1
|
Rojas-Juárez R, Rodríguez-Castelán J, Cuatecontzi-Fuentes I, Mendez-Tepepa M, Cruz-Lumbreras R, Rodríguez-Antolín J, Arroyo-Helguera OE, Cuevas-Romero E. Pancreatic inflammation induced by hypothyroidism in female rabbits is associated with cholesterol accumulation and a reduced expression of CYP51A1, FXRβ, and PPARβ/δ. Anat Rec (Hoboken) 2025; 308:1517-1528. [PMID: 39403034 DOI: 10.1002/ar.25590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 04/04/2025]
Abstract
In women and animal models, hypothyroidism induces hypercholesterolemia, pancreatitis, and insulitis. We investigated whether lipids are involved in the effects of hypothyroidism in the pancreas. Control (n = 6) and hypothyroid (n = 6) adult female rabbits were used. We quantified serum and pancreatic triacylglycerol and total cholesterol levels, the oxidative and antioxidant status, and the expression of low-density lipoprotein cholesterol receptor (LDLR) in the pancreas. Inflammation of the pancreas was evaluated by infiltration of immune cells positive to CD163 and α-farnesoid receptor (FXRα). Other lipid players involved in both inflammation and insulin secretion of the pancreas, such as lanosterol 14-α-demethylase (CYP51A1), β-farnesoid receptor (FXRβ), 3β-hydroxysteroid dehydrogenase (3β-HSD), and peroxisome proliferator-activated receptor β (PPARβ/δ), were quantified. Groups were compared by t-Student or U-Mann-Whitney tests (p ≤ 0.05). Hypothyroidism induced hypercholesterolemia and a high cholesterol accumulation in the pancreas of female rabbits, without affecting oxidative or antioxidative status nor the expression of LDLR. The pancreas of hypothyroid females showed inflammation identified by a great infiltration of immune cells, macrophages CD163+, and loss of expression of FXRα+ in immune cells. Moreover, a reduced expression of CYP51A1, FXRβ, and PPARβ/δ, but not 3β-HSD, in the hypothyroid pancreas was found. Pancreatitis and insulitis promoted by hypothyroidism may be related to the accumulation of cholesterol, lanosterol actions, and the activation of PPARβ/δ. All inflammatory markers evaluated in this study are related to glucose regulation, suggesting the link between hypothyroidism and diabetes.
Collapse
Affiliation(s)
- Rubicela Rojas-Juárez
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
- Programa Educativo de Química Clínica, Facultad de Ciencias de la Salud, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | | | - Ismael Cuatecontzi-Fuentes
- Programa Educativo de Química Clínica, Facultad de Ciencias de la Salud, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
- Maestría en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Maribel Mendez-Tepepa
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Rosalía Cruz-Lumbreras
- Programa Educativo de Química Clínica, Facultad de Ciencias de la Salud, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
- Facultad de Odontología, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Jorge Rodríguez-Antolín
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | | | - Estela Cuevas-Romero
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| |
Collapse
|
2
|
Xu B, Li H, Zheng H, Gao Z, Miao Z, Xu X, Yang H, Yang Y. Interleukin-18 interacts with NKCC1 to mediate brain injury after intracerebral hemorrhage. Brain Behav Immun Health 2024; 42:100890. [PMID: 39507306 PMCID: PMC11538613 DOI: 10.1016/j.bbih.2024.100890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/13/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024] Open
Abstract
Interleukin 18 (IL-18), a proinflammatory cytokine, has been implicated in various neurological disorders, including cerebrovascular disease and psychiatric disorders. In a previous study, IL-18 was observed to activate microglia and enhance the inflammatory response following intracranial hemorrhage (ICH). However, the underlying mechanism remains unclear. In the present study, we found that IL-18 and IL-18 receptor (IL-18 R) are primarily secreted by neurons during the early stages after ICH, with microglia becoming the predominant source at 12-24 h after ICH. Meanwhile, the expression level of IL-18 R increased following ICH, along with an augmentation in the binding affinity of IL-18 R to IL-18. Subsequently, the deficiency of IL-18 R mitigated neurological impairment and subsequent activation of inflammatory pathways in mice post-ICH. Moreover, our findings suggest that IL-18-induced neurological injury after ICH may be mediated by the interaction between IL18R and NKCC1. Significantly, the NKCC1 inhibitor rescued the neurologic injury after ICH. In conclusion, our study suggests that targeting the IL-18/IL-18R/NKCC1 pathway could be an effective therapeutic strategy to attenuate secondary brain injury after ICH.
Collapse
Affiliation(s)
- Beibei Xu
- Departments of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hao Li
- Departments of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Departments of Neurology, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - He Zheng
- Departments of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhongyu Gao
- Computer Science and Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Xingshun Xu
- Departments of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Hao Yang
- Department of Fetology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yi Yang
- Departments of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
3
|
Provasek VE, Bacolla A, Rangaswamy S, Mitra J, Kodavati M, Yusuf IO, Malojirao VH, Vasquez V, Britz GW, Li GM, Xu Z, Mitra S, Garruto RM, Tainer JA, Hegde ML. RNA/DNA Binding Protein TDP43 Regulates DNA Mismatch Repair Genes with Implications for Genome Stability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594552. [PMID: 38798341 PMCID: PMC11118483 DOI: 10.1101/2024.05.16.594552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
TAR DNA-binding protein 43 (TDP43) is increasingly recognized for its involvement in neurodegenerative diseases, particularly amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). TDP43 proteinopathy, characterized by dysregulated nuclear export and cytoplasmic aggregation, is present in most ALS/FTD cases and is associated with a loss of nuclear function and genomic instability in neurons. Building on prior evidence linking TDP43 pathology to DNA double-strand breaks (DSBs), this study identifies a novel regulatory role for TDP43 in the DNA mismatch repair (MMR) pathway. We demonstrate that depletion or overexpression of TDP43 affects the expression of key MMR genes, including MLH1, MSH6, MSH2, MSH3, and PMS2. Specifically, TDP43 modulates the expression of MLH1 and MSH6 proteins through alternative splicing and transcript stability. These findings are validated in ALS mice models, patient-derived neural progenitor cells and autopsied brain tissues from ALS patients. Furthermore, MMR depletion showed a partial rescue of TDP43-induced DNA damage in neuronal cells. Bioinformatics analysis of TCGA cancer database reveals significant correlations between TDP43 and MMR gene expressions and mutational burden across various cancer subtypes. These results collectively establish TDP43 as a critical regulator of the MMR pathway, with broad implications for understanding the genomic instability underlying neurodegenerative and neoplastic diseases.
