR4 dysregulation. This validates the traditional application of O. javanica in inflammation and positions OJlipo1 as a promising therapeutic candidate, enriching our understanding of its molecular underpinnings and therapeutic prospects.
Collapse
Affiliation(s)
- Hyun Soo Kim
- Department of Marine Life Science, College of Life Science, Gangneung-Wonju National University, Gangneung, 25457 Gangwon Republic of Korea
| | - Jeong Won Ahn
- Department of Marine Life Science, College of Life Science, Gangneung-Wonju National University, Gangneung, 25457 Gangwon Republic of Korea
| | - Jung Youl Park
- Glocal University Project Group, Andong National University, 1375 Gyeongdong-ro, Andong, Gyeongsangbuk-do 36729 Republic of Korea
| | - Seong Soo Joo
- Department of Marine Life Science, College of Life Science, Gangneung-Wonju National University, Gangneung, 25457 Gangwon Republic of Korea
- Huscion MAJIC R&D Center, 331 Pangyo-ro, Seongnam, 13488 Gyeonggi Republic of Korea
| |
Collapse
|
4
|
Lei H, Ruan Y, Ding R, Li H, Zhang X, Ji X, Wang Q, Lv S. The role of celastrol in inflammation and diseases. Inflamm Res 2025; 74:23. [PMID: 39862265 DOI: 10.1007/s00011-024-01983-5] [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: 07/27/2024] [Revised: 10/04/2024] [Accepted: 10/18/2024] [Indexed: 01/27/2025] Open
Abstract
Celastrol is one of the main active ingredients extracted from the plant Tripterygium wilfordii Hook F. A growing number of studies have shown that celastrol has various pharmacological effects, including anti-inflammation, anti-rheumatism, treatment of neurodegenerative diseases, and anti-tumor. This article systematically summarized the mechanism and role of celastrol in lipid metabolism and obesity, rheumatoid arthritis (RA), osteoarthritis (OA), gouty arthritis, inflammatory bowel disease, neurodegenerative diseases, and cancer and other diseases (such as diabetes, respiratory-related diseases, atherosclerosis, psoriasis, hearing loss, etc.). The celastrol played roles in inflammation response, cell apoptosis, autophagy, ferroptosis, and lipid metabolism mainly by acting on chondrocytes, macrophages, mitochondria, and endoplasmic reticulum (ER) through NF-κB, STAT, MAPK, TLR, PI3K-AKT-mTOR, and other signal pathways. This review could provide a reference for the clinical application and further development and utilization of celastrol.
Collapse
Affiliation(s)
- Han Lei
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Yantian Ruan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Ruidong Ding
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Haotian Li
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Xiaoguang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475001, Henan, China
| | - Xinying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
- Center for Molecular Medicine, Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Mazhai, Erqi District, Zhengzhou, 450064, Henan, China
| | - Qi Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
| | - Shuangyu Lv
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
- Department of Neurosurgery, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475001, Henan, China.
| |
Collapse
|
5
|
Kim M, Kang S, Oh S. The Anti-Inflammatory Activities of Benzylideneacetophenone Derivatives in LPS Stimulated BV2 Microglia Cells and Mice. Biomol Ther (Seoul) 2025; 33:106-116. [PMID: 39390761 PMCID: PMC11704402 DOI: 10.4062/biomolther.2024.049] [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: 03/22/2024] [Revised: 05/28/2024] [Accepted: 06/09/2024] [Indexed: 10/12/2024] Open
Abstract
A previously reported study highlighted the neuroprotective potential of the novel benzylideneacetophenone derivative, JC3, in mice. In pursuit of compounds with even more robust neuroprotective and anti-inflammatory properties compared to JC3, we synthesized substituted 1,3-diphenyl-2-propen-1-ones based on chalcones. Molecular modeling studies aimed at discerning the chemical structural features conducive to heightened biological activity revealed that JCII-8,10,11 exhibited the widest HOMOLUMO gap within this category, indicating facile electron and radical transfer between HOMO and LUMO in model assessments. From the pool of synthesized compounds, JCII-8,10,11 were selected for the present investigation. The biological assays involving JCII-8,10,11 demonstrated their concentration-dependent suppression of iNOS and COX-2 protein levels, alongside various cytokine mRNA expressions in LPS-induced murine microglial BV2 cells. Furthermore, western blot analyses were conducted to investigate the MAPK pathways and NF-κB/p65 nuclear translocation. These evaluations conclusively confirmed the inflammatory inhibition effects in both in vitro and in vivo inflammation models. These findings establish JCII-8,10,11 as potent anti-inflammatory agents, hindering inflammatory mediators and impeding NF-κB/p65 nuclear translocation via JNK and ERK MAPK phosphorylation in BV2 cells. The study positions them as potential therapeutics for inflammation-related conditions. Additionally, JCII-11 exhibited greater activity compared to other tested JCII compounds.
Collapse
Affiliation(s)
- Mijin Kim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Seungmin Kang
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Seikwan Oh
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| |
Collapse
|
6
|
Zeng C, Zhu Q, Peng W, Huang C, Chen H, Huang H, Zhou Y, Zhao C. The protective effect of amitriptyline on experimental colitis through inhibiting TLR-4/MD-2 signaling pathway. J Pharmacol Exp Ther 2025; 392:100024. [PMID: 39892990 DOI: 10.1124/jpet.124.002207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 10/10/2024] Open
Abstract
Amitriptyline, a pleiotropic tricyclic antidepressant, possesses antioxidant and anti-inflammatory properties. Despite its diverse benefits, the specific effects of amitriptyline on inflammatory bowel disease (IBD) are not yet well defined. To explore this, we used a dextran sulfate sodium (DSS)-induced colitis model to examine the anti-inflammatory effects of amitriptyline and the underlying mechanisms by which it operates. Our research revealed that amitriptyline is effective in alleviating several pathological manifestations associated with colitis. This includes improving body weight retention, reducing disease activity index, lessening of colon length shortening, and repairing of colonic mucosal damage. Treatment with amitriptyline significantly protected mucosal injury by preserving the population of goblet cells and increasing the expression of tight junction proteins. Furthermore, we observed that amitriptyline effectively countered immune cell infiltration, specifically neutrophils and macrophages, while simultaneously lowering the levels of inflammatory cytokines such as tumor necrosis factor α, interleukin (IL)-1β, and IL-6. Additionally, RNA sequencing analysis pointed to the potential involvement of the Toll-like receptor (TLR) pathway in the anticolitic effects induced by amitriptyline. Subsequent Western blot analysis indicated that amitriptyline significantly inhibited the TLR-4-mediated nuclear factor (NF)-κB signaling pathway. To bolster our findings, in vitro studies demonstrated that amitriptyline downregulated the TLR-4/NF-κB/mitogen-activated protein kinase signaling cascades in mouse macrophages stimulated with lipopolysaccharide. Further molecular investigations revealed that amitriptyline was able to suppress the elevated expression of myeloid differentiation factor 2 that lipopolysaccharide stimulation typically induces. In summary, our findings suggest that amitriptyline effectively mitigates DSS-induced colitis in mice through the inhibition of TLR-4/myeloid differentiation 2 pathway signaling, indicating its potential repurposing for IBD treatment. SIGNIFICANCE STATEMENT: The potential of using amitriptyline in treating inflammatory bowel disease appears promising, leveraging its established safety and dosing profile as an antidepressant. The study results show that amitriptyline can alleviate pathological symptoms, inflammation, and intestinal mucosal damage in mice with colitis induced by DSS. The protective effect observed appears to be linked to the inhibition of TLR-4/myeloid differentiation 2 signaling pathway. By exploring novel applications for existing medications, we can optimize amitriptyline's efficacy and broaden its impact in both medical and commercial contexts.
Collapse
Affiliation(s)
- Chengcheng Zeng
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Qingqing Zhu
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Wu Peng
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Chen Huang
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Huiting Chen
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Hongli Huang
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Yongjian Zhou
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China
| | - Chong Zhao
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medical, South China University of Technology, Guangzhou, Guangdong, China; Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, China.
| |
Collapse
|
7
|
Zheng S, Qi W, Xue T, Zao X, Xie J, Zhang P, Li X, Ye Y, Liu A. Chinese medicine in the treatment of chronic hepatitis B: The mechanisms of signal pathway regulation. Heliyon 2024; 10:e39176. [PMID: 39640799 PMCID: PMC11620126 DOI: 10.1016/j.heliyon.2024.e39176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Chronic hepatitis B (CHB) is a chronic inflammatory disease of the liver caused by infection with the hepatitis B virus (HBV), which in later stages can lead to the development of end-stage liver diseases such as cirrhosis and hepatocellular carcinoma in severe cases, jeopardizing long-term quality of life, with a poor prognosis, and placing a serious financial burden on many families around the world. The pathogenesis of the disease is complex and closely related to the immune function of the body, which has not yet been fully elucidated. The development of chronic hepatitis B is closely related to the involvement of various signaling pathways, such as JAK/STAT, PI3K/Akt, Toll-like receptor, NF-κB and MAPK signaling pathways. A large number of studies have shown that Chinese medicine has obvious advantages in anti-hepatitis B virus, and it can effectively treat the disease by modulating relevant signaling pathways, strengthening immune resistance and defense, and inhibiting inflammatory responses, and certain research progress has been made, but there is still a lack of a comprehensive review on the modulation of relevant signaling pathways in Chinese medicine for the treatment of CHB. Therefore, this article systematically combed and elaborated the relevant literature on the modulation of relevant signaling pathways by traditional Chinese medicine in recent years, with a view to providing new ideas for the treatment of CHB and further drug development.
Collapse
Affiliation(s)
- Shihao Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Wenying Qi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Tianyu Xue
- Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, 050000, China
| | - Xiaobin Zao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
| | - Jinchi Xie
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Peng Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Xiaoke Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yongan Ye
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Aimin Liu
- Shangzhuang Township Community Health Service Center, Beijing, 100094, China
| |
Collapse
|
8
|
Mandlem VKK, Rivera A, Khan Z, Quazi SH, Deba F. TLR4 induced TRPM2 mediated neuropathic pain. Front Pharmacol 2024; 15:1472771. [PMID: 39329114 PMCID: PMC11424904 DOI: 10.3389/fphar.2024.1472771] [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: 07/30/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Ion channels play an important role in mediating pain through signal transduction, regulation, and control of responses, particularly in neuropathic pain. Transient receptor potential channel superfamily plays an important role in cation permeability and cellular signaling. Transient receptor potential channel Melastatin 2 (TRPM2) subfamily regulates Ca2+ concentration in response to various chemicals and signals from the surrounding environment. TRPM2 has a role in several physiological functions such as cellular osmosis, temperature sensing, cellular proliferation, as well as the manifestation of many disease processes such as pain process, cancer, apoptosis, endothelial dysfunction, angiogenesis, renal and lung fibrosis, and cerebral ischemic stroke. Toll-like Receptor 4 (TLR4) is a critical initiator of the immune response to inflammatory stimuli, particularly those triggered by Lipopolysaccharide (LPS). It activates downstream pathways leading to the production of oxidative molecules and inflammatory cytokines, which are modulated by basal and store-operated calcium ion signaling. The cytokine production and release cause an imbalance of antioxidant enzymes and redox potential in the Endoplasmic Reticulum and mitochondria due to oxidative stress, which results from TLR-4 activation and consequently induces the production of inflammatory cytokines in neuronal cells, exacerbating the pain process. Very few studies have reported the role of TRPM2 and its association with Toll-like receptors in the context of neuropathic pain. However, the molecular mechanism underlying the interaction between TRPM2 and TLR-4 and the quantum of impact in acute and chronic neuropathic pain remains unclear. Understanding the link between TLR-4 and TRPM2 will provide more insights into pain regulation mechanisms for the development of new therapeutic molecules to address neuropathic pain.
