1
|
Shang J, Huang G, Wang B, Wang J, Wei W, Cui Y, Liu X. Shuxuetong injection inhibits pyroptosis in acute ischemic stroke via CD44/NLRP3/GSDMD signal. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119618. [PMID: 40074097 DOI: 10.1016/j.jep.2025.119618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute ischemic stroke (AIS) is an important cause of death and disability in the world. Based on the blood stasis syndrome of stroke, Shuxuetong Injection (SXT) is a representative prescription for the treatment of AIS, which extracted by modern technology from Whitmania pigra Whitman (Shuizhi) and Pheretima aspergillum E.Perrier (Dilong). AIM OF THE STUDY This study is in order to examine whether SXT regulates pyroptosis in AIS via Cluster of Differentiation 44 (CD44)/NOD-like receptor thermal protein domain associated protein 3 (NLRP3)/gasdermin D (GSDMD) signal. MATERIALS AND METHODS The rats were randomly divided into sham group, model (transient middle cerebral artery occlusion, 24 h) group, SXT low-dose group (0.27 mL/kg), SXT medium-dose group (0.54 mL/kg), SXT high-dose group (1.08 mL/kg) and positive drug group (edaravone injection, 1.35 mL/kg). Transient middle cerebral artery occlusion (tMCAO, 24 h) model of rats was set up. Neurological deficit score, tetrazolium red staining, hematoxylin-eosin staining, and Nissl staining were used to observe and screen out the optimal dosage for improving AIS. Mechanism research indicators included transmission electron microscopy and Western blot. Adeno-associated virus (AAV)-CD44 and small interfering RNA (siRNA) of CD44 were used for knocking down the CD44 expression level to verify whether SXT could resist pyroptosis through CD44. The oxygen and glucose deprivation/re-oxygenation (OGD/R, 24 h) model of PC12 cells was used for in vitro pharmacological validation. Molecular docking, cellular thermal shift assay and drug affinity responsive target stability were employed to assess the binding affinity of critical components for the CD44 protein. RESULTS SXT conspicuously mitigated the neurological function scores and cerebral infarct volume in tMCAO rats, thereby safeguarding nerve cells. In vitro, SXT not only enhanced the viability of PC12 cells subjected to OGD/R but also mitigated cellular swelling and inflammatory infiltration. The optimal dose was 1.08 mL/kg (rats) or 72.56 mg/mL (PC12 cells). SXT reduced pyroptosis and inflammation in tMCAO rats and OGD/R cells by decreasing the expression levels of GSDMD-N, NLRP3 and CD44. In addition, after knocking down the expression level of CD44 by using AAV-CD44 and siRNA-CD44, it was found that the pyroptosis of AIS intervened by SXT was closely related to the CD44/NLRP3/GSDMD signal. The pivotal constituent of SXT, xanthine, exhibited pronounced binding affinity towards the CD44 protein, thereby demonstrating the capacity to stabilize this molecular target. CONCLUSION This study demonstrates that Shuxuetong Injection inhibits pyroptosis in acute ischemic stroke via CD44/NLRP3/GSDMD signal.
Collapse
Affiliation(s)
- Jinfeng Shang
- Beijing University of Chinese Medicine, Beijing, 102488, China.
| | | | - Bohong Wang
- Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jingyi Wang
- Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Wanting Wei
- Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Yiran Cui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Xin Liu
- Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
2
|
Ju Y, Wu R, Wang G, Shen T, Hu J, Kong Y. Vitronectin stimulates hepatic gluconeogenesis by activating the cAMP/PKA/CREB axis in the liver. Mol Cell Endocrinol 2025; 599:112485. [PMID: 39909307 DOI: 10.1016/j.mce.2025.112485] [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: 11/01/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Vitronectin, a protein derived the human placenta, has been identified as an inducer of insulin resistance in trophoblast cells in gestational diabetes mellitus (GDM). As a secreted protein, vitronectin may have systemic effects on dysregulated glucose metabolism in GDM. To address this speculation, we generated a GDM mouse model using high-fat diet-induced obese mice. Consistent with findings in placentas of GDM patients, GDM mouse placentas showed higher vitronectin expression, accompanied by increased serum vitronectin levels. Reduced insulin signaling transduction was observed in both the placentas and livers of GDM mice, along with enhanced hepatic gluconeogenesis. To further explore the role of vitronectin in hepatic gluconeogenesis, we constructed an adeno-associated virus expressing Vtn (AAV-VTN), which was administered to mice via tail vein injection. In AAV-VTN-treated mice, glucose production from exogenous pyruvate increased, and the expression of gluconeogenic genes in the liver was upregulated, indicating that hepatic gluconeogenesis was stimulated by vitronectin. Mechanistically, vitronectin binds to its receptor CD51/61, activating the cAMP/PKA/CREB axis in hepatocytes, thereby promoting hepatic gluconeogenesis. In summary, our findings suggest that placenta-derived vitronectin plays a critical role in inducing insulin resistance in the liver in GDM. Moreover, vitronectin stimulates hepatic gluconeogenesis through activation of the cAMP/PKA/CREB axis. These results point to vitronectin as a potential therapeutic target for managing hyperglycemia in GDM.
Collapse
Affiliation(s)
- Yuejun Ju
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu 215500, Jiangsu, PR China; Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, PR China
| | - Runze Wu
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu 215500, Jiangsu, PR China
| | - Guanyi Wang
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu 215500, Jiangsu, PR China
| | - Ting Shen
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu 215500, Jiangsu, PR China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, PR China.
| | - Yinghong Kong
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu 215500, Jiangsu, PR China.
| |
Collapse
|
3
|
Yousefpour Shahrivar R, Karami F, Karami E. Differential gene expression patterns in Niemann-Pick Type C and Tay-Sachs diseases: Implications for neurodegenerative mechanisms. PLoS One 2025; 20:e0319401. [PMID: 40106490 PMCID: PMC11922228 DOI: 10.1371/journal.pone.0319401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/01/2025] [Indexed: 03/22/2025] Open
Abstract
Lysosomal storage disorders (LSDs) are a group of rare genetic conditions characterized by the impaired function of enzymes responsible for lipid digestion. Among these LSDs, Tay-Sachs disease (TSD) and Niemann-Pick type C (NPC) may share a common gene expression profile. In this study, we conducted a bioinformatics analysis to explore the gene expression profile overlap between TSD and NPC. Analyses were performed on RNA-seq datasets for both TSD and NPC from the Gene Expression Omnibus (GEO) database. Datasets were subjected to differential gene expression analysis utilizing the DESeq2 package in the R programming language. A total of 147 differentially expressed genes (DEG) were found to be shared between the TSD and NPC datasets. Enrichment analysis was then performed on the DEGs. We found that the common DEGs are predominantly associated with processes such as cell adhesion mediated by integrin, cell-substrate adhesion, and urogenital system development. Furthermore, construction of protein-protein interaction (PPI) networks using the Cytoscape software led to the identification of four hub genes: APOE, CD44, SNCA, and ITGB5. Those hub genes not only can unravel the pathogenesis of related neurologic diseases with common impaired pathways, but also may pave the way towards targeted gene therapy of LSDs.In addition, they serve as the potential biomarkers for related neurodegenerative diseases warranting further investigations.
