1
|
Kawakita S, Naito K, Kubota D, Ueno Y, Negishi-Koga T, Yamamoto Y, Suzuki T, Imazu N, Kawamura K, Hattori N, Ishijima M. Glycoprotein 130 improves repressor element‑1 silencing transcription factor‑related axon regenerative capacity in peripheral nerves with aging. Mol Med Rep 2025; 31:121. [PMID: 40052576 PMCID: PMC11920774 DOI: 10.3892/mmr.2025.13486] [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: 10/08/2024] [Accepted: 01/09/2025] [Indexed: 03/21/2025] Open
Abstract
Axon regenerative capacity diminishes with aging and differences in the condition of peripheral nerves between young and elderly individuals have been reported. However, the underlying pathology remains unclear. The expression of repressor element‑1 silencing transcription factor (REST) increases with age and is reported to suppress axon regeneration. The present study investigated the pathology and potential treatment of reduced axon regenerative capacity using REST‑regulated cells and a mouse model. This study examined the molecular expression of the janus kinase 1 (JAK1)/signal transducer and activator of transcription 3 (STAT3) pathway, which is involved in growth‑associated protein 43 (GAP43) expression. In REST‑overexpressed (REST‑OE), glycoprotein 130 (GP130), JAK1 and phosphorylated STAT3 (p‑STAT3) expression was decreased compared with the control (GP130, P=0.004; JAK1, P=0.038; pSTAT3, P=0.015). On the other hand, in REST‑low expressed (siREST), GP130, JAK1 and pSTAT3 expression was increased compared with the control (GP130, P=0.004; JAK1, P=0.003; pSTAT3, P=0.033). It suggested that GP130 plays an important role. Therefore, GP130 agonist was administered to REST‑OE and aged mice and resulted in a significant increase in GAP43 expression (REST‑OE: Protein P=0.018, mRNA P=0.040; aged mice: Protein P=0.016, mRNA P=0.013). The results of this study suggest that the pathology of reduction in peripheral nerve axon regenerative capacity is inhibited by age‑related increase in REST expression, which leads to decreased GP130 expression and inhibition of JAK1/STAT3 pathway activity. These findings suggest that regulating GP130 expression may improve axon regenerative capacity by aging.
Collapse
Affiliation(s)
- So Kawakita
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113‑8421, Japan
| | - Kiyohito Naito
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113‑8421, Japan
| | - Daisuke Kubota
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113‑8421, Japan
| | - Yuji Ueno
- Department of Neurology, Graduate School of Medical Sciences, University of Yamanashi, Chuo, Yamanashi 409‑3898, Japan
| | - Takako Negishi-Koga
- Department of Orthopedics, Juntendo University Faculty of Medicine, Tokyo 113‑8421, Japan
| | - Yasuhiro Yamamoto
- Department of Orthopedics, Juntendo University Faculty of Medicine, Tokyo 113‑8421, Japan
| | - Takamaru Suzuki
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113‑8421, Japan
| | - Norizumi Imazu
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113‑8421, Japan
| | - Kenjiro Kawamura
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113‑8421, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo 113‑8421, Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113‑8421, Japan
| |
Collapse
|
2
|
Yang Y, Gong S, Zhou C, Xin W, Qin S, Yao M, Lan Q, Liao W, Zhao J, Huang Y. REST contributes to renal fibrosis through inducing mitochondrial energy metabolism imbalance in tubular epithelial cells. Cell Commun Signal 2025; 23:176. [PMID: 40200371 PMCID: PMC11980176 DOI: 10.1186/s12964-025-02166-3] [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: 11/20/2024] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Renal fibrosis represents the final common pathological manifestation of chronic kidney disease (CKD), yet the underlying mechanism remains elusive, and there is still a lack of effective targeted therapeutic strategy. Although previous research indicated that repressor element 1-silencing transcription factor (REST) contributed to acute kidney injury (AKI) in renal tubular epithelial cells (RTECs), its specific contribution to renal fibrosis and associated mechanisms remains largely unexplored. METHODS Renal biopsies from CKD patients were collected to evaluate