Collapse
Affiliation(s)
- Venkata Kiran Kumar Mandlem
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| | - Ana Rivera
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| | - Zaina Khan
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
- Departmental of Neuroscience, University of Texas at Dallas, Richardson, TX, United States
| | - Sohel H Quazi
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
- Department of Biology, Division of Natural and Computation Sciences, Texas College, Tyler, TX, United States
| | - Farah Deba
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| |
Collapse
|
9
|
Tang Q, Chen W, Ke H, Lan C. Optical imaging detection of extracellular vesicles of miR-146 modified bone marrow mesenchymal stem cells promoting spinal cord injury repair. SLAS Technol 2024; 29:100172. [PMID: 39067816 DOI: 10.1016/j.slast.2024.100172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/02/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Bone Marrow mesenchymal Stem Cells (BMSCs) are considered as an important source of cells for regenerative medicine, In particular, Bone Marrow mesenchymal Stem Cells Exosomes (BMSCs-EXO) have the most significant effect in the treatment of Spinal Cord Injury (SCI), but the mechanism of action is still unknown. This study found that compared with other SCI groups, BMSCs-EXO loaded with miR-146a could significantly improve the functional recovery of the hind limbs of SCI rats. Hematoxylin and eosin (H&E) indicated that the lesion area of spinal cord injury was less, nissl staining indicated that the number of nissl bodies remained more; the mechanism may be through inhibiting the expression of IRAK1 and TRAF6, blocking the activation of NF-κB p65, reducing the expression of TNF-α, IL-1β and IL-6 inflammatory factors and oxidative stress, improving the SCI microenvironment, and promoting the repair of neural function. In general, we found that BMSCs-EXO loaded with miR-146a could reduce the inflammatory response and oxidative stress in SCI by inhibiting the activation of IRAK1/TRAF6/NF-κB p65 signaling pathway, and promote the recovery of neurological function in SCI rats.
Collapse
Affiliation(s)
- Qianli Tang
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 533000, China
| | - Wu Chen
- Department of Neurosurgery, The Peoples Hospital of Baise, Baise 533009, China
| | - Huang Ke
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 533000, China; Department of Trauma Orthopedics, The Affiliated Hospital of Youjiang Medical University for Nationalities, Bais 533009, China
| | - Changgong Lan
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 533000, China; Department of Trauma Orthopedics, The Affiliated Hospital of Youjiang Medical University for Nationalities, Bais 533009, China; Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Youjiang Medical University for Nationalities, Baise 533009, China; Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Youjiang Medical University for Nationalities, Baise 533009, China; Guangxi Key Laboratory of Clinical Medical Research on Bone and Joint Degenerative Diseases Cohort, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533009, China.
| |
Collapse
|
10
|
Barut Z, Aslan M, Çırçırlı B, Çeker T, Yılmaz Ç. Antiproliferative Effect of 7-Ketositosterol in Breast and Liver Cancer Cells: Possible Impact on Ceramide, Extracellular Signal-Regulated Kinases, and Nuclear Factor Kappa B Signaling Pathways. Pharmaceuticals (Basel) 2024; 17:860. [PMID: 39065711 PMCID: PMC11279788 DOI: 10.3390/ph17070860] [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: 05/28/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
Background: This study aimed to examine the effect of 7-Ketositosterol (7-KSS), on sphingomyelin/ceramide metabolites and apoptosis in human breast MCF-7 and human liver HepG2 cancer cells. Methods: Anti-proliferative effects of 7-KSS treatment were assessed at different concentrations and periods. Cell viability was assessed through MTT analysis, whereas the levels of sphingosine-1-phosphate (S1P), sphingomyelins (SMs), and ceramides (CERs) were measured using LC-MS/MS. Phosphorylated 44/42 ERK1/2 and NF-κB p65 (Ser536) protein levels were measured by Western blot analysis and immunofluorescence staining. Apoptosis was evaluated by TUNEL staining and flow cytometric assessment of annexin-V and propidium iodide (PI) labeling. Results: Treatment with 7-KSS significantly decreased cell survival and S1P, p-44/42 ERK1/2, and p-NF-κB p65 protein levels in cancer cells compared to controls. A substantial rise was detected in intracellular amounts of C16-C24 CERs and apoptosis in cancer cells incubated with 7-KSS. Conclusions: 7-KSS stimulated ceramide accumulation and apoptosis while decreasing cell proliferation via downregulating S1P, p-44/42 ERK1/2, and p-NF-κB p65 protein levels.
Collapse
Affiliation(s)
- Zerrin Barut
- Faculty of Dentistry, Antalya Bilim University, 07190 Antalya, Turkey;
| | - Mutay Aslan
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, 07070 Antalya, Turkey; (T.Ç.); (Ç.Y.)
- Department of Medical Biotechnology, Institute of Health Sciences, Akdeniz University, 07070 Antalya, Turkey;
| | - Bürke Çırçırlı
- Department of Medical Biotechnology, Institute of Health Sciences, Akdeniz University, 07070 Antalya, Turkey;
| | - Tuğçe Çeker
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, 07070 Antalya, Turkey; (T.Ç.); (Ç.Y.)
| | - Çağatay Yılmaz
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, 07070 Antalya, Turkey; (T.Ç.); (Ç.Y.)
| |
Collapse
|
11
|
Tian M, Xie D, Hong Y, Ding F, Wu X, Tang D. Anti-inflammatory effects and related mechanisms in vitro and in vivo of Hedychium coccineum rhizome essential oil. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118103. [PMID: 38527573 DOI: 10.1016/j.jep.2024.118103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedychium coccineum rhizome is an anti-inflammatory ethnomedicine used to remedy inflammation-related swelling and bronchial asthma. AIM OF THE STUDY The study aimed to analyze the phytochemical constituents of H. coccineum rhizome essential oil (EO) and evaluate its in vitro and in vivo anti-inflammatory effects and underlying mechanisms. MATERIALS AND METHODS Phytochemical constituents of H. coccineum rhizome EO were analyzed using GC-FID/MS. In RAW264.7 macrophages induced by LPS, blockade of PGE2, NO, IL-1β, IL-6, and TNF-α secretion by H. coccineum rhizome EO was measured, and then Western blot, qRT-PCR, and immunofluorescent staining were used to evaluate its underlying mechanisms. Moreover, we used the xylene-induced ear edema model for testing anti-inflammatory potential in vivo and examined auricular swelling as well as tissue and serum contents of IL-1β, IL-6, and TNF-α. RESULTS EO's main components were E-nerolidol (40.5%), borneol acetate (24.8%), spathulenol (4.5%), linalool (3.8%), elemol (3.5%), and borneol (3.4%). In RAW264.7 cells stimulated by LPS, EO downregulated the expression of pro-inflammatory enzyme (iNOS and COX-2) genes and proteins, thereby suppressing pro-inflammatory mediators (NO and PGE2) secretion. Simultaneously, it reduced TNF-α, IL-1β, and IL-6 release by downregulating their mRNA expression. Besides, H. coccineum EO attenuated LPS-stimulated activation of NF-κB (by reducing IκBα phosphorylation and degradation to inhibit NF-κB nuclear translocation) and MAPK (by downregulating JNK, p38, and ERK phosphorylation). In xylene-induced mouse ear edema, EO relieved auricular swelling and lowered serum and tissue levels of TNF-α, IL-1β, and IL-6. CONCLUSIONS H. coccineum EO had powerful in vivo and in vitro anti-inflammatory effects by inhibiting MAPK and NF-κB activation. Hence, H. coccineum EO should have great potential for application in the pharmaceutical field as a novel anti-inflammatory agent.
Collapse
Affiliation(s)
- Minyi Tian
- First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, China; National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China; Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou University, Guiyang, 550025, China.
| | - Dan Xie
- First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, China
| | - Yi Hong
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou University, Guiyang, 550025, China
| | - Furong Ding
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Xia Wu
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Dongxin Tang
- First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, China.
| |
Collapse
|
12
|
Zhao H, Becharef S, Dumas E, Carn F, Patriarche G, Mura S, Gazeau F, Serre C, Steunou N. A gold nanocluster/MIL-100(Fe) bimodal nanovector for the therapy of inflammatory disease through attenuation of Toll-like receptor signaling. NANOSCALE 2024; 16:12037-12049. [PMID: 38809107 DOI: 10.1039/d3nr06685a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
A better understanding of the molecular and cellular events involved in the inflammation process has opened novel perspectives in the treatment of inflammatory diseases, particularly through the development of well-designed nanomedicines. Here we describe the design of a novel class of anti-inflammatory nanomedicine (denoted as Au@MIL) synthesized through a one-pot, cost-effective and green approach by coupling a benchmark mesoporous iron(III) carboxylate metal organic framework (MOF) (i.e. MIL-100(Fe)) and glutathionate protected gold nanoclusters (i.e. Au25SG18 NCs). This nano-carrier exhibits low toxicity and excellent colloidal stability combined with the high loading capacity of the glucocorticoid dexamethasone phosphate (DexP) whose pH-dependent delivery was observed. The drug loaded Au@MIL nanocarrier shows high anti-inflammatory activity due to its capacity to specifically hinder inflammatory cell growth, scavenge intracellular reactive oxygen species (ROS) and downregulate pro-inflammatory cytokine secretion. In addition, this formulation has the capacity to inhibit the Toll-like receptor (TLR) signaling cascade namely the nuclear factor kappa B (NF-κB) and the interferon regulatory factor (IRF) pathways. This not only provides a new avenue for the nanotherapy of inflammatory diseases but also enhances our fundamental knowledge of the role of nanoMOF based nanomedicine in the regulation of innate immune signaling.
Collapse
Affiliation(s)
- Heng Zhao
- Institut des Matériaux Poreux de Paris, ENS, ESPCI Paris, CNRS, PSL University, Paris, France.
| | - Sonia Becharef
- Université Paris Cité, MSC UMR CNRS 7057, 75006 Paris, France.
| | - Eddy Dumas
- Institut Lavoisier de Versailles, UMR CNRS 8180, Université de Versailles St Quentin en Yvelines, Université Paris Saclay, Versailles, France
| | - Florent Carn
- Université Paris Cité, MSC UMR CNRS 7057, 75006 Paris, France.
| | - Gilles Patriarche
- Université Paris-Saclay, CNRS, Centre de Nanosciences et de Nanotechnologies, 91120 Palaiseau, France
| | - Simona Mura
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Florence Gazeau
- Université Paris Cité, MSC UMR CNRS 7057, 75006 Paris, France.
| | - Christian Serre
- Institut des Matériaux Poreux de Paris, ENS, ESPCI Paris, CNRS, PSL University, Paris, France.
| | - Nathalie Steunou
- Institut des Matériaux Poreux de Paris, ENS, ESPCI Paris, CNRS, PSL University, Paris, France.
- Institut Lavoisier de Versailles, UMR CNRS 8180, Université de Versailles St Quentin en Yvelines, Université Paris Saclay, Versailles, France
| |
Collapse
|
13
|
Kim J, Kim JY, Byeon HE, Kim JW, Kim HA, Suh CH, Choi S, Linton MF, Jung JY. Inhibition of Toll-like Receptors Alters Macrophage Cholesterol Efflux and Foam Cell Formation. Int J Mol Sci 2024; 25:6808. [PMID: 38928513 PMCID: PMC11203583 DOI: 10.3390/ijms25126808] [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: 05/23/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Arterial macrophage cholesterol accumulation and impaired cholesterol efflux lead to foam cell formation and the development of atherosclerosis. Modified lipoproteins interact with toll-like receptors (TLR), causing an increased inflammatory response and altered cholesterol homeostasis. We aimed to determine the effects of TLR antagonists on cholesterol efflux and foam cell formation in human macrophages. Stimulated monocytes were treated with TLR antagonists (MIP2), and the cholesterol efflux transporter expression and foam cell formation were analyzed. The administration of MIP2 attenuated the foam cell formation induced by lipopolysaccharides (LPS) and oxidized low-density lipoproteins (ox-LDL) in stimulated THP-1 cells (p < 0.001). The expression of ATP-binding cassette transporters A (ABCA)-1, ABCG-1, scavenger receptor (SR)-B1, liver X receptor (LXR)-α, and peroxisome proliferator-activated receptor (PPAR)-γ mRNA and proteins were increased (p < 0.001) following MIP2 administration. A concentration-dependent decrease in the phosphorylation of p65, p38, and JNK was also observed following MIP2 administration. Moreover, an inhibition of p65 phosphorylation enhanced the expression of ABCA1, ABCG1, SR-B1, and LXR-α. TLR inhibition promoted the cholesterol efflux pathway by increasing the expression of ABCA-1, ABCG-1, and SR-B1, thereby reducing foam cell formation. Our results suggest a potential role of the p65/NF-kB/LXR-α/ABCA1 axis in TLR-mediated cholesterol homeostasis.
