1
|
Xing L, Li H, Miao D, Wei H, Zhang S, Xue Q, Wang H, Li J. Intermittent and mild cold stimulation enhances immune function of broilers via co-regulation of CIRP and TRPM8 on NF-κB and MAPK signaling pathways. Poult Sci 2024; 103:103984. [PMID: 38986357 PMCID: PMC11296020 DOI: 10.1016/j.psj.2024.103984] [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: 02/16/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
Improving immune function is an important indicator for establishing cold adaptation in broilers. In the study, to explore the effects and molecular mechanisms of intermittent and mild cold stimulation (IMCS) on the immune function of broilers, CIRP and TRPM8, induced by cold stimulation, as well as the NF-κB and MAPK pathways which play an important role in immune response, were selected to investigate. A total of 192 one-day-old broilers (Ross 308) were selected and randomly divided into the control group (CC) and the cold stimulation group (CS). The broilers in CC were raised at normal feeding temperature from d 1 to 43, while the broilers in CS were subjected to cold stimulation from day 15 to 35, with a temperature 3 °C below that of the CC group for 5 h, at 1 d intervals. The results showed that IMCS had little effect on the broiler hearts, and the myocardial structure was not damaged. On d 22, IMCS significantly increased the mRNA levels of CIRP, TRPM8, P65, P38, COX-2, TNF-α, IFN- γ, IL-6, IL-10, and the protein levels of CIRP, P65, P38, IL-1β and iNOS in the hearts, and the levels of CIRP and all cytokines in the serum (P ≤ 0.05). The mRNA and protein levels of IκB-α were significantly reduced (P ≤ 0.05). On d 36, the mRNA levels of TRPM8, P65, ERK, and IL-10 in the hearts and the content of COX-2 in the serum in CS were increased significantly (P ≤ 0.05), while the mRNA levels of IκB-α, P38, and IL-1β were decreased significantly (P ≤ 0.05). On d 43, IMCS significantly upregulated the mRNA levels of TRPM8, IFN- γ, IL-4, IL-6, IL-10, and the protein levels of IκB-α, P38, and the levels of iNOS, TNF-α, IL6 and IL10 in the serum (P ≤ 0.05); whereas it significantly downregulated CIRP, JNK, P38, iNOS, TNF-α mRNA levels, and CIRP, P65, ERK, JNK, IL1β and iNOS protein levels (P ≤ 0.05). Therefore, IMCS can enhance broiler immune function through co-regulation of CIRP and TRPM8 on the NF-κB and MAPK pathways, which facilitate the cold adaptation in broilers.
Collapse
Affiliation(s)
- Lu Xing
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Haochen Li
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Deyang Miao
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Haidong Wei
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Shijie Zhang
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Qiang Xue
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Hongyu Wang
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, China.
| |
Collapse
|
2
|
Ding C, Gai S, Ma Z, Yang L, Fu Z. The role of potassium ion channels in chronic sinusitis. Front Pharmacol 2024; 15:1431330. [PMID: 39015366 PMCID: PMC11249563 DOI: 10.3389/fphar.2024.1431330] [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: 05/11/2024] [Accepted: 06/12/2024] [Indexed: 07/18/2024] Open
Abstract
Chronic sinusitis is a common inflammatory disease of the nasal and sinus mucosa, leading to symptoms such as nasal congestion, runny nose, decreased sense of smell, and headache. It often recurs and seriously affects the quality of life of patients. However, its pathological and physiological mechanisms are not fully understood. In recent years, the role of potassium ion channels in the regulation of mucosal barrier function and inflammatory cell function has received increasing attention. In chronic sinusitis, there are often changes in the expression and function of potassium channels, leading to mucosal damage and a stronger inflammatory response. However, the related research is still in its early stages. This article will review the role of the potassium channel in the pathological and physiological changes of chronic sinusitis. The studies revealed that BK/TREK-1 potassium channel play a protective role in the nasal mucosal function through p38-MAPK pathway, and KCa3.1/Kv1.3 enhance the inflammatory response of Chronic rhinosinusitis by regulating immune cell function, intracellular Ca2+ signaling and ERK/MAPK/NF-κB pathway. Because ion channels are surface proteins of cell membranes, they are easier to intervene with drugs, and the results of these studies may provide new effective targets for the prevention and treatment of chronic sinusitis.
Collapse
Affiliation(s)
- Changhui Ding
- Department of Otorhinolaryngology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Senxi Gai
- Department of Otorhinolaryngology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhiyong Ma
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lizhuo Yang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhijie Fu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
3
|
Cheng S, Jiang D, Lan X, Liu K, Fan C. Voltage-gated potassium channel 1.3: A promising molecular target in multiple disease therapy. Biomed Pharmacother 2024; 175:116651. [PMID: 38692062 DOI: 10.1016/j.biopha.2024.116651] [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: 02/20/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
Voltage-gated potassium channel 1.3 (Kv1.3) has emerged as a pivotal player in numerous biological processes and pathological conditions, sparking considerable interest as a potential therapeutic target across various diseases. In this review, we present a comprehensive examination of Kv1.3 channels, highlighting their fundamental characteristics and recent advancements in utilizing Kv1.3 inhibitors for treating autoimmune disorders, neuroinflammation, and cancers. Notably, Kv1.3 is prominently expressed in immune cells and implicated in immune responses and inflammation associated with autoimmune diseases and chronic inflammatory conditions. Moreover, its aberrant expression in certain tumors underscores its role in cancer progression. While preclinical studies have demonstrated the efficacy of Kv1.3 inhibitors, their clinical translation remains pending. Molecular imaging techniques offer promising avenues for tracking Kv1.3 inhibitors and assessing their therapeutic efficacy, thereby facilitating their development and clinical application. Challenges and future directions in Kv1.3 inhibitor research are also discussed, emphasizing the significant potential of targeting Kv1.3 as a promising therapeutic strategy across a spectrum of diseases.
