1
|
Muench NA, Schmitt HM, Schlamp CL, Su AJA, Washington K, Nickells RW. Preservation of Murine Whole Eyes With Supplemented UW Cold Storage Solution: Anatomical Considerations. Transl Vis Sci Technol 2024; 13:24. [PMID: 39560629 DOI: 10.1167/tvst.13.11.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024] Open
Abstract
Purpose Retinal ganglion cell (RGC) apoptosis and axon regeneration are the principal obstacles challenging the development of successful whole eye transplantation (WET). The purpose of this study was to create a neuroprotective cocktail that targets early events in the RGC intrinsic apoptotic program to stabilize RGCs in a potential donor eye. Methods University of Wisconsin (UW) solution was augmented with supplements known to protect RGCs. Supplements targeted tyrosine kinase signaling, histone deacetylase activity, K+ ion efflux, macroglial stasis, and provided energy support. Modified UW (mUW) solutions with individual supplements were injected into the vitreous of enucleated mouse eyes, which were then stored in cold UW solution for 24 hours. Histopathology, immunostaining of individual retinal cell types, and analysis of cell-specific messenger RNAs (mRNAs) were used to identify supplements that were combined to create optimal mUW solution. Results UW and mUW solutions reduced ocular edema and focal ischemia in globes stored in cold storage. Two major issues were noted after cold storage, including retinal detachment and reduction in glial fibrillary acidic protein staining in astrocytes. A combination of supplements resolved both these issues and performed better than the individual supplements alone. Cold storage resulted in a reduction in cell-specific mRNAs, even though it preserved the corresponding protein products. Conclusions Eyes treated with optimal mUW solution exhibited preservation of retinal and cellular architecture, but did display a decrease in mRNA levels, suggesting that cold storage induced cellular stasis. Translational Relevance Application of optimal mUW solution lowers an important barrier to the development of a successful whole eye transplantation procedure.
Collapse
Affiliation(s)
- Nicole A Muench
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Heather M Schmitt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Perfuse Therapeutics Inc., Durham, NC, USA
| | - Cassandra L Schlamp
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - An-Jey A Su
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kia Washington
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert W Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- https://orcid.org/0000-0002-2998-5494
| |
Collapse
|
2
|
Ghosh K, Huang Y, Chen SR, Pan HL. Nerve injury augments Cacna2d1 transcription via CK2-mediated phosphorylation of the histone deacetylase HDAC2 in dorsal root ganglia. J Biol Chem 2024; 300:107848. [PMID: 39357831 PMCID: PMC11555424 DOI: 10.1016/j.jbc.2024.107848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
The development of chronic neuropathic pain involves complex synaptic and epigenetic mechanisms. Nerve injury causes sustained upregulation of α2δ-1 (encoded by the Cacna2d1 gene) in the dorsal root ganglion (DRG), contributing to pain hypersensitivity by directly interacting with and augmenting presynaptic NMDA receptor activity in the spinal dorsal horn. Under normal conditions, histone deacetylase 2 (HDAC2) is highly enriched at the Cacna2d1 gene promoter in the DRG, which constitutively suppresses Cacna2d1 transcription. However, nerve injury leads to HDAC2 dissociation from the Cacna2d1 promoter, promoting the enrichment of active histone marks and Cacna2d1 transcription in primary sensory neurons. In this study, we determined the mechanism by which nerve injury diminishes HDAC2 occupancy at the Cacna2d1 promoter in the DRG. Spinal nerve injury in rats increased serine-394 phosphorylation of HDAC2 in the DRG. Coimmunoprecipitation showed that nerve injury enhanced the physical interaction between HDAC2 and casein kinase II (CK2) in the DRG. Furthermore, repeated intrathecal treatment with CX-4945, a potent and specific CK2 inhibitor, markedly reversed nerve injury-induced pain hypersensitivity, HDAC2 phosphorylation, and α2δ-1 expression levels in the DRG. In addition, treatment with CX-4945 largely restored HDAC2 enrichment at the Cacna2d1 promoter and reduced the elevated levels of acetylated H3 and H4 histones, particularly H3K9ac and H4K5ac, at the Cacna2d1 promoter in the injured DRG. These findings suggest that nerve injury increases CK2 activity and CK2-HDAC2 interactions, which enhance HDAC2 phosphorylation in the DRG. This, in turn, diminishes HDAC2 enrichment at the Cacna2d1 promoter, thereby promoting Cacna2d1 transcription.
Collapse
Affiliation(s)
- Krishna Ghosh
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuying Huang
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
3
|
Yamashita S, Okamoto M, Mendonca M, Fujiwara N, Kitamura E, Chang CSS, Brueckner S, Shindo S, Kuriki N, Cooley MA, Gill Dhillon N, Kawai T, Bartlett JD, Everett ET, Suzuki M. Fluoride Alters Gene Expression via Histone H3K27 Acetylation in Ameloblast-like LS8 Cells. Int J Mol Sci 2024; 25:9600. [PMID: 39273544 PMCID: PMC11395493 DOI: 10.3390/ijms25179600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Excessive fluoride ingestion during tooth development can cause dental fluorosis. Previously, we reported that fluoride activates histone acetyltransferase (HAT) to acetylate p53, promoting fluoride toxicity in mouse ameloblast-like LS8 cells. However, the roles of HAT and histone acetylation status in fluoride-mediated gene expression remain unidentified. Here, we demonstrate that fluoride-mediated histone modification causes gene expression alterations in LS8 cells. LS8 cells were treated with or without fluoride followed by ChIP-Seq analysis of H3K27ac. Genes were identified by differential H3K27ac peaks within ±1 kb from transcription start sites. The levels of mRNA of identified genes were assessed using rea-time PCR (qPCR). Fluoride increased H3K27ac peaks associated with Bax, p21, and Mdm2 genes and upregulated their mRNA levels. Fluoride decreased H3K27ac peaks and p53, Bad, and Bcl2 had suppressed transcription. HAT inhibitors (Anacardic acid or MG149) suppressed fluoride-induced mRNA of p21 and Mdm2, while fluoride and the histone deacetylase (HDAC) inhibitor sodium butyrate increased Bad and Bcl2 expression above that of fluoride treatment alone. To our knowledge, this is the first study that demonstrates epigenetic regulation via fluoride treatment via H3 acetylation. Further investigation is required to elucidate epigenetic mechanisms of fluoride toxicity in enamel development.
Collapse
Affiliation(s)
- Shohei Yamashita
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.Y.); (M.O.); (M.M.); (S.B.); (S.S.); (N.K.); (T.K.)
| | - Motoki Okamoto
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.Y.); (M.O.); (M.M.); (S.B.); (S.S.); (N.K.); (T.K.)
| | - Melanie Mendonca
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.Y.); (M.O.); (M.M.); (S.B.); (S.S.); (N.K.); (T.K.)