Collapse
Affiliation(s)
- Jaemi Kim
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| | - Ji-Yun Kim
- Institute of Medical Science, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.-Y.K.)
| | - Hye-Eun Byeon
- Institute of Medical Science, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.-Y.K.)
| | - Ji-Won Kim
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| | - Hyoun-Ah Kim
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| | - Chang-Hee Suh
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea;
| | - MacRae F. Linton
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| | - Ju-Yang Jung
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| |
Collapse
|
14
|
Chuang JMJ, Chen HL, Chang CI, Lin JS, Chang HM, Wu WJ, Lin MY, Chen WF, Lee CH. Nobiletin derivative, 5-acetoxy-6,7,8,3',4'-pentamethoxyflavone, inhibits neuroinflammation through the inhibition of TLR4/MyD88/MAPK signaling pathways and STAT3 in microglia. Immunopharmacol Immunotoxicol 2024:1-11. [PMID: 38800857 DOI: 10.1080/08923973.2024.2360050] [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: 07/12/2023] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE Microglia in the central nervous system regulate neuroinflammation that leads to a wide range of neuropathological alterations. The present study investigated the anti-neuroinflammatory properties of nobiletin (Nob) derivative, 5-acetoxy-6,7,8,3',4'-pentamethoxyflavone (5-Ac-Nob), in lipopolysaccharide (LPS)-activated BV2 microglia. MATERIALS AND METHODS By using the MTT assay, Griess method, flow cytometry, and enzyme-linked immunosorbent assay (ELISA), we determined the cell viability, the levels of nitric oxide (NO), reactive oxygen species (ROS), and pro-inflammatory factors (interleukin 1 beta; IL-1β, interleukin 6; IL-6, tumor necrosis factor alpha; TNF-α and prostaglandin E2; PGE2) in LPS-stimulated BV2 microglia. Toll-like receptor 4 (TLR4)-mediated myeloid differentiation primary response gene 88 (MyD88)/nuclear factor-kappa B (NF-κB), mitogen-activated protein kinase (MAPK) signaling pathway and signal transducer and activator of transcription 3 (STAT3) were measured by western blotting. Analysis of NO generation and mRNA of pro-inflammatory cytokines was confirmed in the zebrafish model. RESULTS 5-Ac-Nob reduced cell death, the levels of NO, ROS, inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2), and pro-inflammatory factors in LPS-activated BV-2 microglial cells. TLR4-mediated MyD88/NF-κB and MAPK pathway (p38, ERK and JNK) after exposure to 5-Ac-Nob was also suppressed. Moreover, 5-Ac-Nob inhibited phosphorylated STAT3 proteins expression in LPS-induced BV-2 microglial cells. Furthermore, we confirmed that 5-Ac-Nob decreased LPS-induced NO generation and mRNA of pro-inflammatory cytokines in the zebrafish model. CONCLUSIONS Our findings suggest that 5-Ac-Nob represses neuroinflammatory responses by inhibiting TLR4-mediated signaling pathway and STAT3. As a result of these findings, 5-Ac-Nob has potential as an anti-inflammatory agent against microglia-mediated neuroinflammatory disorders.
Collapse
Affiliation(s)
- Jimmy Ming-Jung Chuang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Hsien-Lin Chen
- Division of General Surgery, Department of Surgery, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Chi-I Chang
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Research Centre for Active Natural Products Development, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Jia-Syuan Lin
- Department of Pharmacology, School of Post-Baccalaureate Medicine; Division of Pharmacology and Traditional Chinese Medicine, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Min Chang
- Department of Pharmacology, School of Post-Baccalaureate Medicine; Division of Pharmacology and Traditional Chinese Medicine, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Ju Wu
- Department of Pharmacology, School of Post-Baccalaureate Medicine; Division of Pharmacology and Traditional Chinese Medicine, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ying Lin
- Community Health Promotion Center, Kaohsiung Municipal Ci-Jin Hospital, Kaohsiung, Taiwan
| | - Wu-Fu Chen
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chien-Hsing Lee
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Research Centre for Active Natural Products Development, National Pingtung University of Science and Technology, Pingtung, Taiwan
- Department of Pharmacology, School of Post-Baccalaureate Medicine; Division of Pharmacology and Traditional Chinese Medicine, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
15
|
Wang J, Li H, Liu Y, Andrzejczyk NE, Qiao K, Ma Y, Zhou S, Gui W, Zhu G, Li S, Schlenk D. Contribution of Immune Responses to the Cardiotoxicity and Hepatotoxicity of Deltamethrin in Early Life Stage Zebrafish ( Danio rerio). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:9515-9524. [PMID: 38687472 DOI: 10.1021/acs.est.3c10682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Deltamethrin (DM) is a widely used insecticide that has demonstrated developmental toxicity in the early life stages of fish. To better characterize the underlying mechanisms, embryos from Tg(cmlc2:RFP), Tg(apo14:GFP), and Tg(mpx:GFP) transgenic strains of zebrafish were exposed to nominal DM concentrations of 0.1, 1, 10, 25, and 50 μg/L until 120 h post-fertilization (hpf). Heart size increased 56.7%, and liver size was reduced by 17.1% in zebrafish exposed to 22.7 and 24.2 μg/L DM, respectively. RNA sequencing and bioinformatic analyses predicted that key biological processes affected by DM exposure were related to inflammatory responses. Expression of IL-1 protein was increased by 69.0% in the 24.4 μg/L DM treatment, and aggregation of neutrophils in cardiac and hepatic histologic sections was also observed. Coexposure to resatorvid, an anti-inflammatory agent, mitigated inflammatory responses and cardiac toxicity induced by DM and also restored liver biomass. Our data indicated a complex proinflammatory mechanism underlying DM-induced cardiotoxicity and hepatotoxicity which may be important for key events of adverse outcomes and associated risks of DM to early life stages of fish.
Collapse
Affiliation(s)
- Jie Wang
- Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, P. R. China
| | - Hanqing Li
- Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yuanyuan Liu
- Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, P. R. China
| | - Nicolette E Andrzejczyk
- Department of Environmental Sciences,University of California, Riverside, California 92521, United States
| | - Kun Qiao
- Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, P. R. China
- Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, P. R. China
- Department Evolutionary Ecology and Environmental Toxicology, Faculty Biological Sciences, Goethe University 10 Frankfurt, Frankfurt Am Main 60438, Germany
| | - Yongfang Ma
- Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, P. R. China
| | - Shengli Zhou
- Ecological and Environmental Monitoring Center of Zhejiang Province, Hangzhou 310012, P. R. China
| | - Wenjun Gui
- Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, P. R. China
| | - Guonian Zhu
- Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, P. R. China
| | - Shuying Li
- Institute of Pesticide and Environmental Toxicology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, P. R. China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou 310058, P. R. China
| | - Daniel Schlenk
- Department of Environmental Sciences,University of California, Riverside, California 92521, United States
| |
Collapse
|
16
|
Zhao L, Zhang Z, Li P, Gao Y, Shi Y. Bakuchiol regulates TLR4/MyD88/NF-κB and Keap1/Nrf2/HO-1 pathways to protect against LPS-induced acute lung injury in vitro and in vivo. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3301-3312. [PMID: 37930390 DOI: 10.1007/s00210-023-02813-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
Bakuchiol (Bak) possesses a protective effect in acute lung injury (ALI). Nonetheless, the molecular processes that regulate the protective activity of Bak in ALI remain elusive. Lipopolysaccharide (LPS)-treated rats and RLE-6TN cells were used as the ALI models in vivo and in vitro to investigate the function and mechanism of Bak. Rats were divided into four groups: control, LPS, LPS + Bak (30 mg/kg), and LPS + Bak (60 mg/kg). RLE-6TN cells were assigned into four groups: control, LPS, LPS + Bak (10 µM), and LPS + Bak (20 µM). Myeloperoxidase (MPO) and 4-hydroxy-2-nonenal (4-HNE) levels were detected by immunohistochemistry (IHC). The levels of TNF-α, IL-6, and IL-1β were quantified by ELISA. Apoptosis was analyzed by TdT-mediated dUTP nick-end labeling (TUNEL) staining and flow cytometry. Malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and reactive oxygen species (ROS) were assayed to evaluate oxidative stress. In LPS-induced rats, Bak attenuated pathological injury, lung wet/dry weight ratio, MPO expression, and protein concentration and cell number in bronchial alveolar lavage fluid (BALF). Bak decreased the secretion of TNF-α, IL-6, and IL-1β in BALF. Bak reduced MDA content and 4-HNE expression, and increased SOD and GSH-Px activities in lung tissues. Bak also repressed pulmonary apoptosis by decreasing Bax expression and enhancing Bcl-2 expression. In LPS-treated RLE-6TN cells, Bak downregulated the mRNA levels of TNF-α, IL-6, and IL-1β and inhibited the protein expression of iNOS and COX2. Bak decreased MDA level and ROS production and increased SOD and GSH-Px activities. Bak also suppressed cell apoptosis, reduced Bax expression, and increased Bcl-2 expression. Moreover, Bak decreased the expression of TLR4, MyD88, p-IκBα, and p-p65. Additionally, Bak inhibited Keap1 expression and increased Nrf2 and HO-1 levels. Bak protects against LPS-induced inflammation, oxidative stress, and apoptosis in ALI by regulating TLR4/MyD88/NF-κB and Keap1/Nrf2/HO-1 pathways.
Collapse
Affiliation(s)
- Li Zhao
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China
| | - Zhengliang Zhang
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China
| | - Ping Li
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China
| | - Yanxia Gao
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China
| | - Yu Shi
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China.
| |
Collapse
|
17
|
Mirsanei Z, Jamshidi-Adegani F, Vakilian S, Ahangari F, Soufihasanabad S, Al-Riyami K, Soudi S, Ghaffari Khaligh S, Al-Hashmi S, Hashemi SM. Synergistic effects of mesenchymal stem cell-derived extracellular vesicles and dexamethasone on macrophage polarization under inflammatory conditions. Inflammopharmacology 2024; 32:1317-1332. [PMID: 38512654 DOI: 10.1007/s10787-024-01438-7] [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: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 03/23/2024]
Abstract
The undesirable inflammation and the excessive M1 macrophage activity may lead to inflammatory diseases. Corticosteroids and stem cell therapy are used in clinical practice to promote anti-inflammatory responses. However, this protocol has limitations and is associated with numerous side effects. In this study, the synergistic anti-inflammatory effects of dexamethasone (Dex) and mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) were evaluated to enhance the polarization of M1 inflammatory macrophages into the anti-inflammatory (M2) phenotype. Hence, we designed different combinations of Dex and EVs using three methods, including EVs isolated from Dex-preconditioned MSCs (Pre-Dex-EVs), EVs loaded with Dex (L-Dex-EVs), and EVs and Dex co-administration (Dex + EVs). All designed EVs had a significant effect on reducing the expression of M1-related genes (iNOS, Stat1, and IRF5), cytokines (IL6 and TNF-a), and CD markers (CD86) in lipopolysaccharide-stimulated macrophages. On the other hand, these combinations promoted the expression of alternative-activated M2-related genes (Arg-1, Stat6, and IRF4), cytokine (IL10), and CD markers (CD206).The combination of Dex and MSC-EVs enhances the effectiveness of both and synergistically promotes the conversion of inflammatory macrophages into an anti-inflammatory state.
Collapse
Affiliation(s)
- Zahra Mirsanei
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Jamshidi-Adegani
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, P. O. Box: 33, PC, 616, Nizwa, Oman
| | - Saeid Vakilian
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, P. O. Box: 33, PC, 616, Nizwa, Oman
| | - Fateme Ahangari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Soufihasanabad
- Department of Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Khamis Al-Riyami
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, P. O. Box: 33, PC, 616, Nizwa, Oman
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | | | - Sulaiman Al-Hashmi
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, P. O. Box: 33, PC, 616, Nizwa, Oman.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Hussein MA, Shahidul Islam M, Ali AA, Mansour MS, Bondok M, Salem MA, Amein AS, ElGizawy HA. Malva parviflora seed oil; Isolation, gas chromatographic profiling and its cardioprotective activity against myocardial infraction in animal model. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2024; 36:103060. [DOI: 10.1016/j.jksus.2023.103060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
|
19
|
Naidoo K, Khathi A. The Potential Role of Gossypetin in the Treatment of Diabetes Mellitus and Its Associated Complications: A Review. Int J Mol Sci 2023; 24:17609. [PMID: 38139436 PMCID: PMC10743819 DOI: 10.3390/ijms242417609] [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: 11/23/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder caused by insulin resistance and dysfunctional beta (β)-cells in the pancreas. Hyperglycaemia is a characteristic of uncontrolled diabetes which eventually leads to fatal organ system damage. In T2DM, free radicals are continuously produced, causing extensive tissue damage and subsequent macro-and microvascular complications. The standard approach to managing T2DM is pharmacological treatment with anti-diabetic medications. However, patients' adherence to treatment is frequently decreased by the side effects and expense of medications, which has a detrimental impact on their health outcomes. Quercetin, a flavonoid, is a one of the most potent anti-oxidants which ameliorates T2DM. Thus, there is an increased demand to investigate quercetin and its derivatives, as it is hypothesised that similar structured compounds may exhibit similar biological activity. Gossypetin is a hexahydroxylated flavonoid found in the calyx of Hibiscus sabdariffa. Gossypetin has a similar chemical structure to quercetin with an extra hydroxyl group. Furthermore, previous literature has elucidated that gossypetin exhibits neuroprotective, hepatoprotective, reproprotective and nephroprotective properties. The mechanisms underlying gossypetin's therapeutic potential have been linked to its anti-oxidant, anti-inflammatory and immunomodulatory properties. Hence, this review highlights the potential role of gossypetin in the treatment of diabetes and its associated complications.
