1
|
Han J, Zhang J, Yao X, Meng M, Wan Y, Cheng Y. Mechanism of HDAC1 Regulating Iron Overload-Induced Neuronal Oxidative Damage After Cerebral Hemorrhage. Mol Neurobiol 2024; 61:7549-7566. [PMID: 38403721 DOI: 10.1007/s12035-024-04000-2] [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] [Accepted: 01/29/2024] [Indexed: 02/27/2024]
Abstract
Iron overload is associated with brain edema in the context of intracerebral hemorrhage (ICH). Here, we investigated the role of histone deacetylase 1 (HDAC1) in mediating oxidative damage induced by iron overload after ICH. Utilizing ICH mouse models and FeCl2-induced HT-22 cell models, we assessed HDAC1 expression and its impact on iron overload and oxidative damage. We examined the levels of Kruppel like factor 4 (KLF4), RAN binding protein 9 (RANBP9), as well as the acetylation levels of HDAC1 and histones H3 and H4 in the KLF4 promoter, and the KLF4 level in the RANBP9 promoter. Additionally, we investigated the binding relationships between KLF4 and the RANBP9 promoter, HDAC1 and miR-129-5p. Our results demonstrated elevated HDAC1 expression in ICH mice and FeCl2-induced HT-22 cells. HDAC1 silencing improved neurological function in mice, reduced brain edema, and alleviated iron overload and oxidative damage in vitro. HDAC1 downregulated KLF4 expression by reducing acetylation levels in the KLF4 promoter, leading to decreased KLF4 enrichment in the RANBP9 promoter and increased RANBP9 expression. Furthermore, upstream miR-129-5p inhibited HDAC1, and the downregulation of miR-129-5p mitigated the protective effect of HDAC1 silencing. Collectively, our findings highlight the significant role of HDAC1 in exacerbating iron overload-induced oxidative damage following ICH and its regulation by miR-129-5p.
Collapse
Affiliation(s)
- Jing Han
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jinnan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xiaojuan Yao
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Meng Meng
- Department of Neurology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, 300000, China
| | - Yahui Wan
- Department of Neurology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, 300000, China
| | - Yan Cheng
- Department of Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
2
|
Liu D, Hu Z, Lu J, Yi C. Redox-Regulated Iron Metabolism and Ferroptosis in Ovarian Cancer: Molecular Insights and Therapeutic Opportunities. Antioxidants (Basel) 2024; 13:791. [PMID: 39061859 PMCID: PMC11274267 DOI: 10.3390/antiox13070791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Ovarian cancer (OC), known for its lethality and resistance to chemotherapy, is closely associated with iron metabolism and ferroptosis-an iron-dependent cell death process, distinct from both autophagy and apoptosis. Emerging evidence suggests that dysregulation of iron metabolism could play a crucial role in OC by inducing an imbalance in the redox system, which leads to ferroptosis, offering a novel therapeutic approach. This review examines how disruptions in iron metabolism, which affect redox balance, impact OC progression, focusing on its essential cellular functions and potential as a therapeutic target. It highlights the molecular interplay, including the role of non-coding RNAs (ncRNAs), between iron metabolism and ferroptosis, and explores their interactions with key immune cells such as macrophages and T cells, as well as inflammation within the tumor microenvironment. The review also discusses how glycolysis-related iron metabolism influences ferroptosis via reactive oxygen species. Targeting these pathways, especially through agents that modulate iron metabolism and ferroptosis, presents promising therapeutic prospects. The review emphasizes the need for deeper insights into iron metabolism and ferroptosis within the redox-regulated system to enhance OC therapy and advocates for continued research into these mechanisms as potential strategies to combat OC.