- Biology I Halmos College of Arts and Sciences, Behavioral Neuroscience I College of Psychology, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Natsumi Fujiwara
- Department of Oral Health Care Management, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto, Tokushima 770-8504, Japan;
| | - Eiko Kitamura
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (E.K.)
| | | | - Susanne Brueckner
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.Y.); (M.O.); (M.M.); (S.B.); (S.S.); (N.K.); (T.K.)
| | - Satoru Shindo
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.Y.); (M.O.); (M.M.); (S.B.); (S.S.); (N.K.); (T.K.)
| | - Nanako Kuriki
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.Y.); (M.O.); (M.M.); (S.B.); (S.S.); (N.K.); (T.K.)
| | - Marion A. Cooley
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Navi Gill Dhillon
- Department of Biological Sciences, Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
| | - Toshihisa Kawai
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.Y.); (M.O.); (M.M.); (S.B.); (S.S.); (N.K.); (T.K.)
| | - John D. Bartlett
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH 43210, USA;
| | - Eric T. Everett
- Department of Biomedical Sciences, Adams School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Maiko Suzuki
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (S.Y.); (M.O.); (M.M.); (S.B.); (S.S.); (N.K.); (T.K.)
| |
Collapse
|
4
|
Pereira M, Cruz MT, Fortuna A, Bicker J. Restoring the epigenome in Alzheimer's disease: advancing HDAC inhibitors as therapeutic agents. Drug Discov Today 2024; 29:104052. [PMID: 38830501 DOI: 10.1016/j.drudis.2024.104052] [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: 02/05/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Current treatment options for Alzheimer's disease (AD) focus on symptom relief rather than halting disease progression. In this context, targeting histone deacetylation emerges as a promising therapeutic alternative. Dysregulation of histone deacetylase (HDAC) activity is present in AD, contributing to cognitive decline. Pharmacological HDAC inhibition has shown benefits in preclinical models, namely reduced amyloid beta plaque formation, lower phosphorylation and aggregation of tau protein, greater microtubule stability, less neuroinflammation, and improved metabolic homeostasis and cell survival. Nonetheless, clinical trials evidenced limitations such as insufficient selectivity or blood-brain barrier penetration. Hence, future innovative strategies are required to enhance their efficacy/safety.
Collapse
Affiliation(s)
- Márcia Pereira
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - Maria Teresa Cruz
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Center for Neuroscience and Cell Biology, 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
|
5
|
Liu QQ, Wu GH, Wang XC, Xiong XW, Rui-Wang, Yao BL. The role of Foxo3a in neuron-mediated cognitive impairment. Front Mol Neurosci 2024; 17:1424561. [PMID: 38962803 PMCID: PMC11220205 DOI: 10.3389/fnmol.2024.1424561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024] Open
Abstract
Cognitive impairment (COI) is a prevalent complication across a spectrum of brain disorders, underpinned by intricate mechanisms yet to be fully elucidated. Neurons, the principal cell population of the nervous system, orchestrate cognitive processes and govern cognitive balance. Extensive inquiry has spotlighted the involvement of Foxo3a in COI. The regulatory cascade of Foxo3a transactivation implicates multiple downstream signaling pathways encompassing mitochondrial function, oxidative stress, autophagy, and apoptosis, collectively affecting neuronal activity. Notably, the expression and activity profile of neuronal Foxo3a are subject to modulation via various modalities, including methylation of promoter, phosphorylation and acetylation of protein. Furthermore, upstream pathways such as PI3K/AKT, the SIRT family, and diverse micro-RNAs intricately interface with Foxo3a, engendering alterations in neuronal function. Through several downstream routes, Foxo3a regulates neuronal dynamics, thereby modulating the onset or amelioration of COI in Alzheimer's disease, stroke, ischemic brain injury, Parkinson's disease, and traumatic brain injury. Foxo3a is a potential therapeutic cognitive target, and clinical drugs or multiple small molecules have been preliminarily shown to have cognitive-enhancing effects that indirectly affect Foxo3a. Particularly noteworthy are multiple randomized, controlled, placebo clinical trials illustrating the significant cognitive enhancement achievable through autophagy modulation. Here, we discussed the role of Foxo3a in neuron-mediated COI and common cognitively impaired diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Bao-Le Yao
- Department of Rehabilitation Medicine, Ganzhou People’s Hospital, Ganzhou, China
| |
Collapse
|
6
|
Kong Q, Li F, Sun K, Sun X, Ma J. Valproic acid ameliorates cauda equina injury by suppressing HDAC2-mediated ferroptosis. CNS Neurosci Ther 2024; 30:e14524. [PMID: 38105511 PMCID: PMC11017456 DOI: 10.1111/cns.14524] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/24/2023] [Accepted: 10/29/2023] [Indexed: 12/19/2023] Open
Abstract
INTRODUCTION Persistent neuroinflammatory response after cauda equina injury (CEI) lowers nociceptor firing thresholds, accompanied by pathological pain and decreasing extremity dysfunction. Histone deacetylation has been considered a key regulator of immunity, inflammation, and neurological dysfunction. Our previous study suggested that valproic acid (VPA), a histone deacetylase inhibitor, exhibited neuroprotective effects in rat models of CEI, although the underlying mechanism remains elusive. METHODS The cauda equina compression surgery was performed to establish the CEI model. The Basso, Beattie, Bresnahan score, and the von Frey filament test were carried out to measure the animal behavior. Immunofluorescence staining of myelin basic protein and GPX4 was carried out. In addition, transmission electron microscope analysis was used to assess the effect of VPA on the morphological changes of mitochondria. RNA-sequencing was conducted to clarify the underlying mechanism of VPA on CEI protection. RESULTS In this current study, we revealed that the expression level of HDAC1 and HDAC2 was elevated after cauda equina compression model but was reversed by VPA treatment. Meanwhile, HDAC2 knockdown resulted in the improvement of motor functions and pathologic pain, similar to treatment with VPA. Histology analysis also showed that knockdown of histone deacetylase (HDAC)-2, but not HDAC1, remarkably alleviated cauda equina injury and demyelinating lesions. The potential mechanism may be related to lowering oxidative stress and inflammatory response in the injured region. Notably, the transcriptome sequencing indicated that the therapeutic effect of VPA may depend on HDAC2-mediated ferroptosis. Ferroptosis-related genes were analyzed in vivo and DRG cells further validated the reliability of RNA-sequencing results, suggesting HDAC2-H4K12ac axis participated in epigenetic modulation of ferroptosis-related genes. CONCLUSION HDAC2 is critically involved in the ferroptosis and neuroinflammation in cauda equina injury, and VPA ameliorated cauda equina injury by suppressing HDAC2-mediated ferroptosis.