Collapse
Affiliation(s)
- Ramin Yousefpour Shahrivar
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Fatemeh Karami
- Department of Medical Genetics, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ebrahim Karami
- Department of Electrical and Computer Engineering, Faculty of Engineering and Applied Sciences, Memorial University of Newfoundland, St. John's, Canada
| |
Collapse
|
4
|
Chen Y, Zhu Y, Tan Z, Zhang X, Hu J, Zhu R, Xie M, Wang J, Chen L, Guo Z. Jiajian Shuyu pills effectively ameliorate cognitive impairment via regulating the inflammation of microglia in an Alzheimer's disease mouse model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119508. [PMID: 39971019 DOI: 10.1016/j.jep.2025.119508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by progressive cognitive decline and behavioral impairments in the elderly. Microglia, the resident immune cells of the central nervous system, play a crucial role in modulating the pathological processes associated with AD. Jiajian Shuyu Pills (JJSYP) are frequently employed in the treatment of AD, purportedly by enhancing the physiological functions of human tissues and organs to modulate the immune response. Nevertheless, the underlying mechanisms by which JJSYP exert their therapeutic effects in the context of AD remain inadequately elucidated. AIM OF THE STUDY This study aimed to assess the effects of JJSYP on cognitive enhancement and the alleviation of neuroinflammation in the treatment of AD, as well as to explore the underlying mechanisms using mouse models. MATERIALS AND METHODS The components of JJSYP in serum were analyzed using HPLC-Q/TOF-MS. APP/PS1 transgenic mice served as AD models in this investigation. Cognitive function in the AD mice was assessed through the Mirror Water Maze Test and the Novel Object Recognition Test. The quantification of apoptotic hippocampal cells was conducted using Nissl staining and TUNEL staining. Immunofluorescence (IF) and Western blot (WB) analyses were employed to examine microglial activation and the expression of relevant proteins. Transcriptomic sequencing analysis and network pharmacology were administrated to explore the potential mechanisms of JJSYP in AD treatment. Inflammatory cytokine levels in the brain were measured using RT-PCR. RESULTS A total of 74 absorbed prototype components from JJSYP were identified. JJSYP effectively improved cognitive function and neuroapoptosis in AD model mice by modulating the activation of microglia. The JJSYP intervention alleviated neuroinflammation by suppressing microglial activation and reducing the accumulation of amyloid β-protein. Through transcriptome sequencing and WB verification, 34 differentially expressed genes (DEGs) were identified, including ACKR3, NR1H3 and Adra1a. Following treatment with a high dose of JJSYP, both ACKR3 and NR1H3 showed a significant decrease compared to the model group. Conversely, ADRA1A expression was reduced in model group compared to the control group, but increased following high dose JJSYP treatment. Research involving RNA sequencing and network pharmacology indicated that JJSYP altered the activation of CXCL12/ACKR3 signaling pathways in the hippocampus. CONCLUSIONS JJSYP exhibits potential anti-Alzheimer's Disease effects and warrants further investigation and development as a prosper treatment for AD.
Collapse
Affiliation(s)
- Yan Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China.
| | - Yan Zhu
- Wuhan No.1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, Hubei, 430015, China
| | - Zihu Tan
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Xueyi Zhang
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China
| | - Jiafeng Hu
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Ruichi Zhu
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jing Wang
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China
| | - Lizhu Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China
| | - Zhenli Guo
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, No. 11 Lingjiaohu Road, Wuhan, Hubei, 430015, China.
| |
Collapse
|
5
|
Bonet IJM, Araldi D, Khomula EV, Bogen O, Green PG, Levine JD. G-protein-coupled estrogen receptor 30 regulation of signaling downstream of protein kinase Cε mediates sex dimorphism in hyaluronan-induced antihyperalgesia. Pain 2025; 166:539-556. [PMID: 39787533 PMCID: PMC11810595 DOI: 10.1097/j.pain.0000000000003419] [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: 04/24/2024] [Accepted: 07/12/2024] [Indexed: 01/12/2025]
Abstract
ABSTRACT High molecular weight hyaluronan (HMWH) inhibits hyperalgesia induced by diverse pronociceptive inflammatory mediators and their second messengers, in rats of both sexes. However, the hyperalgesia induced by ligands at 3 pattern recognition receptors, lipopolysaccharide (a toll-like receptor 4 agonist), lipoteichoic acid (a toll-like receptor 2/6 agonist), and nigericin (a NOD-like receptor family, pyrin domain containing 3 activator), and oxaliplatin and paclitaxel chemotherapy-induced peripheral neuropathy are only attenuated in males. After gonadectomy or intrathecal administration of an antisense to G-protein-coupled estrogen receptor 30 (GPER) mRNA, HMWH produces antihyperalgesia in females. In nociceptors cultured from rats that had been treated with oxaliplatin, HMWH reverses nociceptor sensitization from male and GPER antisense-treated female, but not from gonad intact females. G-protein-coupled estrogen receptor-dependent sex dimorphism for HMWH-induced antihyperalgesia was also observed for the prolongation of prostaglandin E 2 (PGE 2 )-induced hyperalgesia in primed nociceptors. While in primed rats, HMWH inhibits early, protein kinase A-dependent hyperalgesia, 30 minutes post PGE 2 injection, in both sexes; measured 4 hours post-PGE 2 , HMWH inhibits the protein kinase Cε (PKCε)-dependent prolongation of PGE 2 hyperalgesia only in males and GPER antisense-treated females. In females, hyperalgesia induced by PKCε agonist, ψεRACK, in control but not in primed nociceptors, was inhibited by HMWH. Inhibitors of 2 GPER second messengers, extracellular-regulated kinase 1/2 and nonreceptor tyrosine kinase, also unmasked HMWH antihyperalgesia in females with oxaliplatin chemotherapy-induced peripheral neuropathy, a condition in which nociceptors are primed as well as sensitized. Our results support GPER-dependent sex dimorphism in HMWH-induced antihyperalgesia for pain induced by pattern recognition receptor agonists, and chronic inflammatory and neuropathic pain, mediated by changes in signaling downstream of PKCε in primed nociceptors.
Collapse
Affiliation(s)
- Ivan J. M. Bonet
- Department of Oral & Maxillofacial Surgery, and Division of Neuroscience, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Dionéia Araldi
- Department of Oral & Maxillofacial Surgery, and Division of Neuroscience, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Eugen V. Khomula
- Department of Oral & Maxillofacial Surgery, and Division of Neuroscience, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Oliver Bogen
- Department of Oral & Maxillofacial Surgery, and Division of Neuroscience, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Paul G. Green
- Department of Oral & Maxillofacial Surgery, and Division of Neuroscience, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
- Departments of Preventative & Restorative Dental Sciences and Oral & Maxillofacial Surgery, and Division of Neuroscience, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Jon D. Levine
- Department of Oral & Maxillofacial Surgery, and Division of Neuroscience, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
- Departments of Medicine and Oral & Maxillofacial Surgery, and Division of Neuroscience, UCSF Pain and Addiction Research Center, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| |
Collapse
|
6
|
Lv J, Jiao Y, Zhao X, Kong X, Chen Y, Li L, Chen X, Tao X, Dong D. Examining the Impact of Microglia on Ischemic Stroke With an Emphasis on the Metabolism of Immune Cells. CNS Neurosci Ther 2025; 31:e70229. [PMID: 39945118 PMCID: PMC11822359 DOI: 10.1111/cns.70229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/13/2024] [Accepted: 01/11/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Ischemic stroke, a major cause of disability and the second leading cause of death, poses a significant public health challenge. Post-stroke inflammation can harm the blood-brain barrier and worsen neurological deficits, which are key factors in neuronal damage in patients with ischemic stroke. Microglia are crucial in the central nervous system, involved in inflammation, neuronal damage, and repair after cerebral ischemia. While cellular immune metabolism has been widely studied, its role in ischamic stroke remains unclear. AIM This review aims to examine how inflammation affects the phenotypic characteristics of immune cells after ischemic stroke and to explore the effects of the immune metabolic microenvironment on the phenotypic profiles and functions of microglia in ischemic stroke. METHOD The review refers to the available literature in PubMed, searching for critical terms related to Ischemic stroke, neuroinflammation, microglia, and immunometabolism. RESULT In this review, we found that during stroke progression, microglia can dynamically switch between pro-inflammatory and anti-inflammatory phenotypes. Microglial glycometabolism includes oxidative phosphorylation and glycolysis, and lipid metabolism involves lipid synthesis and breakdown. Modulating the production of inflammatory mediator precursors can induce an anti-inflammatory phenotype in microglia. CONCLUSION Thus, studying microglial metabolic pathways and their products may offer new insights for ischemic stroke treatment.