Collapse
Affiliation(s)
| | - Andile Khathi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
| |
Collapse
|
20
|
Shi XY, Zheng XM, Liu HJ, Han X, Zhang L, Hu B, Li S. Rotundic acid improves nonalcoholic steatohepatitis in mice by regulating glycolysis and the TLR4/AP1 signaling pathway. Lipids Health Dis 2023; 22:214. [PMID: 38049817 PMCID: PMC10694891 DOI: 10.1186/s12944-023-01976-z] [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: 06/27/2023] [Accepted: 11/21/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Steatosis and inflammation are the hallmarks of nonalcoholic steatohepatitis (NASH). Rotundic acid (RA) is among the key triterpenes of Ilicis Rotundae Cortex and has exhibited multipronged effects in terms of lowering the lipid content and alleviating inflammation. The study objective is to systematically evaluate the potential mechanisms through which RA affects the development and progression of NASH. METHODS Transcriptomic and proteomic analyses of primary hepatocytes isolated from the control, high-fat diet-induced NASH, and RA treatment groups were performed through Gene Ontology analysis and pathway enrichment. Hub genes were identified through network analysis. Integrative analysis revealed key RA-regulated pathways, which were verified by gene and protein expression studies and cell assays. RESULTS Hub genes were identified and enriched in the Toll-like receptor 4 (TLR4)/activator protein-1 (AP1) signaling pathway and glycolysis pathway. RA reversed glycolysis and attenuated the TLR4/AP1 pathway, thereby reducing lipid accumulation and inflammation. Additionally, lactate release in L-02 cells increased with NaAsO2-treated and significantly decreased with RA treatment, thus revealing that RA had a major impact on glycolysis. CONCLUSIONS RA is effective in lowering the lipid content and reducing inflammation in mice with NASH by ameliorating glycolysis and TLR4/AP1 pathways, which contributes to the existing knowledge and potentially sheds light on the development of therapeutic interventions for patients with NASH.
Collapse
Affiliation(s)
- Xing-Yang Shi
- MOE International Joint Laboratory for Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Xiao-Min Zheng
- MOE International Joint Laboratory for Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Hui-Jie Liu
- MOE International Joint Laboratory for Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Xue Han
- MOE International Joint Laboratory for Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Lei Zhang
- MOE International Joint Laboratory for Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- NMPA Key Laboratory for Quality Control of Blood Products, Guangdong Institute for Drug Control, Guangzhou, 510663, PR China
| | - Bei Hu
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510030, PR China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, PR China.
| | - Shan Li
- MOE International Joint Laboratory for Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China.
| |
Collapse
|
21
|
Liu X, Wei W, Wu YZ, Wang Y, Zhang WW, Wang YP, Dong XP, Shi Q. Emodin treatment of papillary thyroid cancer cell lines in vitro inhibits proliferation and enhances apoptosis via downregulation of NF‑κB and its upstream TLR4 signaling. Oncol Lett 2023; 26:514. [PMID: 37927413 PMCID: PMC10623093 DOI: 10.3892/ol.2023.14101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/15/2023] [Indexed: 11/07/2023] Open
Abstract
Thyroid cancer is one of the most common types of endocrine malignancy. In addition to surgical treatment, it is very important to find new treatment methods. The aim of the present study was to evaluate the effect of 1,3,8-trihydroxy-6-methylanthraquinone (emodin) on cellular NF-κB components and the upstream regulatory pathway of toll-like receptor 4 (TLR4) signaling, as well as the invasion and migration of papillary thyroid carcinoma (PTC) cells. The protein expression of NF-κB components p65 and p50 and their phosphorylated (p-) forms in the sections of PTC tissues was measured by individual immunohistochemical assays. PTC cell lines TPC-1 and IHH4 were exposed to 20 and 40 µM emodin for 24 h. The levels of the NF-κB components p65, p50, c-Rel, p-p65 and p-p50, elements in TLR4 signaling, including TLR4, MYD88 innate immune signal transduction adaptor (MyD88), interferon regulatory factor 3, AKT and MEK, and proliferative and apoptotic biomarkers, including c-Myc, cyclin D1, proliferating cell nuclear antigen, Bcl-2 and Bax, were evaluated by western blotting and immunofluorescent assays. The invasion and migration of PTC cell lines exposed to emodin were tested by plate colony and wound healing assay. Compared with hyperplasia tissue, the expression levels of NF-κB components p65 and p50, and p-p65 and p-p50 in PTC tissue were significantly increased. Treatment of PTC cell lines with emodin lead to significantly reduced levels of the aforementioned NF-κB components, accompanied by markedly downregulated TLR4 signaling. MYD 88-dependent and -independent pathways, are also significantly down-regulated. Downregulation of proliferative factors and activation of apoptotic factors were observed in the cell lines following treatment with emodin. Consequently, inhibition of the invasion and migration activities were observed in the emodin-treated PTC cells. Emodin could inhibit proliferation and promote apoptosis of PTC cells, which is dependent on the downregulation of cellular NF-κB and the TLR4 signaling pathway.
Collapse
Affiliation(s)
- Xin Liu
- Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, P.R. China
- National Key-Laboratory of Intelligent Tracing and Forecasting for Infectious Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Wei Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Head and Neck Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Yue-Zhang Wu
- National Key-Laboratory of Intelligent Tracing and Forecasting for Infectious Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Yuan Wang
- National Key-Laboratory of Intelligent Tracing and Forecasting for Infectious Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
- Basic Medical College, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Wei-Wei Zhang
- National Key-Laboratory of Intelligent Tracing and Forecasting for Infectious Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
- Basic Medical College, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Yong-Ping Wang
- Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, P.R. China
| | - Xiao-Ping Dong
- National Key-Laboratory of Intelligent Tracing and Forecasting for Infectious Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Qi Shi
- National Key-Laboratory of Intelligent Tracing and Forecasting for Infectious Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
| |
Collapse
|
22
|
Korakaki E, Simos YV, Karouta N, Spyrou K, Zygouri P, Gournis DP, Tsamis KI, Stamatis H, Dounousi E, Vezyraki P, Peschos D. Effect of Highly Hydrophilic Superparamagnetic Iron Oxide Nanoparticles on Macrophage Function and Survival. J Funct Biomater 2023; 14:514. [PMID: 37888179 PMCID: PMC10607831 DOI: 10.3390/jfb14100514] [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: 08/01/2023] [Revised: 09/09/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have garnered significant attention in the medical sector due to their exceptional superparamagnetic properties and reliable tracking capabilities. In this study, we investigated the immunotoxicity of SPIONs with a modified surface to enhance hydrophilicity and prevent aggregate formation. The synthesized SPIONs exhibited a remarkably small size (~4 nm) and underwent surface modification using a novel "haircut" reaction strategy. Experiments were conducted in vitro using a human monocytic cell line (THP-1). SPIONs induced dose-dependent toxicity to THP-1 cells, potentially by generating ROS and initiating the apoptotic pathway in the cells. Concentrations up to 10 μg/mL did not affect the expression of Nrf2, HO-1, NF-κB, or TLR-4 proteins. The results of the present study demonstrated that highly hydrophilic SPIONs were highly toxic to immune cells; however, they did not activate pathways of inflammation and immune response. Further investigation into the mechanisms of cytotoxicity is warranted to develop a synthetic approach for producing effective, highly hydrophilic SPIONs with little to no side effects.
Collapse
Affiliation(s)
- Efterpi Korakaki
- Laboratory of Physiology, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.K.); (K.I.T.); (P.V.); (D.P.)
| | - Yannis Vasileios Simos
- Laboratory of Physiology, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.K.); (K.I.T.); (P.V.); (D.P.)
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
| | - Niki Karouta
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
| | - Konstantinos Spyrou
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
| | - Panagiota Zygouri
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios Panagiotis Gournis
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
- Department of Materials Science and Engineering, University of Ioannina, 45110 Ioannina, Greece
| | - Konstantinos Ioannis Tsamis
- Laboratory of Physiology, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.K.); (K.I.T.); (P.V.); (D.P.)
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
| | - Haralambos Stamatis
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
- Department of Biological Applications and Technologies, University of Ioannina, 45110 Ioannina, Greece
| | - Evangelia Dounousi
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Patra Vezyraki
- Laboratory of Physiology, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.K.); (K.I.T.); (P.V.); (D.P.)
| | - Dimitrios Peschos
- Laboratory of Physiology, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.K.); (K.I.T.); (P.V.); (D.P.)
- Nanomedicine and Nanobiotechnology Research Group, University of Ioannina, 45110 Ioannina, Greece; (N.K.); (P.Z.); (D.P.G.); (H.S.); (E.D.)
| |
Collapse
|
23
|
Yang C, Cheng J, Zhu Q, Pan Q, Ji K, Li J. Review of the Protective Mechanism of Paeonol on Cardiovascular Disease. Drug Des Devel Ther 2023; 17:2193-2208. [PMID: 37525853 PMCID: PMC10387245 DOI: 10.2147/dddt.s414752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/19/2023] [Indexed: 08/02/2023] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death in the world. Paeonol(Pae) is a phenolic component extracted from peony bark, peony root and Xu Changqing. Studies have shown that Pae can protect cardiomyocytes by inhibiting oxidative stress, promoting mitochondrial fusion, regulating mitochondrial autophagy and inhibiting inflammation. In addition, Pae improves ventricular remodeling by inhibiting myocardial apoptosis, hypertrophy and fibrosis. Pae also has a good protective effect on blood vessels by inhibiting vascular inflammation, reducing the expression of adhesion molecules, inhibiting vascular proliferation, and inhibiting oxidative stress and endoplasmic reticulum stress(ERS). Pae also has the effect of anti-endothelial cell senescence, promoting thrombus recanalization and vasodilating. In conclusion, the molecular targets of Pae are very complex, and the relationship between different targets and signaling pathways cannot be clearly explained, which requires us to use systems biology methods to further study specific molecular targets of Pae. It has to be mentioned that the bioavailability of Pae is poor, and some nanotechnology-assisted drug delivery systems improve the therapeutic effect of Pae. We reviewed the protective mechanism of paeonol on the cardiovascular system, hoping to provide help for drug development in the treatment of CVD.