Collapse
Affiliation(s)
- Sixuan Cheng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Kun Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Cheng Fan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
4
|
Wang S, Su W, Wu X, Dong W. Restoring Treg/Th17 cell balance in ulcerative colitis through HRas silencing and MAPK pathway inhibition. Int Immunopharmacol 2024; 130:111608. [PMID: 38428143 DOI: 10.1016/j.intimp.2024.111608] [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: 10/06/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 03/03/2024]
Abstract
This study investigates HRas-dependent mechanisms in the disruption of regulatory T (Treg) cells and T helper 17 (Th17) cells balance in ulcerative colitis (UC). Comprehensive RNA sequencing and bioinformatics analyses revealed elevated HRas and MAPK pathway-related protein expression in UC samples. Using a murine UC model induced by dextran sulfate sodium (DSS), HRas silencing was found to promote Treg cell differentiation and suppress Th17 cell production, effectively restoring balance. Inactivation of the MAPK pathway played a pivotal role in this rebalancing effect. In vivo experiments further confirmed that HRas silencing mitigated colon tissue damage in DSS-induced mice, emphasizing its potential as a therapeutic strategy for UC.
Collapse
Affiliation(s)
- Siwei Wang
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, PR China
| | - Wenhao Su
- Department of Gastroenterology, Renmin Hospital of Wuhan University,Wuhan 430060, PR China
| | - Xiaohan Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University,Wuhan 430060, PR China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University,Wuhan 430060, PR China.
| |
Collapse
|
5
|
Liu L, Xu W, Li K, Hu Y, Shen L, Zhang H, Wang Y. Kv1.3 mediates ox-LDL-induced vascular smooth muscle cell proliferation through JAK2/STAT3 signaling pathway. Arch Biochem Biophys 2023; 746:109719. [PMID: 37591369 DOI: 10.1016/j.abb.2023.109719] [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: 05/26/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/19/2023]
Abstract
Kv1.3 channel has been shown to participate in regulating inflammatory activation, proliferation and apoptosis in several cell types. However, most of those existing studies focused on the ion-conducting properties of Kv1.3 in maintaining the resting potential and regulating Ca2+ influx. The aim of our study was to explore whether the Kv1.3-JAK2/STAT3 signaling pathway was involved in oxidized low density lipoprotein (ox-LDL) induced vascular smooth muscle cell (VSMC) proliferation. VSMCs from mouse aorta were cultured and treated with ox-LDL (25 μg/mL). The cell counting kit-8 was used to assess cell proliferation, and western blotting was performed to detect expression levels of Kv1.3, JAK2/STAT3, phosphorylated JAK2/STAT3, cyclin B1 and cyclin D1 in treated VSMCs. VSMCs were transfected with Kv1.3 small interfering RNA (Kv1.3-siRNA) or infected with a Kv1.3 lentiviral expression vector (Lv-Kv1.3) and treated with a JAK2 inhibitor LY2784544 to assess the role of Kv1.3 and JAK2/STAT3 signaling in mediating VSMC proliferation induced by ox-LDL. Ox-LDL induced cell proliferation and upregulated the expression of Kv1.3 in mouse VSMCs. In VSMCs transfected with Kv1.3-siRNA, ox-LDL was not efficient in inducing cell proliferation or the levels of proliferation associated proteins, cyclin B1 and cyclin D1. However, cell proliferation, cyclin B1 and cyclin D1 levels increased in VSMCs infected with Lv-Kv1.3. Levels of phosphorylated JAK2 and STAT3 were increased in ox-LDL-treated VSMCs, and this increase was prevented in VSMCs transfected with Kv1.3-siRNA. Treatment with the JAK2 inhibitor LY2784544 also prevented the increase in VSMCs proliferation treated with ox-LDL. Our findings demonstrated that Kv1.3 promoted proliferation of VSMCs treated with ox-LDL, and that this effect might be mediated through activation of the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Lin Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Ji'nan, 250012, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Ji'nan, 250012, China
| | - Wei Xu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, 250012, China
| | - Kaiwen Li
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Ji'nan, 250012, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Ji'nan, 250012, China
| | - Yanyan Hu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Ji'nan, 250012, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Ji'nan, 250012, China
| | - Lin Shen
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Ji'nan, 250012, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Ji'nan, 250012, China
| | - Hongyu Zhang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Ji'nan, 250012, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Ji'nan, 250012, China
| | - Yuanyuan Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Ji'nan, 250012, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Ji'nan, 250012, China.
| |
Collapse
|
6
|
Zhang Q, Liu L, Hu Y, Shen L, Li L, Wang Y. Kv1.3 Channel Is Involved In Ox-LDL-induced Macrophage Inflammation Via ERK/NF-κB signaling pathway. Arch Biochem Biophys 2022; 730:109394. [PMID: 36100082 DOI: 10.1016/j.abb.2022.109394] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022]
Abstract
Macrophage inflammatory response is crucial for the initiation and progression of atherosclerosis. The voltage-gated potassium channel Kv1.3 plays an important role in the modulation of macrophage function. The aim of this study was to investigate the effect and possible mechanism of Kv1.3 on inflammation in oxidized low-density lipoprotein (ox-LDL)-induced RAW264.7 macrophages. Treatment with Kv1.3-siRNA attenuated the expression of IL-6 and TNF-α and reduced the phosphorylation of ERK1/2 and NF-κB in ox-LDL-induced macrophages. In contrast, overexpression of Kv1.3 with Lv-Kv1.3 promoted the expression of IL-6 and TNF-α, and increased ERK1/2 and NF-κB phosphorylation in macrophages. PD-98059, a specific inhibitor of ERK, reversed the expression of IL-6 and TNF-α in ox-LDL-treated macrophages. Kv1.3-siRNA did not inhibit inflammation any further when cells were treated with PD-98059. This suggests that ERK acts as a downstream regulator of the Kv1.3 channel. In conclusion, Kv1.3 may be an indispensable membrane protein in ox-LDL-induced RAW264.7 macrophage inflammation in atherosclerosis through the ERK/NF-κB pathway.