Collapse
Affiliation(s)
- Jing Lv
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- College of PharmacyDalian Medical UniversityDalianChina
| | - Yang Jiao
- Department of NeurologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Dalian Innovation Institute of Stem Cell and Precision MedicineDalianChina
| | - Xinya Zhao
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- College of PharmacyDalian Medical UniversityDalianChina
| | - Xin Kong
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Dalian Innovation Institute of Stem Cell and Precision MedicineDalianChina
| | - Yanwei Chen
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Lu Li
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xuyang Chen
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xufeng Tao
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Deshi Dong
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
7
|
Wu Z, Chen J, Kong F, Zhang Y, Yi J, Li Y, Hu M, Wang D. Polypeptide of Inonotus hispidus extracts alleviates periodontitis through suppressing inflammatory bone loss. Int J Biol Macromol 2025; 287:138350. [PMID: 39645101 DOI: 10.1016/j.ijbiomac.2024.138350] [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: 08/22/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
This study aimed to characterize and evaluate the effects of a novel polypeptide isolated from Inonotus hispidus (IH) against periodontitis. The polypeptides extracted and purified from the fruiting body of IH had a uniform molar mass, including 23 types of peptides. IH polypeptide (IHP) exerted antimicrobial activity against Porphyromonas gingivalis (P. gingivalis) by damaging the cell walls and membranes of microorganisms, disturbing energy metabolism, and regulating the expression of virulence factors. IHP significantly inhibited inflammation in lipopolysaccharides (LPS)-stimulated Raw264.7 cells evidenced by the regulation of inflammatory cytokine levels. In rats with ligature-induced periodontitis, IHP treatment ameliorated alveolar bone destruction and preserved the balance between oral flora and gut microbes. The interaction between oral and intestinal flora possibly affected the relevant metabolites. Proteomics combined with confirmation experiment revealed that the β-catenin/ nuclear factor-kappa B (NF-κB) signaling may be involved in IHP-mediated anti-periodontitis in rats, which helps reduce the secretion of pro-inflammatory factors and inhibit inflammatory osteoclastic response in the periodontal tissue. Additionally, IHP improved clinical parameters, including the plaque index (PLI), pocket depth (PD), bleeding on probing (BOP), and average probing depth in individuals with periodontitis. These findings augment the understanding of the potential role of IHP in treating periodontitis.
Collapse
Affiliation(s)
- Zhina Wu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| | - Jianai Chen
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Fange Kong
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China 2 National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Yaqin Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | | | - Yutong Li
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China; School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China.
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun 130012, China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
8
|
Knab F, Guaitoli G, Jarboui MA, von Zweydorf F, Isik FB, Klose F, Rajkumar AP, Gasser T, Gloeckner CJ. The cellular and extracellular proteomic signature of human dopaminergic neurons carrying the LRRK2 G2019S mutation. Front Neurosci 2024; 18:1502246. [PMID: 39726830 PMCID: PMC11669673 DOI: 10.3389/fnins.2024.1502246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Background Extracellular vesicles are easily accessible in various biofluids and allow the assessment of disease-related changes in the proteome. This has made them a promising target for biomarker studies, especially in the field of neurodegeneration where access to diseased tissue is very limited. Genetic variants in the LRRK2 gene have been linked to both familial and sporadic forms of Parkinson's disease. With LRRK2 inhibitors entering clinical trials, there is an unmet need for biomarkers that reflect LRRK2-specific pathology and target engagement. Methods In this study, we used induced pluripotent stem cells derived from a patient with Parkinson's disease carrying the LRRK2 G2019S mutation and an isogenic gene-corrected control to generate human dopaminergic neurons. We isolated extracellular vesicles and neuronal cell lysates and characterized their proteomic signature using data-independent acquisition proteomics. Then, we performed differential expression analysis to identify dysregulated proteins in the mutated line. We used Metascape and gene ontology enrichment analysis on the dysregulated proteomes to identify changes in associated functional networks. Results We identified 595 significantly differentially regulated proteins in extracellular vesicles and 3,205 in cell lysates. We visualized functionally relevant protein-protein interaction networks and identified key regulators within the dysregulated proteomes. Using gene ontology, we found a close association with biological processes relevant to neurodegeneration and Parkinson's disease. Finally, we focused on proteins that were dysregulated in both the extracellular and cellular proteomes. We provide a list of ten biomarker candidates that are functionally relevant to neurodegeneration and linked to LRRK2-associated pathology, for example, the sonic hedgehog signaling molecule, a protein that has tightly been linked to LRRK2-related disruption of cilia function. Conclusion In conclusion, we characterized the cellular and extracellular proteome of dopaminergic neurons carrying the LRRK2 G2019S mutation and proposed an experimentally based list of biomarker candidates for future studies.
Collapse
Affiliation(s)
- Felix Knab
- Hertie Institute for Clinical Brain Research, Department of Neurodegeneration, University of Tübingen, Tübingen, Germany
| | | | - Mohamed Ali Jarboui
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | | | - Fatma Busra Isik
- Institute of Mental Health, Mental Health and Clinical Neurosciences Academic Unit, University of Nottingham, Nottingham, United Kingdom
| | - Franziska Klose
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Anto Praveen Rajkumar
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Thomas Gasser
- Hertie Institute for Clinical Brain Research, Department of Neurodegeneration, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | |
Collapse
|
9
|
Cai X, Xie Z, Zhao J, Lu W, Zhu Z, Chen M, Huang Z, Ying Y, Fu Y, Xu J, Zhu S. FGF20 promotes spinal cord injury repair by inhibiting the formation of necrotic corpuscle P-MLKL/P-RIP1/P-RIP3 in neurons. J Cell Mol Med 2024; 28:e70109. [PMID: 39676730 DOI: 10.1111/jcmm.70109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 12/17/2024] Open
Abstract
The disruption of the local microenvironment subsequent to spinal cord injury (SCI) leads to a substantial loss of neurons in the affected region, which is a major contributing factor to impaired motor function recovery in patients. Fibroblast growth factor 20 (FGF20) is a neurotrophic factor that plays a crucial role in neuronal development and homeostasis. In this study, the recombinant human FGF20 (rhFGF20) was found to mitigate the process of necroptosis in a mouse model of SCI, thereby reducing neural functional deficits and promoting SCI repair. FGF20 protein was injected into the SCI mice via intraperitoneal injection. Using the BMS scale and inclined plane test, we found that FGF20 significantly promoted the recovery of motor function. The Nissl staining revealed the level of neuronal survival within the region of injury. The expression changes of NeuN, GAP43, NF200 and GFAP indicated that FGF20 has the nerve repair ability to delay the formation of glial scar. Through fluorescence detection of Ace-Tubulin and Tyr-Tubulin, FGF20 was revealed to promote the polymerization of axon-regenerated microtubules. Furthermore, FGF20 was also found to reduce the expression levels of necroptosis induced by SCI. These data suggest that FGF20 may exert a neuroprotective effect by inhibiting injury-induced necroptosis, thereby facilitating functional recovery following SCI. Moreover, systemic administration of FGF20 holds promise as a potential therapeutic strategy for repairing the damaged spinal cord. The discovery paves the way for a novel avenue of growth factor research in the field of SCI.
Collapse
Affiliation(s)
- Xiong Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenwen Xie
- The First Clinical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Juan Zhao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenjie Lu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhongwei Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiyang Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yibo Ying
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yining Fu
- The First Clinical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
Zou C, Cai R, Li Y, Xue Y, Zhang G, Alitongbieke G, Pan Y, Zhang S. β-chitosan attenuates hepatic macrophage-driven inflammation and reverses aging-related cognitive impairment. iScience 2024; 27:110766. [PMID: 39280626 PMCID: PMC11401205 DOI: 10.1016/j.isci.2024.110766] [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: 03/20/2024] [Revised: 07/13/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Recently, increasing evidence has shown the association between liver abnormal inflammation and cognition impairment, yet their age-related pathogenesis remains obscure. Here, our study provides a potential mechanistic link between liver macrophage excessive activation and neuroinflammation in aging progression. In aged and LPS-injected C57BL/6J mice, systemic administration of β-chitosan ameliorates hepatic macrophage-driven inflammation and reduces peripheral accumulations of TNF-α and IL-1β. Downregulation of circulatory pro-inflammatory cytokines then decreases vascular VCAM1 expression and neuroinflammation in the hippocampus, leading to cognitive improvement in aged/LPS-stimulated mice. Interestingly, β-chitosan treatment also exhibits the beneficial effects on the behavioral recovery of aged/LPS-stimulated zebrafish and Caenorhabditis elegans. In our cell culture and molecular docking experiments, we found that β-chitosan prefers shielding the MD-2 pocket, thus blocking the activation of TLR4-MD-2 complex to suppress NF-κB signaling pathway activation. Together, our findings highlight the extensive therapeutic potential of β-chitosan in reversing aged-related/LPS-induced cognitive impairment via the liver-brain axis.
