1
|
Zhuo Y, Li WS, Lu W, Li X, Ge LT, Huang Y, Gao QT, Deng YJ, Jiang XC, Lan ZW, Deng Q, Chen YH, Xiao Y, Lu S, Jiang F, Liu Z, Hu L, Liu Y, Ding Y, He ZW, Tan DA, Duan D, Lu M. TGF-β1 mediates hypoxia-preconditioned olfactory mucosa mesenchymal stem cells improved neural functional recovery in Parkinson's disease models and patients. Mil Med Res 2024; 11:48. [PMID: 39034405 PMCID: PMC11265117 DOI: 10.1186/s40779-024-00550-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 07/01/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra (SN). Activation of the neuroinflammatory response has a pivotal role in PD. Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach for various nerve injuries, but there are limited reports on their use in PD and the underlying mechanisms remain unclear. METHODS We investigated the effects of clinical-grade hypoxia-preconditioned olfactory mucosa (hOM)-MSCs on neural functional recovery in both PD models and patients, as well as the preventive effects on mouse models of PD. To assess improvement in neuroinflammatory response and neural functional recovery induced by hOM-MSCs exposure, we employed single-cell RNA sequencing (scRNA-seq), assay for transposase accessible chromatin with high-throughput sequencing (ATAC-seq) combined with full-length transcriptome isoform-sequencing (ISO-seq), and functional assay. Furthermore, we present the findings from an initial cohort of patients enrolled in a phase I first-in-human clinical trial evaluating the safety and efficacy of intraspinal transplantation of hOM-MSC transplantation into severe PD patients. RESULTS A functional assay identified that transforming growth factor-β1 (TGF-β1), secreted from hOM-MSCs, played a critical role in modulating mitochondrial function recovery in dopaminergic neurons. This effect was achieved through improving microglia immune regulation and autophagy homeostasis in the SN, which are closely associated with neuroinflammatory responses. Mechanistically, exposure to hOM-MSCs led to an improvement in neuroinflammation and neural function recovery partially mediated by TGF-β1 via activation of the anaplastic lymphoma kinase/phosphatidylinositol-3-kinase/protein kinase B (ALK/PI3K/Akt) signaling pathway in microglia located in the SN of PD patients. Furthermore, intraspinal transplantation of hOM-MSCs improved the recovery of neurologic function and regulated the neuroinflammatory response without any adverse reactions observed in patients with PD. CONCLUSIONS These findings provide compelling evidence for the involvement of TGF-β1 in mediating the beneficial effects of hOM-MSCs on neural functional recovery in PD. Treatment and prevention of hOM-MSCs could be a promising and effective neuroprotective strategy for PD. Additionally, TGF-β1 may be used alone or combined with hOM-MSCs therapy for treating PD.
Collapse
Affiliation(s)
- Yi Zhuo
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
- Department of Neurosurgery, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410000, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410006, China
| | - Wen-Shui Li
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410006, China
| | - Wen Lu
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xuan Li
- Department of Neurosurgery, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410000, China
| | - Li-Te Ge
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yan Huang
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Qing-Tao Gao
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Yu-Jia Deng
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Xin-Chen Jiang
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410006, China
| | - Zi-Wei Lan
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Que Deng
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410006, China
| | - Yong-Heng Chen
- First Clinical Department of Changsha Medical University, Changsha, 410219, China
| | - Yi Xiao
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Shuo Lu
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Feng Jiang
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Zuo Liu
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Li Hu
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Yu Liu
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Yu Ding
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China
| | - Zheng-Wen He
- Department of Neurosurgery, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410000, China
| | - De-An Tan
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China.
| | - Da Duan
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China.
| | - Ming Lu
- Hunan Provincial Key Laboratory of Neurorestoratology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China, (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410003, China.
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410006, China.
| |
Collapse
|
2
|
Yamaguchi M, Yoshiike K, Watanabe H, Watanabe M. The Marine Factor 3,5-dihydroxy-4-methoxybenzyl Alcohol Suppresses Cell Growth, Inflammatory Cytokine Production, and NF-κB Signaling-enhanced Osteoclastogenesis in In vitro Mouse Macrophages RAW264.7 Cells. Curr Mol Med 2024; 24:813-825. [PMID: 37365791 DOI: 10.2174/1566524023666230626141519] [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: 01/24/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND AND OBJECTIVE The novel marine factor 3,5-dihydroxy-4- methoxybenzyl alcohol (DHMBA) was originally identified in the Pacific oyster Crassostrea Gigas. DHMBA has been shown to prevent oxidative stress by scavenging radicals and enhance the production of antioxidant proteins. However, the pharmacologic role of DHMBA has been poorly understood. Inflammation is implicated in the pathogenesis of many diseases. Inflammatory cytokines are produced in macrophages with stimulation of lipopolysaccharide (LPS) and are used as biomarkers that cause diverse disease conditions. Therefore, this study has been undertaken to elucidate whether DHMBA expresses anti-inflammatory effects in in vitro mouse macrophage RAW264.7 cells. METHODS Mouse macrophage RAW264.7 cells were cultured in a medium containing 10% fetal bovine serum (FBS) with or without DHMBA (1-1000 μM). RESULTS Culturing with DHMBA (1-1000 μM) suppressed the growth and stimulated the death of RAW264.7 cells in vitro, leading to a decrease in cell number. Treatment with DHMBA reduced the levels of Ras, PI3K, Akt, MAPK, phospho-MAPK, and mTOR, which are signalling factors to promote cell proliferation, and it raised the levels of p53, p21, Rb, and regucalcin, which are cell growth suppressors. DHMBA treatment elevated caspase-3 and cleaved caspase-3 levels. Interestingly, DHMBA treatment repressed the production of inflammatory cytokines, including tumor necrosis factor-α, interleukin-6, interleukin-1β, or prostaglandin E2, which were enhanced by LPS stimulation. Notably, the levels of NF-κB p65 were increased by LPS treatment, and this augmentation was repres-sed by DHMBA treatment. Moreover, LPS treatment stimulated osteoclastogenesis of RAW264.7 cells. This stimulation was blocked by DHMBA treatment, and this effect was not caused by the presence of an NF-κB signalling inhibitor. CONCLUSION DHMBA was found to potentially suppress the activity of inflammatory macrophages in vitro, suggesting its therapeutic usefulness in inflammatory conditions.
Collapse
Affiliation(s)
- Masayoshi Yamaguchi
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Honolulu, Hawaii, HI 96813, USA
| | - Kenji Yoshiike
- Watanabe Oyster Laboratory Co. Ltd., 490-3, Shimoongata-cho, Hachioji, 192-0154, Tokyo, Japan
| | - Hideaki Watanabe
- Watanabe Oyster Laboratory Co. Ltd., 490-3, Shimoongata-cho, Hachioji, 192-0154, Tokyo, Japan
| | - Mitsugu Watanabe
- Watanabe Oyster Laboratory Co. Ltd., 490-3, Shimoongata-cho, Hachioji, 192-0154, Tokyo, Japan
- Graduate School of Science and Engineering, Soka University, 1-236 Tangi-machi, Hachioji, Tokyo, 192-8577, Japan
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| |
Collapse
|
3
|
Bourebaba L, Kępska M, Qasem B, Zyzak M, Łyczko J, Klemens M, Mularczyk M, Marycz K. Sex hormone-binding globulin improves lipid metabolism and reduces inflammation in subcutaneous adipose tissue of metabolic syndrome-affected horses. Front Mol Biosci 2023; 10:1214961. [PMID: 38146533 PMCID: PMC10749534 DOI: 10.3389/fmolb.2023.1214961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 11/14/2023] [Indexed: 12/27/2023] Open
Abstract
Equine metabolic syndrome (EMS) is a steadily growing endocrine disorder representing a real challenge in veterinary practice. As a multifactorial condition, EMS is characterized by three main metabolic abnormalities including insulin resistance, increased adiposity or obesity and hoof laminitis. Adipose tissue dysfunction is recognized as a core pathophysiological determinant of EMS, as it strongly participates to lipotoxicity and systemic metaflammation, both of which have been closely linked to the development of generalized insulin resistance. Besides, sex hormone binding globulin (SHBG) is an important sex steroids transporters that has been recently proposed as an important metabolic mediator. Therefore, the aim of this study was to verify whether SHBG treatment may ameliorate subcutaneous adipose tissue metabolic failure under EMS condition in terms of lipidome homeostasis, lipid metabolism programs, insulin signalling and local inflammation. Subcutaneous adipose tissue (SAT) biopsies were collected post-mortem from healthy (n = 3) and EMS (n = 3) slaughtered horses. SHBG protein has been applied to SAT samples from EMS horses for 24 h at a final concentration of 50 nM, while control groups (healthy and untreated EMS) were cultured in the presence of SHBG-vehicle only. Tissues from all groups were afterwards secured for downstream analysis of gene expression using RT-qPCR, protein levels by Western blot and ELISA assay and lipidomics through GC-MS technique. Obtained results showcased that SHBG intervention efficiently normalized the altered fatty acids (FAs) profiles by lowering the accumulation of saturated and trans FAs, as well as the pro-inflammatory arachidonic and linoleic acids. Moreover, SHBG showed promising value for the regulation of adipocyte lipolysis and engorgement by lowering the levels of perilipin-1. SHBG exerted moderated effect toward SCD1 and FASN enzymes expression, but increased the LPL abundance. Interestingly, SHBG exhibited a negative regulatory effect on pro-adipogenic stimulators and induced higher expression of KLF3, IRF3 and β-catenin, known as strong adipogenesis repressors. Finally, SHBG protein showed remarkable ability in restoring the insulin signal transduction, IR/IRS/Pi3K/AKT phosphorylation events and GLUT4 transporter abundance, and further attenuate pro-inflammatory response by lowering IL-6 tissue levels and targeting the PDIA3/ERK axis. Overall, the obtained data clearly demonstrate the benefice of SHBG treatment in the regulation of adipose tissue metabolism in the course of EMS and provide new insights for the development of molecular therapies with potential translational application to human metabolic disorders.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Martyna Kępska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Badr Qasem