Collapse
Affiliation(s)
- Qiujie Zhang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China
| | - Lin Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China
| | - Yanyan Hu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China
| | - Lin Shen
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China
| | - Li Li
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Qilu Hospital of Shandong University, China
| | - Yuanyuan Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, China; Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University, China.
| |
Collapse
|
7
|
Kang YM, Kim HM, Lee M, An HJ. Effects of Echinocystic Acid on Atopic Dermatitis and Allergic Inflammation of the Skin and Lungs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154211. [PMID: 35717808 DOI: 10.1016/j.phymed.2022.154211] [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: 12/29/2021] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Echinocystic acid (ECA), a pentacyclic triterpene enriched in various herbs, promotes anti-inflammatory and antioxidant activity; however, its therapeutic effects on atopic dermatitis (AD) or atopic march and the underlying mechanisms of action have not yet been fully elucidated. PURPOSE This study aimed to elucidate the effects and molecular mechanisms of ECA on AD and allergic inflammation. METHODS We evaluated the inhibitory effects of ECA using a house dust mite (HDM)-induced AD mouse model and human keratinocytes. RESULTS The results revealed that ECA improved AD symptoms by decreasing epidermal/dermal thickness, immune cell infiltration, and restoring skin barrier function, as well as an imbalanced immune response. In addition, repeated epicutaneous HDM challenges aggravated allergic inflammation in mice lungs, which was caused by the infiltration of immune cells and collagen deposition, whereas ECA alleviated these symptoms. Moreover, ECA suppressed the expression of T helper cell-derived cytokines, phosphorylation of extracellular signal-regulated kinase, and signal transducer and activator of transcription 1 in the skin and lungs of mice with HDM-induced AD, as well as inhibited the translocation of nuclear factor-κB in HaCaT keratinocytes. CONCLUSION This is the meaningful study to demonstrate that ECA improves allergic inflammation of the skin and lungs through recovery of the skin barrier, regulation of immune balance, and alleviation of lung inflammation, suggesting that ECA has therapeutic potential as an antiatopic and antiallergic agent that blocks the progression of AD to atopic march.
Collapse
Affiliation(s)
- Yun-Mi Kang
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea; Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Hye-Min Kim
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, Republic of Korea.
| | - Hyo-Jin An
- Department of Pharmacology, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea.
| |
Collapse
|
8
|
Jiang Y, Yan Q, Liu CX, Peng CW, Zheng WJ, Zhuang HF, Huang HT, Liu Q, Liao HL, Zhan SF, Liu XH, Huang XF. Insights into potential mechanisms of asthma patients with COVID-19: A study based on the gene expression profiling of bronchoalveolar lavage fluid. Comput Biol Med 2022; 146:105601. [PMID: 35751199 PMCID: PMC9117163 DOI: 10.1016/j.compbiomed.2022.105601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/21/2022]
Abstract
Background The 2019 novel coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is currently a major challenge threatening the global healthcare system. Respiratory virus infection is the most common cause of asthma attacks, and thus COVID-19 may contribute to an increase in asthma exacerbations. However, the mechanisms of COVID-19/asthma comorbidity remain unclear. Methods The “Limma” package or “DESeq2” package was used to screen differentially expressed genes (DEGs). Alveolar lavage fluid datasets of COVID-19 and asthma were obtained from the GEO and GSV database. A series of analyses of common host factors for COVID-19 and asthma were conducted, including PPI network construction, module analysis, enrichment analysis, inference of the upstream pathway activity of host factors, tissue-specific analysis and drug candidate prediction. Finally, the key host factors were verified in the GSE152418 and GSE164805 datasets. Results 192 overlapping host factors were obtained by analyzing the intersection of asthma and COVID-19. FN1, UBA52, EEF1A1, ITGB1, XPO1, NPM1, EGR1, EIF4E, SRSF1, CCR5, PXN, IRF8 and DDX5 as host factors were tightly connected in the PPI network. Module analysis identified five modules with different biological functions and pathways. According to the degree values ranking in the PPI network, EEF1A1, EGR1, UBA52, DDX5 and IRF8 were considered as the key cohost factors for COVID-19 and asthma. The H2O2, VEGF, IL-1 and Wnt signaling pathways had the strongest activities in the upstream pathways. Tissue-specific enrichment analysis revealed the different expression levels of the five critical host factors. LY294002, wortmannin, PD98059 and heparin might have great potential to evolve into therapeutic drugs for COVID-19 and asthma comorbidity. Finally, the validation dataset confirmed that the expression of five key host factors were statistically significant among COVID-19 groups with different severity and healthy control subjects. Conclusions This study constructed a network of common host factors between asthma and COVID-19 and predicted several drugs with therapeutic potential. Therefore, this study is likely to provide a reference for the management and treatment for COVID-19/asthma comorbidity.