Collapse
Affiliation(s)
- Chunkun Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Jiawen Cheng
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qinwei Zhu
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, People's Republic of China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, People's Republic of China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, People's Republic of China
| | - Jun Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
24
|
Gong P, Jia HY, Li R, Ma Z, Si M, Qian C, Zhu FQ, Sheng-Yong L. Downregulation of Nogo-B ameliorates cerebral ischemia/reperfusion injury in mice through regulating microglia polarization via TLR4/NF-kappaB pathway. Neurochem Int 2023; 167:105553. [PMID: 37230196 DOI: 10.1016/j.neuint.2023.105553] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 05/27/2023]
Abstract
Many studies have shown a close association between Nogo-B and inflammation-related diseases. However, uncertainty does exist, regarding Nogo-B function in the pathological progression of cerebral ischemia/reperfusion (I/R) injury. Middle cerebral artery occlusion/reperfusion (MCAO/R) model was utilized in C57BL/6L mice to mimic ischemic stroke in vivo. Using oxygen-glucose deprivation and reoxygenation (ODG/R) model in microglia cells (BV-2) to establish cerebral I/R injury in vitro. Various methods, including Nogo-B siRNA transfection, mNSS and the rotarod test, TTC, HE and Nissl staining, immunofluorescence staining, immunohistochemistry, Western blot, ELISA, TUNEL and qRT-PCR were employed to probe into the effect of Nogo-B downregulation on cerebral I/R injury and the potential mechanisms. A small amount of Nogo-B expression (protein and mRNA) was observed in cortex and hippocampus before ischemia, then Nogo-B expression increased significantly on day 1, reaching the maximum on day 3, remaining stable on day 14 after I/R, and decreasing gradually after 21 days, but it still rose significantly compared with that observed preischemia. Nogo-B down-regulation could markedly reduce the neurological score and infarct volume, improve the histopathological changes and neuronal apoptosis, lower the number of CD86+/Iba1+ cells and the levels of IL-1β, IL-6, and TNF-α, and raise the density of NeuN fluorescence, the number of CD206+/Iba1+ cells, and the level of IL-4, IL-10 and TGF-β in brain of MCAO/R mice. Treatment with Nogo-B siRNA or TAK-242 in BV-2 cells could obviously decrease the CD86 fluorescence density and the mRNA expression of IL-1β, IL-6 and TNF-α, increase CD206 fluorescence density and the mRNA expression of IL-10 after OGD/R injury. In addition, the expression of TLR4, p-IκBα and p-p65 proteins significantly increased in the brain after MCAO/R and BV-2 cells exposed to OGD/R. Treatment with Nogo-B siRNA or TAK-242 prominently reduced the expression of TLR4, p-IκBα and p-p65. Our findings suggest that the down-regulation of Nogo-B exerts protective effect on cerebral I/R injury by modulating the microglia polarization through inhibiting TLR4/NF-κB signaling pathway. Nogo-B may be a potential therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Peng Gong
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China.
| | - Hui-Yu Jia
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China.
| | - Rui Li
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China.
| | - Zheng Ma
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China.
| | - Min Si
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China.
| | - Can Qian
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China.
| | - Feng-Qin Zhu
- Cancer Hospital, Chinese Academy of Science, Hefei, Anhui, 230032, China.
| | - Luo Sheng-Yong
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China.
| |
Collapse
|
25
|
Atre R, Sharma R, Vadim G, Solanki K, Wadhonkar K, Singh N, Patidar P, Khabiya R, Samaur H, Banerjee S, Baig MS. The indispensability of macrophage adaptor proteins in chronic inflammatory diseases. Int Immunopharmacol 2023; 119:110176. [PMID: 37104916 DOI: 10.1016/j.intimp.2023.110176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/06/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023]
Abstract
Adaptor proteins represent key signalling molecules involved in regulating immune responses. The host's innate immune system recognizes pathogens via various surface and intracellular receptors. Adaptor molecules are centrally involved in different receptor-mediated signalling pathways, acting as bridges between the receptors and other molecules. The presence of adaptors in major signalling pathways involved in the pathogenesis of various chronic inflammatory diseases has drawn attention toward the role of these proteins in such diseases. In this review, we summarize the importance and roles of different adaptor molecules in macrophage-mediated signalling in various chronic disease states. We highlight the mechanistic roles of adaptors and how they are involved in protein-protein interactions (PPI) via different domains to carry out signalling. Hence, we also provide insights into how targeting these adaptor proteins can be a good therapeutic strategy against various chronic inflammatory diseases.
Collapse
Affiliation(s)
- Rajat Atre
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rahul Sharma
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Gaponenko Vadim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Khandu Wadhonkar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Neha Singh
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Pramod Patidar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rakhi Khabiya
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India; School of Pharmacy, Devi Ahilya Vishwavidyalaya, Indore, India
| | - Harshita Samaur
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Sreeparna Banerjee
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey.
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
26
|
CuMV VLPs Containing the RBM from SARS-CoV-2 Spike Protein Drive Dendritic Cell Activation and Th1 Polarization. Pharmaceutics 2023; 15:pharmaceutics15030825. [PMID: 36986686 PMCID: PMC10055701 DOI: 10.3390/pharmaceutics15030825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Dendritic cells (DCs) are the most specialized and proficient antigen-presenting cells. They bridge innate and adaptive immunity and display a powerful capacity to prime antigen-specific T cells. The interaction of DCs with the receptor-binding domain of the spike (S) protein from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a pivotal step to induce effective immunity against the S protein-based vaccination protocols, as well as the SARS-CoV-2 virus. Herein, we describe the cellular and molecular events triggered by virus-like particles (VLPs) containing the receptor-binding motif from the SARS-CoV-2 spike protein in human monocyte-derived dendritic cells, or, as controls, in the presence of the Toll-like receptors (TLR)3 and TLR7/8 agonists, comprehending the events of dendritic cell maturation and their crosstalk with T cells. The results demonstrated that VLPs boosted the expression of major histocompatibility complex molecules and co-stimulatory receptors of DCs, indicating their maturation. Furthermore, DCs’ interaction with VLPs promoted the activation of the NF-kB pathway, a very important intracellular signalling pathway responsible for triggering the expression and secretion of proinflammatory cytokines. Additionally, co-culture of DCs with T cells triggered CD4+ (mainly CD4+Tbet+) and CD8+ T cell proliferation. Our results suggested that VLPs increase cellular immunity, involving DC maturation and T cell polarization towards a type 1 T cells profile. By providing deeper insight into the mechanisms of activation and regulation of the immune system by DCs, these findings will enable the design of effective vaccines against SARS-CoV-2.
Collapse
|
27
|
Liu C, Liu LX, Yang J, Liu YG. Exploration and analysis of the composition and mechanism of efficacy of camel milk. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
|
28
|
Wang R, Li L, Wang J, Zhao X, Shen J. CircBRMS1L Participates in Lipopolysaccharide-induced Chondrocyte Injury via the TLR4/NF-κB Pathway through Serving as a miR-142-5p Decoy. BIOTECHNOL BIOPROC E 2023. [DOI: 10.1007/s12257-021-0224-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
29
|
Huang J, Wu T, Zhong Y, Huang J, Kang Z, Zhou B, Zhao H, Liu D. Effect of curcumin on regulatory B cells in chronic colitis mice involving TLR/MyD88 signaling pathway. Phytother Res 2023; 37:731-742. [PMID: 36196887 DOI: 10.1002/ptr.7656] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/12/2022] [Accepted: 09/15/2022] [Indexed: 11/09/2022]
Abstract
Curcumin (Cur) is a natural active phenolic compound extracted from the root of Curcuma Longa L. It has anti-inflammatory, anti-tumor and other pharmacological activities, and is commonly used to treat ulcerative colitis (UC). However, it is not clear whether curcumin regulates the function and differentiation of Breg cells to treat UC. In this study, mice with chronic colitis were induced by dextran sulfate sodium (DSS), and treated with curcumin for 12 days. Curcumin effectively improved the body weight, colonic weight, colonic length, decreased colonic weight index and pathological injury score under colonoscopy in mice with chronic colitis, and significantly inhibited the production of IL-1β, IL-6, IL-33, CCL-2, IFN-γ, TNF-α, and promoted the secretion of IL-4, IL-10, IL-13 and IgA. Importantly, curcumin markedly upregulated CD3- CD19+ CD1d+ , CD3- CD19+ CD25+ , CD3- CD19+ Foxp3+ Breg cells level and significantly down-regulated CD3- CD19+ PD-L1+ , CD3- CD19+ tim-1+ , CD3- CD19+ CD27+ Breg cells level. In addition, our results also showed that curcumin observably inhibited TLR2, TLR4, TLR5, MyD88, IRAK4, p-IRAK4, NF-κB P65, IRAK1, TRAF6, TAB1, TAB2, TAK1, MKK3, MKK6, p38MAPK, p-p38MAPK and CREB expression in TLR/MyD88 signaling pathway. These results suggest that curcumin can regulate the differentiation and function of Breg cell to alleviate DSS-induced colitis, which may be realized by inhibiting TLR/MyD88 pathway.
Collapse
Affiliation(s)
- Jie Huang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
| | - Tiantian Wu
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
| | - Youbao Zhong
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
| | - Jiaqi Huang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
| | - Zengping Kang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
| | - Bugao Zhou
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
| | - Haimei Zhao
- College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
| | - Duanyong Liu
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People's Republic of China
| |
Collapse
|
30
|
Tian M, Xie D, Yang Y, Tian Y, Jia X, Wang Q, Deng G, Zhou Y. Hedychium flavum flower essential oil: Chemical composition, anti-inflammatory activities and related mechanisms in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115846. [PMID: 36280015 DOI: 10.1016/j.jep.2022.115846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedychium flavum, an ornamental, edible, and medicinal plant, is extensively cultivated as a source of aromatic essential oils (EO). Its flower is a traditional Chinese medicine for treating inflammation-related diseases like indigestion, diarrhea, and stomach pain. In particular, H. flavum flower EO has been used in cosmetics and as an aromatic stomachic to treat chronic gastritis in China. AIM OF THE STUDY This research aimed to analyze H. flavum flower EO's chemical composition and explore its anti-inflammatory activities and related mechanisms in vitro and in vivo. MATERIALS AND METHODS EO's chemical composition was determined by GC-FID/MS analysis. For in vitro test, the anti-inflammatory activity of EO was demonstrated by measuring the LPS-induced release of NO, PGE2, IL-1β, TNF-α, and IL-6 in RAW264.7 macrophages, and then its related mechanisms were explored using qRT-PCR, western blot, and immunofluorescent staining analysis. Next, EO's in vivo anti-inflammatory potential was further evaluated using a xylene-induced ear edema model, in which ear swelling and TNF-α, IL-6, and IL-1β levels in serum and tissue were examined. RESULTS The main components of EO were β-pinene (20.2%), α-pinene (9.3%), α-phellandrene (8.3%), 1,8-cineole (7.1%), E-nerolidol (5.4%), limonene (4.4%), borneol (4.1%), and β-caryophyllene (3.7%). For the anti-inflammatory activities in vitro, EO dramatically reduced the LPS-stimulated NO and PGE2 release by suppressing the mRNA and protein expression of iNOS and COX-2. Meanwhile, it remarkably decreased IL-6, TNF-α, and IL-1β production by inhibiting their mRNA levels. Related mechanism studies indicated that it not only inhibited IκBα phosphorylation and degradation, leading to blockade of NF-κB nuclear transfer but also suppressed MAPKs (ERK, p38, and JNK) phosphorylation in LPS-stimulated RAW264.7 cells. Further in vivo assay showed that EO ameliorated xylene-induced ear edema in mice and reduced TNF-α, IL-6, and IL-1β levels in serum and tissue. CONCLUSIONS H. flavum EO exerted significant anti-inflammatory activity in vivo and in vitro, and its mechanism of action is related to the inhibition of MAPK and NF-κB activation. Thus, H. flavum EO could be considered a novel and promising anti-inflammatory agent and possess high potential for utilization in the pharmaceutical field.