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Magdalena Zyzak
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Jacek Łyczko
- Department of Food Chemistry and Biocatalysis, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Marta Klemens
- Department of Food Chemistry and Biocatalysis, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Malwina Mularczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- International Institute of Translational Medicine, Wisznia Mała, Poland
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- International Institute of Translational Medicine, Wisznia Mała, Poland
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
4
|
Nigdelioglu Dolanbay S, Şirin S, Aslim B. Cocktail of three isoquinoline alkaloids derived from Glaucium grandiflorum Boiss. & A. Huet subsp. refractum (Nábelek) Mory inhibits the production of LPS-induced ROS, pro-inflammatory cytokines, and mediators through the down-regulation of p38 MAPK in BV-2 cells. Fitoterapia 2023; 170:105652. [PMID: 37595642 DOI: 10.1016/j.fitote.2023.105652] [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: 03/06/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Glaucium grandiflorum extracts have traditionally been used to treat brain-related disorders. G. grandiflorum extracts also exhibited inhibitory effects on cholinesterase enzymes, as well as antigenotoxic activity. However, no research has been done on the effect of G. grandiflorum alkaloid extracts on the anti-oxidative and anti-inflammatory mechanisms. In this study we aimed to evaluate the anti-oxidative and anti-inflammatory activities of the alkaloid extract obtained from G. grandiflorum as well as the mechanisms responsible for their neuroprotective effects in neuronal damage caused by LPS in BV2 cells. We used LC-MS/MS and 1H, 13C NMR analysis to determine the presence of major alkaloids (allocryptopine, tetrahydropalmatine, and tetrahydroberberine N-oxide (trans-cannadine-N-oxide) in the alkaloid extracts. We used flow cytometry to study the alkaloid extracts' effects on ROS production; we also employed qRT-PCR and Western Blot to analyze the effects of oxidative stress and inflammation-related genes and proteins. ROS production within the cell was inhibited by chloroform alkaloid extract (CAE). There occurred marked CAE-induced reductions in IL-1β, Cox-2, and iNOS mRNA expressions. We also observed marked reductions in IL-6 and TNF-α mRNA expressions with methanol alkaloid extract (MAE). CAE effectively suppressed IL-1β and iNOS protein levels, especially as in qRT-PCR studies, while MAE effectively reduced IL-6 and TNF-α protein levels. Additionally, MAE was found to be prominent in suppressing the levels of Cox-2 protein, unlike qRT-PCR studies. According to our study findings, oxidative stress brought about by inflammation was suppressed by alkaloid extracts from G. grandiflorum which can be attributed to their suppressor effects on the pro-inflammatory cytokines-mediators, and p38 MAPK. As a result, a drug active substance that suppresses oxidative stress and inflammation has been brought to the neuropharmacological field.
Collapse
Affiliation(s)
| | - Seda Şirin
- Gazi University, Faculty of Science, Department of Biology, 06500, Teknikokullar, Ankara, Turkey
| | - Belma Aslim
- Gazi University, Faculty of Science, Department of Biology, 06500, Teknikokullar, Ankara, Turkey
| |
Collapse
|
5
|
Zhuo Y, Li X, He Z, Lu M. Pathological mechanisms of neuroimmune response and multitarget disease-modifying therapies of mesenchymal stem cells in Parkinson's disease. Stem Cell Res Ther 2023; 14:80. [PMID: 37041580 PMCID: PMC10091615 DOI: 10.1186/s13287-023-03280-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 03/13/2023] [Indexed: 04/13/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by the degeneration of dopaminergic neurons in the substantia nigra (SN); the etiology and pathological mechanism of the disease are still unclear. Recent studies have shown that the activation of a neuroimmune response plays a key role in the development of PD. Alpha-synuclein (α-Syn), the primary pathological marker of PD, can gather in the SN and trigger a neuroinflammatory response by activating microglia which can further activate the dopaminergic neuron's neuroimmune response mediated by reactive T cells through antigen presentation. It has been shown that adaptive immunity and antigen presentation processes are involved in the process of PD and further research on the neuroimmune response mechanism may open new methods for its prevention and therapy. While current therapeutic regimens are still focused on controlling clinical symptoms, applications such as immunoregulatory strategies can delay the symptoms and the process of neurodegeneration. In this review, we summarized the progression of the neuroimmune response in PD based on recent studies and focused on the use of mesenchymal stem cell (MSC) therapy and challenges as a strategy of disease-modifying therapy with multiple targets.
Collapse
Affiliation(s)
- Yi Zhuo
- Department of Neurosurgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410000, Hunan, China
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410006, Hunan, China
| | - Xuan Li
- Department of Neurosurgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410000, Hunan, China
| | - Zhengwen He
- Department of Neurosurgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410000, Hunan, China.
| | - Ming Lu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410006, Hunan, China.
- Hunan Provincial Key Laboratory of Neurorestoratology, The Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha, 410003, Hunan, China.
| |
Collapse
|
6
|
Wang H, Su X, Zhang QQ, Zhang YY, Chu ZY, Sun ZH, Zhang JL, Tang YF. Cystic Fibrosis Transmembrane Conductance Regulator Attenuates Oxidative Stress-Induced Injury in Diabetic Retinopathy Rats. Curr Eye Res 2023; 48:416-424. [PMID: 36476257 DOI: 10.1080/02713683.2022.2156548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To investigate the effects of cystic fibrosis transmembrane conductance regulator (CFTR) on oxidative stress-induced injury of diabetic retinopathy (DR) rats. METHODS DR rat model was constructed treated with Ad-CFTR. Hematoxylin and Eosin (HE) staining was applied for testing the thickness of each layer of retinal tissues. Enzyme-linked immunosorbent assay (ELISA) was used to determine levels of serum inflammatory cytokines and contents of oxidative stress related genes in rats. Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) staining was used to detect retinal cell apoptosis, and western blotting to measure the expression of MAPK/NF-κB pathway-related proteins in retinal tissues. RESULTS Our experiment revealed the remarkable decrease of CFTR protein in retinal tissues of DR rats. DR rats had decreased body weight and increased blood glucose level, with decreased thickness of total retinal thickness (TRT), outer nuclear layer and outer plexiform layer (ONL + OPL), inner nuclear layer (INL), and inner plexiform layer (IPL). Besides, DR rats were apparently up-regulated in the expression of pro-inflammatory cytokines, with increased malondial dehyde (MDA), p-ERK1/2/ERK1/2 and p-JNK1/2/JNK1/2 expressions, decreased superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activity in retinal tissues, as well as up-regulated p65 protein in nucleus and down-regulated p65 protein in cytoplasm. DR rats treated with Ad-CFTR were effectively improved regarding the above parameters except body weight and blood glucose. CONCLUSIONS CFTR can inhibit MAPK/NF-κB signaling pathway to ameliorate inflammatory response and oxidative stress-induced injury of DR rats, thereby reducing retinal cell apoptosis and playing a protective role in retina.
Collapse
Affiliation(s)
- Hui Wang
- Department of Ophthalmology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xian Su
- Department of Ophthalmology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Qian-Qian Zhang
- Outpatient Department, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Ying-Ying Zhang
- Department of Ophthalmology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Zhan-Ya Chu
- Department of Ophthalmology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Zhao-Hui Sun
- Department of Ophthalmology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Jin-Ling Zhang
- Department of Ophthalmology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Yu-Fen Tang
- Department of Outpatient Operating Room, Shijiazhuang People's Hospital, Shijiazhuang, China
| |
Collapse
|
7
|
Yu L, Li Y. Involvement of Intestinal Enteroendocrine Cells in Neurological and Psychiatric Disorders. Biomedicines 2022; 10:biomedicines10102577. [PMID: 36289839 PMCID: PMC9599815 DOI: 10.3390/biomedicines10102577] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022] Open
Abstract
Neurological and psychiatric patients have increased dramatically in number in the past few decades. However, effective treatments for these diseases and disorders are limited due to heterogeneous and unclear pathogenic mechanisms. Therefore, further exploration of the biological aspects of the disease, and the identification of novel targets to develop alternative treatment strategies, is urgently required. Systems-level investigations have indicated the potential involvement of the brain–gut axis and intestinal microbiota in the pathogenesis and regulation of neurological and psychiatric disorders. While intestinal microbiota is crucial for maintaining host physiology, some important sensory and regulatory cells in the host should not be overlooked. Intestinal epithelial enteroendocrine cells (EECs) residing in the epithelium throughout intestine are the key regulators orchestrating the communication along the brain-gut-microbiota axis. On one hand, EECs sense changes in luminal microorganisms via microbial metabolites; on the other hand, they communicate with host body systems via neuroendocrine molecules. Therefore, EECs are believed to play important roles in neurological and psychiatric disorders. This review highlights the involvement of EECs and subtype cells, via secretion of endocrine molecules, in the development and regulation of neurological and psychiatric disorders, including Parkinson’s disease (PD), schizophrenia, visceral pain, neuropathic pain, and depression. Moreover, the current paper summarizes the potential mechanism of EECs in contributing to disease pathogenesis. Examination of these mechanisms may inspire and lead to the development of new aspects of treatment strategies for neurological and psychiatric disorders in the future.