Collapse
Affiliation(s)
- Yong Jiang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, China.
| | - Qian Yan
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, China.
| | - Cheng-Xin Liu
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, China.
| | - Chen-Wen Peng
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, China.
| | - Wen-Jiang Zheng
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, China.
| | - Hong-Fa Zhuang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China.
| | - Hui-Ting Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China.
| | - Qiong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China.
| | - Hui-Li Liao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China.
| | - Shao-Feng Zhan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China.
| | - Xiao-Hong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China.
| | - Xiu-Fang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China.
| |
Collapse
|
9
|
Zeng Q, Lu W, Deng Z, Zhang B, Wu J, Chai J, Chen X, Xu X. The toxin mimic FS48 from the salivary gland of Xenopsylla cheopis functions as a Kv1.3 channel-blocking immunomodulator of T cell activation. J Biol Chem 2022; 298:101497. [PMID: 34919963 PMCID: PMC8732088 DOI: 10.1016/j.jbc.2021.101497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/14/2021] [Accepted: 12/10/2021] [Indexed: 11/29/2022] Open
Abstract
The Kv1.3 channel has been widely demonstrated to play crucial roles in the activation and proliferation of T cells, which suggests that selective blockers could serve as potential therapeutics for autoimmune diseases mediated by T cells. We previously described that the toxin mimic FS48 from salivary gland of Xenopsylla cheopis downregulates the secretion of proinflammatory factors by Raw 264.7 cells by blocking the Kv1.3 channel and the subsequent inactivation of the proinflammatory MAPK/NF-κB pathways. However, the effects of FS48 on human T cells and autoimmune diseases are unclear. Here, we described its immunomodulatory effects on human T cells derived from suppression of Kv1.3 channel. Kv1.3 currents in Jurkat T cells were recorded by whole-cell patch-clamp, and Ca2+ influx, cell proliferation, and TNF-α and IL-2 secretion were measured using Fluo-4, CCK-8, and ELISA assays, respectively. The in vivo immunosuppressive activity of FS48 was evaluated with a rat DTH model. We found that FS48 reduced Kv1.3 currents in Jurkat T cells in a concentration-dependent manner with an IC50 value of about 1.42 μM. FS48 also significantly suppressed Kv1.3 protein expression, Ca2+ influx, MAPK/NF-κB/NFATc1 pathway activation, and TNF-α and IL-2 production in activated Jurkat T cells. Finally, we show that FS48 relieved the DTH response in rats. We therefore conclude that FS48 can block the Kv1.3 channel and inhibit human T cell activation, which most likely contributes to its immunomodulatory actions and highlights the great potential of this evolutionary-guided peptide as a drug template in future studies.
Collapse
Affiliation(s)
- Qingye Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wancheng Lu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Zhenhui Deng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Bei Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Wang C, Huang CF, Li M. Sodium houttuynia alleviates airway inflammation in asthmatic mice by regulating FoxP3/RORγT expression and reversing Treg/Th17 cell imbalance. Int Immunopharmacol 2021; 103:108487. [PMID: 34959187 DOI: 10.1016/j.intimp.2021.108487] [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: 10/29/2021] [Revised: 12/03/2021] [Accepted: 12/16/2021] [Indexed: 11/05/2022]
Abstract
Synthetized from a natural oil of Houttuynia cordata, sodium houttuynia was reported to have anti-inflammatory effects. The present study aimed to investigate whether sodium houttuynia could alleviate the characteristic airway inflammation and Treg/Th17 cell imbalance of asthma in vivo. Experimental mice with neutrophilic asthma were injected with sodium houttuynia or dexamethasone (alone or in combination) intraperitoneally. The airway reactivity was measured, and bronchoalveolar lavage fluid was collected for cell count. Hematoxylin/eosin and periodic acid-Schiff staining were performed to assess pulmonary inflammation and mucus hypersecretion. Immunohistochemical analysis was conducted to determine the expression of IL-10, IL-17A, FoxP3, and RORγT in the lung tissue, and the serum levels of IL-10 and IL-17A were analyzed by ELISA. The proportion of CD4+CD25+FoxP3+ Treg and Th17 cells within the CD4+ T cell subset of splenocytes was analyzed by flow cytometry. FoxP3 and RORγT mRNA and protein expressions in the lung were analyzed by real-time PCR and western blot, respectively. Overall, sodium houttuynia was found to ameliorate the Treg/Th17 cell imbalance and reduce the airway inflammation, hyperresponsiveness, and mucus hypersecretion by increasing the frequency of CD4+CD25+FoxP3+ Treg cells and the secretion of IL-10, while decreasing the proportion of Th17 cells and IL-17A production. Although the regulatory effect of sodium houttuynia was not as good as that achieved with dexamethasone, combination of the two compounds showed improved inhibitory effects on airway hyperresponsiveness, inflammation, and mucus hypersecretion. Hence, sodium houttuynia may be beneficial for the treatment of asthma.
Collapse
Affiliation(s)
- Chao Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Chen-Feng Huang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Miao Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
11
|
Pak SW, Lee AY, Seo YS, Lee SJ, Kim WI, Shin DH, Kim JC, Kim JS, Lim JO, Shin IS. Anti-asthmatic effects of Phlomis umbrosa Turczaninow using ovalbumin induced asthma murine model and network pharmacology analysis. Biomed Pharmacother 2021; 145:112410. [PMID: 34775237 DOI: 10.1016/j.biopha.2021.112410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Phlomis umbrosa Turczaninow has been used as a tradition herbal medicine for treating various inflammatory diseases. PURPOSE In present study, we explored the effects of P. umbrosa on asthma induced by ovalbumin (OVA) and elucidated the mechanism via in vivo verification and network pharmacology prediction. METHODS The animals were intraperitoneally injected OVA on day 1 and 14, followed by OVA inhalation on days 21, 22, and 23. The animals were daily treated P. umbrosa extract (PUE, 20 and 40 mg/kg) by oral gavage from day 18 to day 23. RESULTS PUE significantly decreased airway hyperresponsiveness, eosinophilia, and the production of inflammatory cytokines and OVA specific immunoglobulin E in animals with asthma, along with a reduction in airway inflammation and mucus secretion in lung tissue. In network analysis, antiasthmatic effects of PUE were closely related with suppression of mitogen-activated protein kinases and matrix metalloproteinases (MMPs). Consistent with the results from network analysis, PUE suppressed the phosphorylation of ERK and p65, which was accompanied by a decline in MMP-9 expression. CONCLUSION Administration of PUE effectively reduced allergic responses in asthmatic mice, which was associated with the suppressed phosphorylation of ERK and p65, and expression of MMP-9. These results indicate that PUE has therapeutic potential to treat allergic asthma.