Collapse
Affiliation(s)
- Chenming Zou
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Ruihua Cai
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Yunbing Li
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Yu Xue
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Guoguang Zhang
- School of Biological Science and Biotechnology, Minnan Normal University, Zhangzhou 363000, China
| | - Gulimiran Alitongbieke
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Yutian Pan
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Sanguo Zhang
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| |
Collapse
|
11
|
Zheng L, Zhou C, Mao C, Xie C, You J, Cheng W, Liu C, Huang P, Guan X, Guo T, Wu J, Luo Y, Xu X, Zhang B, Zhang M, Wang L, Feng J. Contrastive machine learning reveals Parkinson's disease specific features associated with disease severity and progression. Commun Biol 2024; 7:954. [PMID: 39112797 PMCID: PMC11306336 DOI: 10.1038/s42003-024-06648-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Parkinson's disease (PD) exhibits heterogeneity in terms of symptoms and prognosis, likely due to diverse neuroanatomical alterations. This study employs a contrastive deep learning approach to analyze Magnetic Resonance Imaging (MRI) data from 932 PD patients and 366 controls, aiming to disentangle PD-specific neuroanatomical alterations. The results reveal that these neuroanatomical alterations in PD are correlated with individual differences in dopamine transporter binding deficit, neurodegeneration biomarkers, and clinical severity and progression. The correlation with clinical severity is verified in an external cohort. Notably, certain proteins in the cerebrospinal fluid are strongly associated with PD-specific features, particularly those involved in the immune function. The most notable neuroanatomical alterations are observed in both subcortical and temporal regions. Our findings provide deeper insights into the patterns of brain atrophy in PD and potential underlying molecular mechanisms, paving the way for earlier patient stratification and the development of treatments to slow down neurodegeneration.
Collapse
Affiliation(s)
- Liping Zheng
- Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Cheng Zhou
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengjie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chao Xie
- Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Jia You
- Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China
| | - Chunfeng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoujun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Guo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Wu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yajun Luo
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Linbo Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
- School of Data Science, Fudan University, Shanghai, China.
- Department of Computer Science, University of Warwick, Coventry, UK.
| |
Collapse
|
12
|
Ayten M, Straub T, Kaplan L, Hauck SM, Grosche A, Koch SF. CD44 signaling in Müller cells impacts photoreceptor function and survival in healthy and diseased retinas. J Neuroinflammation 2024; 21:190. [PMID: 39095775 PMCID: PMC11297696 DOI: 10.1186/s12974-024-03175-8] [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: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
Retinitis pigmentosa (RP), an inherited retinal disease, affects 1,5 million people worldwide. The initial mutation-driven photoreceptor degeneration leads to chronic inflammation, characterized by Müller cell activation and upregulation of CD44. CD44 is a cell surface transmembrane glycoprotein and the primary receptor for hyaluronic acid. It is involved in many pathological processes, but little is known about CD44's retinal functions. CD44 expression is also increased in Müller cells from our Pde6bSTOP/STOP RP mouse model. To gain a more detailed understanding of CD44's role in healthy and diseased retinas, we analyzed Cd44-/- and Cd44-/-Pde6bSTOP/STOP mice, respectively. The loss of CD44 led to enhanced photoreceptor degeneration, reduced retinal function, and increased inflammatory response. To understand the underlying mechanism, we performed proteomic analysis on isolated Müller cells from Cd44-/- and Cd44-/-Pde6bSTOP/STOP retinas and identified a significant downregulation of glutamate transporter 1 (SLC1A2). This downregulation was accompanied by higher glutamate levels, suggesting impaired glutamate homeostasis. These novel findings indicate that CD44 stimulates glutamate uptake via SLC1A2 in Müller cells, which in turn, supports photoreceptor survival and function.
Collapse
Affiliation(s)
- Monika Ayten
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, 81377, Germany
| | - Tobias Straub
- Bioinformatics Unit, Biomedical Center Munich, Ludwig-Maximilians-Universität München, Planegg-Martinsried, 82152, Germany
| | - Lew Kaplan
- Department of Physiological Genomics, Biomedical Center Munich, Ludwig-Maximilians-Universität München, Planegg-Martinsried, 82152, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Biomedical Center Munich, Ludwig-Maximilians-Universität München, Planegg-Martinsried, 82152, Germany
| | - Susanne F Koch
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, 81377, Germany.
| |
Collapse
|
13
|
Lu Z, Liu Z, Wang C, Jiang R, Wang Z, Liao W, Wang W, Chen J, Zhu X, Zhao J, Liu Q, Yang Y, Gong P. CD300LF + microglia impede the neuroinflammation following traumatic brain injury by inhibiting STING pathway. CNS Neurosci Ther 2024; 30:e14824. [PMID: 38965803 PMCID: PMC11224125 DOI: 10.1111/cns.14824] [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: 04/25/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024] Open
Abstract
INTRODUCTION The diversity in microglial phenotypes and functions following traumatic brain injury (TBI) is poorly characterized. The aim of this study was to explore precise targets for improving the prognosis of TBI patients from a microglial perspective. OBJECTIVES To assess whether the prognosis of TBI can be improved by modulating microglia function. RESULTS In CD300LF-deficient mice, we observed an increase in glial cell proliferation, more extensive neuronal loss, and worsened neurological function post-TBI. Transcriptomic comparisons between CD300LF-positive and CD300LF-negative microglia illuminated that the neuroprotective role of CD300LF is principally mediated by the inhibition of the STING signaling pathway. In addition, this protective effect can be augmented using the STING pathway inhibitor C-176. CONCLUSIONS Our research indicates that CD300LF reduces neuroinflammation and promotes neurological recovery after TBI, and that microglia are integral to the protective effects of CD300LF in this context. In summary, our findings highlight CD300LF as a critical molecular regulator modulating the adverse actions of microglia following acute brain injury and propose a novel therapeutic approach to enhance outcomes for patients with TBI.
Collapse
Affiliation(s)
- Zhichao Lu
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongJiangsuChina
- Neuro‐Microscopy and Minimally Invasive Translational Medicine Innovation CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Jiangsu Medical Innovation Centre, Neurological Disease Diagnosis and Treatment CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Zongheng Liu
- Department of Neurosurgery, Zhejiang Provincial Hospital of Chinese MedicineThe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Chenxing Wang
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongJiangsuChina
- Neuro‐Microscopy and Minimally Invasive Translational Medicine Innovation CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Jiangsu Medical Innovation Centre, Neurological Disease Diagnosis and Treatment CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Rui Jiang
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongJiangsuChina
- Neuro‐Microscopy and Minimally Invasive Translational Medicine Innovation CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Jiangsu Medical Innovation Centre, Neurological Disease Diagnosis and Treatment CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Ziheng Wang
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Department of BiobankAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Weiquan Liao
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongJiangsuChina
- Neuro‐Microscopy and Minimally Invasive Translational Medicine Innovation CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Jiangsu Medical Innovation Centre, Neurological Disease Diagnosis and Treatment CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Wei Wang
- Department of PathologyAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Jianfeng Chen
- Department of Orthopedics and TraumatologyWuxi TCM Hospital Affiliated to Nanjing University of Chinese MedicineWuxiJiangsuChina
| | - Xingjia Zhu
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongJiangsuChina
- Neuro‐Microscopy and Minimally Invasive Translational Medicine Innovation CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Jiangsu Medical Innovation Centre, Neurological Disease Diagnosis and Treatment CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Jingwei Zhao
- Department of General Surgery, Shanghai Key Laboratory of Biliary Tract Disease Research, Research Institute of Biliary Tract DiseaseXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qianqian Liu
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongJiangsuChina
- Neuro‐Microscopy and Minimally Invasive Translational Medicine Innovation CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Jiangsu Medical Innovation Centre, Neurological Disease Diagnosis and Treatment CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Yang Yang
- Department of NeurosurgeryWuxi Taihu HosptialWuxiChina
| | - Peipei Gong
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongJiangsuChina
- Neuro‐Microscopy and Minimally Invasive Translational Medicine Innovation CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Jiangsu Medical Innovation Centre, Neurological Disease Diagnosis and Treatment CenterAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| |
Collapse
|
14
|
Singh MK, Shin Y, Ju S, Han S, Kim SS, Kang I. Comprehensive Overview of Alzheimer's Disease: Etiological Insights and Degradation Strategies. Int J Mol Sci 2024; 25:6901. [PMID: 39000011 PMCID: PMC11241648 DOI: 10.3390/ijms25136901] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
15
|
Park HY, Lee GS, Go J, Ryu YK, Lee CH, Moon JH, Kim KS. Angiotensin-converting enzyme inhibition prevents l-dopa-induced dyskinesia in a 6-ohda-induced mouse model of Parkinson's disease. Eur J Pharmacol 2024; 973:176573. [PMID: 38642669 DOI: 10.1016/j.ejphar.2024.176573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/11/2024] [Accepted: 04/09/2024] [Indexed: 04/22/2024]
Abstract
Parkinson's disease (PD) is characterised by severe movement defects and the degeneration of dopaminergic neurones in the midbrain. The symptoms of PD can be managed with dopamine replacement therapy using L-3, 4-dihydroxyphenylalanine (L-dopa), which is the gold standard therapy for PD. However, long-term treatment with L-dopa can lead to motor complications. The central renin-angiotensin system (RAS) is associated with the development of neurodegenerative diseases in the brain. However, the role of the RAS in dopamine replacement therapy for PD remains unclear. Here, we tested the co-treatment of the angiotensin-converting enzyme inhibitor (ACEI) with L-dopa altered L-dopa-induced dyskinesia (LID) in a 6-hydroxydopamine (6-OHDA)-lesioned mouse model of PD. Perindopril, captopril, and enalapril were used as ACEIs. The co-treatment of ACEI with L-dopa significantly decreased LID development in 6-OHDA-lesioned mice. In addition, the astrocyte and microglial transcripts involving Ccl2, C3, Cd44, and Iigp1 were reduced by co-treatment with ACEI and L-dopa in the 6-OHDA-lesioned striatum. In conclusion, co-treatment with ACEIs and L-dopa, such as perindopril, captopril, and enalapril, may mitigate the severity of L-DOPA-induced dyskinesia in a mouse model of PD.