Collapse
Affiliation(s)
- Qingjie Kong
- Department of OrthopedicsShanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Key Laboratory of Medical Immunology & Institute of ImmunologySecond Military Medical UniversityShanghaiChina
| | - Fudong Li
- Department of Orthopedic SurgerySpine Center, Shanghai Changzheng Hospital, Second Military Medical UniversityShanghaiChina
| | - Kaiqiang Sun
- Department of Orthopedic SurgerySpine Center, Shanghai Changzheng Hospital, Second Military Medical UniversityShanghaiChina
| | - Xiaofei Sun
- Department of Orthopedic SurgerySpine Center, Shanghai Changzheng Hospital, Second Military Medical UniversityShanghaiChina
| | - Jun Ma
- Department of OrthopedicsShanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
7
|
Hosseini SA, Ghatrehsamani M, Yaghoobi H, Elahian F, Mirzaei SA. Epigenetic disruption of histone deacetylase-2 accelerated apoptotic signaling and retarded malignancy in gastric cells. Epigenomics 2024; 16:277-292. [PMID: 38356395 DOI: 10.2217/epi-2023-0350] [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] [Indexed: 02/16/2024] Open
Abstract
Background: The objective of this research was to determine whether HDAC2 function is associated with gastric cancer progression. Methods: HDAC2 was knocked out in EPG85.257 cells using CRISPR/Cas9 and tumorigenesis pathways were evaluated. Results: Cell proliferation, colony formation, wound healing and transwell invasion were inhibited in ΔHDAC2:EPG85.257 cells. Quantitative analyses revealed a significant downregulation of MMP1, p53, Bax, MAPK1, MAPK3, pro-Caspase3, ERK1/2, p-ERK1/2, AKT1/2/3, p-AKT1/2/3, p-NF-κB (p65), Twist, Snail and p-FAK transcripts/proteins, while SIRT1, PTEN, p21 and Caspase3 were upregulated in ΔHDAC2:EPG85.257 cells. Conclusion: These results indicated that HDAC2 enhanced migration, colony formation and transmigration ability. HDAC2 inhibition may improve gastric cancer chemotherapy pathways.
Collapse
Affiliation(s)
- Sayedeh Azimeh Hosseini
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahdi Ghatrehsamani
- Cellular & Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hajar Yaghoobi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Elahian
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Abbas Mirzaei
- Cellular & Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
8
|
Wu Z, Qu J, Zhang W, Liu GH. Stress, epigenetics, and aging: Unraveling the intricate crosstalk. Mol Cell 2024; 84:34-54. [PMID: 37963471 DOI: 10.1016/j.molcel.2023.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023]
Abstract
Aging, as a complex process involving multiple cellular and molecular pathways, is known to be exacerbated by various stresses. Because responses to these stresses, such as oxidative stress and genotoxic stress, are known to interplay with the epigenome and thereby contribute to the development of age-related diseases, investigations into how such epigenetic mechanisms alter gene expression and maintenance of cellular homeostasis is an active research area. In this review, we highlight recent studies investigating the intricate relationship between stress and aging, including its underlying epigenetic basis; describe different types of stresses that originate from both internal and external stimuli; and discuss potential interventions aimed at alleviating stress and restoring epigenetic patterns to combat aging or age-related diseases. Additionally, we address the challenges currently limiting advancement in this burgeoning field.
Collapse
Affiliation(s)
- Zeming Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; China National Center for Bioinformation, Beijing 100101, China; CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; The Fifth People's Hospital of Chongqing, Chongqing 400062, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
9
|
Role of FOXO3a Transcription Factor in the Regulation of Liver Oxidative Injury. Antioxidants (Basel) 2022; 11:antiox11122478. [PMID: 36552685 PMCID: PMC9774119 DOI: 10.3390/antiox11122478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress has been identified as a key mechanism in liver damage caused by various chemicals. The transcription factor FOXO3a has emerged as a critical regulator of redox imbalance. Multiple post-translational changes and epigenetic processes closely regulate the activity of FOXO3a, resulting in synergistic or competing impacts on its subcellular localization, stability, protein-protein interactions, DNA binding affinity, and transcriptional programs. Depending on the chemical nature and subcellular context, the oxidative-stress-mediated activation of FOXO3a can induce multiple transcriptional programs that play crucial roles in oxidative injury to the liver by chemicals. Here, we mainly review the role of FOXO3a in coordinating programs of genes that are essential for cellular homeostasis, with an emphasis on exploring the regulatory mechanisms and potential application of FOXO3a as a therapeutic target to prevent and treat liver oxidative injury.
Collapse
|
10
|
Zhang J, Chen SR, Zhou MH, Jin D, Chen H, Wang L, DePinho RA, Pan HL. HDAC2 in Primary Sensory Neurons Constitutively Restrains Chronic Pain by Repressing α2δ-1 Expression and Associated NMDA Receptor Activity. J Neurosci 2022; 42:8918-8935. [PMID: 36257688 PMCID: PMC9732832 DOI: 10.1523/jneurosci.0735-22.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 01/05/2023] Open
Abstract
α2δ-1 (encoded by the Cacna2d1 gene) is a newly discovered NMDA receptor-interacting protein and is the therapeutic target of gabapentinoids (e.g., gabapentin and pregabalin) frequently used for treating patients with neuropathic pain. Nerve injury causes sustained α2δ-1 upregulation in the dorsal root ganglion (DRG), which promotes NMDA receptor synaptic trafficking and activation in the spinal dorsal horn, a hallmark of chronic neuropathic pain. However, little is known about how nerve injury initiates and maintains the high expression level of α2δ-1 to sustain chronic pain. Here, we show that nerve injury caused histone hyperacetylation and diminished enrichment of histone deacetylase-2 (HDAC2), but not HDAC3, at the Cacna2d1 promoter in the DRG. Strikingly, Hdac2 knockdown or conditional knockout in DRG neurons in male and female mice consistently induced long-lasting mechanical pain hypersensitivity, which was readily reversed by blocking NMDA receptors, inhibiting α2δ-1 with gabapentin or disrupting the α2δ-1-NMDA receptor interaction at the spinal cord level. Hdac2 deletion in DRG neurons increased histone acetylation levels at the Cacna2d1 promoter, upregulated α2δ-1 in the DRG, and potentiated α2δ-1-dependent NMDA receptor activity at primary afferent central terminals in the spinal dorsal horn. Correspondingly, Hdac2 knockdown-induced pain hypersensitivity was blunted in Cacna2d1 knockout mice. Thus, our findings reveal that HDAC2 functions as a pivotal transcriptional repressor of neuropathic pain via constitutively suppressing α2δ-1 expression and ensuing presynaptic NMDA receptor activity in the spinal cord. HDAC2 enrichment levels at the Cacna2d1 promoter in DRG neurons constitute a unique epigenetic mechanism that governs acute-to-chronic pain transition.SIGNIFICANCE STATEMENT Excess α2δ-1 proteins produced after nerve injury directly interact with glutamate NMDA receptors to potentiate synaptic NMDA receptor activity in the spinal cord, a prominent mechanism of nerve pain. Because α2δ-1 upregulation after nerve injury is long lasting, gabapentinoids relieve pain symptoms only temporarily. Our study demonstrates for the first time the unexpected role of intrinsic HDAC2 activity at the α2δ-1 gene promoter in limiting α2δ-1 gene transcription, NMDA receptor-dependent synaptic plasticity, and chronic pain development after nerve injury. These findings challenge the prevailing view about the role of general HDAC activity in promoting chronic pain. Restoring the repressive HDAC2 function and/or reducing histone acetylation at the α2δ-1 gene promoter in primary sensory neurons could lead to long-lasting relief of nerve pain.