Collapse
Affiliation(s)
- So-Won Pak
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea
| | - A Yeong Lee
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 111 Geonjae-ro, Naju-si, Jeollanam-do 58245, South Korea
| | - Yun-Soo Seo
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 111 Geonjae-ro, Naju-si, Jeollanam-do 58245, South Korea
| | - Se-Jin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea
| | - Woong-Il Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea
| | - Dong-Ho Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea
| | - Joong-Sun Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea
| | - Je-Oh Lim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea.
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, South Korea.
| |
Collapse
|
12
|
Papain Ameliorates Lipid Accumulation and Inflammation in High-Fat Diet-Induced Obesity Mice and 3T3-L1 Adipocytes via AMPK Activation. Int J Mol Sci 2021; 22:ijms22189885. [PMID: 34576066 PMCID: PMC8468764 DOI: 10.3390/ijms22189885] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/19/2022] Open
Abstract
Papain is a proteolytic enzyme present in the leaves, fruits, roots, and latex of the Carica papaya (papaya) plant. Although it exhibits a wide range of activities, there are no reports on the anti-obesity effects of papain. This study examined the anti-obesity effect and obesity-involved anti-inflammatory mechanism of papain in in vivo and in vitro models using high-fat diet (HFD)-induced obese mice and 3T3-L1 preadipocytes. Oral administration of papain reduced HFD-induced weight of the body, liver, and adipose tissues of mice. Papain also reduced hepatic lipid accumulation and adipocyte size. Moreover, serum total cholesterol and triglyceride levels were markedly reduced in papain-treated mice. In addition, papain inhibited the differentiation of preadipocytes and oil accumulation in 3T3-L1 preadipocytes and rat primary preadipocytes. Mechanistically, papain significantly downregulated the protein levels of key adipogenesis regulators and reversed the expression of pro-inflammatory cytokines and adipokines in HFD-induced obese mice and 3T3-L1 preadipocytes. Papain also markedly enhanced activation of the AMP-activated protein kinase pathway in both models. Collectively, these results suggest that papain exerts anti-obesity effects in HFD-induced mice and 3T3-L1 preadipocytes by regulating levels of adipogenic factors involved in lipid metabolism and inflammation; thus, it could be useful in the prevention and treatment of obesity.
Collapse
|
13
|
Mathie A, Veale EL, Golluscio A, Holden RG, Walsh Y. Pharmacological Approaches to Studying Potassium Channels. Handb Exp Pharmacol 2021; 267:83-111. [PMID: 34195873 DOI: 10.1007/164_2021_502] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this review, we consider the pharmacology of potassium channels from the perspective of these channels as therapeutic targets. Firstly, we describe the three main families of potassium channels in humans and disease states where they are implicated. Secondly, we describe the existing therapeutic agents which act on potassium channels and outline why these channels represent an under-exploited therapeutic target with potential for future drug development. Thirdly, we consider the evidence desired in order to embark on a drug discovery programme targeting a particular potassium channel. We have chosen two "case studies": activators of the two-pore domain potassium (K2P) channel TREK-2 (K2P10.1), for the treatment of pain and inhibitors of the voltage-gated potassium channel KV1.3, for use in autoimmune diseases such as multiple sclerosis. We describe the evidence base to suggest why these are viable therapeutic targets. Finally, we detail the main technical approaches available to characterise the pharmacology of potassium channels and identify novel regulatory compounds. We draw particular attention to the Comprehensive in vitro Proarrhythmia Assay initiative (CiPA, https://cipaproject.org ) project for cardiac safety, as an example of what might be both desirable and possible in the future, for ion channel regulator discovery projects.
Collapse
Affiliation(s)
- Alistair Mathie
- Medway School of Pharmacy, University of Kent, Kent, UK. .,Medway School of Pharmacy, University of Greenwich, London, UK. .,School of Engineering, Arts, Science and Technology, University of Suffolk, Ipswich, UK.
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Alessia Golluscio
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Robyn G Holden
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Yvonne Walsh
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| |
Collapse
|
14
|
Xie Y, Ma J, Yang M, Fan L, Chen W. Extracellular signal-regulated kinase signaling pathway and silicosis. Toxicol Res (Camb) 2021; 10:487-494. [PMID: 34141162 DOI: 10.1093/toxres/tfaa109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/16/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Silicosis is a scarring lung disease caused by inhaling fine particles of crystalline silica in the workplace of many industries. Due to the lack of effective treatment and management, the continued high incidence of silicosis remains a major public health concern worldwide, especially in the developing countries. Till now, related molecular mechanisms underlying silicosis are still not completely understood. Multiple pathways have been reported to be participated in the pathological process of silicosis, and more complex signaling pathways are receiving attention. The activated extracellular signal-regulated kinase (ERK) signaling pathway has been recognized to control some functions in the cell. Recent studies have identified that the ERK signaling pathway contributes to the formation and development of silicosis through regulating the processes of oxidative stress, inflammatory response, proliferation and activation of fibroblasts, epithelial-mesenchymal transformation, autophagy, and apoptosis of cells. In this review article, we summarize the latest findings on the role of ERK signaling pathway in silica-induced experimental models of silicosis, as well as clinical perspectives.