Collapse
Affiliation(s)
- Hye-Yeon Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Ga Seul Lee
- Core Research Facility & Analysis Center, KRIBB, Daejeon 34141, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jun Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Young-Kyoung Ryu
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; KRIBB School, University of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jeong Hee Moon
- Core Research Facility & Analysis Center, KRIBB, Daejeon 34141, Republic of Korea.
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; KRIBB School, University of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
16
|
Bradford BM, Walmsley-Rowe L, Reynolds J, Verity N, Mabbott NA. Cell adhesion molecule CD44 is dispensable for reactive astrocyte activation during prion disease. Sci Rep 2024; 14:13749. [PMID: 38877012 PMCID: PMC11178777 DOI: 10.1038/s41598-024-63464-3] [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: 02/22/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024] Open
Abstract
Prion diseases are fatal, infectious, neurodegenerative disorders resulting from accumulation of misfolded cellular prion protein in the brain. Early pathological changes during CNS prion disease also include reactive astrocyte activation with increased CD44 expression, microgliosis, as well as loss of dendritic spines and synapses. CD44 is a multifunctional cell surface adhesion and signalling molecule which is considered to play roles in astrocyte morphology and the maintenance of dendritic spine integrity and synaptic plasticity. However, the role of CD44 in prion disease was unknown. Here we used mice deficient in CD44 to determine the role of CD44 during prion disease. We show that CD44-deficient mice displayed no difference in their response to CNS prion infection when compared to wild type mice. Furthermore, the reactive astrocyte activation and microgliosis that accompanies CNS prion infection was unimpaired in the absence of CD44. Together, our data show that although CD44 expression is upregulated in reactive astrocytes during CNS prion disease, it is dispensable for astrocyte and microglial activation and the development of prion neuropathogenesis.
Collapse
Affiliation(s)
- Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| | - Lauryn Walmsley-Rowe
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Joe Reynolds
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
- Maurice Wohl Basic and Clinical Neuroscience Institute, King's College London, Denmark Hill, London, SE5 9NU, UK
| | - Nicholas Verity
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| |
Collapse
|
17
|
Nguyen HD, Kim WK, Huong Vu G. Molecular mechanisms implicated in protein changes in the Alzheimer's disease human hippocampus. Mech Ageing Dev 2024; 219:111930. [PMID: 38554950 DOI: 10.1016/j.mad.2024.111930] [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: 12/03/2023] [Revised: 01/18/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
This study aimed to elucidate the specific biochemical pathways linked to changes in proteins in the Alzheimer's disease (AD) human hippocampus. Our data demonstrate a constant rise in the expression of four proteins (VGF, GFAP, HSPB1, and APP) across all eleven studies. Notably, UBC was the most centrally involved and had increased expression in the hippocampus tissue of individuals with AD. Modified proteins in the hippocampal tissue were found to activate the innate immune system and disrupt communication across chemical synapses. Four hub proteins (CD44, APP, ITGB2, and APOE) are connected to amyloid plaques, whereas two hub proteins (RPL24 and RPS23) are related to neurofibrillary tangles (NFTs). The presence of modified proteins was discovered to trigger the activation of microglia and decrease the functioning of ribosomes and mitochondria in the hippocampus. Three significant microRNAs (hsa-miR-106b-5p, hsa-miR-17-5p, and hsa-miR-16-5p) and transcription factors (MYT1L, PIN1, and CSRNP3) have been discovered to improve our understanding of the alterations in proteins within the hippocampal tissues that lead to the progression of AD. These findings establish a path for possible treatments for AD to employ therapeutic strategies that specifically focus on the proteins or processes linked to the illness.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea; Division of Microbiology, Tulane National Primate Research Center, Tulane University, Louisiana, USA.
| | - Woong-Ki Kim
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Louisiana, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Giang Huong Vu
- Department of Public Health, Hong Bang Health Center, Hai Phong, Vietnam
| |
Collapse
|
18
|
Ju Y, Shen T, Guo Z, Kong Y, Huang Y, Hu J. Vitronectin promotes insulin resistance in trophoblast cells by activating JNK in gestational diabetes mellitus. Cell Biol Int 2024. [PMID: 38654431 DOI: 10.1002/cbin.12167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 03/13/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
Gestational diabetes mellitus (GDM) is a common disorder in the clinic, which may lead to severe detrimental outcomes both for mothers and infants. However, the underlying mechanisms for GDM are still not clear. In the present study, we performed label-free proteomics using placentas from GDM patients and normal controls. Vitronectin caused our attention among differentially expressed proteins due to its potential role in the pathological progression of GDM. Vitronectin was increased in the placentas of GDM patients, which was confirmed by Western blot analysis. Vitronectin represses insulin signal transduction in trophoblast cells, whereas the knockdown of vitronectin further potentiates insulin-evoked events. Neutralization of CD51/61 abolishes the repressed insulin signal transduction in vitronectin-treated trophoblast cells. Moreover, vitronectin activates JNK in a CD51/61-depedent manner. Inhibition of JNK rescues impaired insulin signal transduction induced by vitronectin. Overall, our data indicate that vitronectin binds CD51/61 in trophoblast cells to activate JNK, and thus induces insulin resistance. In this regard, increased expression of vitronectin is likely a risk factor for the pathological progression of GDM. Moreover, blockade of vitronectin production or its receptors (CD51/61) may have therapeutic potential for dealing with GDM.
Collapse
Affiliation(s)
- Yuejun Ju
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, P.R. China
| | - Ting Shen
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, P.R. China
| | - Zhanhong Guo
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, P.R. China
| | - Yinghong Kong
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, P.R. China
| | - Yun Huang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
19
|
Chen C, He Y, Ni Y, Tang Z, Zhang W. Identification of crosstalk genes relating to ECM-receptor interaction genes in MASH and DN using bioinformatics and machine learning. J Cell Mol Med 2024; 28:e18156. [PMID: 38429902 PMCID: PMC10907849 DOI: 10.1111/jcmm.18156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 01/01/2024] [Accepted: 01/12/2024] [Indexed: 03/03/2024] Open
Abstract
This study aimed to identify genes shared by metabolic dysfunction-associated fatty liver disease (MASH) and diabetic nephropathy (DN) and the effect of extracellular matrix (ECM) receptor interaction genes on them. Datasets with MASH and DN were downloaded from the Gene Expression Omnibus (GEO) database. Pearson's coefficients assessed the correlation between ECM-receptor interaction genes and cross talk genes. The coexpression network of co-expression pairs (CP) genes was integrated with its protein-protein interaction (PPI) network, and machine learning was employed to identify essential disease-representing genes. Finally, immuno-penetration analysis was performed on the MASH and DN gene datasets using the CIBERSORT algorithm to evaluate the plausibility of these genes in diseases. We found 19 key CP genes. Fos proto-oncogene (FOS), belonging to the IL-17 signalling pathway, showed greater centrality PPI network; Hyaluronan Mediated Motility Receptor (HMMR), belonging to ECM-receptor interaction genes, showed most critical in the co-expression network map of 19 CP genes; Forkhead Box C1 (FOXC1), like FOS, showed a high ability to predict disease in XGBoost analysis. Further immune infiltration showed a clear positive correlation between FOS/FOXC1 and mast cells that secrete IL-17 during inflammation. Combining the results of previous studies, we suggest a FOS/FOXC1/HMMR regulatory axis in MASH and DN may be associated with mast cells in the acting IL-17 signalling pathway. Extracellular HMMR may regulate the IL-17 pathway represented by FOS through the Mitogen-Activated Protein Kinase 1 (ERK) or PI3K-Akt-mTOR pathway. HMMR may serve as a signalling carrier between MASH and DN and could be targeted for therapeutic development.