the expression of REST. Kidney-specific Rest conditional knockout (Cdh16-Cre/Restflox/flox) mice were generated and employed unilateral ureter obstruction (UUO) models to investigate the role of REST in renal fibrosis. RNA sequencing was performed to elucidate the mechanism. Mitochondrial function was evaluated by transmission electron microscopy (TEM), reactive oxygen species (ROS), oxygen consumption rates (OCR), extracellular acidifcation rate (ECAR) and adenosine triphosphate (ATP). The severity of renal fibrosis was assessed through Western blot, immunofluorescent staining and immumohistochemical staining. Bioinformatic prediction, dual luciferase reporter gene assay, point mutation and chromatin immunoprecipitation (ChIP) assay were utilized to clarify the molecular mechanism. RESULTS REST was significantly up-regulated in the kidney tissues from CKD patients, UUO-induced fibrotic mouse models and TGF-β1-incubated RTECs. Notably, kidney-specific knockout of Rest prominently alleviated renal fibrosis by improving mitochondrial energy metabolism and restoring fatty acid oxidation. Mechanically, REST disturbed mitochondrial energy metabolism through repressing the transcription of oxoglutarate dehydrogenase-like (OGDHL) via directly binding to its promotor region. Further, pharmacological inhibition of REST using the specific REST inhibitor, X5050, significantly ameliorated the progression of renal fibrosis both in vitro and in vivo. CONCLUSIONS Our explorations revealed the upregulation of REST in renal fibrosis disrupts mitochondrial energy metabolism through transcriptionally suppressing OGDHL, which may act as a promising therapeutic target for renal fibrosis.
Collapse
Affiliation(s)
- Yingxian Yang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Shuiqin Gong
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Chun Zhou
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Wang Xin
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Shaozong Qin
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Mengying Yao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Qigang Lan
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Wenhao Liao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Jinghong Zhao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China.
| | - Yinghui Huang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
3
|
Al-Abbasi Z, Bhuiyan SA, Renthal W, Molliver DC. A Transcriptomic Comparison of the HD10.6 Human Sensory Neuron-Derived Cell Line with Primary and iPSC Sensory Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.643725. [PMID: 40236231 PMCID: PMC11996562 DOI: 10.1101/2025.04.03.643725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
A key concern in early-stage analgesic discovery efforts is the extent to which mechanisms identified in rodents will translate to humans. To evaluate an alternative approach to the use of rodent dissociated DRG neurons for in vitro analyses of nociceptive signaling, we performed a transcriptomic analysis of the HD10.6 human dorsal root ganglion (DRG)-derived immortalized cell line. We conducted RNA-seq on proliferating and mature HD10.6 cells to characterize transcriptional changes associated with maturation. We then compared the transcriptomes of HD10.6 cells and several recently developed lines of human induced pluripotent stem cell-derived sensory neurons (iPSC-SN) to single-nucleus RNA-seq data from human DRGs. HD10.6 cells showed the highest correlation with 3 human sensory neuron subtypes associated with nociception and pruriception. Each of the iPSC-SN lines evaluated showed a distinct pattern of correlation with human sensory neuron subtypes. We identified G protein-coupled receptors (GPCRs) and ion channels that are expressed in both HD10.6 cells and human DRG neurons, as well as numerous genes that are expressed in human DRG but not in rodent, underscoring the need for human sensory neuron in vitro models. Proof-of-concept evaluations of protein kinase A, protein kinase C and Erk signaling provide examples of scalable assays using HD10.6 cells to investigate well-established GPCR signaling pathways. We conclude that HD10.6 cells provide a versatile model for exploring human neuronal signaling mechanisms.