Collapse
Affiliation(s)
- Liangen Yu
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA
| | - Yihang Li
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Correspondence:
| |
Collapse
|
8
|
Qiu M, Feng L, Zhao C, Gao S, Bao L, Zhao Y, Fu Y, Hu X. Commensal Bacillus subtilis from cow milk inhibits Staphylococcus aureus biofilm formation and mastitis in mice. FEMS Microbiol Ecol 2022; 98:6596871. [PMID: 35648454 DOI: 10.1093/femsec/fiac065] [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: 11/23/2021] [Revised: 05/15/2022] [Accepted: 05/30/2022] [Indexed: 11/12/2022] Open
Abstract
The colonization and virulence production of Staphylococcus aureus (S. aureus), a known pathogen that induces mastitis, depend on its quorum-sensing (QS) system and biofilm formation. It has been reported that Bacillus can inhibit the QS system of S. aureus, thereby reducing S. aureus colonization in the intestine. However, whether Bacillus affects S. aureus biofilm formation and consequent colonization during mastitis is still unknown. In this study, the differences in the colonization of S. aureus and Bacillus were first analyzed by isolating and culturing bacteria from milk samples. It was found that the colonization of Bacillus and S. aureus in cow mammary glands was negatively correlated. Secondly, we found that although Bacillus did not affect S. aureus growth, it inhibited the biofilm formation of S. aureus by interfering its QS signaling. The most significant anti-biofilm effect was found in Bacillus subtilis H28 (B. subtilis H28). Finally, we found that B. subtilis H28 treatment alleviated S. aureus-induced mastitis in a mice model. Our results rerealed that bovine milk derived commensal Bacillus inhibited S. aureus colonization and alleviated S. aureus-induced mastitis by influencing biofilm formation, suggesting a potential targeted strategy to limit the colonization of S. aureus in vivo.
Collapse
Affiliation(s)
- Min Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Lianjun Feng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Siyuan Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Lijuan Bao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Yihong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| | - Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province 130062, China
| |
Collapse
|
9
|
Butt S, Gagnon J, Saleh M. A Protective Role for Glucagon-like Peptide-2 in Heat-stable Enterotoxin b (STb)-Induced L-Cell Toxicity. Endocrinology 2022; 163:6546206. [PMID: 35266539 DOI: 10.1210/endocr/bqac029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Indexed: 11/19/2022]
Abstract
Enterotoxigenic Escherichia coli (ETEC)-derived purified heat-stable enterotoxin b (STb) is responsible for secretory diarrhea in livestock and humans. STb disrupts intestinal fluid homeostasis, epithelial barrier function, and promotes cell death. Glucagon-like peptide-2 (GLP-2) is a potent intestinotrophic hormone secreted by enteroendocrine L cells. GLP-2 enhances crypt cell proliferation, epithelial barrier function, and inhibits enterocyte apoptosis. Whether STb can affect GLP-2 producing L cells remains to be elucidated. First, secreted-His-labeled STb from transformed E coli was collected and purified. When incubated with L-cell models (GLUTag, NCI-H716, and secretin tumor cell line [STC-1]), fluorescent immunocytochemistry revealed STb was internalized and was differentially localized in the cytoplasm and nucleus. Cell viability experiments with neutral red and resazurin revealed that STb was toxic in all but the GLUTag cells. STb stimulated 2-hour GLP-2 secretion in all cell models. Interestingly, GLUTag cells produced the highest amount of GLP-2 when treated with STb, demonstrating an inverse relationship in GLP-2 secretion and cell toxicity. To demonstrate a protective role for GLP-2, GLUTag-conditioned media (rich in GLP-2) blocked STb toxicity in STC-1 cells. Confirming a protective role of GLP-2, teduglutide was able to improve cell viability in cells treated with H2O2. In conclusion, STb interacts with the L cell, stimulates secretion, and may induce toxicity if GLP-2 is not produced at high levels. GLP-2 or receptor agonists have the ability to improve cell viability in response to toxins. These results suggest that GLP-2 secretion can play a protective role during STb intoxication. This work supports future investigation into the use of GLP-2 therapies in enterotoxigenic-related diseases.
Collapse
Affiliation(s)
- Shahnawaz Butt
- Laurentian University, School of Natural Sciences, Sudbury, Ontario P3E 2C6, Canada
| | - Jeffrey Gagnon
- Laurentian University, School of Natural Sciences, Sudbury, Ontario P3E 2C6, Canada
| | - Mazen Saleh
- Laurentian University, School of Natural Sciences, Sudbury, Ontario P3E 2C6, Canada
| |
Collapse
|
10
|
Song S, Guo R, Mehmood A, Zhang L, Yin B, Yuan C, Zhang H, Guo L, Li B. Liraglutide attenuate central nervous inflammation and demyelination through AMPK and pyroptosis-related NLRP3 pathway. CNS Neurosci Ther 2022; 28:422-434. [PMID: 34985189 PMCID: PMC8841291 DOI: 10.1111/cns.13791] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 12/22/2022] Open
Abstract
Aims Multiple sclerosis (MS) still maintains increasing prevalence and poor prognosis, while glucagon‐like peptide‐1 receptor (GLP‐1R) agonists show excellent neuroprotective capacities recently. Thus, we aim to evaluate whether the GLP‐1R agonist liraglutide (Lira) could ameliorate central nervous system demyelination and inflammation. Methods The therapeutic effect of Lira was tested on experimental autoimmune encephalitis (EAE) in vivo and a microglia cell line BV2 in vitro. Results Lira administration could ameliorate the disease score of EAE mice, delay the disease onset, ameliorate pathological demyelination and inflammation score in lumbar spinal cord, reduce pathogenic T helper cell transcription in spleen, restore phosphorylated adenosine monophosphate‐activated protein kinase (pAMPK) level, autophagy level, and inhibit pyroptosis‐related NLR family, pyrin domain‐containing protein 3 (NLRP3) pathway in lumbar spinal cord. Additionally, cell viability test, lactate dehydrogenase release test, and dead/live cell staining test for BV2 cells showed Lira could not salvage BV2 from nigericin‐induced pyroptosis significantly. Conclusion Lira has anti‐inflammation and anti‐demyelination effect on EAE mice, and the protective effect of Lira in the EAE model may be related to regulation of pAMPK pathway, autophagy, and NLRP3 pathway. However, Lira treatment cannot significantly inhibit pyroptosis of BV2 cells in vitro. Our study provides Lira as a potential candidate for Multiple Sclerosis treatment.
Collapse
Affiliation(s)
- Shuang Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Ruoyi Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Lu Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Bowen Yin
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China.,Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Congcong Yuan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China.,Department of Neurology, Baoding First Central Hospital, Baoding, China
| | - Huining Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Li Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| | - Bin Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Neurology of Hebei Province, Shijiazhuang, China
| |
Collapse
|
11
|
Wang Z, Wang G, Wang Y, Liu Q, Li H, Xie P, Wang Z. Omp31 of Brucella Inhibits NF-κB p65 Signaling Pathway by Inducing Autophagy in BV-2 Microglia. Neurochem Res 2021; 46:3264-3272. [PMID: 34536195 DOI: 10.1007/s11064-021-03429-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 02/04/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Neurobrucellosis is a serious central nervous system (CNS) inflammatory disorder caused by Brucella, and outer membrane protein-31 (Omp31) plays an important role in Brucella infection. This study aims to determine whether Omp31 can induce autophagy in BV-2 microglia. Another goal of the study is to further examine the effect of autophagy on the nuclear transcription factor κB (NF-κB) p65 signaling pathway. We observed that Omp31 stimulated autophagy by increasing microtubule-associated protein 1 light chain 3B (LC3B-II) levels and inducing autophagosome formation at 6 h and 12 h. Concomitantly, Omp31 induced tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) expression in a time-dependent manner but reduced the expression of TNF-α at 6 h. We utilized Omp31 with or without rapamycin or 3-methyladenine (3-MA) to treat BV-2 microglia, and it demonstrated further that Omp31 induced autophagy by promoting LC3B-II, Beclin-1 proteins expression and inhibiting the p62 protein levels. Furthermore, we explored the effects of autophagy on the NF-κB p65 pathway through western blot analysis, RT-qPCR assay, enzyme-linked immunosorbent assay (ELISA) and immunofluorescence. The data suggest that Omp31 as well as rapamycin, the autophagy inducer, can decrease TNF-α levels through the inhibition of the NF-κB p65 signaling pathway. Taken together, Omp31 can function as a catalyst in both autophagy induction and NF-κB p65 signal inhibition. Furthermore, Omp31-induced autophagy may inhibit the expression of TNF-α by negatively regulating NF-κB p65 signaling pathway.
Collapse
Affiliation(s)
- Zhao Wang
- Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Guowei Wang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yanbai Wang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qiang Liu
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Haining Li
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Peng Xie
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Zhenhai Wang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China. .,Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
12
|
Targeting the TLR4/NF-κB pathway in β-amyloid-stimulated microglial cells: A possible mechanism that oxysophoridine exerts anti-oxidative and anti-inflammatory effects in an in vitro model of Alzheimer's disease. Brain Res Bull 2021; 175:150-157. [PMID: 34329731 DOI: 10.1016/j.brainresbull.2021.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022]
Abstract
β-amyloid (Aβ) accumulation is a major neuropathological characteristic of Alzheimer's disease (AD) and serves as an inflammatory stimulus for microglial cells. Oxysophoridine has multiple pharmacological effects, including anti-inflammatory and anti-oxidative activities. In view of this, the current study aimed to investigate the effects of oxysophoridine on Aβ-induced activation of microglial BV-2 cells. Cell Counting Kit-8 assay showed that oxysophoridine concentration-dependently attenuated Aβ-induced viability reduction of BV-2 cells. Aβ stimulation reduced the activities of glutathione peroxidase (GPx), catalase (CAT), and superoxide dismutase (SOD) and elevated malondialdehyde (MDA) content in BV-2 cells, but these effects were attenuated by oxysophoridine. Oxysophoridine abolished Aβ-induced increase of mRNA expression, secretion, and protein expression of tumor necrosis factor-α (TNF-α) and interleukin 1β (IL-1β) in BV-2 cells. Additionally, western blot suggested that oxysophoridine inhibited Aβ-induced activation of the toll-like receptor 4 (TLR4) and nuclear factor-kappa B (NF-κB) pathways in BV-2 cells. Inhibition of the TLR4/NF-κB pathway by TAK-242 enhanced the effects of oxysophoridine on Aβ-induced viability reduction, oxidative stress, and inflammation in BV-2 cells. Taken together, oxysophoridine suppressed Aβ-induced oxidative stress and inflammation in BV-2 cells by inhibition of the TLR4/NF-κB pathway.