Collapse
Affiliation(s)
- Dan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| | - Zewen Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| | - Jinzhi Lu
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
- Department of Laboratory Medicine, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China
| | - Cunjian Yi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| |
Collapse
|
3
|
Elkjaer ML, Hartebrodt A, Oubounyt M, Weber A, Vitved L, Reynolds R, Thomassen M, Rottger R, Baumbach J, Illes Z. Single-Cell Multi-Omics Map of Cell Type-Specific Mechanistic Drivers of Multiple Sclerosis Lesions. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200213. [PMID: 38564686 PMCID: PMC11073880 DOI: 10.1212/nxi.0000000000200213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/19/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND AND OBJECTIVES In progressive multiple sclerosis (MS), compartmentalized inflammation plays a pivotal role in the complex pathology of tissue damage. The interplay between epigenetic regulation, transcriptional modifications, and location-specific alterations within white matter (WM) lesions at the single-cell level remains underexplored. METHODS We examined intracellular and intercellular pathways in the MS brain WM using a novel dataset obtained by integrated single-cell multi-omics techniques from 3 active lesions, 3 chronic active lesions, 3 remyelinating lesions, and 3 control WM of 6 patients with progressive MS and 3 non-neurologic controls. Single-nucleus RNA-seq and ATAC-seq were combined and additionally enriched with newly conducted spatial transcriptomics from 1 chronic active lesion. Functional gene modules were then validated in our previously published bulk tissue transcriptome data obtained from 73 WM lesions of patients with progressive MS and 25 WM of non-neurologic disease controls. RESULTS Our analysis uncovered an MS-specific oligodendrocyte genetic signature influenced by the KLF/SP gene family. This modulation has potential associations with the autocrine iron uptake signaling observed in transcripts of transferrin and its receptor LRP2. In addition, an inflammatory profile emerged within these oligodendrocytes. We observed unique cellular endophenotypes both at the periphery and within the chronic active lesion. These include a distinct metabolic astrocyte phenotype, the importance of FGF signaling among astrocytes and neurons, and a notable enrichment of mitochondrial genes at the lesion edge populated predominantly by astrocytes. Our study also identified B-cell coexpression networks indicating different functional B-cell subsets with differential location and specific tendencies toward certain lesion types. DISCUSSION The use of single-cell multi-omics has offered a detailed perspective into the cellular dynamics and interactions in MS. These nuanced findings might pave the way for deeper insights into lesion pathogenesis in progressive MS.
Collapse
Affiliation(s)
- Maria L Elkjaer
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Anne Hartebrodt
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mhaned Oubounyt
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Anna Weber
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lars Vitved
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Richard Reynolds
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mads Thomassen
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Richard Rottger
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jan Baumbach
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
4
|
Zhao K, Liu J, Sun T, Zeng L, Cai Z, Li Z, Liu R. The miR-25802/KLF4/NF-κB signaling axis regulates microglia-mediated neuroinflammation in Alzheimer's disease. Brain Behav Immun 2024; 118:31-48. [PMID: 38360375 DOI: 10.1016/j.bbi.2024.02.016] [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: 08/09/2023] [Revised: 01/15/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024] Open
Abstract
Microglia-mediated neuroinflammation plays a critical role in the occurrence and progression of Alzheimer's disease (AD). In recent years, studies have increasingly explored microRNAs as biomarkers and treatment interventions for AD. This study identified a novel microRNA termed miR-25802 from our high-throughput sequencing dataset of an AD model and explored its role and the underlying mechanism. The results confirmed the miRNA properties of miR-25802 based on bioinformatics and experimental verification. Expression of miR-25802 was increased in the plasma of AD patients and in the hippocampus of APP/PS1 and 5 × FAD mice carrying two and five familial AD gene mutations. Functional studies suggested that overexpression or inhibition of miR-25802 respectively aggravated or ameliorated AD-related pathology, including cognitive disability, Aβ deposition, microglial pro-inflammatory phenotype activation, and neuroinflammation, in 5 × FAD mice and homeostatic or LPS/IFN-γ-stimulated EOC20 microglia. Mechanistically, miR-25802 negatively regulates KLF4 by directly binding to KLF4 mRNA, thus stimulating microglia polarization toward the pro-inflammatory M1 phenotype by promoting the NF-κB-mediated inflammatory response. The results also showed that inhibition of miR-25802 increased microglial anti-inflammatory M2 phenotype activity and suppressed NF-κB-mediated inflammatory reactions in the brains of 5 × FAD mice, while overexpression of miR-25802 exacerbated microglial pro-inflammatory M1 activity by enhancing NF-κB pathways. Of note, AD-associated manifestations induced by inhibition or overexpression of miR-25802 via the NF-κB signaling pathway were reversed by KLF4 silencing or upregulation. Collectively, these results provide the first evidence that miR-25802 is a regulator of microglial activity and establish the role of miR-25802/KLF4/NF-κB signaling in microglia-mediated neuroinflammation, suggesting potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Kaiyue Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Jianghong Liu