Collapse
Affiliation(s)
- Vincent E Provasek
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
- School of Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Albino Bacolla
- Department of Molecular and Cellular Oncology, Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Suganya Rangaswamy
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Joy Mitra
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Manohar Kodavati
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Issa O Yusuf
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Vikas H Malojirao
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Velmarini Vasquez
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Gavin W Britz
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Neurosurgery and Department of Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Guo-Min Li
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zuoshang Xu
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Sankar Mitra
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Ralph M Garruto
- Department of Biological Sciences, Binghamton University, State University of New York, Binghamton, NY 13902
| | - John A Tainer
- Department of Molecular and Cellular Oncology, Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Muralidhar L Hegde
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
4
|
Girdhar M, Sen A, Nigam A, Oswalia J, Kumar S, Gupta R. Antimicrobial peptide-based strategies to overcome antimicrobial resistance. Arch Microbiol 2024; 206:411. [PMID: 39311963 DOI: 10.1007/s00203-024-04133-x] [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: 07/13/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024]
Abstract
Antibiotic resistance has emerged as a global threat, rendering the existing conventional treatment strategies ineffective. In view of this, antimicrobial peptides (AMPs) have proven to be potent alternative therapeutic interventions with a wide range of applications in clinical health. AMPs are small peptides produced naturally as a part of the innate immune responses against a broad range of bacterial, fungal and viral pathogens. AMPs present a myriad of advantages over traditional antibiotics, including their ability to target multiple sites, reduced susceptibility to resistance development, and high efficacy at low doses. These peptides have demonstrated notable potential in inhibiting microbes resistant to traditional antibiotics, including the notorious ESKAPE pathogens, recognized as the primary culprits behind nosocomial infections. AMPs, with their multifaceted benefits, emerge as promising candidates in the ongoing efforts to combat the escalating challenges posed by antibiotic resistance. This in-depth review provides a detailed discussion on AMPs, encompassing their classification, mechanism of action, and diverse clinical applications. Focus has been laid on combating newly emerging drug-resistant organisms, emphasizing the significance of AMPs in mitigating this pressing challenge. The review also illuminates potential future strategies that may be implemented to improve AMP efficacy, such as structural modifications and using AMPs in combination with antibiotics and matrix-inhibiting compounds.
Collapse
Affiliation(s)
| | - Aparajita Sen
- Department of Genetics, University of Delhi, South Campus, New Delhi, 110021, India
| | - Arti Nigam
- Department of Microbiology, Institute of Home Economics, University of Delhi, New Delhi, 110016, India
| | - Jyoti Oswalia
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sachin Kumar
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India
| | - Rashi Gupta
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India.
| |
Collapse
|
5
|
Zubareva OE, Kharisova AR, Roginskaya AI, Kovalenko AA, Zakharova MV, Schwarz AP, Sinyak DS, Zaitsev AV. PPARβ/δ Agonist GW0742 Modulates Microglial and Astroglial Gene Expression in a Rat Model of Temporal Lobe Epilepsy. Int J Mol Sci 2024; 25:10015. [PMID: 39337503 PMCID: PMC11432388 DOI: 10.3390/ijms251810015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
The role of astroglial and microglial cells in the pathogenesis of epilepsy is currently under active investigation. It has been proposed that the activity of these cells may be regulated by the agonists of peroxisome proliferator-activated nuclear receptors (PPARs). This study investigated the effects of a seven-day treatment with the PPAR β/δ agonist GW0742 (Fitorine, 5 mg/kg/day) on the behavior and gene expression of the astroglial and microglial proteins involved in the regulation of epileptogenesis in the rat brain within a lithium-pilocarpine model of temporal lobe epilepsy (TLE). TLE resulted in decreased social and increased locomotor activity in the rats, increased expression of astro- and microglial activation marker genes (Gfap, Aif1), pro- and anti-inflammatory cytokine genes (Tnfa, Il1b, Il1rn), and altered expression of other microglial (Nlrp3, Arg1) and astroglial (Lcn2, S100a10) genes in the dorsal hippocampus and cerebral cortex. GW0742 attenuated, but did not completely block, some of these impairments. Specifically, the treatment affected Gfap gene expression in the dorsal hippocampus and Aif1 gene expression in the cortex. The GW0742 injections attenuated the TLE-specific enhancement of Nlrp3 and Il1rn gene expression in the cortex. These results suggest that GW0742 may affect the expression of some genes involved in the regulation of epileptogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Aleksey V. Zaitsev
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 194223 Saint Petersburg, Russia; (O.E.Z.); (A.R.K.); (A.I.R.); (A.A.K.); (M.V.Z.); (A.P.S.); (D.S.S.)