Collapse
Affiliation(s)
- Jixiang Zhang
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Meng-Hua Zhou
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Daozhong Jin
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hong Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Li Wang
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
11
|
Zhao Y, Guo Y, Sun M, Hussion S, Zheng Y, Huang H, Huo X, Zhao Y, Zhang F, Han Y, Ning Q, Xu P, Sun J, Lu S. Selenium-sensitive histone deacetylase 2 is required for forkhead box O3A and regulates extracellular matrix metabolism in cartilage. J Bone Miner Metab 2022; 40:914-926. [PMID: 36156740 DOI: 10.1007/s00774-022-01369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/24/2022] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Selenium (Se) as well as selenoproteins are vital for osteochondral system development. Se deficiency (SeD) has a definite impact on the expression and activity of histone deacetylases (HDACs). Abnormal expression of some HDACs affects cartilage development. This current study aims to explore the relationship between differentially expressed HDACs and cartilage development, especially extracellular matrix (ECM) homeostasis maintenance, under SeD conditions. MATERIALS AND METHODS Dark Agouti rats and C28/I2 cell line under SeD states were used to detect the differently expressed HDAC by RT-qPCR, western blotting and IHC staining. Meanwhile, the biological roles of the above HDAC in cartilage development and homeostasis maintenance were confirmed by siRNA transfection, western blotting, RNA sequence and inhibitor treatment experiments. RESULTS HDAC2 exhibited lower expression at protein level in both animals and chondrocytes during SeD condition. The results of cell-level experiments indicated that forkhead box O3A (FOXO3A), which was required to maintain metabolic homeostasis of cartilage matrix, was reduced by HDAC2 knockdown. Meanwhile, induced HDAC2 was positively associated with FOXO3A in rat SeD model. Meanwhile, knockdown of HDAC2 and FOXO3A led to an increase of intracellular ROS level, which activated NF-κB pathway. Se supplementary significantly inhibited the activation of NF-κB pathway with IL-1β treatment. CONCLUSION Our results suggested that low expression of HDAC2 under SeD condition increased ROS content by decreasing FOXO3A in chondrocytes, which led to the activation of NF-κB pathway and ECM homeostasis imbalance.
Collapse
Affiliation(s)
- Yitong Zhao
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yuanxu Guo
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Mengyao Sun
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Safdar Hussion
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Ying Zheng
- Department of Digestive Disease and Gastrointestinal Motility Research Room, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xi Wu Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Huang Huang
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Xinyu Huo
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yutong Zhao
- Department of Software Engineering, Xinjiang University School of Software, Urumqi, 830000, Xinjiang, People's Republic of China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yan Han
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Qilan Ning
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Honghui Hospital, Easter Youyi Road No. 555, Xi'an, 710054, Shaanxi, People's Republic of China
| | - Jian Sun
- Key Laboratory of Trace Elements and Endemic Diseases, Xi'an Jiaotong University School of Public Health, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
12
|
DL-3-N-Butylphthalide Promotes Cartilage Extracellular Matrix Synthesis and Inhibits Osteoarthritis Development by Regulating FoxO3a. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9468040. [PMID: 35910845 PMCID: PMC9329036 DOI: 10.1155/2022/9468040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/23/2022] [Indexed: 12/04/2022]
Abstract
Osteoarthritis (OA) has been reported as a progressive disease in the elderly, primarily characterized by degenerated articular cartilage. There has been no satisfactory drug for the treatment of OA. DL-3-n-butylphthalide (NBP), a small molecule compound extracted from celery seeds, may have antiapoptotic, antioxidant, and anti-inflammatory activities in numerous studies. However, the effects of NBP on OA and its mechanisms have been rarely reported. In this study, the effect of NBP on OA in vitro and in vivo and its possible mechanism were investigated. The results showed that NBP injection into the knee joint inhibited osteoarthritis development in a rat model of osteoarthritis induced by DMM+ACLT. NBP could increase the expressions of extracellular matrix-related components (such as type II collagen, aggrecan, proteoglycan 4, and SRY-box 9) in human osteoarthritic chondrocytes and cartilage explants. Moreover, NBP promoted the expressions of SOD and CAT. NBP upregulated the expression of FoxO3a by inhibiting the PI3K/AKT pathway, which subsequently inhibited the apoptosis of human OA chondrocytes. In conclusion, NBP promotes cartilage extracellular matrix synthesis and inhibits osteoarthritis development and the underlying mechanism related to the activation of FoxO3a.
Collapse
|
13
|
Xun T, Lin Z, Zhang M, Mo L, Chen Y, Wang X, Zhao J, Ye C, Feng H, Yang X. Advanced oxidation protein products upregulate ABCB1 expression and activity via HDAC2-Foxo3α-mediated signaling in vitro and in vivo. Toxicol Appl Pharmacol 2022; 449:116140. [PMID: 35753429 DOI: 10.1016/j.taap.2022.116140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 11/28/2022]
Abstract
The unpredictable pharmacokinetics of non-renal cleared drugs in chronic kidney disease (CKD) patients is associated with the activity of drug transporters. However, the mechanisms underlying regulation of drug transporters are yet to be established. In this study, we demonstrated the involvement of a HDAC2-Foxo3α pathway in advanced oxidation protein products (AOPPs)-induced ATP-binding cassette subfamily B member 1 (ABCB1) expression and activity. The correlation of AOPPs accumulation with concentration of cyclosporine in plasma was evaluated in 194 patients with transplantation. Molecular changes in acetylation of various histones and related regulatory molecules were examined in HepG2 cell cultures treated with AOPPs. Accumulation of AOPPs in serum in relation to molecular changes in HDAC2-Foxo3α in vivo were evaluated in 5/6 nephrectomy (5/6 nx) and oral adenine (Adenine) CKD rat models. Interestingly, the cyclosporine level was negatively correlated with AOPPs in plasma. In addition, AOPPs markedly suppressed the expression of histone deacetylase 2 (HDAC2), inducing ABCB1 expression and activity in vitro and in vivo. Importantly, AOPPs modulated phosphorylation of Foxo3α and the upstream Akt protein. Our findings indicate that AOPPs regulate the expression and activity of ABCB1 via reducing HDAC2 expression and activating Foxo3α-dependent signaling. The collective results support the utility of AOPPs as a potential target for drug and/or dosage adjustment in CKD patients. Targeting of AOPPs presents a novel approach to regulate non-renal clearance.