Collapse
|
4
|
Messingschlager M, Mackowiak SD, Voelker MT, Bieg M, Loske J, Chua RL, Liebig J, Lukassen S, Thürmann L, Seegebarth A, Twardziok S, Doncevic D, Herrmann C, Lorenz S, Klages S, Steinbeis F, Witzenrath M, Kurth F, Conrad C, Sander LE, Ishaque N, Eils R, Lehmann I, Laudi S, Trump S. DNA methylation changes during acute COVID-19 are associated with long-term transcriptional dysregulation in patients' airway epithelial cells. EMBO Mol Med 2025:10.1038/s44321-025-00215-5. [PMID: 40119174 DOI: 10.1038/s44321-025-00215-5] [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: 01/30/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Molecular changes underlying the persistent health effects after SARS-CoV-2 infection remain poorly understood. To discern the gene regulatory landscape in the upper respiratory tract of COVID-19 patients, we performed enzymatic DNA methylome and single-cell RNA sequencing in nasal cells of COVID-19 patients (n = 19, scRNA-seq n = 14) and controls (n = 14, scRNA-seq n = 10). In addition, we resampled a subset of these patients for transcriptome analyses at 3 (n = 7) and 12 months (n = 5) post infection and followed the expression of differentially regulated genes over time. Genome-wide DNA methylation analysis revealed 3112 differentially methylated regions between COVID-19 patients and controls. Hypomethylated regions affected immune regulatory genes, while hypermethylated regions were associated with genes governing ciliary function. These genes were not only downregulated in the acute phase of the disease but sustained repressed up to 12 months post infection in ciliated cells. Validation in an independent cohort collected 6 months post infection (n = 15) indicated symptom-dependent transcriptional repression of ciliary genes. We therefore propose that hypermethylation observed in the acute phase may exert a long-term effect on gene expression, possibly contributing to post-acute COVID-19 sequelae.
Collapse
Affiliation(s)
- Marey Messingschlager
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
- Freie Universität Berlin, Institute of Biology, Berlin, Germany
| | - Sebastian D Mackowiak
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Maria Theresa Voelker
- Department of Anesthesiology and Intensive Care, University Hospital Leipzig, Leipzig, Germany
| | - Matthias Bieg
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Jennifer Loske
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
- Freie Universität Berlin, Institute of Biology, Berlin, Germany
| | - Robert Lorenz Chua
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Johannes Liebig
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Sören Lukassen
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Loreen Thürmann
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
| | - Anke Seegebarth
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
| | - Sven Twardziok
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Daria Doncevic
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Carl Herrmann
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Stephan Lorenz
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Sven Klages
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Fridolin Steinbeis
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
| | - Martin Witzenrath
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Florian Kurth
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Medicine, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Conrad
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Leif E Sander
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Naveed Ishaque
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Roland Eils
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, University of Heidelberg, Heidelberg, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Irina Lehmann
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Sven Laudi
- Department of Anesthesiology and Intensive Care, University Hospital Leipzig, Leipzig, Germany
| | - Saskia Trump
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany.
| |
Collapse
|
5
|
Edwards DL, Huang M, Wang TT. Soluble Factors and Mechanisms Regulated by Sialylated IgG Signaling. Immunol Rev 2025; 330:e70021. [PMID: 40084926 DOI: 10.1111/imr.70021] [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: 11/01/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Inflammation is a complex biological response that can be both induced and actively suppressed by IgG-Fc gamma receptor (FcγR) interactions. This review explores the role of IgG sialylation in reducing or blocking inflammatory responses. We first revisit foundational studies that established the anti-inflammatory properties of sialylated IgG1 Fc. These early investigations revealed that the sialylated fraction is crucial for intravenous immunoglobulin's (IVIg's) ability to reduce inflammation in many autoinflammatory diseases and defined a paracrine signaling mechanism underlying this activity. Next, we discuss a recently identified mechanism whereby sialylated IgG directly induces RE1-Silencing Transcription Factor (REST) which functions as a transcriptional repressor of NF-κB1. This mechanism suggests a very broad role for sialylated IgG signaling in inflammation control since NF-κB is a central mediator of responses downstream of diverse activating receptors on both adaptive and innate immune cells. Finally, we review a set of soluble factors that are suppressed by sialylated IgG signaling in the murine airway and in purified human macrophages, providing additional insight into mechanisms by which sialylated IgG contributes to broad inflammatory control.