| |
Collapse
|
6
|
Pan HC, Yang CN, Lee WJ, Sheehan J, Wu SM, Chen HS, Lin MH, Shen LW, Lee SH, Shen CC, Pan LY, Liu SH, Sheu ML. Melatonin Enhanced Microglia M2 Polarization in Rat Model of Neuro-inflammation Via Regulating ER Stress/PPARδ/SIRT1 Signaling Axis. J Neuroimmune Pharmacol 2024; 19:11. [PMID: 38530514 DOI: 10.1007/s11481-024-10108-y] [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: 08/10/2022] [Accepted: 02/15/2024] [Indexed: 03/28/2024]
Abstract
Neuro-inflammation involves distinct alterations of microglial phenotypes, containing nocuous pro-inflammatory M1-phenotype and neuroprotective anti-inflammatory M-phenotype. Currently, there is no effective treatment for modulating such alterations. M1/M2 marker of primary microglia influenced by Melatonin were detected via qPCR. Functional activities were explored by western blotting, luciferase activity, EMSA, and ChIP assay. Structure interaction was assessed by molecular docking and LIGPLOT analysis. ER-stress detection was examined by ultrastructure TEM, calapin activity, and ERSE assay. The functional neurobehavioral evaluations were used for investigation of Melatonin on the neuroinflammation in vivo. Melatonin had targeted on Peroxisome Proliferator Activated Receptor Delta (PPARδ) activity, boosted LPS-stimulated alterations in polarization from the M1 to the M2 phenotype, and thereby inhibited NFκB-IKKβ activation in primary microglia. The PPARδ agonist L-165,041 or over-expression of PPARδ plasmid (ov-PPARδ) showed similar results. Molecular docking screening, dynamic simulation approaches, and biological studies of Melatonin showed that the activated site was located at PPARδ (phospho-Thr256-PPARδ). Activated microglia had lowered PPARδ activity as well as the downstream SIRT1 formation via enhancing ER-stress. Melatonin, PPARδ agonist and ov-PPARδ all effectively reversed the above-mentioned effects. Melatonin blocked ER-stress by regulating calapin activity and expression in LPS-activated microglia. Additionally, Melatonin or L-165,041 ameliorated the neurobehavioral deficits in LPS-aggravated neuroinflammatory mice through blocking microglia activities, and also promoted phenotype changes to M2-predominant microglia. Melatonin suppressed neuro-inflammation in vitro and in vivo by tuning microglial activation through the ER-stress-dependent PPARδ/SIRT1 signaling cascade. This treatment strategy is an encouraging pharmacological approach for the remedy of neuro-inflammation associated disorders.
Collapse
Affiliation(s)
- Hung-Chuan Pan
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Doctoral Program in Biotechnology Industrial Management and Innovation, National Chung Hsing University, Taichung, Taiwan
- College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| | - Sheng-Mao Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Hong-Shiu Chen
- Department of Neurosurgery, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Mao-Hsun Lin
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Li-Wei Shen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Hua Lee
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chin-Chang Shen
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Liang-Yi Pan
- School of Medicine, Kaohsiung Medical University, Taichung, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Meei-Ling Sheu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Sciences, College of Life Sciences, National Chung Hsing University, 250, Kuo Kuang Road, Taichung, 402, Taiwan.
| |
Collapse
|
7
|
Chen Y, Tang W, Huang X, An Y, Li J, Yuan S, Shan H, Zhang M. Mitophagy in intracerebral hemorrhage: a new target for therapeutic intervention. Neural Regen Res 2024; 19:316-323. [PMID: 37488884 PMCID: PMC10503626 DOI: 10.4103/1673-5374.379019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/12/2023] [Accepted: 05/18/2023] [Indexed: 07/26/2023] Open
Abstract
Intracerebral hemorrhage is a life-threatening condition with a high fatality rate and severe sequelae. However, there is currently no treatment available for intracerebral hemorrhage, unlike for other stroke subtypes. Recent studies have indicated that mitochondrial dysfunction and mitophagy likely relate to the pathophysiology of intracerebral hemorrhage. Mitophagy, or selective autophagy of mitochondria, is an essential pathway to preserve mitochondrial homeostasis by clearing up damaged mitochondria. Mitophagy markedly contributes to the reduction of secondary brain injury caused by mitochondrial dysfunction after intracerebral hemorrhage. This review provides an overview of the mitochondrial dysfunction that occurs after intracerebral hemorrhage and the underlying mechanisms regarding how mitophagy regulates it, and discusses the new direction of therapeutic strategies targeting mitophagy for intracerebral hemorrhage, aiming to determine the close connection between mitophagy and intracerebral hemorrhage and identify new therapies to modulate mitophagy after intracerebral hemorrhage. In conclusion, although only a small number of drugs modulating mitophagy in intracerebral hemorrhage have been found thus far, most of which are in the preclinical stage and require further investigation, mitophagy is still a very valid and promising therapeutic target for intracerebral hemorrhage in the long run.