Collapse
Affiliation(s)
- Yujia Xie
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meng Yang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lieyang Fan
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weihong Chen
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
15
|
Deng Z, Zeng Q, Tang J, Zhang B, Chai J, Andersen JF, Chen X, Xu X. Anti-inflammatory effects of FS48, the first potassium channel inhibitor from the salivary glands of the flea Xenopsylla cheopis. J Biol Chem 2021; 296:100670. [PMID: 33864815 PMCID: PMC8131326 DOI: 10.1016/j.jbc.2021.100670] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 11/29/2022] Open
Abstract
The voltage-gated potassium (Kv) 1.3 channel plays a crucial role in the immune responsiveness of T-lymphocytes and macrophages, presenting a potential target for treatment of immune- and inflammation related-diseases. FS48, a protein from the rodent flea Xenopsylla cheopis, shares the three disulfide bond feature of scorpion toxins. However, its three-dimensional structure and biological function are still unclear. In the present study, the structure of FS48 was evaluated by circular dichroism and homology modeling. We also described its in vitro ion channel activity using patch clamp recording and investigated its anti-inflammatory activity in LPS-induced Raw 264.7 macrophage cells and carrageenan-induced paw edema in mice. FS48 was found to adopt a common αββ structure and contain an atypical dyad motif. It dose-dependently exhibited the Kv1.3 channel in Raw 264.7 and HEK 293T cells, and its ability to block the channel pore was demonstrated by the kinetics of activation and competition binding with tetraethylammonium. FS48 also downregulated the secretion of proinflammatory molecules NO, IL-1β, TNF-α, and IL-6 by Raw 264.7 cells in a manner dependent on Kv1.3 channel blockage and the subsequent inactivation of the MAPK/NF-κB pathways. Finally, we observed that FS48 inhibited the paw edema formation, tissue myeloperoxidase activity, and inflammatory cell infiltrations in carrageenan-treated mice. We therefore conclude that FS48 identified from the flea saliva is a novel potassium channel inhibitor displaying anti-inflammatory activity. This discovery will promote understanding of the bloodsucking mechanism of the flea and provide a new template molecule for the design of Kv1.3 channel blockers.
Collapse
Affiliation(s)
- Zhenhui Deng
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Qingye Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Tang
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bei Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - John F Andersen
- Laboratory of Malaria and Vector Research, NIAID, National Intitutes of Health, Bethesda, Maryland, USA
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
16
|
Hernandez-Santini AC, Mitha AN, Chow D, Hamed MF, Gucwa AL, Vaval V, Martinez LR. Methamphetamine facilitates pulmonary and splenic tissue injury and reduces T cell infiltration in C57BL/6 mice after antigenic challenge. Sci Rep 2021; 11:8207. [PMID: 33859291 PMCID: PMC8050260 DOI: 10.1038/s41598-021-87728-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/01/2021] [Indexed: 12/21/2022] Open
Abstract
Methamphetamine (METH) is a strong addictive central nervous system stimulant. METH abuse can alter biological processes and immune functions necessary for host defense. The acquisition and transmission of HIV, hepatitis, and other communicable diseases are possible serious infectious consequences of METH use. METH also accumulates extensively in major organs. Despite METH being a major public health and safety problem globally, there are limited studies addressing the impact of this popular recreational psychostimulant on tissue adaptive immune responses after exposure to T cell dependent [ovalbumin (OVA)] and independent [lipopolysaccharide (LPS)] antigens. We hypothesized that METH administration causes pulmonary and splenic tissue alterations and reduces T cell responses to OVA and LPS in vivo, suggesting the increased susceptibility of users to infection. Using a murine model of METH administration, we showed that METH causes tissue injury, apoptosis, and alters helper and cytotoxic T cell recruitment in antigen challenged mice. METH also reduces the expression and distribution of CD3 and CD28 molecules on the surface of human Jurkat T cells. In addition, METH decreases the production of IL-2 in these T-like cells, suggesting a negative impact on T lymphocyte activation and proliferation. Our findings demonstrate the pleotropic effects of METH on cell-mediated immunity. These alterations have notable implications on tissue homeostasis and the capacity of the host to respond to infection.
Collapse
Affiliation(s)
| | - Anum N Mitha
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, USA
| | - Daniela Chow
- Department of Biological Sciences, The Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA
| | - Mohamed F Hamed
- Department of Oral Biology, University of Florida College of Dentistry, 1395 Center Drive, Room DG-48, P.O. Box 100424, Gainesville, FL, 32610, USA.,Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Azad L Gucwa
- Department of Biology, Farmingdale State College, Farmingdale, NY, USA
| | - Valerie Vaval
- Department of Biomedical Sciences, Long Island University, C. W. Post, Brookville, NY, USA
| | - Luis R Martinez
- Department of Oral Biology, University of Florida College of Dentistry, 1395 Center Drive, Room DG-48, P.O. Box 100424, Gainesville, FL, 32610, USA.