Collapse
Affiliation(s)
- Tianrong Xun
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhufen Lin
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Mimi Zhang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Liqian Mo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Chen
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaokang Wang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jingqian Zhao
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chunxiao Ye
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Haixing Feng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xixiao Yang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China.
| |
Collapse
|
14
|
Lingling D, Miaomiao Q, Yili L, Hongyun H, Yihao D. Attenuation of histone H4 lysine 16 acetylation (H4K16ac) elicits a neuroprotection against ischemic stroke by alleviating the autophagic/lysosomal dysfunction in neurons at the penumbra. Brain Res Bull 2022; 184:24-33. [DOI: 10.1016/j.brainresbull.2022.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/06/2022] [Accepted: 03/24/2022] [Indexed: 11/02/2022]
|
15
|
Goodin BR, Overstreet DS, Penn TM, Bakshi R, Quinn TL, Sims A, Ptacek T, Jackson P, Long DL, Aroke EN. Epigenome-wide DNA methylation profiling of conditioned pain modulation in individuals with non-specific chronic low back pain. Clin Epigenetics 2022; 14:45. [PMID: 35346352 PMCID: PMC8962463 DOI: 10.1186/s13148-022-01265-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 03/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The pathoanatomic cause of chronic low back pain (cLBP) cannot be identified for up to 90% of individuals. However, dysfunctional processing of endogenous nociceptive input, measured as conditioned pain modulation (CPM), has been associated with cLBP and may involve changes in neuronal gene expression. Epigenetic-induced changes such as DNA methylation (DNAm) have been associated with cLBP. METHODS In the present study, the relationship between CPM and DNAm changes in a sample of community-dwelling adults with nonspecific cLBP (n = 48) and pain-free controls (PFC; n = 50) was examined using reduced representation bisulfite sequencing. Gene ontology (GO) term enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were applied to identify key pathways involved in efficient versus deficient CPM. RESULTS Based on CPM efficiency, we identified 6006 and 18,305 differentially methylated CpG sites (DMCs) with q values < 0.01 among individuals with cLBP and PFCs, respectively. Most of the DMCs were hypomethylated and annotated to genes of relevance to pain, including OPRM1, ADRB2, CACNA2D3, GNA12, LPL, NAXD, and ASPHD1. In both cLBP and PFC groups, the DMCs annotated genes enriched many GO terms relevant to pain processing, including transcription regulation by RNA polymerase II, nervous system development, generation of neurons, neuron differentiation, and neurogenesis. Both groups also enriched the pathways involved in Rap1-signaling, cancer, and dopaminergic neurogenesis. However, MAPK-Ras signaling pathways were enriched in the cLBP, not the PFC group. CONCLUSIONS This is the first study to investigate the genome-scale DNA methylation profiles of CPM phenotype in adults with cLBP and PFCs. Based on CPM efficiency, fewer DMC enrichment pathways were unique to the cLBP than the PFCs group. Our results suggest that epigenetically induced modification of neuronal development/differentiation pathways may affect CPM efficiency, suggesting novel potential therapeutic targets for central sensitization. However, CPM efficiency and the experience of nonspecific cLBP may be independent. Further mechanistic studies are required to confirm the relationship between CPM, central sensitization, and nonspecific cLBP.
Collapse
Affiliation(s)
- Burel R Goodin
- Department of Psychology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Addiction and Pain Prevention and Intervention (CAPPI), University of Alabama at Birmingham, Birmingham, AL, USA
| | - Demario S Overstreet
- Department of Psychology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Terence M Penn
- Department of Psychology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rahm Bakshi
- Department of Psychology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tammie L Quinn
- Department of Psychology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew Sims
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Travis Ptacek
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Pamela Jackson
- Department of Acute, Chronic and Continuing Care, School of Nursing, University of Alabama at Birmingham, 1701 University Boulevard, Birmingham, AL, 35294, USA
| | - D Leann Long
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Edwin N Aroke
- Department of Acute, Chronic and Continuing Care, School of Nursing, University of Alabama at Birmingham, 1701 University Boulevard, Birmingham, AL, 35294, USA.
| |
Collapse
|
16
|
Kwak S, Park SH, Kim SH, Sung GJ, Song JH, Jeong JH, Kim H, Ha CH, Kim SW, Choi KC. miR-3189-targeted GLUT3 repression by HDAC2 knockdown inhibits glioblastoma tumorigenesis through regulating glucose metabolism and proliferation. J Exp Clin Cancer Res 2022; 41:87. [PMID: 35260183 PMCID: PMC8903173 DOI: 10.1186/s13046-022-02305-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epigenetic regulations frequently appear in Glioblastoma (GBM) and are highly associated with metabolic alterations. Especially, Histone deacetylases (HDACs) correlates with the regulation of tumorigenesis and cell metabolism in GBM progression, and HDAC inhibitors report to have therapeutic efficacy in GBM and other neurological diseases; however, GBM prevention and therapy by HDAC inhibition lacks a mechanism in the focus of metabolic reprogramming. METHODS HDAC2 highly express in GBM and is analyzed in TCGA/GEPIA databases. Therefore, HDAC2 knockdown affects GBM cell death. Analysis of RNA sequencing and qRT-PCR reveals that miR-3189 increases and GLUT3 decreases by HDAC2 knockdown. GBM tumorigenesis also examines by using in vivo orthotopic xenograft tumor models. The metabolism change in HDAC2 knockdown GBM cells measures by glucose uptake, lactate production, and OCR/ECAR analysis, indicating that HDAC2 knockdown induces GBM cell death by inhibiting GLUT3. RESULTS Notably, GLUT3 was suppressed by increasing miR-3189, demonstrating that miR-3189-mediated GLUT3 inhibition shows an anti-tumorigenic effect and cell death by regulating glucose metabolism in HDAC2 knockdown GBM. CONCLUSIONS Our findings will demonstrate the central role of HDAC2 in GBM tumorigenesis through the reprogramming of glucose metabolism by controlling miR-3189-inhibited GLUT3 expression, providing a potential new therapeutic strategy for GBM treatment.