Collapse
Affiliation(s)
- Yiyang Chen
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
- Shanghai Key Lab of Forensic Medicine, Key Lab of Forensic Science, Ministry of Justice (Academy of Forensic Science), Shanghai, China
| | - Wenxuan Tang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Xinqi Huang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Yumei An
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiawen Li
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Shengye Yuan
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Mingyang Zhang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
- Shanghai Key Lab of Forensic Medicine, Key Lab of Forensic Science, Ministry of Justice (Academy of Forensic Science), Shanghai, China
| |
Collapse
|
8
|
Lu W, Huang J, Flores J, Li P, Wang W, Liu S, Zhang JH, Tang J. GW0742 reduces mast cells degranulation and attenuates neurological impairments via PPAR β/δ/CD300a/SHP1 pathway after GMH in neonatal rats. Exp Neurol 2024; 372:114615. [PMID: 37995951 PMCID: PMC10842885 DOI: 10.1016/j.expneurol.2023.114615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/03/2023] [Accepted: 11/19/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Activation of mast cells plays an important role in brain inflammation. CD300a, an inhibitory receptor located on mast cell surfaces, has been reported to reduce the production of pro-inflammatory cytokines and exert protective effects in inflammation-related diseases. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ), a ligand-activated nuclear receptor, activation upregulates the transcription of CD300a. In this study, we aim to investigate the role of PPARβ/δ in the attenuation of germinal matrix hemorrhage (GMH)-induced mast cell activation via CD300a/SHP1 pathway. METHODS GMH model was induced by intraparenchymal injection of bacterial collagenase into the right hemispheric ganglionic eminence in P7 Sprague Dawley rats. GW0742, a PPARβ/δ agonist, was administered intranasally at 1 h post-ictus. CD300a small interfering RNA (siRNA) and PPARβ/δ siRNA were injected intracerebroventricularly 5 days and 2 days before GMH induction. Behavioral tests, Western blot, immunofluorescence, Toluidine Blue staining, and Nissl staining were applied to assess post-GMH evaluation. RESULTS Results demonstrated that endogenous protein levels of PPARβ/δ and CD300a were decreased, whereas chymase, tryptase, IL-17A and transforming growth factor β1 (TGF-β1) were elevated after GMH. GMH induced significant short- and long-term neurobehavioral deficits in rat pups. GW0742 decreased mast cell degranulation, improved neurological outcomes, and attenuated ventriculomegaly after GMH. Additionally, GW0742 increased expression of PPARβ/δ, CD300a and phosphorylation of SHP1, decreased phosphorylation of Syk, chymase, tryptase, IL-17A and TGF-β1 levels. PPARβ/δ siRNA and CD300a siRNA abolished the beneficial effects of GW0742. CONCLUSIONS GW0742 inhibited mast cell-induced inflammation and improved neurobehavior after GMH, which is mediated by PPARβ/δ/CD300a/SHP1 pathway. GW0742 may serve as a potential treatment to reduce brain injury for GMH patients.
Collapse
Affiliation(s)
- Weitian Lu
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing 400016, China; Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Juan Huang
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing 400016, China; Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jerry Flores
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Peng Li
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Wenna Wang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Shengpeng Liu
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
9
|
Li Z, Hu E, Zheng F, Wang S, Zhang W, Luo J, Tang T, Huang Q, Wang Y. The effects of astragaloside IV on gut microbiota and serum metabolism in a mice model of intracerebral hemorrhage. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155086. [PMID: 37783132 DOI: 10.1016/j.phymed.2023.155086] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 10/04/2023]
Abstract
BACKGROUND Astragaloside IV (AS-IV) is the main active component of "Astragalus membranaceus (Fisch.) Bunge, a synonym of Astragalus propinquus Schischkin (Fabaceae)", which demonstrated to be useful for the treatment of intracerebral hemorrhage (ICH). However, due to the low bioavailability and barrier permeability of AS-IV, the gut microbiota may be an important key regulator for AS-IV to work. OBJECTIVE To explore the influences of gut microbiota on the effects of AS-IV on ICH. METHODS Mice were randomly divided into five groups: sham, ICH, and AS-IV-treated groups (25 mg/kg, 50 mg/kg, and 100 mg/kg). Behavioral tests, brain histopathology, and immunohistochemistry analysis were used to evaluate the degree of brain injury. Western blot was employed to verify peri‑hematoma inflammation. The plasma lipopolysaccharide (LPS) leakage, the fluorescein isothiocyanate-dextran permeability, the colonic histopathology, and immunohistochemistry were detected to evaluate the barrier function of intestinal mucosal. Moreover, 16S rDNA sequencing and metabolomic analysis was applied to screen differential bacteria and metabolites, respectively. The correlation analysis was adopted to determine the potential relationship between differential bacteria and critical metabolites or neurological deficits. RESULTS AS-IV alleviated neurological deficits, neuronal injury and apoptosis, and blood-brain barrier disruption. This compound reduced tumor necrosis factor (TNF)-α expression, increased arginase (Arg)-1 and interleukin (IL)-33 levels around the hematoma. Next, 16S rRNA sequencing indicated that AS-IV altered the gut microbiota, and inhibited the production of conditional pathogenic bacteria. Metabolomic analysis demonstrated that AS-IV regulated the serum metabolic profiles, especially the aminoacid metabolism and peroxisome proliferator-activated receptor (PPAR) signaling pathway. Additionally, AS-IV mitigated intestinal barrier damage and LPS leakage. CONCLUSION This study provides a new perspective on the use of AS-IV for the treatment of ICH. Among them, gut microbiota and its metabolites may be the key regulator of AS-IV in treating ICH.