Collapse
Affiliation(s)
- Minyi Tian
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China; Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou University, Guiyang, 550025, China.
| | - Dan Xie
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Yao Yang
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Yufeng Tian
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Xiaoyan Jia
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Qinqin Wang
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Guodong Deng
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Ying Zhou
- National & Local Joint Engineering Research Center for the Exploitation of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China; College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| |
Collapse
|
31
|
Muscat SM, Deems NP, Butler MJ, Scaria EA, Bettes MN, Cleary SP, Bockbrader RH, Maier SF, Barrientos RM. Selective TLR4 Antagonism Prevents and Reverses Morphine-Induced Persistent Postoperative Cognitive Dysfunction, Dysregulation of Synaptic Elements, and Impaired BDNF Signaling in Aged Male Rats. J Neurosci 2023; 43:155-172. [PMID: 36384680 PMCID: PMC9838714 DOI: 10.1523/jneurosci.1151-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 11/07/2022] [Accepted: 11/10/2023] [Indexed: 11/18/2022] Open
Abstract
Perioperative neurocognitive disorders (PNDs) are characterized by confusion, difficulty with executive function, and episodic memory impairment in the hours to months following a surgical procedure. Postoperative cognitive dysfunction (POCD) represents such impairments that last beyond 30 d postsurgery and is associated with increased risk of comorbidities, progression to dementia, and higher mortality. While it is clear that neuroinflammation plays a key role in PND development, what factors underlie shorter self-resolving versus persistent PNDs remains unclear. We have previously shown that postoperative morphine treatment extends POCD from 4 d (without morphine) to at least 8 weeks (with morphine) in aged male rats, and that this effect is likely dependent on the proinflammatory capabilities of morphine via activation of toll-like receptor 4 (TLR4). Here, we extend these findings to show that TLR4 blockade, using the selective TLR4 antagonist lipopolysaccharide from the bacterium Rhodobacter sphaeroides (LPS-RS Ultrapure), ameliorates morphine-induced POCD in aged male rats. Using either a single central preoperative treatment or a 1 week postoperative central treatment regimen, we demonstrate that TLR4 antagonism (1) prevents and reverses the long-term memory impairment associated with surgery and morphine treatment, (2) ameliorates morphine-induced dysregulation of the postsynaptic proteins postsynaptic density 95 and synaptopodin, (3) mitigates reductions in mature BDNF, and (4) prevents decreased activation of the BDNF receptor TrkB (tropomyosin-related kinase B), all at 4 weeks postsurgery. We also reveal that LPS-RS Ultrapure likely exerts its beneficial effects by preventing endogenous danger signal HMGB1 (high-mobility group box 1) from activating TLR4, rather than by blocking continuous activation by morphine or its metabolites. These findings suggest TLR4 as a promising therapeutic target to prevent or treat PNDs.SIGNIFICANCE STATEMENT With humans living longer than ever, it is crucial that we identify mechanisms that contribute to aging-related vulnerability to cognitive impairment. Here, we show that the innate immune receptor toll-like receptor 4 (TLR4) is a key mediator of cognitive dysfunction in aged rodents following surgery and postoperative morphine treatment. Inhibition of TLR4 both prevented and reversed surgery plus morphine-associated memory impairment, dysregulation of synaptic elements, and reduced BDNF signaling. Together, these findings implicate TLR4 in the development of postoperative cognitive dysfunction, providing mechanistic insight and novel therapeutic targets for the treatment of cognitive impairments following immune challenges such as surgery in older individuals.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio 43210
| | - Nicholas P Deems
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio 43210
| | - Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Emmanuel A Scaria
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Sean P Cleary
- Campus Chemical Instrumentation Center, The Ohio State University, Columbus, Ohio 43210
| | - Ross H Bockbrader
- Pharmaceutical Sciences Graduate Program, Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210
| | - Steven F Maier
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado 80309
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, Ohio 43210
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
32
|
ATP2B1-AS1 exacerbates sepsis-induced cell apoptosis and inflammation by regulating miR-23a-3p/TLR4 axis. Allergol Immunopathol (Madr) 2023; 51:17-26. [PMID: 36916084 DOI: 10.15586/aei.v51i2.782] [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: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 03/07/2023]
Abstract
BACKGROUND Sepsis is a life-threatening disease with dominant mortality. Its early diagnosis and treatment can improve prognosis and reduce mortality. Long noncoding RNAs (lncRNAs) ATPase plasma membrane Ca2+ transporting 1 antisense RNA 1 (ATP2B1-AS1) is dysregulated and is involved in the progression of various diseases. Nevertheless, the role of ATP2B1-AS1 in sepsis remains unclear. METHODS A human monocytic cell line, THP-1 cells, was stimulated to induce a model of sepsis in vitro. The levels of ATP2B1-AS1, miR-23a-3p, and TLR4 were assessed by real-time quantitative polymerase chain reaction. The role of ATP2B1-AS1 in cell apoptosis and inflammation was explored by flow cytometry, Western blot analysis and enzyme-linked immunosorbent serologic assay. The binding sites between ATP2B1-AS1 and miR-23a-3p, and between miR-23a-3p and TLR4 were predicted by BiBiServ and the Encyclopedia of RNA Interactomes (ENCORI) online sites, respectively, and confirmed by the luciferase assay. RESULTS The level of ATP2B1-AS1 was increased in lipopolysaccharide (LPS)-treated THP-1 cells. LPS increased apoptosis ratio, relative protein expressions of pro-apoptotic factors, and relative messenger RNA (mRNA) level and concentrations of pro-inflammatory cytokines, but decreased the relative expression of anti-apoptosis protein and relative mRNA level and concentrations of anti-inflammatory factor. All these alterations were reversed with transfection of shATP2B1-AS1 into THP-1 cells. Moreover, ATP2B1-AS1 directly bound miR-23a-3p and negatively modulated the level of miR-23a-3p. Meanwhile, TLR4 was directly targeted by miR-23a-3p, and negatively and positively modulated by miR-23a-3p and ATP2B1-AS1, respectively. CONCLUSION ATP2B1-AS1 aggravated apoptosis and inflammation by modulating miR-23a-3p/TLR4 axis in LPS-treated THP-1 cells.
Collapse
|
33
|
Wang L, Geng G, Zhu T, Chen W, Li X, Gu J, Jiang E. Progress in Research on TLR4-Mediated Inflammatory Response Mechanisms in Brain Injury after Subarachnoid Hemorrhage. Cells 2022; 11:cells11233781. [PMID: 36497041 PMCID: PMC9740134 DOI: 10.3390/cells11233781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is one of the common clinical neurological emergencies. Its incidence accounts for about 5-9% of cerebral stroke patients. Even surviving patients often suffer from severe adverse prognoses such as hemiplegia, aphasia, cognitive dysfunction and even death. Inflammatory response plays an important role during early nerve injury in SAH. Toll-like receptors (TLRs), pattern recognition receptors, are important components of the body's innate immune system, and they are usually activated by damage-associated molecular pattern molecules. Studies have shown that with TLR 4 as an essential member of the TLRs family, the inflammatory transduction pathway mediated by it plays a vital role in brain injury after SAH. After SAH occurrence, large amounts of blood enter the subarachnoid space. This can produce massive damage-associated molecular pattern molecules that bind to TLR4, which activates inflammatory response and causes early brain injury, thus resulting in serious adverse prognoses. In this paper, the process in research on TLR4-mediated inflammatory response mechanism in brain injury after SAH was reviewed to provide a new thought for clinical treatment.
Collapse
Affiliation(s)
- Lintao Wang
- Institute of Nursing and Health, Henan University, Kaifeng 475004, China
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
- Department of Neurology, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Guangping Geng
- Henan Technician College of Medicine and Health, Kaifeng 475000, China
| | - Tao Zhu
- Department of Geriatrics, Kaifeng Traditional Chinese Medicine Hospital, Kaifeng 475001, China
| | - Wenwu Chen
- Department of Neurology, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Xiaohui Li
- Department of Neurology, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Jianjun Gu
- Department of Neurosurgery, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Enshe Jiang
- Institute of Nursing and Health, Henan University, Kaifeng 475004, China
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng 475004, China
- Correspondence:
| |
Collapse
|
34
|
Li WQ, Liu WH, Qian D, Liu J, Zhou SQ, Zhang L, Peng W, Su L, Zhang H. Traditional Chinese medicine: An important source for discovering candidate agents against hepatic fibrosis. Front Pharmacol 2022; 13:962525. [PMID: 36081936 PMCID: PMC9445813 DOI: 10.3389/fphar.2022.962525] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/28/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatic fibrosis (HF) refers to the pathophysiological process of connective tissue dysplasia in the liver caused by various pathogenic factors. Nowadays, HF is becoming a severe threat to the health of human being. However, the drugs available for treating HF are limited. Currently, increasing natural agents derived from traditional Chinese medicines (TCMs) have been found to be beneficial for HF. A systemic literature search was conducted from PubMed, GeenMedical, Sci-Hub, CNKI, Google Scholar and Baidu Scholar, with the keywords of "traditional Chinese medicine," "herbal medicine," "natural agents," "liver diseases," and "hepatic fibrosis." So far, more than 76 natural monomers have been isolated and identified from the TCMs with inhibitory effect on HF, including alkaloids, flavones, quinones, terpenoids, saponins, phenylpropanoids, and polysaccharides, etc. The anti-hepatic fibrosis effects of these compounds include hepatoprotection, inhibition of hepatic stellate cells (HSC) activation, regulation of extracellular matrix (ECM) synthesis & secretion, regulation of autophagy, and antioxidant & anti-inflammation, etc. Natural compounds and extracts from TCMs are promising agents for the prevention and treatment of HF, and this review would be of great significance to development of novel drugs for treating HF.
Collapse
Affiliation(s)
- Wen-Qing Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Hao Liu
- Department of Pharmacy, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Die Qian
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shi-Qiong Zhou
- Hospital of Nursing, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Lei Zhang
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Hong Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
35
|
Sophorolipid Suppresses LPS-Induced Inflammation in RAW264.7 Cells through the NF-κB Signaling Pathway. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27155037. [PMID: 35956987 PMCID: PMC9370320 DOI: 10.3390/molecules27155037] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022]
Abstract
Objectives: Biosurfactants with anti-inflammatory activity may alleviate skin irritation caused by synthetic surfactants in cleaning products. Sophorolipid (SL) is a promising alternative to synthetic surfactants. However, there are few reports on the anti-inflammatory activity of SL and the underlying mechanism. The purpose of this work is to verify that lipopolysaccharide (LPS)-induced inflammation could be inhibited through targeting the pathway of nuclear factor-κB (NF-κB) in RAW264.7 cells. Methods: The influence of SL on cytokine release was investigated by LPS-induced RAW264.7 cells using ELISA. The quantification of the protein expression of corresponding molecular markers was realized by Western blot analysis. Flow cytometry was employed to determine the levels of Ca2+ and reactive oxygen species (ROS). The relative expression of inducible nitric oxide synthase (INOS) and cyclooxygenase-2 (COX-2) was determined by RT-PCR. An immunofluorescence assay and confocal microscope were used to observe the NF-κB/p65 translocation from the cytoplasm into the nucleus. The likely targets of SL were predicted by molecular docking analysis. Results: SL showed anti-inflammatory activity and reduced the release of inflammatory cytokines including interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and nitric oxide (NO). The experimental results show that SL suppressed the Ca2+ and ROS levels influx in the LPS-induced RAW264.7 cells and alleviated the LPS-induced expression of iNOS and COX-2, the LPS-induced translocation of NF-κB (p65) from the cytoplasm into the nucleus, and the expression of phosphorylated proteins such as p65 and IκBα. Furthermore, molecular docking analysis showed that SL may inhibit inflammatory signaling by competing with LPS to bind TLR4/MD-2 through hydrophobic interactions and by inhibiting IKKβ activation through the hydrogen bonding and hydrophobic interactions. Conclusion: This study demonstrated that SL exerted anti-inflammatory activity via the pathway of NF-κB in RAW264.7 cells.
Collapse
|
36
|
V. Lima B, Oliveira MJ, Barbosa MA, Gonçalves RM, Castro F. Harnessing chitosan and poly-(γ-glutamic acid)-based biomaterials towards cancer immunotherapy. MATERIALS TODAY ADVANCES 2022; 15:100252. [DOI: 10.1016/j.mtadv.2022.100252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
37
|
Zhuang C, Chen R, Zheng Z, Lu J, Hong C. Toll-Like Receptor 3 in Cardiovascular Diseases. Heart Lung Circ 2022; 31:e93-e109. [PMID: 35367134 DOI: 10.1016/j.hlc.2022.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
Toll-like receptor 3 (TLR3) is an important member of the innate immune response receptor toll-like receptors (TLRs) family, which plays a vital role in regulating immune response, promoting the maturation and differentiation of immune cells, and participating in the response of pro-inflammatory factors. TLR3 is activated by pathogen-associated molecular patterns and damage-associated molecular patterns, which support the pathophysiology of many diseases related to inflammation. An increasing number of studies have confirmed that TLR3, as a crucial medium of innate immunity, participates in the occurrence and development of cardiovascular diseases (CVDs) by regulating the transcription and translation of various cytokines, thus affecting the structure and physiological function of resident cells in the cardiovascular system, including vascular endothelial cells, vascular smooth muscle cells, cardiomyocytes, fibroblasts and macrophages. The dysfunction and structural damage of vascular endothelial cells and proliferation of vascular smooth muscle cells are the key factors in the occurrence of vascular diseases such as pulmonary arterial hypertension, atherosclerosis, myocardial hypertrophy, myocardial infarction, ischaemia/reperfusion injury, and heart failure. Meanwhile, cardiomyocytes, fibroblasts, and macrophages are involved in the development of CVDs. Therefore, the purpose of this review was to explore the latest research published on TLR3 in CVDs and discuss current understanding of potential mechanisms by which TLR3 contributes to CVDs. Even though TLR3 is a developing area, it has strong treatment potential as an immunomodulator and deserves further study for clinical translation.