Collapse
Affiliation(s)
- Sungmin Kwak
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Seung-Ho Park
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Sung-Hak Kim
- Department of Animal Science, Chonnam National University, Gwangju, Republic of Korea
| | - Gi-Jun Sung
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Ji-Hye Song
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.,Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Ji-Hoon Jeong
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Hyunhee Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Chang Hoon Ha
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Seong Who Kim
- Departments of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Kyung-Chul Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
17
|
Kaur D, Behl T, Sehgal A, Singh S, Sharma N, Badavath VN, Ul Hassan SS, Hasan MM, Bhatia S, Al-Harassi A, Khan H, Bungau S. Unravelling the potential neuroprotective facets of erythropoietin for the treatment of Alzheimer's disease. Metab Brain Dis 2022; 37:1-16. [PMID: 34436747 DOI: 10.1007/s11011-021-00820-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023]
Abstract
During the last three decades, recombinant DNA technology has produced a wide range of hematopoietic and neurotrophic growth factors, including erythropoietin (EPO), which has emerged as a promising protein drug in the treatment of several diseases. Cumulative studies have recently indicated the neuroprotective role of EPO in preclinical models of acute and chronic neurodegenerative disorders, including Alzheimer's disease (AD). AD is one of the most prevalent neurodegenerative illnesses in the elderly, characterized by the accumulation of extracellular amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles (NFTs), which serve as the disease's two hallmarks. Unfortunately, AD lacks a successful treatment strategy due to its multifaceted and complex pathology. Various clinical studies, both in vitro and in vivo, have been conducted to identify the various mechanisms by which erythropoietin exerts its neuroprotective effects. The results of clinical trials in patients with AD are also promising. Herein, it is summarized and reviews all such studies demonstrating erythropoietin's potential therapeutic benefits as a pleiotropic neuroprotective agent in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Syed Shams Ul Hassan
- School of Medicine and Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mohammad Mehedi Hasan
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
- Amity Institute of Pharmacy, Amity University, Noida, Haryana, India
| | - Ahmed Al-Harassi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
18
|
Epigenetic Alterations in Pediatric Sleep Apnea. Int J Mol Sci 2021; 22:ijms22179523. [PMID: 34502428 PMCID: PMC8430725 DOI: 10.3390/ijms22179523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/03/2022] Open
Abstract
Pediatric obstructive sleep apnea has significant negative effects on health and behavior in childhood including depression, failure to thrive, neurocognitive impairment, and behavioral issues. It is strongly associated with an increased risk for chronic adult disease such as obesity and diabetes, accelerated atherosclerosis, and endothelial dysfunction. Accumulating evidence suggests that adult-onset non-communicable diseases may originate from early life through a process by which an insult applied at a critical developmental window causes long-term effects on the structure or function of an organism. In recent years, there has been increased interest in the role of epigenetic mechanisms in the pathogenesis of adult disease susceptibility. Epigenetic mechanisms that influence adaptive variability include histone modifications, non-coding RNAs, and DNA methylation. This review will highlight what is currently known about the phenotypic associations of epigenetic modifications in pediatric obstructive sleep apnea and will emphasize the importance of epigenetic changes as both modulators of chronic disease and potential therapeutic targets.
Collapse
|
19
|
Qu Y, Xu Y, Jiang Y, Yu D, Jiang X, Zhao L. Macrophage-derived extracellular vesicles regulates USP5-mediated HDAC2/NRF2 axis to ameliorate inflammatory pain. FASEB J 2021; 35:e21332. [PMID: 34423867 DOI: 10.1096/fj.202001185rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/11/2022]
Abstract
Emerging research has highlighted the capacity of microRNA-23a-3p (miR-23a-3p) to alleviate inflammatory pain. However, the molecular mechanism by which miR-23a-3p attenuates inflammatory pain is yet to be fully understood. Hence, the current study aimed to elucidate the mechanism by which miR-23a-3p influences inflammatory pain. Bioinformatics was initially performed to predict the inflammatory pain related downstream targets of miR-23a-3p in macrophage-derived extracellular vesicles (EVs). An animal inflammatory pain model was established using Complete Freund's Adjuvant (CFA). The miR-23a-3p expression was downregulated in the microglia of CFA-induced mice, after which the inflammatory factors were determined by ELISA. FISH and immunofluorescence were performed to analyze the co-localization of miR-23a-3p and microglia. Interestingly, miR-23a-3p was transported to the microglia via M2 macrophage-EVs, which elevated the mechanical allodynia and the thermal hyperalgesia thresholds in mice model. The miR-23a-3p downstream target, USP5, was found to stabilize HDAC2 via deubiquitination to promote its expression while inhibiting the expression of NRF2. Taken together, the key findings of the current study demonstrate that macrophage-derived EVs containing miR-23a-3p regulates the HDAC2/NRF2 axis by decreasing USP5 expression to alleviate inflammatory pain, which may provide novel therapeutic targets for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Yao Qu
- Department of Pain Management, The First Hospital of Jilin University, Changchun, P.R. China
| | - Yunhe Xu
- Department of Stomatology, The First Hospital of Jilin University, Changchun, P.R. China
| | - Yuncheng Jiang
- Department of Anesthesiology, Dehui People's Hospital, Dehui, P.R. China
| | - Dehai Yu
- The Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, P.R. China
| | - Xi Jiang
- Health Promotion Center, The First Hospital of Jilin University, Changchun, P.R. China
| | - Ling Zhao
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
20
|
Zou D, Mou Z, Wu W, Liu H. TRIM33 protects osteoblasts from oxidative stress-induced apoptosis in osteoporosis by inhibiting FOXO3a ubiquitylation and degradation. Aging Cell 2021; 20:e13367. [PMID: 34101965 PMCID: PMC8282270 DOI: 10.1111/acel.13367] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/04/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
This study aimed to probe into the effect of TRIM33 on oxidative stress‐induced apoptosis of osteoblasts in osteoporosis and to probe into the underlying mechanism. The apoptosis of osteoblasts was induced by H2O2 treatment and tested by flow cytometry. A mouse osteoporosis model was conducted by ovariectomy (OVX). The function of TRIM33 was assessed by in vitro and in vivo experiments. The mechanism of TRIM33 was determined by immunoprecipitation, immunofluorescent staining and co‐transfection experiments. Here, we found that TRIM33 expression was lessened in the osteoblasts of patients with osteoporosis and was positively correlated with the bone mineral density of these patients. FOXO3a and TRIM33 were co‐localized in the osteoblasts nuclei. TRIM33 silence boosted FOXO3a degradation in normal osteoblasts, while TRIM33 overexpression restrained FOXO3a degradation in H2O2‐treated osteoblasts. The binding of TRIM33 to CBP and its overexpression restrained CBP‐mediated FOXO3a acetylation, thereby attenuating FOXO3a ubiquitylation. The H2O2‐induced apoptosis of osteoblasts was restrained by TRIM33 overexpression, while the FOXO3a knockdown reversed this trend. The in vivo experiments corroborated that TRIM33 overexpression attenuated the OVX‐driven impacts in mice. In general, our findings expounded that TRIM33 protected osteoblasts against oxidative stress‐induced apoptosis in osteoporosis and that the underlying mechanism was the restraint of FOXO3a ubiquitylation and degradation.