Collapse
Affiliation(s)
- Zhilin Li
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - En Hu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya Hospital, Central South University, Jiangxi 330004, China
| | - Fei Zheng
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Song Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Stroke Center, Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jiekun Luo
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya Hospital, Central South University, Jiangxi 330004, China
| | - Tao Tang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya Hospital, Central South University, Jiangxi 330004, China
| | - Qing Huang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Stroke Center, Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yang Wang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya Hospital, Central South University, Jiangxi 330004, China.
| |
Collapse
|
10
|
Gasparyan A, Navarro D, Navarrete F, Austrich-Olivares A, Scoma ER, Hambardikar VD, Acosta GB, Solesio ME, Manzanares J. Cannabidiol repairs behavioral and brain disturbances in a model of fetal alcohol spectrum disorder. Pharmacol Res 2023; 188:106655. [PMID: 36642113 DOI: 10.1016/j.phrs.2023.106655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/31/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Fetal alcohol spectrum disorder (FASD) includes neuropsychiatric disturbances related to gestational and lactational ethanol exposure. Available treatments are minimal and do not modulate ethanol-induced damage. Developing animal models simulating FASD is essential for understanding the underlying brain alterations and searching for efficient therapeutic approaches. The main goal of this study was to evaluate the effects of early and chronic cannabidiol (CBD) administration on offspring exposed to an animal model of FASD. Ethanol gavage (3 g/kg/12 h, p.o.) was administered to C57BL/6 J female mice, with a previous history of alcohol consumption, between gestational day 7 and postnatal day 21. On the weaning day, pups were separated by sex, and CBD administration began (30 mg/kg/day, i.p.). After 4-6 weeks of treatment, behavioral and neurobiological changes were analyzed. Mice exposed to the animal model of FASD showed higher anxiogenic and depressive-like behaviors and cognitive impairment that were evaluated through several experimental tests. These behaviors were accompanied by alterations in the gene, cellular and metabolomic targets. CBD administration normalized FASD model-induced emotional and cognitive disturbances, gene expression, and cellular changes with sex-dependent differences. CBD modulates the metabolomic changes detected in the hippocampus and prefrontal cortex. Interestingly, no changes were found in mitochondria or the oxidative status of the cells. These results suggest that the early and repeated administration of CBD modulated the long-lasting behavioral, gene and protein alterations induced by the FASD model, encouraging the possibility of performing clinical trials to evaluate the effects of CBD in children affected with FASD.
Collapse
Affiliation(s)
- Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Amaya Austrich-Olivares
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain
| | - Ernest R Scoma
- Rutgers University, Department of Biology and CCIB, Camden, NJ, USA
| | | | - Gabriela B Acosta
- Instituto de Neurociencias Cognitiva y Traslacional (INCYT), CONICET, INECO, Universidad Favaloro, Ciudad Autónoma de Buenos Aires C1079ABE, Argentina
| | - María E Solesio
- Rutgers University, Department of Biology and CCIB, Camden, NJ, USA
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernandez-CSIC, San Juan de Alicante, Alicante, Spain; Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| |
Collapse
|
11
|
Cisplatin-induced changes in calcitonin gene-related peptide or TNF-α release in rat dorsal root ganglia in vitro model of neurotoxicity are not reverted by rosiglitazone. Neurotoxicology 2022; 93:211-221. [DOI: 10.1016/j.neuro.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/24/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
|
12
|
Roginskaya AI, Dyomina AV, Kovalenko AA, Zakharova MV, Schwarz AP, Melik-Kasumov TB, Zubareva OE. Effect of Anakinra on the Gene Expression of Receptors Activated by the Peroxisome Proliferator in the Rat Brain in the Lithium Pilocarpine Model of Epilepsy. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Li H, Tian J, Yin Y, Diao S, Zhang X, Zuo T, Miao Z, Yang Y. Interleukin-18 mediated inflammatory brain injury after intracerebral hemorrhage in male mice. J Neurosci Res 2022; 100:1359-1369. [PMID: 35316547 DOI: 10.1002/jnr.25044] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 12/11/2022]
Abstract
Interleukin-18 (IL-18), a pro-inflammatory cytokine, is thought to be associated with inflammation in many neurological diseases such as ischemic stroke and poststroke depression, but the role of IL-18 in inflammatory injury after intracerebral hemorrhage (ICH) remains unclear. In this study, we established the ICH model in male mice and found that IL-18 expression including protein and mRNA levels was significantly increased in brain tissues after ICH. Meanwhile, exogenous IL-18 exacerbated cerebral hematoma and neurological deficits following ICH. In the IL-18 knockout group, the size of hematoma and neurological functions after ICH was decreased compared with the wild-type group, suggesting the critical role of IL-18 on the modulation of brain injury after ICH. Importantly, exogenous IL-18 increased microglial activation in brain tissues after ICH. Furthermore, IL-18 knockout resulted in the reduction of activated microglia after ICH. These results indicated that IL-18 may regulate the inflammatory response after ICH through the activation of microglia. Thus, IL-18 is expected to be a promising therapeutic target for secondary brain injury after ICH.