Collapse
|
13
|
Kim SK, Ko YH, Lee Y, Lee SY, Jang CG. Antineuroinflammatory Effects of 7,3',4'-Trihydroxyisoflavone in Lipopolysaccharide-Stimulated BV2 Microglial Cells through MAPK and NF-κB Signaling Suppression. Biomol Ther (Seoul) 2021; 29:127-134. [PMID: 32812529 PMCID: PMC7921860 DOI: 10.4062/biomolther.2020.093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/09/2020] [Accepted: 07/24/2020] [Indexed: 01/30/2023] Open
Abstract
Neuroinflammation—a common pathological feature of neurodegenerative disorders such as Alzheimer’s disease—is mediated by microglial activation. Thus, inhibiting microglial activation is vital for treating various neurological disorders. 7,3’,4’-Trihydroxyisoflavone (THIF)—a secondary metabolite of the soybean compound daidzein—possesses antioxidant and anticancer properties. However, the effects of 7,3’,4’-THIF on microglial activation have not been explored. In this study, antineuroinflammatory effects of 7,3’,4’-THIF in lipopolysaccharide (LPS)-stimulated BV2 microglial cells were examined. 7,3’,4’-THIF significantly suppressed the production of the proinflammatory mediators nitric oxide (NO), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) as well as of the proinflammatory cytokine interleukin-6 (IL-6) in LPS-stimulated BV2 microglial cells. Moreover, 7,3’,4’-THIF markedly inhibited reactive oxygen species (ROS) generation. Western blotting revealed that 7,3’,4’-THIF diminished LPS-induced phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), glycogen synthase kinase-3β (GSK-3β), and nuclear factor kappa B (NF-κB). Overall, 7,3’,4’-THIF exerts antineuroinflammatory effects against LPS-induced microglial activation by suppressing mitogen-activated protein kinase (MAPK) and NF-κB signaling, ultimately reducing proinflammatory responses. Therefore, these antineuroinflammatory effects of 7,3’,4’-THIF suggest its potential as a therapeutic agent for neurodegenerative disorders.
Collapse
Affiliation(s)
- Seon-Kyung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yong-Hyun Ko
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
14
|
Zhang Z, Hao L, Shi M, Yu Z, Shao S, Yuan Y, Zhang Z, Hölscher C. Neuroprotective Effects of a GLP-2 Analogue in the MPTP Parkinson's Disease Mouse Model. JOURNAL OF PARKINSONS DISEASE 2021; 11:529-543. [PMID: 33523018 DOI: 10.3233/jpd-202318] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Glucagon-like peptide 2 (GLP-2) is a peptide hormone derived from the proglucagon gene expressed in the intestines, pancreas and brain. Some previous studies showed that GLP-2 improved aging and Alzheimer's disease related memory impairments. Parkinson's disease (PD) is a progressive neurodegenerative disorder, and to date, there is no particular medicine reversed PD symptoms effectively. OBJECTIVE The aim of this study was to evaluate neuroprotective effects of a GLP-2 analogue in the 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) PD mouse model. METHODS In the present study, the protease resistant Gly(2)-GLP-2 (50 nmol/kg ip.) analogue has been tested for 14 days by behavioral assessment, transmission electron microscope, immunofluorescence histochemistry, enzyme-linked immunosorbent assay and western blot in an acute PD mouse model induced by MPTP. For comparison, the incretin receptor dual agonist DA5-CH was tested in a separate group. RESULTS The GLP-2 analogue treatment improved the locomotor and exploratory activity of mice, and improved bradykinesia and movement imbalance of mice. Gly(2)-GLP-2 treatment also protected dopaminergic neurons and restored tyrosine hydroxylase expression levels in the substantia nigra. Gly(2)-GLP-2 furthermore reduced the inflammation response as seen in lower microglia activation, and decreased NLRP3 and interleukin-1β pro-inflammatory cytokine expression levels. In addition, the GLP-2 analogue improved MPTP-induced mitochondrial dysfunction in the substantia nigra. The protective effects were comparable to those of the dual agonist DA5-CH. CONCLUSION The present results demonstrate that Gly(2)-GLP-2 can attenuate NLRP3 inflammasome-mediated inflammation and mitochondrial damage in the substantia nigra induced by MPTP, and Gly(2)-GLP-2 shows neuroprotective effects in this PD animal model.
Collapse
Affiliation(s)
- Zijuan Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China.,Basic Medical Collenge, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Li Hao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China.,Basic Medical Collenge, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ming Shi
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ziyang Yu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Simai Shao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ye Yuan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Christian Hölscher
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| |
Collapse
|
15
|
Singh RP, Bhaiyya R, Khandare K, Tingirikari JMR. Macroalgal dietary glycans: potential source for human gut bacteria and enhancing immune system for better health. Crit Rev Food Sci Nutr 2020; 62:1674-1695. [PMID: 33190530 DOI: 10.1080/10408398.2020.1845605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Macroalgae are the diverse group of photosynthetic algae found at the intertidal regions of oceans. Recent advances suggest that macroalgal derived glycans have tremendous potential to maintain gut microbiome and immune system. The human gut bacteria harbor unique arsenals for utilizing a variety of macroalgal glycans, and produce a variety of oligosaccharides in vivo. Those oligosaccharides interact with immune cell receptors, and also are available for microbial fermentation, thus play magnificent roles in balancing the gut homeostasis. However, this area of research is still in infancy condition in term to understand their molecular interactions. For wooing this area, we urge to emphasize more studies on mechanistic level sympathetic of depolymerizing marine dietary glycans by gut bacteria and elucidating molecular aspect of glycans to cell receptors interactions. This will invent new nutraceutical strategies to purposefully manipulate the microbial composition to improve health. Therefore, review focuses on the recent development of mechanistic understanding of human gut bacterial communities for utilizing macroalgal derived glycans. Recent trends of application of glycans in modulating immune system at mechanistic level and their available evidences are discussed.
Collapse
Affiliation(s)
- Ravindra Pal Singh
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), Punjab, India
| | - Raja Bhaiyya
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), Punjab, India
| | - Kiran Khandare
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), Punjab, India
| | | |
Collapse
|
16
|
Chang Y, Deng Q, Zhang Z, Zhao H, Tang J, Chen X, Liu G, Tian G, Cai J, Jia G. Glucagon-like peptide 2 attenuates intestinal mucosal barrier injury through the MLCK/pMLC signaling pathway in a piglet model. J Cell Physiol 2020; 236:3015-3032. [PMID: 32960454 DOI: 10.1002/jcp.30068] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
Glucagon-like peptide-2 (GLP-2), an intestinotrophic hormone, has drawn considerable attention worldwide due to its potential to promote intestinal development. We investigated the effects and mechanisms of GLP-2 against lipopolysaccharide (LPS)-induced intestinal inflammation and injury both in vitro and in vivo. Forty healthy piglets weaned at the age of 28 days with similar body weight (BW) were assigned to four in vivo treatments with ten piglets each: (i) nonchallenged control; (ii) LPS-challenged control; (iii) LPS + low dose GLP-2; and (iv) LPS + high dose GLP-2. Piglets were subcutaneously injected with phosphate-buffered saline supplemented with GLP-2 at doses of 0, 0, 2, and 10 nmol/kg BW per day for seven consecutive days. The piglets were challenged with an intraperitoneal injection with 100 μg/kg LPS on day 14 to induce intestinal damage. After that, the gene and protein expression levels of representative tight junction proteins and myosin light-chain kinase (MLCK)/phosphorylated myosin light chain (pMLC), as well as proinflammatory cytokine levels were determined using quantitative reverse transcription polymerase chain reaction, western blot, and enzyme-linked immunosorbent assay methods. A high dose of GLP-2 pretreatment increased intestinal permeability by downregulating and redistributing tight junction proteins (p < .05), for example, zona occluden-1 (ZO-1) and occludin. GLP-2 decreased the transcription of proinflammatory cytokines genes including interleukin-1β (IL-1β), IL-6, IL-8, and tumor necrosis factor-α in small intestines (p < .05). GLP-2 prevented the LPS-induced increase in the expression of MLCK dose-dependently and the increase in pMLC levels in the duodenum, jejunum, and ileum. To assess further the protective effect of GLP-2 on LPS-induced intestinal barrier injury after weaning and its possible mechanism, an in vitro intestinal epithelial barrier model was established with IPEC-J2 monolayers and treated with 100 μg/ml LPS with or without 1 × 10-8 mol/L GLP-2 pretreatment. The in vitro analysis included control, LPS, and GLP-2 + LPS treatments. GLP-2 treatment alleviated the destructive effect of LPS on barrier permeability by restoring the expression and ultrastructure of ZO-1 and occludin (p < .05). In addition, GLP-2 reversed the LPS-induced MLCK hyperexpression and pMLC hyperphosphorylation (p < .05). Taken together, our findings revealed a mechanism by which GLP-2 alleviated LPS-challenged intestinal barrier injury and inflammation in weaned piglets and IPEC-J2 cells via the MLCK/pMLC signaling pathway.