Collapse
Affiliation(s)
- Desmond L Edwards
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
| | - Min Huang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
| | - Taia T Wang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
- Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
6
|
Walters K, Baldwin A, Liu Z, Larsen M, Mukherjee N, Kumar TR. Identification of FSH-regulated and estrous stage-specific transcriptional networks in mouse ovaries. Proc Natl Acad Sci U S A 2025; 122:e2411977122. [PMID: 39928863 PMCID: PMC11848299 DOI: 10.1073/pnas.2411977122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/03/2025] [Indexed: 02/12/2025] Open
Abstract
Follicle-stimulating hormone (FSH) acts by binding to FSHRs expressed on ovarian granulosa cells and produces estradiol. FSH is essential for female fertility because mice lacking FSH (Fshb KO) are anestrous and infertile. Although several in vitro cell culture and ex vivo approaches combined with pharmacological hormone treatment were used to identify FSH-regulated genes, how FSH orchestrates ovarian gene networks in vivo has not been investigated. Whether FSH-regulated genes display estrous stage-specific expression changes has also not been studied. Here, we functionally rescued Fshb null mice with a gonadotrope-targeted HFSHB transgene and performed RNA-Seq analysis on ovarian RNAs obtained from FSH-intact (WT), FSH-deficient (Fshb KO), and FSH-rescue (HFSHB+ rescue) mice. By comparing WT vs. Fshb KO and Fshb KO vs. HFSHB+ rescue ovarian gene expression datasets, we identified FSH-responsive genes in vivo. Cross interrogation of these datasets further allowed us to identify several transcription factors (TFs) and RNA-binding proteins specific to FSH-regulated genes. In an independent set of experiments, we performed RNA-Seq analysis on ovarian RNAs from mice in diestrous (DE), proestrous (PE), and estrous (E) and identified estrous stage-specific ovarian gene expression patterns. Interestingly, many of the FSH-regulated TFs themselves were estrous-stage specifically expressed. We found that ESR2 and GATA6, two known FSH-responsive TFs, and their target genes are reciprocally regulated with distinct patterns of expression in estrous stages. Together, our in vivo models and RNA-Seq analyses identify FSH-regulated ovarian genes in specific estrous stages that are under transcriptional and posttranscriptional control.
Collapse
Affiliation(s)
- Kathryn Walters
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO80045
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Amber Baldwin
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO80045
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Zhenghui Liu
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Mark Larsen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Neelanjan Mukherjee
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO80045
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - T. Rajendra Kumar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| |
Collapse
|
7
|
Kedzierski L, Kedzierska K. IgG sialylation puts lung inflammation to REST. Immunity 2025; 58:8-10. [PMID: 39813995 DOI: 10.1016/j.immuni.2024.12.001] [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: 11/21/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/18/2025]
Abstract
The mechanisms underpinning susceptibility to influenza virus infection, resulting in life-threatening disease, are not well understood. In this issue of Immunity, Chakraborty et al. demonstrate that sialylated IgG suppresses NF-κB-driven inflammatory responses in the lungs by inducing repressor element-1 silencing transcription factor (REST) to prevent excessive inflammation without impacting viral replication.