Collapse
Affiliation(s)
- Hao Li
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingluan Tian
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yin Yin
- Laboratory Animal Center, Soochow University, Suzhou, China
| | - Shanshan Diao
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ximeng Zhang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tao Zuo
- Department of Orthopedics, Xuzhou Medical University Affiliated Hospital, Xuzhou City, China
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Yi Yang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Diet-Induced High Serum Levels of Trimethylamine-N-oxide Enhance the Cellular Inflammatory Response without Exacerbating Acute Intracerebral Hemorrhage Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1599747. [PMID: 35242275 PMCID: PMC8886754 DOI: 10.1155/2022/1599747] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/27/2022] [Indexed: 12/14/2022]
Abstract
Trimethylamine-N-oxide (TMAO), an intestinal flora metabolite of choline, may aggravate atherosclerosis by inducing a chronic inflammatory response and thereby promoting the occurrence of cerebrovascular diseases. Knowledge about the influence of TMAO-related inflammatory response on the pathological process of acute stroke is limited. This study was designed to explore the effects of TMAO on neuroinflammation, brain injury severity, and long-term neurologic function in mice with acute intracerebral hemorrhage (ICH). We fed mice with either a regular chow diet or a chow diet supplemented with 1.2% choline pre- and post-ICH. In this study, we measured serum levels of TMAO with ultrahigh-performance liquid chromatography-tandem mass spectrometry at 24 h and 72 h post-ICH. The expression level of P38-mitogen-protein kinase (P38-MAPK), myeloid differentiation factor 88 (MyD88), high-mobility group box1 protein (HMGB1), and interleukin-1β (IL-1β) around hematoma was examined by western blotting at 24 h. Microglial and astrocyte activation and neutrophil infiltration were examined at 72 h. The lesion was examined on days 3 and 28. Neurologic deficits were examined for 28 days. A long-term choline diet significantly increased serum levels of TMAO compared with a regular diet at 24 h and 72 h after sham operation or ICH. Choline diet-induced high serum levels of TMAO did not enhance the expression of P38-MAPK, MyD88, HMGB1, or IL-1β at 24 h. However, it did increase the number of activated microglia and astrocytes around the hematoma at 72 h. Contrary to our expectations, it did not aggravate acute or long-term histologic damage or neurologic deficits after ICH. In summary, choline diet-induced high serum levels of TMAO increased the cellular inflammatory response probably by activating microglia and astrocytes. However, it did not aggravate brain injury or worsen long-term neurologic deficits. Although TMAO might be a potential risk factor for cerebrovascular diseases, this exploratory study did not support that TMAO is a promising target for ICH therapy.
Collapse
|
15
|
Wei Y, Song X, Gao Y, Gao Y, Li Y, Gu L. Iron toxicity in intracerebral hemorrhage: Physiopathological and therapeutic implications. Brain Res Bull 2021; 178:144-154. [PMID: 34838852 DOI: 10.1016/j.brainresbull.2021.11.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 01/09/2023]
Abstract
Intracerebral hemorrhage (ICH)-induced brain injury is a continuous pathological process that involves the deterioration of neurological functions, such as sensory, cognitive or motor functions. Cytotoxic byproducts of red blood cell lysis, especially free iron, appear to be a significant pathophysiologic mechanism leading to ICH-induced injury. Free iron has a crucial role in secondary brain injury after ICH. Chelating iron may attenuate iron-induced neurotoxicity and may be developed as a therapeutic candidate for ICH treatment. In this review, we focused on the potential role of iron toxicity in ICH-induced injury and iron chelation therapy in the management of ICH. It will hopefully advance our understanding of the pathogenesis of ICH and lead to new approaches for treatment.
Collapse
Affiliation(s)
- Yufei Wei
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, China
| | - Xiaoxiao Song
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, China
| | - Ying Gao
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing 100010, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100010, China
| | - Yuanyuan Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100010, China
| | - Lian Gu
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, China.
| |
Collapse
|
16
|
The neuroprotective effect of phillyrin in intracerebral hemorrhagic mice is produced by activation of the Nrf2 signaling pathway. Eur J Pharmacol 2021; 909:174439. [PMID: 34425100 DOI: 10.1016/j.ejphar.2021.174439] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022]
Abstract
Phillyrin, a natural plant extract, has significant antioxidant and anti-apoptotic effects. However, its effect on intracerebral hemorrhage (ICH) remains unclear. In this study, we investigated a potential role for phillyrin in the regulation of the oxidative stress and apoptosis induced by ICH. A model of ICH was induced by collagenase IV (0.2 U in 1 μl sterile normal saline) in male C57BL/6J (B6) mice and different doses of phillyrin (5, 15, or 30 mg/kg) were intraperitoneally (i.p.) injected at 30 min, 6 h, and 22 h after modeling. We found that phillyrin significantly reduced neural function and lesion volume, improved injury of white and grey matter around the lesion, decreased apoptosis and oxidative stress, increased the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase 1(HO-1), NADPH quinone oxidoreductase 1 (NQO1) and Superoxide Dismutase-1(SOD-1) in vitro and in vivo, and protected neurons from the stimulation of hemin by promoting Nrf2 nuclear translocation. Treatment with ML385 (Nrf2 inhibitor) completely reversed the protective effects of phillyrin in vivo after ICH injury. Based on our findings, we conclude that phillyrin treatment alleviates ICH injury-induced apoptosis and oxidative stress via activation of the Nrf2 signaling pathway, highlighting a potential role for phillyrin as an ICH therapeutic.
Collapse
|
17
|
Li DD, Wang Y, Kim EL, Hong J, Jung JH. Neuroprotective Effect of Cyclo-(L-Pro-L-Phe) Isolated from the Jellyfish-Derived Fungus Aspergillus flavus. Mar Drugs 2021; 19:md19080417. [PMID: 34436256 PMCID: PMC8401322 DOI: 10.3390/md19080417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR) expression has been implicated in pathological states such as cancer, inflammation, diabetes, and neurodegeneration. We isolated natural PPAR agonists—eight 2,5-diketopiperazines—from the jellyfish-derived fungus Aspergillus flavus. Cyclo-(L-Pro-L-Phe) was the most potent PPAR-γ activator among the eight 2,5-DKPs identified. Cyclo-(L-Pro-L-Phe) activated PPAR-γ in Ac2F rat liver cells and SH-SY5Y human neuroblastoma cells. The neuroprotective effect of this partial PPAR-γ agonist was examined using the 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, lactate dehydrogenase release, and the Hoechst 33342 staining assay in SH-SY5Y cells. Our findings revealed that cyclo-(L-Pro-L-Phe) reduced hydrogen peroxide-induced apoptosis as well as the generation of reactive oxygen species. Rhodamine 123 staining and western blotting revealed that cyclo-(L-Pro-L-Phe) prevented the loss of mitochondrial membrane potential and inhibited the activation of mitochondria-related apoptotic proteins, such as caspase 3 and poly (ADP-ribose) polymerase. Moreover, cyclo-(L-Pro-L-Phe) inhibited the activation and translocation of nuclear factor-kappa B. Thus, the partial PPAR-γ agonist cyclo-(L-Pro-L-Phe) demonstrated potential neuroprotective activity against oxidative stress-induced neurodegeneration in SH-SY5Y cells.