Collapse
Affiliation(s)
- Chao Chen
- Instrumentation and Service Center for Science and TechnologyBeijing Normal UniversityZhuhaiChina
| | - Yuxi He
- Pediatric Research InstituteThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ying Ni
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Advanced Institute of Natural SciencesBeijing Normal UniversityZhuhaiChina
- Engineering Research Center of Natural Medicine, Ministry of Education, Advanced Institute of Natural SciencesBeijing Normal UniversityZhuhaiChina
| | - Zhanming Tang
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Advanced Institute of Natural SciencesBeijing Normal UniversityZhuhaiChina
- Engineering Research Center of Natural Medicine, Ministry of Education, Advanced Institute of Natural SciencesBeijing Normal UniversityZhuhaiChina
| | - Wensheng Zhang
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Advanced Institute of Natural SciencesBeijing Normal UniversityZhuhaiChina
- Engineering Research Center of Natural Medicine, Ministry of Education, Advanced Institute of Natural SciencesBeijing Normal UniversityZhuhaiChina
| |
Collapse
|
20
|
Han B, Zhou S, Zhang Y, Chen S, Xi W, Liu C, Zhou X, Yuan M, Yu X, Li L, Wang Y, Ren H, Xie J, Li B, Ju M, Zhou Y, Liu Z, Xiong Z, Shen L, Zhang Y, Bai Y, Chen J, Jiang W, Yao H. Integrating spatial and single-cell transcriptomics to characterize the molecular and cellular architecture of the ischemic mouse brain. Sci Transl Med 2024; 16:eadg1323. [PMID: 38324639 DOI: 10.1126/scitranslmed.adg1323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
Neuroinflammation is acknowledged as a pivotal pathological event after cerebral ischemia. However, there is limited knowledge of the molecular and spatial characteristics of nonneuronal cells, as well as of the interactions between cell types in the ischemic brain. Here, we used spatial transcriptomics to study the ischemic hemisphere in mice after stroke and sequenced the transcriptomes of 19,777 spots, allowing us to both visualize the transcriptional landscape within the tissue and identify gene expression profiles linked to specific histologic entities. Cell types identified by single-cell RNA sequencing confirmed and enriched the spatial annotation of ischemia-associated gene expression in the peri-infarct area of the ischemic hemisphere. Analysis of ligand-receptor interactions in cell communication revealed galectin-9 to cell-surface glycoprotein CD44 (LGALS9-CD44) as a critical signaling pathway after ischemic injury and identified microglia and macrophages as the main source of galectins after stroke. Extracellular vesicle-mediated Lgals9 delivery improved the long-term functional recovery in photothrombotic stroke mice. Knockdown of Cd44 partially reversed these therapeutic effects, inhibiting oligodendrocyte differentiation and remyelination. In summary, our study provides a detailed molecular and cellular characterization of the peri-infact area in a murine stroke model and revealed Lgals9 as potential treatment target that warrants further investigation.
Collapse
Affiliation(s)
- Bing Han
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shunheng Zhou
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Yuan Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Sina Chen
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Wen Xi
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenchen Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xu Zhou
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Mengqin Yuan
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Xiaoyu Yu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lu Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yu Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hui Ren
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jian Xie
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Bin Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Minzi Ju
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - You Zhou
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ziqi Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhongli Xiong
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ling Shen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuan Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Wei Jiang
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Honghong Yao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China
| |
Collapse
|
21
|
Qian Y, Yang L, Chen J, Zhou C, Zong N, Geng Y, Xia S, Yang H, Bao X, Chen Y, Xu Y. SRGN amplifies microglia-mediated neuroinflammation and exacerbates ischemic brain injury. J Neuroinflammation 2024; 21:35. [PMID: 38287411 PMCID: PMC10826034 DOI: 10.1186/s12974-024-03026-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Microglia is the major contributor of post-stroke neuroinflammation cascade and the crucial cellular target for the treatment of ischemic stroke. Currently, the endogenous mechanism underlying microglial activation following ischemic stroke remains elusive. Serglycin (SRGN) is a proteoglycan expressed in immune cells. Up to now, the role of SRGN on microglial activation and ischemic stroke is largely unexplored. METHODS Srgn knockout (KO), Cd44-KO and wild-type (WT) mice were subjected to middle cerebral artery occlusion (MCAO) to mimic ischemic stroke. Exogenous SRGN supplementation was achieved by stereotactic injection of recombinant mouse SRGN (rSRGN). Cerebral infarction was measured by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Neurological functions were evaluated by the modified neurological severity score (mNSS) and grip strength. Microglial activation was detected by Iba1 immunostaining, morphological analysis and cytokines' production. Neuronal death was examined by MAP2 immunostaining and FJB staining. RESULTS The expression of SRGN and its receptor CD44 was significantly elevated in the ischemic mouse brains, especially in microglia. In addition, lipopolysaccharide (LPS) induced SRGN upregulation in microglia in vitro. rSRGN worsened ischemic brain injury in mice and amplified post-stroke neuroinflammation, while gene knockout of Srgn exerted reverse impacts. rSRGN promoted microglial proinflammatory activation both in vivo and in vitro, whereas Srgn-deficiency alleviated microglia-mediated inflammatory response. Moreover, the genetic deletion of Cd44 partially rescued rSRGN-induced excessed neuroinflammation and ischemic brain injury in mice. Mechanistically, SRGN boosted the activation of NF-κB signal, and increased glycolysis in microglia. CONCLUSION SRGN acts as a novel therapeutic target in microglia-boosted proinflammatory response following ischemic stroke.
Collapse
Affiliation(s)
- Yi Qian
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Lixuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jian Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Chao Zhou
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Ningning Zong
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yang Geng
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Haiyan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yan Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
- Nanjing Neurology Medical Center, Nanjing, 210008, China.