| |
Collapse
|
17
|
Uhlorn JA, Husband NA, Romero‐Aleshire MJ, Moffett C, Lindsey ML, Langlais PR, Brooks HL. CD4 + T Cell-Specific Proteomic Pathways Identified in Progression of Hypertension Across Postmenopausal Transition. J Am Heart Assoc 2021; 10:e018038. [PMID: 33410333 PMCID: PMC7955317 DOI: 10.1161/jaha.120.018038] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
Background Menopause is associated with an increase in the prevalence and severity of hypertension in women. Although premenopausal females are protected against T cell-dependent immune activation and development of angiotensin II (Ang II) hypertension, this protection is lost in postmenopausal females. Therefore, the current study hypothesized that specific CD4+ T cell pathways are regulated by sex hormones and Ang II to mediate progression from premenopausal protection to postmenopausal hypertension. Methods and Results Menopause was induced in C57BL/6 mice via repeated 4-vinylcyclohexene diepoxide injections, while premenopausal females received sesame oil vehicle. A subset of premenopausal mice and all menopausal mice were infused with Ang II for 14 days (Control, Ang II, Meno/Ang II). Proteomic and phosphoproteomic profiles of CD4+ T cells isolated from spleens were examined. Ang II markedly increased CD4+ T cell protein abundance and phosphorylation associated with DNA and histone methylation in both premenopausal and postmenopausal females. Compared with premenopausal T cells, Ang II infusion in menopausal mice increased T cell phosphorylation of MP2K2, an upstream regulator of ERK, and was associated with upregulated phosphorylation at ERK targeted sites. Additionally, Ang II infusion in menopausal mice decreased T cell phosphorylation of TLN1, a key regulator of IL-2Rα and FOXP3 expression. Conclusions These findings identify novel, distinct T cell pathways that influence T cell-mediated inflammation during postmenopausal hypertension.
Collapse
Affiliation(s)
- Joshua A. Uhlorn
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| | | | | | - Caitlin Moffett
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| | - Merry L. Lindsey
- Department of Cellular and Integrative PhysiologyCenter for Heart and Vascular ResearchNebraska‐Western Iowa Health Care SystemUniversity of Nebraska Medical Center and Research ServiceOmahaNE
| | - Paul R. Langlais
- Department of MedicineCollege of MedicineUniversity of ArizonaTucsonAZ
| | - Heddwen L. Brooks
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| |
Collapse
|
18
|
Protective effects of Clostridium butyricum against oxidative stress induced by food processing and lipid-derived aldehydes in Caco-2 cells. Appl Microbiol Biotechnol 2020; 104:9343-9361. [PMID: 32965561 DOI: 10.1007/s00253-020-10896-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/18/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023]
Abstract
The human body is almost always facing the oxidative stress caused by foodborne aldehydes such as glyoxal (GO) and methylglyoxal (MGO), 4-hydroxyhexenal (HHE), and 4-hydroxynonenal (HNE). When these aldehydes build up, they can cause a range of harm. However, a probiotic, Clostridium butyricum, can increase nuclear factor erythroid-2 related factor 2 (Nrf2) and may have the potential to relieve oxidative stress. If C. butyricum is indeed resistant to aldehydes, the advantages (accessibility, convenience, and safety) will be of great significance compared with drugs. Unfortunately, whether C. butyricum can play a role in alleviating toxic effects of foodborne aldehydes in the intestine (the first line of defense against food-derived toxin) was unclear. To investigate these, we measured the viability, ROS, autophagy, and inflammatory cytokine expression of Caco-2 which were co-cultured with C. butyricum and stimulated by the four aldehydes via Nrf2 pathway (Staphylococcus aureus and Enterococcus faecium as controls). Then, we explored the link among C. butyricum, NLRP6, and Nrf2 signaling pathways when facing the stimuli. In the present study, we demonstrated that Clostridium butyricum relieved the oxidative stress induced by the aldehydes in Caco-2. Most interestingly, we found a "complementary" relationship between NLRP6 and Nrf2 in C. butyricum treatment under aldehyde stress. Our research not only makes a contribution to the popularization of C. butyricum as a probiotic-rich food instead of medicines but also sheds new light on the application of subsequent microecological formulation of C. butyricum. KEY POINTS: • The adverse effects are caused in a dose-dependent manner by foodborne aldehydes. • Clostridium butyricum can significantly ameliorate oxidative stress. • There is a "complementary" relationship between the NLRP6 and Nrf2 signaling pathways. • Using Clostridium butyricum foods to alleviate oxidative stress shows great prospects.
Collapse
|
19
|
Ye M, Guo X, Wang H, Wang Y, Qian X, Deng H, Wang W, Yang S, Ni Q, Chen J, Lv L, Zhao Y, Xue G, Li Y, Zhang L. Mutual regulation between β-TRCP mediated REST protein degradation and Kv1.3 expression controls vascular smooth muscle cell phenotype switch. Atherosclerosis 2020; 313:102-110. [PMID: 33038663 DOI: 10.1016/j.atherosclerosis.2020.08.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/16/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS Phenotypic switch of vascular smooth muscle cells (VSMC) plays a key role in the pathogenesis of atherosclerosis and restenosis after artery intervention. Transcription repressor element 1-silencing transcription factor (REST) has been identified as key regulator of VSMC proliferation. In the present study, we sought to investigate the potential association of E3-ubiquitin ligase β-TRCP mediated REST protein degradation with Kv1.3 expression during VSMC phenotypic switch. METHODS Protein and mRNA expression was measured in ex vivo and in vitro models. Protein interaction and ubiquitination were analyzed by immunoprecipitation assays. ChIP assays were performed to assess the relationship between REST and targeted DNA binding site. RESULTS We found that the expression level of E3-ubiquitin ligase β-TRCP is significantly increased during VSMC phenotypic switch. REST protein ubiquitination mediated by β-TRCP is critical for VSMC proliferation and migration. We also found that the gene KCNA3 encoding potassium channel protein Kv1.3 contains a functional REST binding site and is repressed by REST. Downregulation of REST by β-TRCP and consequently upregulation of Kv1.3 are important events during VSMC phenotypic switch. Furthermore, upregulated Kv1.3 accelerates β-TRCP modulated REST degradation through Erk1/2 signaling. CONCLUSIONS Our results reveal a fundamental role for regulatory interactions between β-TRCP modulated REST degradation and Kv1.3 in the control of the multilayered regulatory programs required for VSMC phenotype switch.