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Ting Sun
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Li Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Zhongdi Cai
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Zhuorong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
5
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
6
|
Kouhnavardi S, Cabatic M, Mañas-Padilla MC, Malabanan MA, Smani T, Cicvaric A, Muñoz Aranzalez EA, Koenig X, Urban E, Lubec G, Castilla-Ortega E, Monje FJ. miRNA-132/212 Deficiency Disrupts Selective Corticosterone Modulation of Dorsal vs. Ventral Hippocampal Metaplasticity. Int J Mol Sci 2023; 24:9565. [PMID: 37298523 PMCID: PMC10253409 DOI: 10.3390/ijms24119565] [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: 05/08/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Cortisol is a potent human steroid hormone that plays key roles in the central nervous system, influencing processes such as brain neuronal synaptic plasticity and regulating the expression of emotional and behavioral responses. The relevance of cortisol stands out in the disease, as its dysregulation is associated with debilitating conditions such as Alzheimer's Disease, chronic stress, anxiety and depression. Among other brain regions, cortisol importantly influences the function of the hippocampus, a structure central for memory and emotional information processing. The mechanisms fine-tuning the different synaptic responses of the hippocampus to steroid hormone signaling remain, however, poorly understood. Using ex vivo electrophysiology and wild type (WT) and miR-132/miR-212 microRNAs knockout (miRNA-132/212-/-) mice, we examined the effects of corticosterone (the rodent's equivalent to cortisol in humans) on the synaptic properties of the dorsal and ventral hippocampus. In WT mice, corticosterone predominantly inhibited metaplasticity in the dorsal WT hippocampi, whereas it significantly dysregulated both synaptic transmission and metaplasticity at dorsal and ventral regions of miR-132/212-/- hippocampi. Western blotting further revealed significantly augmented levels of endogenous CREB and a significant CREB reduction in response to corticosterone only in miR-132/212-/- hippocampi. Sirt1 levels were also endogenously enhanced in the miR-132/212-/- hippocampi but unaltered by corticosterone, whereas the levels of phospo-MSK1 were only reduced by corticosterone in WT, not in miR-132/212-/- hippocampi. In behavioral studies using the elevated plus maze, miRNA-132/212-/- mice further showed reduced anxiety-like behavior. These observations propose miRNA-132/212 as potential region-selective regulators of the effects of steroid hormones on hippocampal functions, thus likely fine-tuning hippocampus-dependent memory and emotional processing.
Collapse
Affiliation(s)
- Shima Kouhnavardi
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Maureen Cabatic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Marife-Astrid Malabanan
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, University of Seville, 41013 Seville, Spain
| | - Ana Cicvaric
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Edison Alejandro Muñoz Aranzalez
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Xaver Koenig
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Ernst Urban
- Department for Pharmaceutical Sciences, Josef-Holaubek-Platz 2, 2D 303, 1090 Vienna, Austria
| | - Gert Lubec
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria
| | | | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
7
|
Tang Y, Wang D, Zhang H, Zhang Y, Wang J, Qi R, Yang J, Shen H, Xu Y, Li M. Rapid responses of adipocytes to iron overload increase serum TG level by decreasing adiponectin. J Cell Physiol 2021; 236:7544-7553. [PMID: 33855731 DOI: 10.1002/jcp.30391] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/14/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022]
Abstract
Iron overload is tightly connected with metabolic disorders. Excess iron in the adipose and its roles in dyslipidemia are of interest to be identified. In acute iron overload mice receiving intraperitoneal injection of 100 mg/kg/day dextran-iron for 5 days, the epididymis adipose showed a remarkable increase in iron. Serum triglyceride and low-density lipoprotein cholesterol (LDL-C) levels were increased and high-density lipoprotein cholesterol (HDL-C) level was decreased, while serum alkaline phosphatase, aspartate aminotransferase, glucose, and insulin were not affected. The serum-cytokine-microarray showed that adipocytokines, including adiponectin, leptin, and resistin were significantly decreased. Other serum cytokines, including pro-insulin cytokines, inflammatory cytokines, chemokines, and growth factors were not changed, except that ghrelin and chemokine RANTES were increased. Iron overload decreased expressions of adiponectin and leptin both in vivo and in vitro. Intraperitoneal injection of recombinant leptin at 1 μg/g in acute iron overload mice had no significant effects on serum levels of TC, TG, HDL-C, and LDL-C, while intraperitoneal injection of recombinant adiponectin at 3 μg/g partially restored serum TG level through improving activities of lipoprotein lipase and hepatic lipase, but abnormal serum LDL-C and HDL-C were not redressed, suggesting other mechanisms also existed. In conclusion, the adipose responds to iron overload at an early stage to interfere with lipid metabolism by secreting adipocytokines, which may further affect glucose metabolism, inflammation, and other iron overload-induced effects on the body.