Collapse
Affiliation(s)
- Lukasz Kedzierski
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| |
Collapse
|
8
|
Chakraborty S, Cheng BYL, Edwards DL, Gonzalez JC, Chiu DKC, Zheng H, Scallan C, Guo X, Tan GS, Coffey GP, Conley PB, Hume PS, Janssen WJ, Byers DE, Mudd PA, Taubenberger J, Memoli M, Davis MM, Chua KF, Diamond MS, Andreakos E, Khatri P, Wang TT. Sialylated IgG induces the transcription factor REST in alveolar macrophages to protect against lung inflammation and severe influenza disease. Immunity 2025; 58:182-196.e10. [PMID: 39541970 PMCID: PMC11735284 DOI: 10.1016/j.immuni.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/15/2024] [Accepted: 10/04/2024] [Indexed: 11/17/2024]
Abstract
While most respiratory viral infections resolve with little harm to the host, severe symptoms arise when infection triggers an aberrant inflammatory response that damages lung tissue. Host regulators of virally induced lung inflammation have not been well defined. Here, we show that enrichment for sialylated, but not asialylated immunoglobulin G (IgG), predicted mild influenza disease in humans and was broadly protective against heterologous influenza viruses in a murine challenge model. Mechanistic studies show that sialylated IgG mediated this protection by inducing the transcription factor repressor element-1 silencing transcription factor (REST), which repressed nuclear factor κB (NF-κB)-driven responses, preventing severe lung inflammation and protecting lung function during influenza infection. Therapeutic administration of a recombinant, sialylated Fc molecule in clinical development similarly activated REST and protected against severe influenza disease, demonstrating that this pathway could be clinically harnessed. Overall, induction of REST through sialylated IgG signaling is a strategy to limit inflammatory disease sequelae in infections caused by antigenically distinct influenza strains.
Collapse
Affiliation(s)
- Saborni Chakraborty
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bowie Yik-Ling Cheng
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Desmond L Edwards
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Gonzalez
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David Kung-Chun Chiu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hong Zheng
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Courtney Scallan
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinrong Guo
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gene S Tan
- J. Craig Venter Institute, La Jolla, San Diego, CA 92037, USA; Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA 92037, USA
| | - Greg P Coffey
- Nuvig Therapeutics Inc., Redwood City, CA 94061, USA
| | | | - Patrick S Hume
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA
| | - William J Janssen
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Derek E Byers
- Department of Medicine, Division of Pulmonology and Critical Care Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Philip A Mudd
- Department of Emergency Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jeffery Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Matthew Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; HHMI, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katrin F Chua
- Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, CA 94305, USA; Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Purvesh Khatri
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Center for Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taia T Wang
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
9
|
Yu W, Han S, Hu S, Ru L, Hua C, Xue G, Zhang G, Lv K, Ge H, Wang M, Zheng L, Zhou J, Hou S, Teng Y, Deng W, Guo W. KIF15 promotes human glioblastoma progression under the synergistic transactivation of REST and P300. Int J Biol Sci 2024; 20:5127-5144. [PMID: 39430242 PMCID: PMC11488581 DOI: 10.7150/ijbs.98668] [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: 05/20/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Glioblastoma (GBM) is highly invasive and lethal. The failure to cure GBM highlights the necessity of developing more effective targeted therapeutic strategies. KIF15 is a motor protein to be involved in cell mitosis promotion, cell structure assembly and cell signal transduction. The precise biological function and the potential upstream regulatory mechanisms of KIF15 in GBM remain elusive. Here, we demonstrated that KIF15 was abnormally up-regulated in GBM and predicted poor prognosis of GBM patients. KIF15 promotes GBM cell proliferation, metastasis and cell cycle progression. REST could bind to KIF15 promoter and transactivate KIF15. Furthermore, REST interacts with P300 and depends on its histone acetyltransferase (HAT) activity to co-regulate KIF15 expression. Both REST and P300 were highly expressed in GBM and predicted poor prognosis of GBM patients alone or in combination with KIF15. The tumorigenic function of KIF15 in GBM was regulated by REST in vitro and in vivo and the combinational treatment of cell cycle inhibitor Palbociclib with P300 HAT inhibitor inhibited GBM xenografts survival more significantly. Our findings indicate that KIF15 promotes GBM progression under the synergistic transactivation of REST and P300. P300/REST/KIF15 signaling axis is expected to be served as a cascade of candidate therapeutic targets in anti-GBM.