Collapse
Affiliation(s)
- Yaqi Chang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Qiuhong Deng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Zhenyu Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China.,Meishan Vocational Technical College, Meishan, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Jiayong Tang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Gang Tian
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Jingyi Cai
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| |
Collapse
|
17
|
Hu L, Ye H, Liao J. LncRNA TUG1 reverses LPS-induced cell apoptosis and inflammation of macrophage via targeting MiR-221-3p/SPRED2 axis. Biosci Biotechnol Biochem 2020; 84:2458-2465. [PMID: 32841583 DOI: 10.1080/09168451.2020.1806704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study aimed to identify the role of lncRNA TUG1 with miR-221-3p on mice with lipopolysaccharide (LPS)-induced acute respiratory distress syndrome (ARDS). Animal model was established, and lung tissue histopathologic status and permeability were detected by hematoxylin-eosin (HE) or Evans blue dye assay respectively. Levels of inflammation cytokines, lncRNA TUG1, miR-221-3p, sprouty related EVH1 domain-containing 2 (SPRED2), and phosphorylated (p)-ERK1/2 were determined by ELISA, qRT-PCR or Western blot. Pulmonary impairment and apoptosis were examined by flow cytometry. We observed that LPS up-regulated levels of tumor necrosis factor-α (TNF-α), Interleukin-1β (1L-1β), and ERK1/2 phosphorylation, and reduced SPRED2 levels, which were rescued by overexpressed lncRNA TUG1. StarBase and dual-luciferase reporter assay verified that miR-221-3p was targeted by lncRNA TUG1. MiR-221-3p could reverse the effect of lncRNA TUG1 on cell apoptosis, levels of TNF-α, IL-1β, SPRED2, and p-ERK1/2. Therefore, overexpressed lncRNA TUG1 attenuated LPS-induced pulmonary impairment in ARDS mice via regulating miR-221-3p/SPRED2 axis.
Collapse
Affiliation(s)
- Lili Hu
- Department of ICU, Shenzhen Hospital, Southern Medical University , Shenzhen, China
| | - Hongwei Ye
- Department of Critical Care Medicine, Changshu Hospital Affiliated to Soochow University , Suzhou, China
| | - Jianjun Liao
- Department of Respiratory, Xi'an International Medical Center Hospital , Xi'an, China
| |
Collapse
|
18
|
Zhuang W, Cai M, Li W, Chen C, Wang Y, Lv E, Fu W. Polyphenols from Toona sinensiss Seeds Alleviate Neuroinflammation Induced by 6-Hydroxydopamine Through Suppressing p38 MAPK Signaling Pathway in a Rat Model of Parkinson's Disease. Neurochem Res 2020; 45:2052-2064. [PMID: 32556929 DOI: 10.1007/s11064-020-03067-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
Abstract
Polyphenols from Toona sinensis seeds (PTSS) have demonstrated anti-inflammatory effects in various diseases, while the anti-neuroinflammatory effects still remain to be investigated. We aimed to investigate the effects of PTSS on Parkinson's disease and underlying mechanisms using a rat model. We employed 6-hydroxydopamine (6-OHDA) to male Sprague Dawley (SD) rats and PC12 cells to construct the in vivo and vitro models of PD and dopaminergic (DA) neuron injury, respectively. Cell viability was detected by cell counting kit-8 (CCK-8) assay and protein levels of inflammatory mediators and some p38 MAPK pathway molecules were investigated by immunohistochemistry and Western blot analyses. The results showed that 6-OHDA significantly increased protein levels of inflammatory mediators, such as cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and tumor necrosis factor α (TNF-α), which could be reversed by PTSS through suppressing the p38 MAPK pathway. The anti-inflammatory effects of PTSS were significantly enhanced by the specific p38 inhibitor of SB203580 in vitro. The present work suggests that PTSS can exert anti-inflammatory effects on PD models, which may be attributed to the suppression of p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Wenxin Zhuang
- Center for Experimental Medical Research, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Meiyun Cai
- Department of Histology and Embryology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Wanzhong Li
- Department of Pharmaceutics, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Chao Chen
- Department of Histology and Embryology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Yanqiang Wang
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang, 261053, Shandong, China
| | - E Lv
- Department of Histology and Embryology, Weifang Medical University, Weifang, 261053, Shandong, China.
| | - Wenyu Fu
- Department of Histology and Embryology, Weifang Medical University, Weifang, 261053, Shandong, China.
| |
Collapse
|
19
|
Baldassano S, Amato A, Terzo S, Caldara GF, Lentini L, Mulè F. Glucagon-like peptide-2 analog and inflammatory state in obese mice. Endocrine 2020; 68:695-698. [PMID: 32172484 DOI: 10.1007/s12020-020-02261-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/03/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Sara Baldassano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Antonella Amato
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Simona Terzo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
- Department of Neuroscience and Cell Biology, University of Palermo, 90127, Palermo, Italy
| | - Gaetano Felice Caldara
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Laura Lentini
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Flavia Mulè
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy.
| |
Collapse
|
20
|
Sipe LM, Chaib M, Pingili AK, Pierre JF, Makowski L. Microbiome, bile acids, and obesity: How microbially modified metabolites shape anti-tumor immunity. Immunol Rev 2020; 295:220-239. [PMID: 32320071 PMCID: PMC7841960 DOI: 10.1111/imr.12856] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
Bile acids (BAs) are known facilitators of nutrient absorption but recent paradigm shifts now recognize BAs as signaling molecules regulating both innate and adaptive immunity. Bile acids are synthesized from cholesterol in the liver with subsequent microbial modification and fermentation adding complexity to pool composition. Bile acids act on several receptors such as Farnesoid X Receptor and the G protein-coupled BA receptor 1 (TGR5). Interestingly, BA receptors (BARs) are expressed on immune cells and activation either by BAs or BAR agonists modulates innate and adaptive immune cell populations skewing their polarization toward a more tolerogenic anti-inflammatory phenotype. Intriguingly, recent evidence also suggests that BAs promote anti-tumor immune response through activation and recruitment of tumoricidal immune cells such as natural killer T cells. These exciting findings have redefined BA signaling in health and disease wherein they may suppress inflammation on the one hand, yet promote anti-tumor immunity on the other hand. In this review, we provide our readers with the most recent understanding of the interaction of BAs with the host microbiome, their effect on innate and adaptive immunity in health and disease with a special focus on obesity, bariatric surgery-induced weight loss, and immune checkpoint blockade in cancer.
Collapse
Affiliation(s)
- Laura M. Sipe
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ajeeth K. Pingili
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joseph F. Pierre
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Liza Makowski
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
21
|
Do HTT, Bui BP, Sim S, Jung JK, Lee H, Cho J. Anti-Inflammatory and Anti-Migratory Activities of Isoquinoline-1-Carboxamide Derivatives in LPS-Treated BV2 Microglial Cells via Inhibition of MAPKs/NF-κB Pathway. Int J Mol Sci 2020; 21:ijms21072319. [PMID: 32230861 PMCID: PMC7177615 DOI: 10.3390/ijms21072319] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/17/2022] Open
Abstract
Eleven novel isoquinoline-1-carboxamides (HSR1101~1111) were synthesized and evaluated for their effects on lipopolysaccharide (LPS)-induced production of pro-inflammatory mediators and cell migration in BV2 microglial cells. Three compounds (HSR1101~1103) exhibited the most potent suppression of LPS-induced pro-inflammatory mediators, including interleukin (IL)-6, tumor necrosis factor-alpha, and nitric oxide (NO), without significant cytotoxicity. Among them, only N-(2-hydroxyphenyl) isoquinoline-1-carboxamide (HSR1101) was found to reverse LPS-suppressed anti-inflammatory cytokine IL-10, so it was selected for further characterization. HSR1101 attenuated LPS-induced expression of inducible NO synthase and cyclooxygenase-2. Particularly, HSR1101 abated LPS-induced nuclear translocation of NF-κB through inhibition of IκB phosphorylation. Furthermore, HSR1101 inhibited LPS-induced cell migration and phosphorylation of mitogen-activated protein kinases (MAPKs) including extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase, and p38 MAPK. The specific MAPK inhibitors, U0126, SP600125, and SB203580, suppressed LPS-stimulated pro-inflammatory mediators, cell migration, and NF-κB nuclear translocation, indicating that MAPKs may be the upstream kinase of NF-κB signaling. Collectively, these results demonstrate that HSR1101 is a potent and promising compound suppressing LPS-induced inflammation and cell migration in BV2 microglial cells, and that inhibition of the MAPKs/NF-κB pathway mediates its anti-inflammatory and anti-migratory effects. Based on our findings, HSR1101 may have beneficial impacts on various neurodegenerative disorders associated with neuroinflammation and microglial activation.
Collapse
Affiliation(s)
- Ha Thi Thu Do
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, Korea; (H.T.T.D.); (B.P.B.)
| | - Bich Phuong Bui
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, Korea; (H.T.T.D.); (B.P.B.)
| | - Seongrak Sim
- College of Pharmacy, Chungbuk National University, Osong, Cheongju 28160, Korea; (S.S.); (J.-K.J.)
| | - Jae-Kyung Jung
- College of Pharmacy, Chungbuk National University, Osong, Cheongju 28160, Korea; (S.S.); (J.-K.J.)
| | - Heesoon Lee
- College of Pharmacy, Chungbuk National University, Osong, Cheongju 28160, Korea; (S.S.); (J.-K.J.)
- Correspondence: (H.L.); (J.C.)
| | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, Korea; (H.T.T.D.); (B.P.B.)