Collapse
Affiliation(s)
- Chunying Zhuang
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; First Clinical School, Guangzhou Medical University, Guangzhou, China
| | - Riken Chen
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenzhen Zheng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Guangzhou, China
| | - Jianmin Lu
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cheng Hong
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Sadeghian I, Heidari R, Raee MJ, Negahdaripour M. Cell-penetrating peptide-mediated delivery of therapeutic peptides/proteins to manage the diseases involving oxidative stress, inflammatory response and apoptosis. J Pharm Pharmacol 2022; 74:1085-1116. [PMID: 35728949 DOI: 10.1093/jpp/rgac038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/22/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Peptides and proteins represent great potential for modulating various cellular processes including oxidative stress, inflammatory response, apoptosis and consequently the treatment of related diseases. However, their therapeutic effects are limited by their inability to cross cellular barriers. Cell-penetrating peptides (CPPs), which can transport cargoes into the cell, could resolve this issue, as would be discussed in this review. KEY FINDINGS CPPs have been successfully exploited in vitro and in vivo for peptide/protein delivery to treat a wide range of diseases involving oxidative stress, inflammatory processes and apoptosis. Their in vivo applications are still limited due to some fundamental issues of CPPs, including nonspecificity, proteolytic instability, potential toxicity and immunogenicity. SUMMARY Totally, CPPs could potentially help to manage the diseases involving oxidative stress, inflammatory response and apoptosis by delivering peptides/proteins that could selectively reach proper intracellular targets. More studies to overcome related CPP limitations and confirm the efficacy and safety of this strategy are needed before their clinical usage.
Collapse
Affiliation(s)
- Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Biotechnology Incubator, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Raee
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
39
|
Guo K, Chang Y, Jin Y, Yuan H, Che P. circ-Ncam2 (mmu_circ_0006413) Participates in LPS-Induced Microglia Activation and Neuronal Apoptosis via the TLR4/NF-κB Pathway. J Mol Neurosci 2022; 72:1738-1748. [PMID: 35687299 DOI: 10.1007/s12031-022-02018-6] [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: 01/20/2022] [Accepted: 04/27/2022] [Indexed: 11/30/2022]
Abstract
Spinal cord injury (SCI) can cause permanent neurological deficits. Circular RNA Ncam2 (circ-Ncam2 also termed mmu_circ_0006413) has been reported to be overexpressed in SCI mouse models. However, the function of circ-Ncam2 in SCI has not been validated. Lipopolysaccharide (LPS) was used to activate mouse microglia (BV2 cells). Expression levels of circ-Ncam2 were determined by RT-qPCR. Relative protein levels were evaluated by western blotting. Cytokines were determined by ELISA. The regulatory mechanism of circ-Ncam2 was validated by dual-luciferase reporter and RNA pull-down assays. Effects of LPS-induced BV2 cells on mouse neuronal (HT22 cells) viability, proliferation, and apoptosis were analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium Bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays. LPS stimulation promoted circ-Ncam2 expression in BV2 cells. Inhibition of circ-Ncam2 mitigated LPS-induced BV2 cell activation and inflammation. Mechanically, circ-Ncam2 adsorbed miR-544-3p to regulate TLR4 expression. Also, either miR-544-3p inhibition or TLR4 overexpression weakened circ-Ncam2 silencing-mediated effects on LPS-induced BV2 cell activation and inflammation. Furthermore, LPS-induced BV2 cells suppressed HT22 cell proliferation and promoted HT22 cell apoptosis through the circ-Ncam2/miR-544-3p axis. Importantly, circ-Ncam2 activated the NF-κB signaling via the miR-544-3p/TLR4axis. circ-Ncam2 silencing lowered LPS-induced microglia activation and neuronal apoptosis via blocking the TLR4/NF-κB pathway through acting as a miR-544-3p sponge, suggesting that circ-Ncam2 may be involved in secondary SCI.
Collapse
Affiliation(s)
- Kai Guo
- Department of Orthopedics, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yulin Chang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yutao Jin
- Department of Orthopedics, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Hong Yuan
- Department of Orthopedics, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ping Che
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
40
|
Ethanol Extract of Sargassum siliquastrum Inhibits Lipopolysaccharide-Induced Nitric Oxide Generation by Downregulating the Nuclear Factor-Kappa B Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6160010. [PMID: 35722164 PMCID: PMC9205721 DOI: 10.1155/2022/6160010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/04/2022] [Indexed: 11/17/2022]
Abstract
Sargassum siliquastrum (SS) is an edible brown seaweed widely consumed in Korea and considered a functional food source. Previous studies have reported various biological activities of SS extracts, including antioxidant and hepatoprotective properties. In the present study, we examined the anti-inflammatory effects of the SS extract and assessed the underlying mechanism of action. The SS extract significantly inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO) production in a dose-dependent manner (% of NO production at 500 μg/mL: 60.1 ± 0.9%), with no obvious toxicity. Furthermore, the SS extract inhibited mRNA and protein expression levels of inducible NO synthase, as well as LPS-induced expression and production of proinflammatory cytokines such as IL-1β, IL-6, or TNF-α (IL-6 production (ng/mL) : LPS−: 0.7 ± 0.3; LPS+: 68.1 ± 2.8; LPS + SS extract: 51.9 ± 1.2; TNF-α production (ng/mL) : LPS−: 0.3 ± 0.1; LPS+: 23.0 ± 0.1; LPS + SS extract: 18.2 ± 10.8). Mechanistically, the SS extract attenuated LPS-induced activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (nuclear factor-kappa B, NF-κB) signaling pathway such as phosphorylation of NF-κB p65 and degradation of IκB-α, thereby blocking LPS-induced activation of NF-κB transcriptional activity. The SS extract also enhanced LPS-induced heme oxygenase-1 expression and attenuated LPS-induced cellular reactive oxygen species production (% of ROS production at 500 μg/mL: 52.2 ± 1.3%). Collectively, these findings suggest that the SS extract elicits anti-inflammatory effects in mouse macrophage cells.
Collapse
|
41
|
Sorafenib inhibits LPS-induced inflammation by regulating Lyn-MAPK-NF-kB/AP-1 pathway and TLR4 expression. Cell Death Dis 2022; 8:281. [PMID: 35680841 PMCID: PMC9184561 DOI: 10.1038/s41420-022-01073-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/07/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022]
Abstract
Sorafenib is an anti-tumor drug widely used in clinical treatment, which can inhibit tyrosine kinase receptor on cell surface and serine/threonine kinase in downstream Ras/MAPK cascade signaling pathway of cells. Tyrosine kinase phosphorylation plays an important role in inflammatory mechanism, such as TLR4 tyrosine phosphorylation, MAPK pathway protein activation, and activation of downstream NF-кB. However, the effects of sorafenib on LPS-induced inflammatory reaction and its specific mechanism have still remained unknown. We found that sorafenib inhibited the phosphorylation of tyrosine kinase Lyn induced by LPS, thereby reducing the phosphorylation level of p38 and JNK, inhibiting the activation of c-Jun and NF-κB, and then inhibiting the expression of inflammatory factors IL-6, IL-1β, and TNF-α. Furthermore, sorafenib also decreased the expression of TLR4 on the macrophage membrane to inhibit the expression of inflammatory factors latterly, which may be related to the inactivation of Lyn. These results provide a new perspective and direction for the clinical treatment of sepsis.
Collapse
|
42
|
Cao Z, Yang F, Lin Y, Shan J, Cao H, Zhang C, Zhuang Y, Xing C, Hu G. Selenium Antagonizes Cadmium-Induced Inflammation and Oxidative Stress via Suppressing the Interplay between NLRP3 Inflammasome and HMGB1/NF-κB Pathway in Duck Hepatocytes. Int J Mol Sci 2022; 23:ijms23116252. [PMID: 35682929 PMCID: PMC9181349 DOI: 10.3390/ijms23116252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/14/2022] Open
Abstract
Cadmium (Cd) is a toxic heavy metal that can accumulate in the liver of animals, damaging liver function. Inflammation and oxidative stress are considered primary causes of Cd-induced liver damage. Selenium (Se) is an antioxidant and can resist the detrimental impacts of Cd on the liver. To elucidate the antagonism of Se on Cd against hepatocyte injury and its mechanism, duck embryo hepatocytes were treated with Cd (4 μM) and/or Se (0.4 μM) for 24 h. Then, the hepatocyte viability, oxidative stress and inflammatory status were assessed. The findings manifested that the accumulation of reactive oxygen species (ROS) and the levels of pro-inflammatory factors were elevated in the Cd group. Simultaneously, immunofluorescence staining revealed that the interaction between NOD-like receptor pyran domain containing 3 (NLRP3) and apoptosis-associated speck-like protein (ASC) was enhanced, the movement of high-mobility group box 1 (HMGB1) from nucleus to cytoplasm was increased and the inflammatory response was further amplified. Nevertheless, the addition of Se relieved the above-mentioned effects, thereby alleviating cellular oxidative stress and inflammation. Collectively, the results suggested that Se could mitigate Cd-stimulated oxidative stress and inflammation in hepatocytes, which might be correlated with the NLRP3 inflammasome and HMGB1/nuclear factor-κB (NF-κB) signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chenghong Xing
- Correspondence: (C.X.); (G.H.); Tel.: +86-18770046182 (C.X.); +86-13807089905 (G.H.)
| | - Guoliang Hu
- Correspondence: (C.X.); (G.H.); Tel.: +86-18770046182 (C.X.); +86-13807089905 (G.H.)
| |
Collapse
|
43
|
Tong KL, Tan KE, Lim YY, Tien XY, Wong PF. CircRNA-miRNA interactions in atherogenesis. Mol Cell Biochem 2022; 477:2703-2733. [PMID: 35604519 DOI: 10.1007/s11010-022-04455-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/27/2022] [Indexed: 11/30/2022]
Abstract
Atherosclerosis is the major cause of coronary artery disease (CAD) which includes unstable angina, myocardial infarction, and heart failure. The onset of atherogenesis, a process of atherosclerotic lesion formation in the intima of arteries, is driven by lipid accumulation, a vicious cycle of reactive oxygen species (ROS)-induced oxidative stress and inflammatory reactions leading to endothelial cell (EC) dysfunction, vascular smooth muscle cell (VSMC) activation, and foam cell formation which further fuel plaque formation and destabilization. In recent years, there is a surge in the number of publications reporting the involvement of circular RNAs (circRNAs) in the pathogenesis of cardiovascular diseases, cancers, and metabolic syndromes. These studies have advanced our understanding on the biological functions of circRNAs. One of the most common mechanism of action of circRNAs reported is the sponging of microRNAs (miRNAs) by binding to the miRNAs response element (MRE), thereby indirectly increases the transcription of their target messenger RNAs (mRNAs). Individual networks of circRNA-miRNA-mRNA associated with atherogenesis have been extensively reported, however, there is a need to connect these findings for a complete overview. This review aims to provide an update on atherogenesis-related circRNAs and analyze the circRNA-miRNA-mRNA interactions in atherogenesis. The atherogenic mechanisms and clinical relevance of each atherogenesis-related circRNA were systematically discussed for better understanding of the knowledge gap in this area.
Collapse
Affiliation(s)
- Kind-Leng Tong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ke-En Tan
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yat-Yuen Lim
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Xin-Yi Tien
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
44
|
Chang CC, Peng SY, Tsao HH, Huang HT, Lai XY, Hsu HJ, Jiang SJ. A Multitarget Therapeutic Peptide Derived From Cytokine Receptors Based on in Silico Analysis Alleviates Cytokine-Stimulated Inflammation. Front Pharmacol 2022; 13:853818. [PMID: 35370629 PMCID: PMC8965626 DOI: 10.3389/fphar.2022.853818] [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: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Septicemia is a severe inflammatory response caused by the invasion of foreign pathogens. Severe sepsis-induced shock and multiple organ failure are the two main causes of patient death. The overexpression of many proinflammatory cytokines, such as TNF-α, IL-1β, and IL-6, is closely related to severe sepsis. Although the treatment of sepsis has been subject to many major breakthroughs of late, the treatment of patients with septic shock is still accompanied by a high mortality rate. In our previous research, we used computer simulations to design the multifunctional peptide KCF18 that can bind to TNF-α, IL-1β, and IL-6 based on the binding regions of receptors and proinflammatory cytokines. In this study, proinflammatory cytokines were used to stimulate human monocytes to trigger an inflammatory response, and the anti-inflammatory ability of the multifunctional KCF18 peptide was further investigated. Cell experiments demonstrated that KCF18 significantly reduced the binding of proinflammatory cytokines to their cognate receptors and inhibited the mRNA and protein expressions of TNF-α, IL-1β, and IL-6. It could also reduce the expression of reactive oxygen species induced by cytokines in human monocytes. KCF18 could effectively decrease the p65 nucleus translocation induced by cytokines, and a mice endotoxemia experiment demonstrated that KCF18 could reduce the expression of IL-6 and the increase of white blood cells in the blood stimulated by lipopolysaccharides. According to our study of tissue sections, KCF18 alleviated liver inflammation. By reducing the release of cytokines in plasma and directly affecting vascular cells, KCF18 is believed to significantly reduce the risk of vascular inflammation.