Collapse
Affiliation(s)
- Dan-dan Li
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (D.-d.L.); (Y.W.); (E.L.K.)
| | - Ying Wang
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (D.-d.L.); (Y.W.); (E.L.K.)
| | - Eun La Kim
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (D.-d.L.); (Y.W.); (E.L.K.)
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, Seoul 02447, Korea;
| | - Jee H. Jung
- College of Pharmacy, Pusan National University, Busan 46241, Korea; (D.-d.L.); (Y.W.); (E.L.K.)
- Correspondence:
| |
Collapse
|
18
|
Wang X, Liu H, Liao X, Qiao L, Zhu L, Wu S, Zhou Y, Zhang Y, Li B, Lin L, Ma J, Gu Q, Shu J. Dissecting the Roles of LncRNAs in the Development of Periventricular White Matter Damage. Front Genet 2021; 12:641526. [PMID: 33995480 PMCID: PMC8120246 DOI: 10.3389/fgene.2021.641526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/29/2021] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNA (LncRNA) has high expression in the brain. Animal studies have shown that lncRNA plays an important role in brain functions and mediates the development of many neurological diseases. However, data on the expression of lncRNAs and the clinical significance in prematurely born infants with diseases such as periventricular white matter damage (PWMD) remains scant. Here, we compared the expression of the lncRNAs in whole blood samples obtained from prematurely born infants with PWMD with samples from prematurely born infants without PWMD. Our data demonstrated differential expression of the lncRNAs between the two groups. Further, we showed that the lncRNAs play important roles in the development of PWMD. Our findings give insights into the functions of the lncRNAs in PWMD and provide evidence for the improvement of diagnostic and treatment strategies in infants with PWMD.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Heng Liu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xiaoli Liao
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Lixing Qiao
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Lihua Zhu
- Institute of Clinical, Jiangsu Health Vocational College, Nanjing, China
| | - Shun Wu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yan Zhou
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yi Zhang
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Bangbang Li
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Lili Lin
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jingjing Ma
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Qianying Gu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jiaping Shu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
19
|
Doroshenko ER, Drohomyrecky PC, Gower A, Whetstone H, Cahill LS, Ganguly M, Spring S, Yi TJ, Sled JG, Dunn SE. Peroxisome Proliferator-Activated Receptor-δ Deficiency in Microglia Results in Exacerbated Axonal Injury and Tissue Loss in Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 12:570425. [PMID: 33732230 PMCID: PMC7959796 DOI: 10.3389/fimmu.2021.570425] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 01/28/2021] [Indexed: 12/23/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR)-δ is a nuclear receptor that functions to maintain metabolic homeostasis, regulate cell growth, and limit the development of excessive inflammation during immune responses. Previously, we reported that PPAR-δ-deficient mice develop a more severe clinical course of experimental autoimmune encephalomyelitis (EAE); however, it was difficult to delineate the role that microglia played in this disease phenotype since PPAR-δ-deficient mice exhibited a number of immune defects that enhanced CNS inflammation upstream of microglia activation. Here, we specifically investigated the role of PPAR-δ in microglia during EAE by using mice where excision of a floxed Ppard allele was driven by expression of a tamoxifen (TAM)-inducible CX3C chemokine receptor 1 promoter-Cre recombinase transgene (Cx3cr1CreERT2: Ppardfl/fl). We observed that by 30 days of TAM treatment, Cx3cr1CreERT2: Ppardfl/fl mice exhibited Cre-mediated deletion primarily in microglia and this was accompanied by efficient knockdown of Ppard expression in these cells. Upon induction of EAE, TAM-treated Cx3cr1CreERT2: Ppardfl/fl mice presented with an exacerbated course of disease compared to TAM-treated Ppardfl/fl controls. Histopathological and magnetic resonance (MR) studies on the spinal cord and brains of EAE mice revealed increased Iba-1 immunoreactivity, axonal injury and CNS tissue loss in the TAM-treated Cx3cr1CreERT2: Ppardfl/fl group compared to controls. In early EAE, a time when clinical scores and the infiltration of CD45+ leukocytes was equivalent between Cx3cr1CreERT2: Ppardfl/fl and Ppardfl/fl mice, Ppard-deficient microglia exhibited a more reactive phenotype as evidenced by a shorter maximum process length and lower expression of genes associated with a homeostatic microglia gene signature. In addition, Ppard-deficient microglia exhibited increased expression of genes associated with reactive oxygen species generation, phagocytosis and lipid clearance, M2-activation, and promotion of inflammation. Our results therefore suggest that PPAR-δ has an important role in microglia in limiting bystander tissue damage during neuroinflammation.