| |
Collapse
|
22
|
Deng H, Gao J, Cao B, Qiu Z, Li T, Zhao R, Li H, Wei B. LncRNA CCAT2 promotes malignant progression of metastatic gastric cancer through regulating CD44 alternative splicing. Cell Oncol (Dordr) 2023; 46:1675-1690. [PMID: 37354353 DOI: 10.1007/s13402-023-00835-4] [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] [Accepted: 05/30/2023] [Indexed: 06/26/2023] Open
Abstract
OBJECTIVE Gastric cancer (GC) is one of the most malignant tumors worldwide. Thus, it is necessary to explore the underlying mechanisms of GC progression and develop novel therapeutic regimens. Long non-coding RNAs (lncRNAs) have been demonstrated to be abnormally expressed and regulate the malignant behaviors of cancer cells. Our previous research demonstrated that lncRNA colon cancer-associated transcript 2 (CCAT2) has potential value for GC diagnosis and discrimination. However, the functional mechanisms of lncRNA CCAT2 in GC development remain to be explored. METHODS GC and normal adjacent tissues were collected to detect the expression of lncRNA CCAT2, ESRP1 and CD44 in clinical specimens and their clinical significance for GC patients. Cell counting kit-8, wound healing and transwell assays were conducted to investigate the malignant behaviors in vitro. The generation of nude mouse xenografts by subcutaneous, intraperitoneal and tail vein injection was performed to examine GC growth and metastasis in vivo. Co-immunoprecipitation, RNA-binding protein pull-down assay and fluorescence in situ hybridization were performed to reveal the binding relationships between ESRP1 and CD44. RESULTS In the present study, lncRNA CCAT2 was overexpressed in GC tissues compared to adjacent normal tissues and correlated with short survival time of patients. lncRNA CCAT2 promoted the proliferation, migration and invasion of GC cells. Its overexpression modulates alternative splicing of Cluster of differentiation 44 (CD44) variants and facilitates the conversion from the standard form to variable CD44 isoform 6 (CD44v6). Mechanistically, lncRNA CCAT2 upregulated CD44v6 expression by binding to epithelial splicing regulatory protein 1 (ESRP1), which subsequently mediates CD44 alternative splicing. The oncogenic role of the lncRNA CCAT2/ESRP1/CD44 axis in the promotion of malignant behaviors was verified by both in vivo and in vitro experiments. CONCLUSIONS Our findings identified a novel mechanism by which lncRNA CCAT2, as a type of protein-binding RNA, regulates alternative splicing of CD44 and promotes GC progression. This axis may become an effective target for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Huan Deng
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, China
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, 28 Fuxing Rd, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jingwang Gao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, 28 Fuxing Rd, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, 28 Fuxing Rd, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Ziyu Qiu
- Health Service Department of the Guard Bureau of the General Office of the Central Committee of the Communist Party of China, Beijing, 100091, China
| | - Tian Li
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, 710021, China
| | - Ruiyang Zhao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, 28 Fuxing Rd, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Hanghang Li
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, 28 Fuxing Rd, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Bo Wei
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, 28 Fuxing Rd, Beijing, 100853, China.
- Medical School of Chinese PLA, Beijing, 100853, China.
| |
Collapse
|
23
|
Du Y, Bradshaw WJ, Leisner TM, Annor-Gyamfi JK, Qian K, Bashore FM, Sikdar A, Nwogbo FO, Ivanov AA, Frye SV, Gileadi O, Brennan PE, Levey AI, Axtman AD, Pearce KH, Fu H, Katis VL. Discovery of FERM domain protein-protein interaction inhibitors for MSN and CD44 as a potential therapeutic approach for Alzheimer's disease. J Biol Chem 2023; 299:105382. [PMID: 37866628 PMCID: PMC10692723 DOI: 10.1016/j.jbc.2023.105382] [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: 05/22/2023] [Revised: 09/11/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
Proteomic studies have identified moesin (MSN), a protein containing a four-point-one, ezrin, radixin, moesin (FERM) domain, and the receptor CD44 as hub proteins found within a coexpression module strongly linked to Alzheimer's disease (AD) traits and microglia. These proteins are more abundant in Alzheimer's patient brains, and their levels are positively correlated with cognitive decline, amyloid plaque deposition, and neurofibrillary tangle burden. The MSN FERM domain interacts with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) and the cytoplasmic tail of CD44. Inhibiting the MSN-CD44 interaction may help limit AD-associated neuronal damage. Here, we investigated the feasibility of developing inhibitors that target this protein-protein interaction. We have employed structural, mutational, and phage-display studies to examine how CD44 binds to the FERM domain of MSN. Interestingly, we have identified an allosteric site located close to the PIP2 binding pocket that influences CD44 binding. These findings suggest a mechanism in which PIP2 binding to the FERM domain stimulates CD44 binding through an allosteric effect, leading to the formation of a neighboring pocket capable of accommodating a receptor tail. Furthermore, high-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction. One compound series was further optimized for biochemical activity, specificity, and solubility. Our results suggest that the FERM domain holds potential as a drug development target. Small molecule preliminary leads generated from this study could serve as a foundation for additional medicinal chemistry efforts with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.
Collapse
Affiliation(s)
- Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - William J Bradshaw
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Oxford, UK
| | - Tina M Leisner
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA
| | - Joel K Annor-Gyamfi
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Structural Genomics Consortium, Chapel Hill, North Carolina, USA
| | - Kun Qian
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Frances M Bashore
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Structural Genomics Consortium, Chapel Hill, North Carolina, USA
| | - Arunima Sikdar
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA
| | - Felix O Nwogbo
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Stephen V Frye
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA
| | - Opher Gileadi
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Oxford, UK
| | - Paul E Brennan
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Oxford, UK
| | - Allan I Levey
- Department of Neurology, Emory Goizueta Alzheimer's Disease Research Center, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alison D Axtman
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Structural Genomics Consortium, Chapel Hill, North Carolina, USA.
| | - Kenneth H Pearce
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, North Carolina, USA.
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, USA.
| | - Vittorio L Katis
- Alzheimer's Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Oxford, UK.
| |
Collapse
|
24
|
Zhang F, Liu M, Tuo J, Zhang L, Zhang J, Yu C, Xu Z. Levodopa-induced dyskinesia: interplay between the N-methyl-D-aspartic acid receptor and neuroinflammation. Front Immunol 2023; 14:1253273. [PMID: 37860013 PMCID: PMC10582719 DOI: 10.3389/fimmu.2023.1253273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder of middle-aged and elderly people, clinically characterized by resting tremor, myotonia, reduced movement, and impaired postural balance. Clinically, patients with PD are often administered levodopa (L-DOPA) to improve their symptoms. However, after years of L-DOPA treatment, most patients experience complications of varying severity, including the "on-off phenomenon", decreased efficacy, and levodopa-induced dyskinesia (LID). The development of LID can seriously affect the quality of life of patients, but its pathogenesis is unclear and effective treatments are lacking. Glutamic acid (Glu)-mediated changes in synaptic plasticity play a major role in LID. The N-methyl-D-aspartic acid receptor (NMDAR), an ionotropic glutamate receptor, is closely associated with synaptic plasticity, and neuroinflammation can modulate NMDAR activation or expression; in addition, neuroinflammation may be involved in the development of LID. However, it is not clear whether NMDA receptors are co-regulated with neuroinflammation during LID formation. Here we review how neuroinflammation mediates the development of LID through the regulation of NMDA receptors, and assess whether common anti-inflammatory drugs and NMDA receptor antagonists may be able to mitigate the development of LID through the regulation of central neuroinflammation, thereby providing a new theoretical basis for finding new therapeutic targets for LID.
Collapse
Affiliation(s)
- Fanshi Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Mei Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jinmei Tuo
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Li Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| |
Collapse
|
25
|
Du Y, Bradshaw WJ, Leisner TM, Annor-Gyamfi JK, Qian K, Bashore FM, Sikdar A, Nwogbo FO, Ivanov AA, Frye SV, Gileadi O, Brennan PE, Levey AI, Axtman AD, Pearce KH, Fu H, Katis VL. Development of FERM domain protein-protein interaction inhibitors for MSN and CD44 as a potential therapeutic strategy for Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541727. [PMID: 37292860 PMCID: PMC10245921 DOI: 10.1101/2023.05.22.541727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Recent genome-wide association studies have revealed genetic risk factors for Alzheimer's disease (AD) that are exclusively expressed in microglia within the brain. A proteomics approach identified moesin (MSN), a FERM (four-point-one ezrin radixin moesin) domain protein, and the receptor CD44 as hub proteins found within a co-expression module strongly linked to AD clinical and pathological traits as well as microglia. The FERM domain of MSN interacts with the phospholipid PIP2 and the cytoplasmic tails of receptors such as CD44. This study explored the feasibility of developing protein-protein interaction inhibitors that target the MSN-CD44 interaction. Structural and mutational analyses revealed that the FERM domain of MSN binds to CD44 by incorporating a beta strand within the F3 lobe. Phage-display studies identified an allosteric site located close to the PIP2 binding site in the FERM domain that affects CD44 binding within the F3 lobe. These findings support a model in which PIP2 binding to the FERM domain stimulates receptor tail binding through an allosteric mechanism that causes the F3 lobe to adopt an open conformation permissive for binding. High-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction, and one compound series was further optimized for biochemical activity, specificity, and solubility. The results suggest that the FERM domain holds potential as a drug development target. The small molecule preliminary leads generated from the study could serve as a foundation for additional medicinal chemistry effort with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.