Collapse
Affiliation(s)
- Yuxiao Tang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Dongyao Wang
- School of Pharmacy, Second Military Medical University, Shanghai, China.,Faculty of Pharmacy, Shanghai University, Shanghai, China
| | - Hongwei Zhang
- Department of Nutrition, Second Military Medical University, Shanghai, China.,Department of Clinical Nutrition, Zhumadian Second People's Hospital, Henan, China
| | - Yinyin Zhang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Jie Wang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Ruirui Qi
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Jianxin Yang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Hui Shen
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Yan Xu
- Institute of International Medical Science and Technology, Sanda University, Shanghai, China
| | - Min Li
- Department of Nutrition, Second Military Medical University, Shanghai, China.,Institute of International Medical Science and Technology, Sanda University, Shanghai, China
| |
Collapse
|
8
|
Tang Y, Wang D, Niu X, Wu H, Yang J, Zhang Y, Song S, Lv D, Chai Y, Lu H, Shen H, Ling C, Li M. Mild iron overload induces TRIP12-mediated degradation of YY1 to trigger hepatic inflammation. Free Radic Biol Med 2020; 161:187-197. [PMID: 33080340 DOI: 10.1016/j.freeradbiomed.2020.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022]
Abstract
Increasing populations are found to bear mild hepatic iron overload (HIO) due to unhealthy lifestyles, metabolic diseases, etc., whether this mild but chronic HIO induces hepatic inflammation is unknown. In the present study, mice receiving a 12-months 0.3% dextran-iron diet show mild HIO with no detectable oxidative damages in the liver but have infiltrated macrophages and increased IL-6, TNFα, AST and ALT since 6-months. The HNF4α/miR-122/CCL2 pathway, identified by our previous studies to induce macrophages infiltration, is initiated by chronic mild HIO. After excluding the role of DNA methylation, a modified transcription factor microarray is applied to find that transcription factor YY1 is responsible for HIO-decreased HNF4α expression. Then the E3 ubiquitin ligase TRIP12 is identified by an immunoprecipitation coupled LC-MS/MS and proved to bind and ubiquitinate YY1, leading to its degradation. The overexpression or silence of YY1 in the liver regulates the HNF4α/miR-122/CCL2 pathway. More importantly, YY1 overexpression alleviates chronic mild HIO induced hepatic inflammatory responses. In conclusion, these results elucidate an oxidative-stress-independent, TRIP12/YY1/HNF4α/miR-122/CCL2 pathway of chronic mild HIO inducing hepatic inflammation, implying that effective measures in addition to antioxidants are needed for individuals at the risk of chronic mild HIO.
Collapse
Affiliation(s)
- Yuxiao Tang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Dongyao Wang
- School of Pharmacy, Second Military Medical University, Shanghai, China; Faculty of Pharmacy, Shanghai University, Shanghai, China
| | - Xiaowen Niu
- Shanghai Dermatology Hospital, Tongji University, Shanghai, China
| | - Huiwen Wu
- Department of Nutrition, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianxin Yang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Yinyin Zhang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Shangjin Song
- School of Traditional Chinese Medicine & Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Diya Lv
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Hongtao Lu
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Hui Shen
- Department of Nutrition, Second Military Medical University, Shanghai, China.
| | - Chen Ling
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China; Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.
| | - Min Li
- Department of Nutrition, Second Military Medical University, Shanghai, China; Institute of International Medical Science and Technology, Sanda University, Shanghai, China.