Collapse
Affiliation(s)
| | | | - Annette Gower
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
| | - Heather Whetstone
- Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Milan Ganguly
- Histology Core, The Centre for Phenogenomics, Toronto, ON, Canada
| | - Shoshana Spring
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Tae Joon Yi
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada
| | - John G Sled
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Shannon E Dunn
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada.,Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
| |
Collapse
|
20
|
Chen W, Guo C, Feng H, Chen Y. Mitochondria: Novel Mechanisms and Therapeutic Targets for Secondary Brain Injury After Intracerebral Hemorrhage. Front Aging Neurosci 2021; 12:615451. [PMID: 33584246 PMCID: PMC7873050 DOI: 10.3389/fnagi.2020.615451] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a destructive form of stroke that often results in death or disability. However, the survivors usually experience sequelae of neurological impairments and psychiatric disorders, which affect their daily functionality and working capacity. The recent MISTIE III and STICH II trials have confirmed that early surgical clearance of hematomas does not improve the prognosis of survivors of ICH, so it is vital to find the intervention target of secondary brain injury (SBI) after ICH. Mitochondrial dysfunction, which may be induced by oxidative stress, neuroinflammation, and autophagy, among others, is considered to be a novel pathological mechanism of ICH. Moreover, mitochondria play an important role in promoting neuronal survival and improving neurological function after a hemorrhagic stroke. This review summarizes the mitochondrial mechanism involved in cell death, reactive oxygen species (ROS) production, inflammatory activation, blood–brain barrier (BBB) disruption, and brain edema underlying ICH. We emphasize the potential of mitochondrial protection as a potential therapeutic target for SBI after stroke and provide valuable insight into clinical strategies.
Collapse
Affiliation(s)
- Weixiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Medical University), Chongqing, China.,Collaborative Innovation Center for Brain Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chao Guo
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Medical University), Chongqing, China.,Collaborative Innovation Center for Brain Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Medical University), Chongqing, China.,Collaborative Innovation Center for Brain Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Medical University), Chongqing, China.,Collaborative Innovation Center for Brain Science, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
21
|
Peroxisome proliferator-activated receptors in the pathogenesis and therapies of liver fibrosis. Pharmacol Ther 2020; 222:107791. [PMID: 33321113 DOI: 10.1016/j.pharmthera.2020.107791] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is a dynamic wound-healing process associated with the deposition of extracellular matrix produced by myofibroblasts. HSCs activation, inflammation, oxidative stress, steatosis and aging play critical roles in the progression of liver fibrosis, which is correlated with the regulation of the peroxisome proliferator-activated receptor (PPAR) pathway. As nuclear receptors, PPARs reduce inflammatory response, regulate lipid metabolism, and inhibit fibrogenesis in the liver associated with aging. Thus, PPAR ligands have been investigated as possible therapeutic agents. Mounting evidence indicated that some PPAR agonists could reverse steatohepatitis and liver fibrosis. Consequently, targeting PPARs might be a promising and novel therapeutic option against liver fibrosis. This review summarizes recent studies on the role of PPARs on the pathogenesis and treatment of liver fibrosis.
Collapse
|
22
|
Strosznajder AK, Wójtowicz S, Jeżyna MJ, Sun GY, Strosznajder JB. Recent Insights on the Role of PPAR-β/δ in Neuroinflammation and Neurodegeneration, and Its Potential Target for Therapy. Neuromolecular Med 2020; 23:86-98. [PMID: 33210212 PMCID: PMC7929960 DOI: 10.1007/s12017-020-08629-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) β/δ belongs to the family of hormone and lipid-activated nuclear receptors, which are involved in metabolism of long-chain fatty acids, cholesterol, and sphingolipids. Similar to PPAR-α and PPAR-γ, PPAR-β/δ also acts as a transcription factor activated by dietary lipids and endogenous ligands, such as long-chain saturated and polyunsaturated fatty acids, and selected lipid metabolic products, such as eicosanoids, leukotrienes, lipoxins, and hydroxyeicosatetraenoic acids. Together with other PPARs, PPAR-β/δ displays transcriptional activity through interaction with retinoid X receptor (RXR). In general, PPARs have been shown to regulate cell differentiation, proliferation, and development and significantly modulate glucose, lipid metabolism, mitochondrial function, and biogenesis. PPAR-β/δ appears to play a special role in inflammatory processes and due to its proangiogenic and anti-/pro-carcinogenic properties, this receptor has been considered as a therapeutic target for treating metabolic syndrome, dyslipidemia, carcinogenesis, and diabetes. Until now, most studies were carried out in the peripheral organs, and despite of its presence in brain cells and in different brain regions, its role in neurodegeneration and neuroinflammation remains poorly understood. This review is intended to describe recent insights on the impact of PPAR-β/δ and its novel agonists on neuroinflammation and neurodegenerative disorders, including Alzheimer’s and Parkinson’s, Huntington’s diseases, multiple sclerosis, stroke, and traumatic injury. An important goal is to obtain new insights to better understand the dietary and pharmacological regulations of PPAR-β/δ and to find promising therapeutic strategies that could mitigate these neurological disorders.
Collapse
Affiliation(s)
- Anna K Strosznajder
- Faculty of Medicine, Medical University of Bialystok, 1 Kilinskiego st., 15-089, Białystok, Poland
| | - Sylwia Wójtowicz
- Department of Cellular Signaling, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawińskiego st., 02-106, Warsaw, Poland
| | - Mieszko J Jeżyna
- Faculty of Medicine, Medical University of Bialystok, 1 Kilinskiego st., 15-089, Białystok, Poland
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, 65211, USA
| | - Joanna B Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawińskiego st., 02-106, Warsaw, Poland.
| |
Collapse
|