Collapse
Affiliation(s)
- Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - William J. Bradshaw
- Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford, OX3 7FZ, UK
| | - Tina M. Leisner
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Joel K. Annor-Gyamfi
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, Chapel Hill, NC 27599, USA
| | - Kun Qian
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Current address: Chemical Biology Consortium Sweden, Division of Chemical Biology and Genome Engineering, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 65 Solna, Sweden
| | - Frances M. Bashore
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, Chapel Hill, NC 27599, USA
| | - Arunima Sikdar
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Felix O. Nwogbo
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Andrey A. Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stephen V. Frye
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Opher Gileadi
- Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford, OX3 7FZ, UK
- Current address: Structural Genomics Consortium, Department of Medicine, Karolinska Hospital and Karolinska Institute, 171 76 Stockholm, Sweden
| | - Paul E. Brennan
- Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford, OX3 7FZ, UK
| | - Allan I. Levey
- Department of Neurology, Emory Goizueta Alzheimer’s Disease Research Center, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Alison D. Axtman
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, Chapel Hill, NC 27599, USA
| | - Kenneth H. Pearce
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, Chapel Hill, NC 27599, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Vittorio L. Katis
- Alzheimer’s Research UK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford, OX3 7FZ, UK
| |
Collapse
|
26
|
Exosomes from Inflamed Macrophages Promote the Progression of Parkinson's Disease by Inducing Neuroinflammation. Mol Neurobiol 2023; 60:1914-1928. [PMID: 36596964 DOI: 10.1007/s12035-022-03179-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023]
Abstract
Inflammation is a common feature both for Parkinson's disease (PD) and obesity-associated metabolic syndromes. Inflammation mediated by inflamed macrophages in white adipose tissue plays a pivotal role for the pathogenesis of metabolic syndromes. Exosomes are important carriers connecting peripheral tissues and the central nervous system (CNS). Therefore, we speculate that exosomes derived from inflamed macrophages may be involved in the pathological progression of PD. Here, we prepared exosomes from lipopolysaccharide (LPS) or interferon gamma (IFNγ) treated macrophages (inflamed macrophages) and examined their potential roles in PD. Our data showed that exosomes from inflamed macrophages stimulate proinflammatory cytokine expression in primary microglia and astrocytes. In vivo, inflamed macrophage exosomes induce behavioral defects in mice as evidenced by shortened duration in the rotarod test and prolonged latency in the pole test. The treatment of exosomes also reduces tyrosine hydroxylase (TH) positive cells in the substantia nigra pars compacta (SNpc) and striatum. All these PD-like phenotypes are likely due to the activation of microglia and astrocytes induced by exosomes from inflamed macrophages. Exosome sequencing, together with bioinformatics analysis and functional studies, revealed that exosomal miRNAs such as miR-155-5p are likely a key factor for inducing an inflammatory response in glial cells. These results indicate that exosomes derived from inflamed macrophages are likely a causative factor for developing PD. In this regard, inflamed macrophage exosomes might be a linker transducing the peripheral tissue inflammation into the CNS.
Collapse
|
27
|
Batiha GES, Al-kuraishy HM, Al-Gareeb AI, Elekhnawy E. SIRT1 pathway in Parkinson's disease: a faraway snapshot but so close. Inflammopharmacology 2023; 31:37-56. [PMID: 36580159 PMCID: PMC9957916 DOI: 10.1007/s10787-022-01125-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 12/19/2022] [Indexed: 12/30/2022]
Abstract
Silent information regulator (SIRT) has distinctive enzymatic activities and physiological functions to control cell-cycle progression, gene expression, and DNA stability by targeting histone and non-histone proteins. SIRT1 enhances synaptic formation and synaptic activity, and therefore, can reduce the progression of various degenerative brain diseases including Parkinson's disease (PD). SIRT1 activity is decreased by aging with a subsequent increased risk for the development of degenerative brain diseases. Inhibition of SIRT1 promotes inflammatory reactions since SIRT1 inhibits transcription of nuclear factor kappa B (NF-κB) which also inhibits SIRT1 activation via activation of microRNA and miR-34a which reduce NAD synthesis. SIRT1 is highly expressed in microglia as well as neurons, and has antioxidant and anti-inflammatory effects. Therefore, this review aimed to find the possible role of SIRT1 in PD neuropathology. SIRT1 has neuroprotective effects; therefore, downregulation of SIRT1 during aging promotes p53 expression and may increase the vulnerability of neuronal cell deaths. PD neuropathology is linked with the sequence of inflammatory changes and the release of pro-inflammatory cytokines due to the activation of inflammatory signaling pathways. In addition, oxidative stress, inflammatory disorders, mitochondrial dysfunction, and apoptosis contribute mutually to PD neuropathology. Thus, SIRT1 and SIRT1 activators play a crucial role in the mitigation of PD neuropathology through the amelioration of oxidative stress, inflammatory disorders, mitochondrial dysfunction, apoptosis, and inflammatory signaling pathways.
Collapse
Affiliation(s)
- Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 AlBeheira Egypt
| | - Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132 Iraq
| | - Ali I. Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132 Iraq
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527 Egypt
| |
Collapse
|
28
|
Chen KW, Hsu PH, Huang HL, Liu HL, Lin YT, Hsu CY, Lin JH, Lin YH. Targeting nanoparticle-conjugated microbubbles combined with ultrasound-mediated microbubble destruction for enhanced tumor therapy. Pharmacol Res 2022; 186:106532. [DOI: 10.1016/j.phrs.2022.106532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
|
29
|
Dong X, Fan J, Lin D, Wang X, Kuang H, Gong L, Chen C, Jiang J, Xia N, He D, Shen W, Jiang P, Kuang R, Zeng L, Xie Y. Captopril alleviates epilepsy and cognitive impairment by attenuation of C3-mediated inflammation and synaptic phagocytosis. J Neuroinflammation 2022; 19:226. [PMID: 36104755 PMCID: PMC9476304 DOI: 10.1186/s12974-022-02587-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/04/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractEvidence from experimental and clinical studies implicates immuno-inflammatory responses as playing an important role in epilepsy-induced brain injury. Captopril, an angiotensin-converting enzyme inhibitor (ACEi), has previously been shown to suppress immuno-inflammatory responses in a variety of neurological diseases. However, the therapeutic potential of captopril on epilepsy remains unclear. In the present study, Sprague Dawley (SD) rats were intraperitoneally subjected to kainic acid (KA) to establish a status epilepticus. Captopril (50 mg/kg, i.p.) was administered daily following the KA administration from day 3 to 49. We found that captopril efficiently suppressed the KA-induced epilepsy, as measured by electroencephalography. Moreover, captopril ameliorated the epilepsy-induced cognitive deficits, with improved performance in the Morris water maze, Y-maze and novel objective test. RNA sequencing (RNA-seq) analysis indicated that captopril reversed a wide range of epilepsy-related biological processes, particularly the glial activation, complement system-mediated phagocytosis and the production of inflammatory factors. Interestingly, captopril suppressed the epilepsy-induced activation and abnormal contact between astrocytes and microglia. Immunohistochemical experiments demonstrated that captopril attenuated microglia-dependent synaptic remodeling presumably through C3–C3ar-mediated phagocytosis in the hippocampus. Finally, the above effects of captopril were partially blocked by an intranasal application of recombinant C3a (1.3 μg/kg/day). Our findings demonstrated that captopril reduced the occurrence of epilepsy and cognitive impairment by attenuation of inflammation and C3-mediated synaptic phagocytosis. This approach can easily be adapted to long-term efficacy and safety in clinical practice.
Graphical Abstract
Collapse
|
30
|
A 2A Adenosine Receptor Antagonists: Are Triazolotriazine and Purine Scaffolds Interchangeable? Molecules 2022; 27:molecules27082386. [PMID: 35458588 PMCID: PMC9032385 DOI: 10.3390/molecules27082386] [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: 03/09/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 12/10/2022] Open
Abstract
The A2A adenosine receptor (A2AAR) is one of the four subtypes activated by nucleoside adenosine, and the molecules able to selectively counteract its action are attractive tools for neurodegenerative disorders. In order to find novel A2AAR ligands, two series of compounds based on purine and triazolotriazine scaffolds were synthesized and tested at ARs. Compound 13 was also tested in an in vitro model of neuroinflammation. Some compounds were found to possess high affinity for A2AAR, and it was observed that compound 13 exerted anti-inflammatory properties in microglial cells. Molecular modeling studies results were in good agreement with the binding affinity data and underlined that triazolotriazine and purine scaffolds are interchangeable only when 5- and 2-positions of the triazolotriazine moiety (corresponding to the purine 2- and 8-positions) are substituted.
Collapse
|