Collapse
Affiliation(s)
- De‐bo Zou
- Department of Orthopaedics The First Affiliated Hospital of Shandong First Medical University Jinan, Shandong China
| | - Zongyou Mou
- Department of Orthopedics Dezhou People’s Hospital Dezhou, Shandong China
| | - Wenliang Wu
- Department of Orthopedics Qilu Hospital of Shandong University Jinan, Shandong China
| | - Haichun Liu
- Department of Orthopedics Qilu Hospital of Shandong University Jinan, Shandong China
| |
Collapse
|
21
|
Sun J, Wang Q, Yuan Y, Hussain S, Zhao Y, Guo Y, Sun M, Huang H, Huo X, Zhang F, Ning Q, Han Y, Xu P, Lu S. Identification of a cartilage specific novel miRNA which directly targets PRMT3 in rats. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100161. [PMID: 36474992 PMCID: PMC9718250 DOI: 10.1016/j.ocarto.2021.100161] [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: 12/17/2020] [Accepted: 04/01/2021] [Indexed: 11/23/2022] Open
Abstract
UNLABELLED Through experiments to testify a candidate novel miRNA previously discovered by us is a real miRNA and involved in cartilage development. DESIGN The miR-novel and the newly hairpin miRNA transcribed sequence (pre-miR-novel) was verified as a genuinely existing miRNA by northern blotting. The predicted secondary structure, sequence alignment and targets of pre-miR-novel were performed by "RNAstructure 5.3" program, LASTN2.8.0+/miRbase22 program and RNA hybird program, respective. GO/KEGG pathway analysis also were performed. The miR-novel expression in cartilage tissue during development was detected by RT-qPCR and dot blotting. The chondrocyte differentiation model was established to examine whether miR-novel is involved in cartilage development. The regulation of PRMT3 expression by novel miRNA was determined with the luciferase reporter gene assay and Western blotting after novel miRNA mimic or inhibitor transfection. RESULTS It's potential role in specifically regulating rodent cartilage development and associated cellular processes. Furthermore, the expression of protein arginine N-methyltransferase 3 (PRMT3), as a predicted target of the novel miRNA, was found consistently downregulated at rat cartilage during developmental stages and RCJ3.1C5.18 (C5.18) cells during the proliferating and hypertrophic phases of the cartilage development, where the miR-novel expression was significantly up-regulated. Both the dual-luciferase reporter gene assay and the up- or down-regulation of miR-novel suggest that the later can specifically bind with the Prmt3 3'-UTR. CONCLUSION Overall, this study provides the first comprehensive evidence that a genuine cartilage-specific novel miRNA directly targets PRMT3 and may regulate multitudinous cellular processes and signal transduction during cartilage development.
Collapse
Affiliation(s)
- Jian Sun
- Endemic of Endemic Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of PR of China, Xi’an, Shaanxi, 710061, PR China
| | - Quancheng Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Ying Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Safdar Hussain
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Yitong Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Yuanxu Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Mengyao Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Huang Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Xinyu Huo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Qilan Ning
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Yan Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| | - Peng Xu
- Department of Orthopedics, Honghui Hospital, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710054, PR China
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, PR China
| |
Collapse
|
22
|
Gao X, Li S, Cong C, Wang Y, Xu L. A Network Pharmacology Approach to Estimate Potential Targets of the Active Ingredients of Epimedium for Alleviating Mild Cognitive Impairment and Treating Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:2302680. [PMID: 33574879 PMCID: PMC7861915 DOI: 10.1155/2021/2302680] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/21/2020] [Accepted: 01/15/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND The present study made use of a network pharmacological approach to evaluate the mechanisms and potential targets of the active ingredients of Epimedium for alleviating mild cognitive impairment (MCI) and treating Alzheimer's disease (AD). METHODS The active ingredients of Epimedium were acquired from the Traditional Chinese Medicine System Pharmacology database, and potential targets were predicted using the TCMSP target module, SwissTargetPrediction, and PharmMapper database. Target proteins correlating with MCI and AD were downloaded from the GeneCards, DisGeNet, and OMIM databases. The common targets of Epimedium, MCI, and AD were identified using the Jvenn online tool, and a protein-protein interaction (PPI) network was constructed using the String database and Cytoscape. Finally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of the common targets was performed using DAVID, and molecular docking between active ingredients and target genes was modeled using AutoDock Vina. RESULTS A total of 20 active ingredients were analyzed, and 337 compound-related targets were identified for Epimedium. Out of 236 proteins associated with MCI and AD, 54 overlapped with the targets of Epimedium. The top 30 interacting proteins in this set were ranked by topological analysis. GO and KEGG enrichment analysis suggested that the common targets participated in diverse biological processes and pathways, including cell proliferation and apoptosis, inflammatory response, signal transduction, and protein phosphorylation through cancer pathway, MAPK signaling pathway, PI3K-Akt signaling pathway, HIF-1 signaling pathway, sphingolipid signaling pathway, FoxO signaling pathway, and TNF signaling pathway. Molecular docking analysis suggested that the 20 active ingredients could bind to the top 5 protein targets. CONCLUSIONS The present study provides theoretical evidence for in-depth analysis of the mechanisms and molecular targets by which Epimedium protects against MCI, AD, and other neurodegenerative diseases and lays the foundation for pragmatic clinical applications and potential new drug development.
Collapse
Affiliation(s)
- Xianwei Gao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shengnan Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Chao Cong
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yuejiao Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lianwei Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
23
|
Protective Effect of Biobran/MGN-3 against Sporadic Alzheimer's Disease Mouse Model: Possible Role of Oxidative Stress and Apoptotic Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8845064. [PMID: 33574982 PMCID: PMC7857904 DOI: 10.1155/2021/8845064] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/14/2020] [Accepted: 01/03/2021] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a debilitating and irreversible brain disease that affects an increasing number of aged individuals, mandating the development of protective nutraceuticals. Biobran/MGN-3, an arabinoxylan from rice bran, has potent antioxidant, antiaging, and immunomodulatory effects. The aim of the present study was to investigate the protective effect of Biobran against sporadic Alzheimer's disease (SAD). SAD was induced in mice via intracerebroventricular injection of streptozotocin (STZ) (3 mg/kg). STZ-treated mice were administered with Biobran for 21 days. The effects of Biobran on memory and learning were measured via the Morris water maze, novel object recognition, and Y-maze tests. Biomarkers for apoptosis, oxidative stress, and amyloidogenesis were measured using ELISA and western blot analysis. Histopathological examination was performed to confirm neuronal damage and amyloid-beta deposition. Biobran reversed the spatial memory deficit in SAD-induced mice, and it increased the expression of glutathione, reduced malondialdehyde, decreased IL-6, decreased intercellular adhesion molecule-1 (ICAM-1), and significantly increased nuclear factor erythroid 2-related factor 2 (Nrf2) and antioxidant response element (ARE). Moreover, Biobran exerted a protective effect against amyloid-beta-induced apoptosis via the suppression of both cleaved caspase-3 and the proapoptotic protein Bax and via the upregulation of the antiapoptotic protein Bcl-2. Furthermore, it reduced the expression of forkhead box class O proteins. It could be concluded from this study that Biobran may be a useful nutritional antioxidant agent for protection against SAD through its activation of the gene expression of Nrf2/ARE, which in turn modulates the apoptotic and amyloidogenic pathways.