Collapse
Affiliation(s)
- Meng Ye
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Xiangjiang Guo
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Han Wang
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yuli Wang
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Xin Qian
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Haoyu Deng
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Weilun Wang
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Qihong Ni
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Jiaquan Chen
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Lei Lv
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yiping Zhao
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Guanhua Xue
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yinan Li
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.
| | - Lan Zhang
- Department of Vascular Surgery, Renji Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
20
|
Blocking Kv1.3 potassium channels prevents postoperative neuroinflammation and cognitive decline without impairing wound healing in mice. Br J Anaesth 2020; 125:298-307. [PMID: 32624183 DOI: 10.1016/j.bja.2020.05.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/15/2020] [Accepted: 05/06/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Postoperative cognitive decline (PCD) requires microglial activation. Voltage-gated Kv1.3 potassium channels are involved in microglial activation. We determined the role of Kv1.3 in PCD and the efficacy and safety of inhibiting Kv1.3 with phenoxyalkoxypsoralen-1 (PAP-1) in preventing PCD in a mouse model. METHODS After institutional approval, we assessed whether Kv1.3-deficient mice (Kv1.3-/-) exhibited PCD, evidenced by tibial-fracture surgery-induced decline in aversive freezing behaviour, and whether PAP-1 could prevent PCD and postoperative neuroinflammation in PCD-vulnerable diet-induced obese (DIO) mice. We also evaluated whether PAP-1 altered either postoperative peripheral inflammation or tibial-fracture healing. RESULTS Freezing behaviour was unaltered in postoperative Kv1.3-/- mice. In DIO mice, PAP-1 prevented postoperative (i) attenuation of freezing behaviour (54 [17.3]% vs 33.4 [12.7]%; P=0.03), (ii) hippocampal microglial activation by size (130 [31] pixels vs 249 [49]; P<0.001) and fluorescence intensity (12 000 [2260] vs 20 800 [5080] absorbance units; P<0.001), and (iii) hippocampal upregulation of interleukin-6 (IL-6) (14.9 [5.7] vs 25.6 [10.4] pg mg-1; P=0.011). Phenoxyalkoxypsoralen-1 neither affected surgery-induced upregulation of plasma IL-6 nor cartilage and bone components of the surgical fracture callus. CONCLUSIONS Microglial-mediated PCD requires Kv1.3 activity, determined by genetic and pharmacological targeting approaches. Phenoxyalkoxypsoralen-1 blockade of Kv1.3 prevented surgery-induced hippocampal microglial activation and neuroinflammation in mice known to be vulnerable to PCD. Regarding perioperative safety, these beneficial effects of PAP-1 treatment occurred without impacting fracture healing. Kv1.3 blockers, currently undergoing clinical trials for other conditions, may represent an effective and safe intervention to prevent PCD.
Collapse
|
21
|
Wang X, Li G, Guo J, Zhang Z, Zhang S, Zhu Y, Cheng J, Yu L, Ji Y, Tao J. Kv1.3 Channel as a Key Therapeutic Target for Neuroinflammatory Diseases: State of the Art and Beyond. Front Neurosci 2020; 13:1393. [PMID: 31992966 PMCID: PMC6971160 DOI: 10.3389/fnins.2019.01393] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/10/2019] [Indexed: 12/26/2022] Open
Abstract
It remains a challenge for the effective treatment of neuroinflammatory disease, including multiple sclerosis (MS), stroke, epilepsy, and Alzheimer’s and Parkinson’s disease. The voltage-gated potassium Kv1.3 channel is of interest, which is considered as a novel therapeutic target for treating neuroinflammatory disorders due to its crucial role in subsets of T lymphocytes as well as microglial cells. Toxic animals, such as sea anemones, scorpions, spiders, snakes, and cone snails, can produce a variety of toxins that act on the Kv1.3 channel. The Stichodactyla helianthus K+ channel blocking toxin (ShK) from the sea anemone S. helianthus is proved as a classical blocker of Kv1.3. One of the synthetic analogs ShK-186, being developed as a therapeutic for autoimmune diseases, has successfully completed first-in-man Phase 1 trials. In addition to addressing the recent progress on the studies underlying the pharmacological characterizations of ShK on MS, the review will also explore the possibility for clinical treatment of ShK-like Kv1.3 blocking polypeptides on other neuroinflammatory diseases.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Institute of Biomembrane and Biopharmaceutics, Shanghai University, Shanghai, China
| | - Guoyi Li
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingkang Guo
- Institute of Biomembrane and Biopharmaceutics, Shanghai University, Shanghai, China
| | - Zhiping Zhang
- Institute of Biomembrane and Biopharmaceutics, Shanghai University, Shanghai, China
| | - Shuzhang Zhang
- Institute of Biomembrane and Biopharmaceutics, Shanghai University, Shanghai, China
| | - Yudan Zhu
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiwei Cheng
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Yu
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yonghua Ji
- Institute of Biomembrane and Biopharmaceutics, Shanghai University, Shanghai, China,Xinhua Translational Institute for Cancer Pain, Shanghai, China
| | - Jie Tao
- Department of Neurology and Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Putuo Clinical Medical School, Anhui Medical University, Shanghai, China
| |
Collapse
|