- Correspondence: (H.L.); (J.C.)
| |
Collapse
|
22
|
Chen D, Cao D, Sui P. Tetramethylpyrazine relieves LPS-induced pancreatic β-cell Min6 injury via regulation of miR-101/MKP-1. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2545-2552. [PMID: 31213095 DOI: 10.1080/21691401.2019.1628039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tetramethylpyrazine (TMP) is a traditional Chinese medicine with anti-inflammation and immunomodulatory effects. In this context, our purpose was to investigate the associated regulatory mechanisms of TMP against lipopolysaccharide (LPS)-caused pancreatic β cell Min6 injury. The injury of Min6 cells was induced by 10 μg/mL of LPS. Viability of Min6 cells was detected through CCK-8 assay, apoptosis process through flow cytometry, and the proteins involved in apoptosis through western blot. Insulin secretion was valued through the glucose-stimulated insulin secretion (GSIS) assay. microRNA-101 (miR-101) was measured through qRT-PCR. Mitogen-activated protein kinase phosphatase 1 (MKP-1) and signaling regulators was measured through western blot. We found that, TMP treatment effectively attenuated LPS-induced injury in Min6 cells by suppressing cell apoptosis and promoting insulin secretion. Further investigation revealed that TMP exerted protective effect through down-regulating miR-101, and MKP-1 was demonstrated as a target of miR-101. Moreover, TMP attenuated LPS-triggered inflammation by inactivating the JNK1/2 and NF-κB through the down-regulation of miR-101. In conclusion, our present study revealed that TMP alleviated LPS-induced injury in pancreatic β-cell Min6 injury via regulation of miR-101/MKP-1 with the bluntness of JNK1/2 and NF-κB pathways.
Collapse
Affiliation(s)
- Dong Chen
- a Department of Nuclear Medicine, Qingdao Municipal Hospital , Qingdao , China
| | - Dong Cao
- b Department of Pharmacy, Qingdao Municipal Hospital , Qingdao , China
| | - Ping Sui
- c Jining Medical University , Jining , China
| |
Collapse
|
23
|
Mo Y, Deng S, Zhang L, Huang Y, Li W, Peng Q, Liu Z, Ai Y. SS-31 reduces inflammation and oxidative stress through the inhibition of Fis1 expression in lipopolysaccharide-stimulated microglia. Biochem Biophys Res Commun 2019; 520:171-178. [PMID: 31582222 DOI: 10.1016/j.bbrc.2019.09.077] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 09/19/2019] [Indexed: 12/14/2022]
Abstract
SS-31 is a kind of mitochondrion-targeted peptide. Recent studies indicated significant neuroprotective effects of SS-31. In this study, we investigated that SS-31 protected the murine cultured microglial cells (BV-2) against lipopolysaccharide (LPS)-induced inflammation and oxidative stress through stabilizing mitochondrial morphology. The morphological study showed that SS-31 preserved LPS-induced mitochondrial ultrastructure by reducing the fission protein 1 (Fis1) expression. Flow cytometry and Western blot verified that SS-31 defended the BV-2 cells against LPS-stimulated inflammation and oxidative stress via suppressing Fis1. To sum up, our study represents that SS-31 preserves BV-2 cells from LPS-stimulated inflammation and oxidative stress by down-regulating the Fis1 expression.
Collapse
Affiliation(s)
- Yunan Mo
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| | - Songyun Deng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| | - Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| | - Yan Huang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| | - Wenchao Li
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| | - Qianyi Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| | - Zhiyong Liu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| | - Yuhang Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| |
Collapse
|
24
|
Role of glucagon-like peptides in inflammatory bowel diseases-current knowledge and future perspectives. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1321-1330. [PMID: 31359088 DOI: 10.1007/s00210-019-01698-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), are chronic, relapsing, intestinal inflammatory disorders with complex and yet unrevealed pathogenesis in which genetic, immunological, and environmental factors play a role. Nowadays, a higher proportion of elderly IBD patients with coexisting conditions, such as cardiovascular disease and/or diabetes is recorded, who require more complex treatment and became a great challenge for gastroenterologists. Furthermore, some patients do not respond to anti-IBD therapy. These facts, together with increasing comorbidities in patients with IBD, imply that urgent, more complex, novel therapeutic strategies in the treatment are needed. Glucagon-like peptides (GLPs) possess numerous functions in the human body such as lowering blood glucose level, controlling body weight, inhibiting gastric emptying, reducing food ingestion, increasing crypt cell proliferation, and improving intestinal growth and nutrient absorption. Thus, GLPs and dipeptidyl peptidase IV (DPP-IV) inhibitors have recently gained attention in IBD research. Several animal models showed that treatment with GLPs may lead to improvement of colitis. This review presents data on the multitude effects of GLPs in the inflammatory intestinal diseases and summarizes the current knowledge on GLPs, which have the potential to become a novel therapeutic option in IBD therapy.
Collapse
|
25
|
Lee J, Kim M, Kim DW. Dose-dependent elevation in amylase and lipase in response to teduglutide administration. Clin Case Rep 2019; 7:960-963. [PMID: 31110724 PMCID: PMC6509918 DOI: 10.1002/ccr3.2127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/26/2019] [Accepted: 03/04/2019] [Indexed: 01/12/2023] Open
Abstract
Teduglutide has been used for patients with short bowel syndrome. Teduglutide can be a dose-dependent triggering factor to increased amylase and lipase. Serum amylase and lipase levels should be monitored routinely for patients on teduglutide, and they should be screened before the initiation of treatment.
Collapse
Affiliation(s)
- Joshua Lee
- Boston University School of MedicineBostonMassachusetts
| | - Myoung Kim
- Boston University School of MedicineBostonMassachusetts
| | - Dong Wook Kim
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight ManagementBoston University School of MedicineBostonMassachusetts
| |
Collapse
|
26
|
Yamaguchi M, Levy RM. The combination of catechin, baicalin and β-caryophyllene potentially suppresses the production of inflammatory cytokines in mouse macrophages in vitro. Exp Ther Med 2019; 17:4312-4318. [PMID: 31007758 DOI: 10.3892/etm.2019.7452] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
It has been demonstrated that the combination of three botanical factors of (+)-catechin, baicalin and β-caryophyllene, which exhibit anti-inflammatory effects, with comparatively lower concentrations of each factor, demonstrating a potent synergistic-suppressive effect on the growth of mouse macrophage RAW264.7 cells in vitro, and suggesting it may function as a pharmacologic tool for managing inflammatory diseases. The present study was undertaken to determine the suppressive effects of (+)-catechin, baicalin or β-caryophyllene on the production of inflammatory cytokines, including TNF-α, IL-6 and IL-1β, which was enhanced by lipopolysaccharide (LPS) in RAW264.7 cells in vitro. The cells were cultured for 3 days without botanical factors, followed by incubation for 5 h in the presence of either vehicle, (+)-catechin [1 µg/ml (3.45 µM)], baicalin [1 µg/ml (2.24 µM)], or β-caryophyllene [1 µg/ml (5 µM)] with or without LPS (100 ng/ml); this did not have significant effects on the number of RAW264.7 cells. The production of TNF-α, IL-6 and IL-1β was not altered by the addition of (+)-catechin, baicalin, β-caryophyllene, or the three combined factors in RAW264.7 cells without LPS. LPS treatment caused a marked production of TNF-α, IL-6, and IL-1β. This enhancement was suppressed by the addition of (+)-catechin, baicalin or β-caryophyllene. Of note, the production of these cytokines was additively suppressed by the combination of the three factors in macrophages. Thus, the combination of (+)-catechin, baicalin and β-caryophyllene was found to reveal a potent suppressive effect on cytokine production in macrophages in vitro. This composition may be a useful tool as a potent anti-inflammatory agent.
Collapse
Affiliation(s)
- Masayoshi Yamaguchi
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1732, USA
| | - Robert M Levy
- Department of Clinical Development, Primus Pharmaceuticals, Inc., Scottsdale, AZ 85251, USA
| |
Collapse
|
27
|
6-Gingerol Attenuates Ischemia-Reperfusion-Induced Cell Apoptosis in Human AC16 Cardiomyocytes through HMGB2-JNK1/2-NF- κB Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:8798653. [PMID: 30886640 PMCID: PMC6388356 DOI: 10.1155/2019/8798653] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/02/2019] [Indexed: 12/15/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a key factor in deterioration of myocardial function. The c-Jun NH2-terminal kinase (JNK) activation and the transcription factor nuclear factor-kappaB (NF-κB) nuclear translocation have been found in I/R injury. 6-Gingerol, an important bioactive ingredient of ginger, has been reported to have cardiovascular pharmacological effects. However, the molecular mechanism through which it is beneficial is unclear. In this work, I/R induced the increase in the apoptosis and reactive oxygen species level in AC16 cardiomyocytes. 6-Gingerol administration decreased cardiomyocyte apoptosis and improved oxidative stress indexes. 6-Gingerol administration also inhibited I/R-induced HMGB2 expression upregulation and JNK activation and reduced Cleaved Poly(ADP-ribose) polymerases (PARP) and Caspase-3 expression. HMGB2 treatment mimicked the effect of I/R-induced cell damage, which was reversed by 6-gingerol administration. On the other hand, transcriptional activity of NF-κB was reduced in 6-gingerol treated cells. Thus, overall results indicated that 6-gingerol administration protected I/R-induced cardiomyocytes apoptosis via JNK/NF-κB pathway in the regulation of HMGB2. This work supported the efficacy of 6-gingerol on cardiovascular disease and partially revealed its mechanism, which was helpful for understanding the therapeutic effects of this natural drug.