Collapse
Affiliation(s)
- Wendan Yu
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shilong Han
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Sheng Hu
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Liyuan Ru
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunyu Hua
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Guoqing Xue
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Guohui Zhang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Kuan Lv
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Hanxiao Ge
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Meiyi Wang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Lina Zheng
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jie Zhou
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shuai Hou
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yun Teng
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, China
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangdong, China
| | - Wei Guo
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
10
|
Santos AB, Carona A, Ettcheto M, Camins A, Falcão A, Fortuna A, Bicker J. Krüppel-like factors: potential roles in blood-brain barrier dysfunction and epileptogenesis. Acta Pharmacol Sin 2024; 45:1765-1776. [PMID: 38684799 PMCID: PMC11335766 DOI: 10.1038/s41401-024-01285-w] [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: 12/22/2023] [Accepted: 04/07/2024] [Indexed: 05/02/2024]
Abstract
Epilepsy is a chronic and debilitating neurological disorder, known for the occurrence of spontaneous and recurrent seizures. Despite the availability of antiseizure drugs, 30% of people with epilepsy experience uncontrolled seizures and drug resistance, evidencing that new therapeutic options are required. The process of epileptogenesis involves the development and expansion of tissue capable of generating spontaneous recurrent seizures, during which numerous events take place, namely blood-brain barrier (BBB) dysfunction, and neuroinflammation. The consequent cerebrovascular dysfunction results in a lower seizure threshold, seizure recurrence, and chronic epilepsy. This suggests that improving cerebrovascular health may interrupt the pathological cycle responsible for disease development and progression. Krüppel-like factors (KLFs) are a family of zinc-finger transcription factors, encountered in brain endothelial cells, glial cells, and neurons. KLFs are known to regulate vascular function and changes in their expression are associated with neuroinflammation and human diseases, including epilepsy. Hence, KLFs have demonstrated various roles in cerebrovascular dysfunction and epileptogenesis. This review critically discusses the purpose of KLFs in epileptogenic mechanisms and BBB dysfunction, as well as the potential of their pharmacological modulation as therapeutic approach for epilepsy treatment.
Collapse
Affiliation(s)
| | - Andreia Carona
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Miren Ettcheto
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Antoni Camins
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Amílcar Falcão
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal.
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| |
Collapse
|
11
|
Grossi E, Nguyen CB, Carcamo S, Moran S, Callaú VK, Filipescu D, Hasson D, Bernstein E. The SWI/SNF PBAF complex facilitates REST occupancy at repressive chromatin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609212. [PMID: 39229151 PMCID: PMC11370600 DOI: 10.1101/2024.08.23.609212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Multimeric SWI/SNF chromatin remodelers assemble into discrete conformations with unique complex functionalities difficult to dissect. Distinct cancers harbor mutations in specific subunits, altering the chromatin landscape, such as the PBAF-specific component ARID2 in melanoma. Here, we performed comprehensive epigenomic profiling of SWI/SNF complexes and their associated chromatin states in melanoma and melanocytes and uncovered a subset of PBAF-exclusive regions that coexist with PRC2 and repressive chromatin. Time-resolved approaches revealed that PBAF regions are generally less sensitive to ATPase-mediated remodeling than BAF sites. Moreover, PBAF/PRC2-bound loci are enriched for REST, a transcription factor that represses neuronal genes. In turn, absence of ARID2 and consequent PBAF complex disruption hinders the ability of REST to bind and inactivate its targets, leading to upregulation of synaptic transcripts. Remarkably, this gene signature is conserved in melanoma patients with ARID2 mutations. In sum, we demonstrate a unique role for PBAF in generating accessibility for a silencing transcription factor at repressed chromatin, with important implications for disease.