| |
Collapse
|
9
|
Li H, Wang D, Wu H, Shen H, Lv D, Zhang Y, Lu H, Yang J, Tang Y, Li M. SLC46A1 contributes to hepatic iron metabolism by importing heme in hepatocytes. Metabolism 2020; 110:154306. [PMID: 32621820 DOI: 10.1016/j.metabol.2020.154306] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Iron is finely regulated due to its vital roles in organisms and the peroxidase reactivity if excess. Solute Carrier Family 46 Member 1 (SLC46A1), also named PCFT or HCP1, is the main importer of heme‑iron in the intestine, but has a high abundance in the liver. Since the liver has a central role in iron homeostasis, whether SLC46A1 regulates hepatic iron metabolism is of interest to be identified. METHODS The recombinant adeno-associated virus vectors were used to hepatic-specifically inhibit SLC46A1 expression to observe its effects on hepatic iron metabolism. Then the abilities of SLC46A1 in importing heme and folate, and consequent alterations of iron content in hepatocytes were determined. Furthermore, effects of iron on SLC46A1 expression were investigated both in vitro and in vivo. RESULTS The hepatocyte-specific inhibition of SLC46A1 decreases iron content in the liver and increases iron content in serum. Expressions of iron-related molecules, transferrin receptor 1, hepcidin and ferroportin, are correspondingly altered. Interestingly, free heme concentration in serum is increased, indicating a decreased import of heme by the liver. In hepatocytes, SLC46A1 is capable of importing hemin, increasing intracellular iron content. The import of hemin by SLC46A1 is unaffected by its other substrate, folate. Instead, hemin treatment decreases SLC46A1 expression, reducing the import of folate. In addition, SLC46A1 itself shows to be iron-responsive both in vivo and in vitro, making it available for regulating iron metabolism. CONCLUSION The results elucidate that SLC46A1 regulates iron metabolism in the liver through a folate-independent manner of importing heme. The iron-responsive characters of SLC46A1 give us a new clue to link heme or iron overload with folate deficiency diseases.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Dongyao Wang
- School of Pharmacy, Second Military Medical University, Shanghai, China; Faculty of Pharmacy, Shanghai University, Shanghai, China
| | - Huiwen Wu
- Department of Nutrition, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Shen
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Diya Lv
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yinyin Zhang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Hongtao Lu
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Jianxin Yang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Yuxiao Tang
- Department of Nutrition, Second Military Medical University, Shanghai, China.
| | - Min Li
- Department of Nutrition, Second Military Medical University, Shanghai, China; Institute of International Medical Science and Technology, Sanda University, Shanghai, China.
| |
Collapse
|
10
|
KLF4 Exerts Sedative Effects in Pentobarbital-Treated Mice. J Mol Neurosci 2020; 71:596-606. [PMID: 32789565 DOI: 10.1007/s12031-020-01680-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/05/2020] [Indexed: 10/23/2022]
Abstract
KLF4 is a zinc-finger transcription factor that plays an essential role in many biological processes, including neuroinflammation, neuron regeneration, cell proliferation, and apoptosis. Through effects on these processes, KLF4 has likely roles in Alzheimer's disease, Parkinson's disease, and traumatic brain injury. However, little is known about the role of KLF4 in more immediate behavioral processes that similarly depend upon broad changes in brain excitability, such as the sleep process. Here, behavioral approaches, western blot, and immunohistochemical experiments were used to explore the role of KLF4 on sedation and the potential mechanisms of those effects. The results showed that overexpression of KLF4 prolonged loss of righting reflex (LORR) duration in pentobarbital-treated mice and increased c-Fos expression in the lateral hypothalamus (LH) and the ventrolateral preoptic nucleus (VLPO), while it decreased c-Fos expression in the tuberomammillary nucleus (TMN). Moreover, overexpression of KLF4 reduced the expression of p53 in the hypothalamus and increased the expression of STAT3 in the hypothalamus. Therefore, these results suggest that KLF4 exerts sedative effects through the regulation of p53 and STAT3 expression, and it indicates a role of KLF4 ligands in the treatment of sleep disorders.
Collapse
|
11
|
Cheng Z, Zou X, Jin Y, Gao S, Lv J, Li B, Cui R. The Role of KLF 4 in Alzheimer's Disease. Front Cell Neurosci 2018; 12:325. [PMID: 30297986 PMCID: PMC6160590 DOI: 10.3389/fncel.2018.00325] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/07/2018] [Indexed: 01/30/2023] Open
Abstract
Krüppel-like factor 4 (KLF4), a member of the family of zinc-finger transcription factors, is widely expressed in range of tissues that play multiple functions. Emerging evidence suggest KLF4’s critical regulatory effect on the neurophysiological and neuropathological processes of Alzheimer’s disease (AD), indicating that KLF4 might be a potential therapeutic target of neurodegenerative diseases. In this review, we will summarize relevant studies and illuminate the regulatory role of KLF4 in the neuroinflammation, neuronal apoptosis, axon regeneration and iron accumulation to clarify KLF4’s status in the pathogenesis of AD.
Collapse
Affiliation(s)
- Ziqian Cheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Xiaohan Zou
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Yang Jin
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Shuohui Gao
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiayin Lv
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Neurobehavioral and biochemical modulation following administration of MgO and ZnO nanoparticles in the presence and absence of acute stress. Life Sci 2018; 203:72-82. [DOI: 10.1016/j.lfs.2018.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/12/2018] [Accepted: 04/15/2018] [Indexed: 12/11/2022]
|