Collapse
|
28
|
Tang Y, Su G, Li N, Li W, Chen G, Chen R, Zhou D, Hou Y. Preventive agents for neurodegenerative diseases from resin of Dracaena cochinchinensis attenuate LPS-induced microglia over-activation. J Nat Med 2018; 73:318-330. [PMID: 30426288 DOI: 10.1007/s11418-018-1266-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 10/05/2018] [Indexed: 12/11/2022]
|
29
|
Sandberg JC, Björck IME, Nilsson AC. Impact of rye-based evening meals on cognitive functions, mood and cardiometabolic risk factors: a randomized controlled study in healthy middle-aged subjects. Nutr J 2018; 17:102. [PMID: 30400947 PMCID: PMC6220522 DOI: 10.1186/s12937-018-0412-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 10/28/2018] [Indexed: 12/25/2022] Open
Abstract
Background Whole grain (WG) intake is associated with reduced risk of obesity, type 2 diabetes and cardiovascular disease, whereas type 2 diabetes increases the risk of cognitive decline and dementia. The purpose of this study was to investigate the effects of short-term intervention with WG rye on cognitive functions, mood and cardiometabolic risk markers in middle-aged test subjects. Method Rye-based breads were provided to 38 healthy test subjects (aged 52-70y) during three consecutive days in a crossover study design, using white wheat flour bread (WWB) as a reference. The rye-based bread consisted of a WG rye kernel/flour mixture (1:1 ratio) supplemented with resistant starch type 2 (RS2) (RB + RS2). The last bread portion was ingested at 2100 h, and cognitive function, mood and cardiometabolic risk markers were determined the following morning, 11 − 14 h post intake. Results In comparison to WWB, the RB + RS2 product increased ratings of mood parameters (valance, P < 0.001; activation P < 0.05). No differences were seen in the cognitive tests depending on intervention (P > 0.05). RB + RS2 increased insulin sensitivity (P < 0.05), fasting levels of gut hormones (PYY, P < 0.05; GLP-2, P < 0.01) and fasting concentrations of plasma acetate, butyrate and total SCFA (P < 0.001). In contrast, fasting levels of IL − 1β were decreased (P < 0.05). Insulin sensitivity was positively correlated with working memory test performance (P < 0.05). Conclusions This study display novel findings regarding effects of WG rye products on mood, and glucose and appetite regulation in middle-aged subjects, indicating anti-diabetic properties of WG rye. The beneficial effects are suggested to be mediated through gut fermentation of dietary fiber in the RB + RS2 product. Trial registration The study was retrospectively registered at ClinicalTrials.gov, register number NCT03275948. Registered September 8 2017.
Collapse
Affiliation(s)
- Jonna C Sandberg
- Department of Food Technology, Engineering and Nutrition, Lund University, P.O. Box 124, SE-221 00, Lund, Sweden.,Food for Health Science Centre, Lund University, P.O. Box 124, SE-221 00, Lund, Sweden
| | - Inger M E Björck
- Food for Health Science Centre, Lund University, P.O. Box 124, SE-221 00, Lund, Sweden
| | - Anne C Nilsson
- Department of Food Technology, Engineering and Nutrition, Lund University, P.O. Box 124, SE-221 00, Lund, Sweden. .,Food for Health Science Centre, Lund University, P.O. Box 124, SE-221 00, Lund, Sweden.
| |
Collapse
|
30
|
Nuzzo D, Baldassano S, Amato A, Picone P, Galizzi G, Caldara GF, Di Carlo M, Mulè F. Glucagon-like peptide-2 reduces the obesity-associated inflammation in the brain. Neurobiol Dis 2018; 121:296-304. [PMID: 30347266 DOI: 10.1016/j.nbd.2018.10.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/12/2018] [Accepted: 10/17/2018] [Indexed: 02/08/2023] Open
Abstract
Growing evidence suggests a link between obesity and neurodegeneration. The purpose of the present study was to explore the neuroprotective potential of glucagon-like peptide-2 (GLP-2) in the brain of high fat diet (HFD)-fed mice. Markers of inflammation and oxidative stress were analysed in the brains of obese mice chronically treated with [Gly2]-GLP-2 (teduglutide), the stable analogue of the GLP-2, and they were compared to age-matched untreated obese and lean animals. Neurodegeneration was examined by TUNEL assay. HFD feeding increased the expression of pro-inflammatory mediators (NF-kB, IL-8, TNF-α, IL-1β and IL-6), glial fibrillary acidic protein (GFAP), index of gliosis and neurodegeneration, stress marker proteins (p-ERK, Hsp60 and i-NOS), amyloid-β precursor protein (APP). [Gly2]-GLP-2 treatment significantly attenuated the HFD-induced increased expression of the various markers, as well as the higher levels of reactive oxygen species found in brains of untreated-HFD mice. Immunofluorescence confirmed that the increase of GFAP or APP in the brain cortex of HFD mice were less prominent in the [Gly2]-GLP-2 treated group. TUNEL-positive cell number in brain sections of [Gly2]-GLP-2-treated HFD-fed mice was significantly lesser in comparison with untreated-HFD animals and similar to STD fed mice. In conclusion, the results of the present study suggest that GLP-2 stable analogue improves the obesity-associated neuroinflammation and the central stress conditions, it reduces the neuronal apoptotic death, providing evidence for a neuroprotective role of the peptide.
Collapse
Affiliation(s)
- Domenico Nuzzo
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy
| | - Sara Baldassano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
| | - Antonella Amato
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
| | - Pasquale Picone
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy
| | - Giacoma Galizzi
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy
| | - Gaetano Felice Caldara
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
| | - Marta Di Carlo
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy
| | - Flavia Mulè
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy.
| |
Collapse
|
31
|
Gao Y, Zhong J, Jiang L. Raf kinase inhibitor protein protects microglial cells against 1-methyl-4-phenylpyridinium-induced neuroinflammation in vitro. Exp Cell Res 2018; 372:108-117. [PMID: 30244177 DOI: 10.1016/j.yexcr.2018.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/15/2018] [Accepted: 09/20/2018] [Indexed: 11/17/2022]
Abstract
The Raf kinase inhibitor protein (RKIP), belonging to a member of the phosphatidylethanolamine-binding protein (PEBP) family, is involved in regulating neural development. However, the role of RKIP in microglial cells stimulated with 1-methyl-4-phenylpyridinium (MPP+) has not been determined. Thus, in the present study, we investigated the role of RKIP and its underlying mechanism in Parkinson's disease (PD). Our results showed that the expression of RKIP was significantly reduced in BV-2 cells treated with MPP+. Overexpression of RKIP markedly rescued cell viability and inhibited cell apoptosis in BV-2 cells exposed to MPP+. In addition, overexpression of RKIP inhibited MPP+-induced the production of pro-inflammatory molecules in BV-2 cells. Similar results were observed in primary microglial cells isolated from neonatal mice. Exploration of the underlying mechanisms of its action indicated that overexpression of RKIP prevented the activation of NF-κB and MEK/ERK pathways in MPP+-stimulated BV-2 cells. Taken together, these findings indicated that RKIP suppresses apoptosis and inflammation in MPP+-treated microglial cells through the inactivation of NF-κB and MEK/ERK signaling pathways. Thus, RKIP may be a promising target molecular involving in the pathogenesis of PD.
Collapse
Affiliation(s)
- Yuanlin Gao
- The First ward of Neurology Department, Kaifeng Central Hospital, Kaifeng 475000, Henan Province, China
| | - Jie Zhong
- Department of Nursing, Kaifeng Central Hospital, Kaifeng 475000, Henan Province, China.
| | - Lei Jiang
- The First ward of Neurology Department, Kaifeng Central Hospital, Kaifeng 475000, Henan Province, China
| |
Collapse
|
32
|
Gu J, Liu J, Huang T, Zhang W, Jia B, Mu N, Zhang K, Hao Q, Li W, Liu W, Zhang W, Zhang Y, Xue X, Zhang C, Li M. The protective and anti-inflammatory effects of a modified glucagon-like peptide-2 dimer in inflammatory bowel disease. Biochem Pharmacol 2018; 155:425-433. [PMID: 30040929 DOI: 10.1016/j.bcp.2018.07.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/20/2018] [Indexed: 12/26/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent, and remitting inflammatory disease resulting from immune dysregulation in the gut. As a clinically frequent disease, it can affect individuals throughout their lives, with multiple complications. Glucagon-like peptide 2 (GLP-2) is a potent epithelium-specific intestinal growth factor. However, native GLP-2 has a relatively short half-life in human circulation because of extensive renal clearance and rapid degradation by the proteolytic enzyme dipeptidyl peptidase-IV (DPP-IV). Previously, We prepared a recombinant GLP-2 variant (GLP-2②), which has increased half-life and activity as compared to the [Gly2]GLP-2 monomer. The aim of the present study was to investigate the protective potential of GLP-2② in IBD models. LPS-induced in vitro model and dextran sulfate sodium (DSS)-induced in vivo model were used to study the anti-inflammatory and therapeutic effect of GLP-2②. We found that treated with GLP-2② showed a significantly reduction in the secretion of inflammatory cytokines. Furthermore, GLP-2② alleviated symptoms of DSS-induced colitis. GLP-2② treated mice displayed an increase in body weight, lower colitis scores, and fewer mucosal damage compared with GLP-2 treated mice. MPO activities, protein expression of NLRP3 and COX2 in the colon tissues were significantly reduced in GLP-2② groups. Importantly, the ameliorative effect of GLP-2② was related to anti-apoptosis effect in colon tissues. These findings demonstrated that GLP-2② may offer a superior therapeutic benefit over [Gly2]GLP-2 monomer for treatment of IBD.
Collapse
Affiliation(s)
- Jintao Gu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Jun Liu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Tonglie Huang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Wangqian Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Bo Jia
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Nan Mu
- Department of Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Kuo Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Liu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaochang Xue
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
33
|
Qin S, Yang C, Huang W, Du S, Mai H, Xiao J, Lü T. Sulforaphane attenuates microglia-mediated neuronal necroptosis through down-regulation of MAPK/NF-κB signaling pathways in LPS-activated BV-2 microglia. Pharmacol Res 2018; 133:218-235. [DOI: 10.1016/j.phrs.2018.01.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/13/2018] [Accepted: 01/22/2018] [Indexed: 12/17/2022]
|
34
|
Chen S, Fang Y, Xu S, Reis C, Zhang J. Mammalian Sterile20-like Kinases: Signalings and Roles in Central Nervous System. Aging Dis 2018; 9:537-552. [PMID: 29896440 PMCID: PMC5988607 DOI: 10.14336/ad.2017.0702] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/02/2017] [Indexed: 12/25/2022] Open
Abstract
Mammalian Sterile20-like (MST) kinases are located upstream in the mitogen-activated protein kinase pathway, and play an important role in cell proliferation, differentiation, renewal, polarization and migration. Generally, five MST kinases exist in mammalian signal transduction pathways, including MST1, MST2, MST3, MST4 and YSK1. The central nervous system (CNS) is a sophisticated entity that takes charge of information reception, integration and response. Recently, accumulating evidence proposes that MST kinases are critical in the development of disease in different systems involving the CNS. In this review, we summarized the signal transduction pathways and interacting proteins of MST kinases. The potential biological function of each MST kinase and the commonly reported MST-related diseases in the neural system are also reviewed. Further investigation of MST kinases and their interaction with CNS diseases would provide the medical community with new therapeutic targets for human diseases.