Collapse
Affiliation(s)
- Elena Grossi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christie B. Nguyen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saul Carcamo
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing (BiNGS) Shared Resource Facility, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shannon Moran
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Valentina Kirigin Callaú
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan Filipescu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan Hasson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Bioinformatics for Next Generation Sequencing (BiNGS) Shared Resource Facility, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Bernstein
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
12
|
Qin P, Pan Z, Zhang W, Wang R, Li X, Lu J, Xu S, Gong X, Ye J, Yan X, Liu Y, Li Y, Zhang Y, Fang F. Integrative proteomic and transcriptomic analysis in the female goat ovary to explore the onset of puberty. J Proteomics 2024; 301:105183. [PMID: 38688390 DOI: 10.1016/j.jprot.2024.105183] [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: 12/10/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Puberty is considered a prerequisite for affecting reproductive performance and productivity. Little was known about molecular changes in pubertal goat ovaries. Therefore, we measured and performed a correlation analysis of the mRNA and proteins changes in the pre-pubertal and pubertal goat ovaries. The results showed that only six differentially expressed genes and differentially abundant proteins out of 18,139 genes and 7550 proteins quantified had significant correlations. CNTN2 and THBS1, discovered in the mRNA-mRNA interaction network, probably participated in pubertal and reproductive regulation by influencing GnRH receptor signals, follicular development, and ovulation. The predicted core transcription factors may either promote or inhibit the expression of reproductive genes and act synergistically to maintain normal reproductive function in animals. The interaction between PKM and TIMP3 with other proteins may impact animal puberty through energy metabolism and ovarian hormone secretion. Pathway enrichment analyses revealed that the co-associated key pathways between ovarian genes and proteins at puberty included calcium signalling pathway and olfactory transduction. These pathways were associated with gonadotropin-releasing hormone synthesis and secretion, signal transmission, and cell proliferation. In summary, these results enriched the potential molecules and signalling pathways that affect puberty and provided new insights for regulating and promoting the onset of puberty. SIGNIFICANCE: This study conducted the first transcriptomic and proteomic correlation analysis of pre-pubertal and pubertal goat ovaries and identified six significantly correlated molecules at both the gene and protein levels. Meanwhile, we were drawn to several molecules and signalling pathways that may play a regulatory role in the onset of puberty and reproduction by influencing reproductive-related gene expression, GnRH receptor signals, energy metabolism, ovarian hormone secretion, follicular development, and ovulation. This information contributed to identify potential biomarkers in pubertal goat ovaries, which was vital for predicting the onset of puberty and improving livestock performance.
Collapse
Affiliation(s)
- Ping Qin
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zhihao Pan
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Wei Zhang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Rui Wang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xiaoqian Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Juntai Lu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Shuangshuang Xu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xinbao Gong
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jing Ye
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xu Yan
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ya Liu
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunsheng Li
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunhai Zhang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Fugui Fang
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui 230036, China; Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
13
|
Chen S, Huang M, Xu D, Li M. Epigenetic regulation in epilepsy: A novel mechanism and therapeutic strategy for epilepsy. Neurochem Int 2024; 173:105657. [PMID: 38145842 DOI: 10.1016/j.neuint.2023.105657] [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: 08/23/2023] [Revised: 12/02/2023] [Accepted: 12/14/2023] [Indexed: 12/27/2023]
Abstract
Epilepsy is a common neurological disorder characterized by recurrent seizures with excessive and abnormal neuronal discharges. Epileptogenesis is usually involved in neuropathological processes such as ion channel dysfunction, neuronal injury, inflammatory response, synaptic plasticity, gliocyte proliferation and mossy fiber sprouting, currently the pathogenesis of epilepsy is not yet completely understood. A growing body of studies have shown that epigenetic regulation, such as histone modifications, DNA methylation, noncoding RNAs (ncRNAs), N6-methyladenosine (m6A) and restrictive element-1 silencing transcription factor/neuron-restrictive silencing factor (REST/NRSF) are also involved in epilepsy. Through epigenetic studies, we found that the synaptic dysfunction, nerve damage, cognitive dysfunction and brain development abnormalities are affected by epigenetic regulation of epilepsy-related genes in patients with epilepsy. However, the functional roles of epigenetics in pathogenesis and treatment of epilepsy are still to be explored. Therefore, profiling the array of genes that are epigenetically dysregulated in epileptogenesis is likely to advance our understanding of the mechanisms underlying the pathophysiology of epilepsy and may for the amelioration of these serious human conditions provide novel insight into therapeutic strategies and diagnostic biomarkers for epilepsy to improve serious human condition.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Hubei University of Chinese Medicine, Wuhan, 430000, China
| | - Ming Huang
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Hubei University of Chinese Medicine, Wuhan, 430000, China
| | - Da Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Man Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| |
Collapse
|