Collapse
Affiliation(s)
- Chun-Chun Chang
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hao-Hsiang Tsao
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hsin-Ting Huang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Xing-Yan Lai
- Department of Life Sciences, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hao-Jen Hsu
- Department of Life Sciences, College of Medicine, Tzu Chi University, Hualien, Taiwan
- *Correspondence: Hao-Jen Hsu, ; Shinn-Jong Jiang,
| | - Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
- *Correspondence: Hao-Jen Hsu, ; Shinn-Jong Jiang,
| |
Collapse
|
45
|
Anti-Inflammatory Functions of Methanol Extract from Malus baccata (L.) Borkh. Leaves and Shoots by Targeting the NF-κB Pathway. PLANTS 2022; 11:plants11050646. [PMID: 35270116 PMCID: PMC8912290 DOI: 10.3390/plants11050646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023]
Abstract
Malus baccata (L.) Borkh. is a widely used medical plant in Asia. Since the anti-inflammatory mechanism of this plant is not fully understood, the aim of this study was to explore the anti-inflammatory function and mechanism of Malus baccata (L.) Borkh. methanol extract (Mb-ME). For in vitro experiments, nitric oxide production assay, PCR, overexpression strategy, immunoblotting, luciferase reporter assay, and immunoprecipitation were employed to explore the molecular mechanism and the target proteins of Mb-ME. For in vivo experiments, an HCl/EtOH-induced gastritis mouse model was used to confirm the anti-inflammatory function. Mb-ME showed a strong ability to inhibit the production of nitric oxide and the expression of inflammatory genes. Mb-ME decreased NF-κB luciferase activity mediated by MyD88 and TRIF. Moreover, Mb-ME blocked the activation of Src, Syk, p85, Akt, p50, p60, IKKα/β, and IκBα in LPS-induced RAW264.7 cells. Overexpression and immunoprecipitation analyses suggested Syk and Src as the target enzymes of Mb-ME. In vitro results showed that Mb-ME could alleviate gastritis and relieve the protein expression of p-Src, p-Syk, and COX-2, as well as the gene expression of COX-2 and TNF-α. In summary, this study implied that Mb-ME performs an anti-inflammatory role by suppressing Syk and Src in the NF-κB signaling pathway, both in vivo and in vitro.
Collapse
|
46
|
Protective Effects of the Wenfei Buqi Tongluo Formula on the Inflammation in Idiopathic Pulmonary Fibrosis through Inhibiting the TLR4/MyD88/NF-κB Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8752325. [PMID: 35178456 PMCID: PMC8843962 DOI: 10.1155/2022/8752325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/14/2021] [Accepted: 12/28/2021] [Indexed: 11/17/2022]
Abstract
Background. Idiopathic pulmonary fibrosis (IPF) is a progressive disease with high mortality and poor prognosis. The prognostic signatures related to conventional therapy response remain limited. The Wenfei Buqi Tongluo (WBT) formula, a traditional Chinese medicine (TCM) formula, has been widely utilized to treat respiratory diseases in China, which is particularly effective in promoting inflammatory absorption. In this study, we aim to explore the mechanism of the WBT formula in the inhibition of inflammatory response during IPF, based on network pharmacology and in vivo experiments. Methods. Network pharmacology was applied to predict the changes of biological processes and potential pathways for the WBT formula against IPF. Histopathological changes, inflammatory factors (IL-6, IL-1β, and TNF-α), and the proteins of the TLR4/MyD88/NF-κB pathway in bleomycin- (BLM-) induced mice model were examined by hematoxylin-eosin (H&E), Masson or immunohistochemistry staining, Western blot, and enzyme-linked immunosorbent assay analysis. Results. A total of 163 possible components and 167 potential targets between the WBT formula and IPF were obtained. The enrichments of network pharmacology showed that inflammation response, TNF, and NF-κB pathways were involved in the treatment of WBT against IPF. The in vivo experiments indicated that the WBT formula could ameliorate inflammatory exudation and collagen deposition at a histopathology level in the BLM-induced mice model. The levels of IL-6, IL-1β, and TNF-α were reduced after the WBT formula treatment. Moreover, the expressions of phosphorylated-NF-κB p65, TLR4, and MyD88 were significantly downregulated by the WBT formula, compared with the BLM-induced group. Conclusion. These results indicated that the WBT formula can suppress BLM-induced IPF in a mouse model by inhibiting the inflammation via the TLR4/MyD88/NF-κB pathway. This study provides a new insight into the molecular mechanisms of the WBT formula in the application at the clinic.
Collapse
|
47
|
Li CQ, Sun QX, Xu SY, Li LD, Xiao H, Zhang QN. Nebulized Mycobacterium vaccae protects against asthma by attenuating the imbalance of IRF4/IRF8 expression in dendritic cells. Asian Pac J Trop Biomed 2022. [DOI: 10.4103/2221-1691.363878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
48
|
Hernandez-Leon A, Moreno-Pérez GF, Martínez-Gordillo M, Aguirre-Hernández E, Valle-Dorado MG, Díaz-Reval MI, González-Trujano ME, Pellicer F. Lamiaceae in Mexican Species, a Great but Scarcely Explored Source of Secondary Metabolites with Potential Pharmacological Effects in Pain Relief. Molecules 2021; 26:7632. [PMID: 34946714 PMCID: PMC8705283 DOI: 10.3390/molecules26247632] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/05/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023] Open
Abstract
The search for molecules that contribute to the relief of pain is a field of research in constant development. Lamiaceae is one of the most recognized families world-wide for its use in traditional medicine to treat diseases that include pain and inflammation. Mexico can be considered one of the most important centers of diversification, and due to the high endemism of this family, it is crucial for the in situ conservation of this family. Information about the most common genera and species found in this country and their uses in folk medicine are scarcely reported in the literature. After an extensive inspection in bibliographic databases, mainly Sciencedirect, Pubmed and Springer, almost 1200 articles describing aspects of Lamiaceae were found; however, 217 articles were selected because they recognize the Mexican genera and species with antinociceptive and/or anti-inflammatory potential to relieve pain, such as Salvia and Agastache. The bioactive constituents of these genera were mainly terpenes (volatile and non-volatile) and phenolic compounds such as flavonoids (glycosides and aglycone). The aim of this review is to analyze important aspects of Mexican genera of Lamiaceae, scarcely explored as a potential source of secondary metabolites responsible for the analgesic and anti-inflammatory properties of these species. In addition, we point out the possible mechanisms of action involved and the modulatory pathways investigated in different experimental models. As a result of this review, it is important to mention that scarce information has been reported regarding species of this family from Mexican genera. In fact, despite Calosphace being one of the largest subgenera of Salvia in the world, found mainly in Mexico, it has been barely investigated regarding its potential biological activities and recognized bioactive constituents. The scientific evidence regarding the different bioactive constituents found in species of Lamiaceae demonstrates that several species require further investigation in preclinical studies, and of course also in controlled clinical trials evaluating the efficacy and safety of these natural products to support their therapeutic potential in pain relief and/or inflammation, among other health conditions. Since Mexico is one of the most important centers of diversification, and due to the high endemism of species of this family, it is crucial their rescue, in situ conservation, and investigation of their health benefits.
Collapse
Affiliation(s)
- Alberto Hernandez-Leon
- Laboratorio de Neurofarmacología de Productos Naturales, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (A.H.-L.); (G.F.M.-P.); (F.P.)
| | - Gabriel Fernando Moreno-Pérez
- Laboratorio de Neurofarmacología de Productos Naturales, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (A.H.-L.); (G.F.M.-P.); (F.P.)
- Programa de Posgrado en Ciencias Biológicas, Facultad de Medicina, Universidad Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - Martha Martínez-Gordillo
- Herbario de la Facultad de Ciencias, Departamento de Biología Comparada, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico;
| | - Eva Aguirre-Hernández
- Laboratorio de Productos Naturales, Departamento de Ecología y Recursos Naturales, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico;
| | - María Guadalupe Valle-Dorado
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico;
| | - María Irene Díaz-Reval
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Mexico;
| | - María Eva González-Trujano
- Laboratorio de Neurofarmacología de Productos Naturales, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (A.H.-L.); (G.F.M.-P.); (F.P.)
| | - Francisco Pellicer
- Laboratorio de Neurofarmacología de Productos Naturales, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (A.H.-L.); (G.F.M.-P.); (F.P.)
| |
Collapse
|
49
|
Potential of cell-penetrating peptides (CPPs) in delivery of antiviral therapeutics and vaccines. Eur J Pharm Sci 2021; 169:106094. [PMID: 34896590 DOI: 10.1016/j.ejps.2021.106094] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
Viral infections are a great threat to human health. Currently, there are no effective vaccines and antiviral drugs against the majority of viral diseases, suggesting the need to develop novel and effective antiviral agents. Since the intracellular delivery of antiviral agents, particularly the impermeable molecules, such as peptides, proteins, and nucleic acids, are essential to exert their therapeutic effects, using a delivery system is highly required. Among various delivery systems, cell-penetrating peptides (CPPs), a group of short peptides with the unique ability of crossing cell membrane, offer great potential for the intracellular delivery of various biologically active cargoes. The results of numerous in vitro and in vivo studies with CPP conjugates demonstrate their promise as therapeutic agents in various medical fields including antiviral therapy. The CPP-mediated delivery of various antiviral agents including peptides, proteins, nucleic acids, and nanocarriers have been associated with therapeutic efficacy both in vitro and in vivo. This review describes various aspects of viruses including their biology, pathogenesis, and therapy and briefly discusses the concept of CPP and its potential in drug delivery. Particularly, it will highlight a variety of CPP applications in the management of viral infections.
Collapse
|
50
|
Jin J, Zhong Y, Long J, Wu T, Jiang Q, Wang H, Ge W, Zhao H, Liu D. Ginsenoside Rg1 relieves experimental colitis by regulating balanced differentiation of Tfh/Treg cells. Int Immunopharmacol 2021; 100:108133. [PMID: 34543978 DOI: 10.1016/j.intimp.2021.108133] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/21/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Inflammatory bowel disease (IBD) is typically characterized by the dysregulation of Tfh cell differentiation. we sought to explore the potential mechanism of Ginsenoside Rg1 (G-Rg1) treated IBD by observing the level of the Tfh/Treg cells and the activation of PI3K/Akt signaling pathway in the colitis mice. In the present study, G-Rg1 significantly inhibited the inflammatory response to mice colitis induced by dextran sodium sulfate (DSS), as evidenced by increased body weight and colon length, decreased colon weight, reduced colon weight index and histopathological scores, lower levels of IL-6 and TNF-α, and increased IL-10 levels. Significantly, G-Rg1 effectively decreased the amounts of CD4+CXCR5+IL-9+(Tfh9), CD4+ CXCR5+IL-17+(Tfh17), and increased CD4+CXCR5+Foxp3+(Tfr) and CD4+CD25+ Foxp3+(Treg) cells. Furthermore, G-Rg1 markedly down-regulated PI3K and p-Akt level, and upregulated PTEN expression. These results indicated that G-Rg1 could effectively regulate the balance of Tfh/Treg cells to relieve experimental colitis, which could be potentially related to PI3K/Akt signaling pathway inhibition.
Collapse
Affiliation(s)
- Jing Jin
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Youbao Zhong
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Jian Long
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Tiantian Wu
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Qingqing Jiang
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Haiyan Wang
- Party and School Office, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wei Ge
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - Haimei Zhao
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| | - Duanyong Liu
- Science and Technology College, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| |
Collapse
|