Collapse
Affiliation(s)
- Sheng Chen
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuanjian Fang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shenbin Xu
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cesar Reis
- 2Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA.,3Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,4Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Peiminine Protects Dopaminergic Neurons from Inflammation-Induced Cell Death by Inhibiting the ERK1/2 and NF-κB Signalling Pathways. Int J Mol Sci 2018. [PMID: 29534526 PMCID: PMC5877682 DOI: 10.3390/ijms19030821] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation, characterized marked by microglial activation, plays a very important role in the pathogenesis of Parkinson's disease (PD). Upon activation, pro-inflammatory mediators are produced by microglia, triggering excessive inflammatory responses and ultimately damaging dopaminergic neurons. Therefore, the identification of agents that inhibit neuroinflammation may be an effective approach for developing novel treatments for PD. In this study, we sought to investigate whether peiminine protects dopaminergic neurons by inhibiting neuroinflammation. We evaluated the effects of peiminine on behavioural dysfunction, microglial activation and the loss of dopaminergic neurons in a rat model of lipopolysaccharide (LPS)-induced PD. BV-2 cells were pretreated with peiminine for 1 h and then stimulated with LPS for different times. Then, inflammatory responses and the related signalling pathways were analysed. Peiminine markedly attenuated behavioural dysfunction and inhibited the loss of dopaminergic neurons and microglial activation in the LPS-induced PD rat model. In BV-2 cells, peiminine significantly decreased LPS-induced expression of the pro-inflammatory mediators TNF-α, IL-6 and IL-1β, COX-2 and iNOS by inhibiting the phosphorylation of ERK1/2, AKT and NF-κB p65. Based on these results demonstrated that peiminine has a role in protecting dopaminergic neurons in the LPS-induced PD rat model by inhibiting neuroinflammation.
Collapse
|
36
|
Haque A, Hsieh MF, Hassan SI, Haque Faizi MS, Saha A, Dege N, Rather JA, Khan MS. Synthesis, characterization, and pharmacological studies of ferrocene-1H-1,2,3-triazole hybrids. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.06.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
37
|
Isoastragaloside I inhibits NF-κB activation and inflammatory responses in BV-2 microglial cells stimulated with lipopolysaccharide. Int J Mol Med 2017; 40:1270-1276. [PMID: 28902359 DOI: 10.3892/ijmm.2017.3114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/25/2017] [Indexed: 11/05/2022] Open
Abstract
The excessive activation of microglia in many neurodegenerative diseases is detrimental to neuronal survival. Isoastragaloside I (ISO I) is a natural saponin molecule found within the roots of Astragalus membranaceus, a famous traditional Chinese medicine. In the present study, the anti‑inflammatory effects and the mechanisms of action of ISO I on activated BV-2 cells stimulated with lipopolysaccharide (LPS) were investigated. ISO I dose‑dependently inhibited the excessive release of nitric oxide (NO) and tumor necrosis factor (TNF)-α in the LPS-stimulated BV-2 cells. Moreover, it decreased the production of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), and mitigated the gene expression of interleukin (IL)-1β, TNF-α and iNOS induced by LPS. Further experiments revealed that ISO I decreased the phosphorylation levels of nuclear factor-κB (NF-κB), and suppressed its nuclear translocation and transactivation activity. In addition, it inhibited the activation of signaling pathway molecules, such as PI3K, Akt and mitogen-activated protein kinases (MAPKs). Taken together, our findings suggest that ISO I prevents LPS-induced microglial activation probably by inhibiting the activation of the NF-κB via PI3K/Akt and MAPK signaling pathways, indicating its therapeutic potential for neurological diseases relevant to neuroinflammation.
Collapse
|
38
|
Yan X, Liu DF, Zhang XY, Liu D, Xu SY, Chen GX, Huang BX, Ren WZ, Wang W, Fu SP, Liu JX. Vanillin Protects Dopaminergic Neurons against Inflammation-Mediated Cell Death by Inhibiting ERK1/2, P38 and the NF-κB Signaling Pathway. Int J Mol Sci 2017; 18:ijms18020389. [PMID: 28208679 PMCID: PMC5343924 DOI: 10.3390/ijms18020389] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 01/22/2017] [Accepted: 02/06/2017] [Indexed: 11/16/2022] Open
Abstract
Neuroinflammation plays a very important role in the pathogenesis of Parkinson’s disease (PD). After activation, microglia produce pro-inflammatory mediators that damage surrounding neurons. Consequently, the inhibition of microglial activation might represent a new therapeutic approach of PD. Vanillin has been shown to protect dopaminergic neurons, but the mechanism is still unclear. Herein, we further study the underlying mechanisms in lipopolysaccharide (LPS)-induced PD models. In vivo, we firstly established rat models of PD by unilateral injection of LPS into substantia nigra (SN), and then examined the role of vanillin in motor dysfunction, microglial activation and degeneration of dopaminergic neurons. In vitro, murine microglial BV-2 cells were treated with vanillin prior to the incubation of LPS, and then the inflammatory responses and the related signaling pathways were analyzed. The in vivo results showed that vanillin markedly improved the motor dysfunction, suppressed degeneration of dopaminergic neurons and inhibited microglial over-activation induced by LPS intranigral injection. The in vitro studies demonstrated that vanillin reduces LPS-induced expression of inducible nitric oxide (iNOS), cyclooxygenase-2 (COX-2), IL-1β, and IL-6 through regulating ERK1/2, p38 and NF-κB signaling. Collectively, these data indicated that vanillin has a role in protecting dopaminergic neurons via inhibiting inflammatory activation.
Collapse
Affiliation(s)
- Xuan Yan
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Dian-Feng Liu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xiang-Yang Zhang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Dong Liu
- Animal Husbandry and Veterinary Medicine, Cangzhou Technic College, Cangzhou 061001, China.
| | - Shi-Yao Xu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Guang-Xin Chen
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Bing-Xu Huang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Wen-Zhi Ren
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Wei Wang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Shou-Peng Fu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Ju-Xiong Liu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
39
|
Zhao JW, Chen DS, Deng CS, Wang Q, Zhu W, Lin L. Evaluation of anti-inflammatory activity of compounds isolated from the rhizome of Ophiopogon japonicas. Altern Ther Health Med 2017; 17:7. [PMID: 28056939 PMCID: PMC5217338 DOI: 10.1186/s12906-016-1539-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Background Ophiopogon japonicas (L.f) Ker-Gawl has been used as a traditional Chinese medicine to cure acute and chronic inflammation and cardiovascular diseases including thrombotic diseases for thousands of years. Previous phytochemical studies showed that O. japonicus contained compounds with anti-inflammatory activity. The aim of this study was to identify and isolate compounds with anti-inflammatory activity from the rhizome of O. japonicas. Methods Compounds were isolated by various column chromatography and their structures were identified in terms of nuclear magnetic resonance spectrum (NMR) and mass spectrum (MS). To measure the anti-inflammatory effects of thirteen compounds in LPS-induced RAW 264.7 macrophage cells, we used the following methods: cell viability assay, nitric oxide assay, enzyme-linked immunosorbent assay, quantitative real-time PCR analysis and western blotting analysis. Results One new and twelve known compounds (mainly homoisoflavonoids) were extracted from O. japonicas, in which 4′-O-Demethylophiopogonanone E (10) was considered as a new compound, additionally, compounds 4-O-(2-Hydroxy-1- hydroxymethylethyl)-dihydroconiferyl alcohol (2) and 5,7-dihydroxy-6-methyl-3-(2′, 4′-dihydroxybenzyl) chroman-4-one (12) were isolated from the rhizome of O. japonicas for the first time. The isolated compounds Oleic acid (3), Palmitic acid (4), desmethylisoophiopogonone B [5,7-dihydroxy-3-(4′-hydroxybenzyl)-8- methyl- chromone] (5), 5,7-dihydroxy-6-methyl-3-(4′-hydroxybenzyl) chromone (7) and 10 significantly suppressed the production of NO in LPS-induced RAW 264.7 cells. Especially compound 10 showed the strongest effect against the production of the pro-inflammatory cytokine IL-1β and IL-6 with the IC50 value of 32.5 ± 3.5 μg/mL and 13.4 ± 2.3 μg/mL, respectively. Further analysis elucidated that the anti-inflammatory activity of compound 10 might be exerted through inhibiting the phosphorylation of ERK1/2 and JNK in MAPK signaling pathways to decrease NO and pro-inflammatory cytokines production. Conclusions Our results indicated that 4′-O-Demethylophiopogonanone E can be considered as a potential source of therapeutic medicine for inflammatory diseases.
Collapse
|
40
|
N-Acetyl-chitobiose ameliorates metabolism dysfunction through Erk/p38 MAPK and histone H3 phosphorylation in type 2 diabetes mice. J Funct Foods 2017. [DOI: 10.1016/j.jff.2016.11.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
41
|
Yamaguchi M, Levy RM. The combination of β-caryophyllene, baicalin and catechin synergistically suppresses the proliferation and promotes the death of RAW267.4 macrophages in vitro. Int J Mol Med 2016; 38:1940-1946. [DOI: 10.3892/ijmm.2016.2801] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/04/2016] [Indexed: 11/05/2022] Open
|