Collapse
|
24
|
Abstract
The global increase in lifespan noted not only in developed nations, but also in large developing countries parallels an observed increase in a significant number of non-communicable diseases, most notable neurodegenerative disorders. Neurodegenerative disorders present a number of challenges for treatment options that do not resolve disease progression. Furthermore, it is believed by the year 2030, the services required to treat cognitive disorders in the United States alone will exceed $2 trillion annually. Mammalian forkhead transcription factors, silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae), the mechanistic target of rapamycin, and the pathways of autophagy and apoptosis offer exciting avenues to address these challenges by focusing upon core cellular mechanisms that may significantly impact nervous system disease. These pathways are intimately linked such as through cell signaling pathways involving protein kinase B and can foster, sometimes in conjunction with trophic factors, enhanced neuronal survival, reduction in toxic intracellular accumulations, and mitochondrial stability. Feedback mechanisms among these pathways also exist that can oversee reparative processes in the nervous system. However, mammalian forkhead transcription factors, silent mating type information regulation 2 homolog 1, mechanistic target of rapamycin, and autophagy can lead to cellular demise under some scenarios that may be dependent upon the precise cellular environment, warranting future studies to effectively translate these core pathways into successful clinical treatment strategies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling New York, New York, NY, USA
| |
Collapse
|
25
|
Zaidi SAH, Guzman W, Singh S, Mehrotra S, Husain S. Changes in Class I and IIb HDACs by δ-Opioid in Chronic Rat Glaucoma Model. Invest Ophthalmol Vis Sci 2020; 61:4. [PMID: 33263714 PMCID: PMC7718808 DOI: 10.1167/iovs.61.14.4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose This study determines if δ-opioid receptor agonist (i.e. SNC-121)-induced epigenetic changes via regulation of histone deacetylases (HDACs) for retinal ganglion cell (RGC) neuroprotection in glaucoma model. Methods Intraocular pressure was raised in rat eyes by injecting 2M hypertonic saline into the limbal veins. SNC-121 (1 mg/kg; i.p.) was administered to the animals for 7 days. Retinas were collected at days 7 and 42, post-injury followed by measurement of HDAC activities, mRNA, and protein expression by enzyme assay, quantitative real-time PCR (qRT-PCR), Western blotting, and immunohistochemistry. Results The visual acuity, contrast sensitivity, and pattern electroretinograms (ERGs) were declined in ocular hypertensive animals, which were significantly improved by SNC-121 treatment. Class I and IIb HDACs activities were significantly increased at days 7 and 42 in ocular hypertensive animals. The mRNA and protein expression of HDAC 1 was increased by 1.33 ± 0.07-fold and 20.2 ± 2.7%, HDAC 2 by 1.4 ± 0.05-fold and 17.0 ± 2.4%, HDAC 3 by 1.4 ± 0.06-fold and 17.4 ± 3.4%, and HDAC 6 by 1.5 ± 0.09-fold and 15.1 ± 3.3% at day 7, post-injury. Both the mRNA and protein expression of HDACs were potentiated further at day 42 in ocular hypertensive animals. HDAC activities, mRNA, and protein expression were blocked by SNC-121 treatment at days 7 and 42 in ocular hypertensive animals. Conclusions Data suggests that class I and IIb HDACs are activated and upregulated during early stages of glaucoma. Early intervention with δ-opioid receptor activation resulted in the prolonged suppression of class I and IIb HDACs activities and expression, which may, in part, play a crucial role in RGC neuroprotection.
Collapse
Affiliation(s)
- Syed A H Zaidi
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Wendy Guzman
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Sudha Singh
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Shikhar Mehrotra
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Shahid Husain
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, South Carolina, United States
| |
Collapse
|
26
|
Goswami S, Kareem O, Goyal RK, Mumtaz SM, Tonk RK, Gupta R, Pottoo FH. Role of Forkhead Transcription Factors of the O Class (FoxO) in Development and Progression of Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:709-721. [PMID: 33001019 DOI: 10.2174/1871527319666201001105553] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 07/20/2020] [Accepted: 08/31/2020] [Indexed: 11/22/2022]
Abstract
In the Central Nervous System (CNS), a specific loss of focal neurons leads to mental and neurological disorders like dementia, Alzheimer's Disease (AD), Huntington's disease, Parkinson's disease, etc. AD is a neurological degenerative disorder, which is progressive and irreversible in nature and is the widely recognized reason for dementia in the geriatric populace. It affects 10% of people above the age of 65 and is the fourth driving reason for death in the United States. Numerous evidence suggests that the neuronal compartment is not the only genesis of AD, but transcription factors also hold significant importance in the occurrence and advancement of the disease. It is the need of the time to find the novel molecular targets and new techniques for treating or slowing down the progression of neurological disorders, especially AD. In this article, we summarised a conceivable association between transcriptional factors and their defensive measures against neurodegeneration and AD. The mammalian forkhead transcription factors of the class O (FoxO) illustrate one of the potential objectives for the development of new methodologies against AD and other neurocognitive disorders. The presence of FoxO is easily noticeable in the "cognitive centers" of the brain, specifically in the amygdala, hippocampus, and the nucleus accumbens. FoxO proteins are the prominent and necessary factors in memory formation and cognitive functions. FoxO also assumes a pertinent role in the protection of multiple cells in the brain by controlling the involving mechanism of autophagy and apoptosis and also modulates the process of phosphorylation of the targeted protein, thus FoxO must be a putative target in the mitigation of AD. This review features the role of FoxO as an important biomarker and potential new targets for the treatment of AD.
Collapse
Affiliation(s)
- Shikha Goswami
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Ozaifa Kareem
- Department of Pharmaceutical Sciences, Faculty of Applied Sciences and Technology, University of Kashmir, Srinagar, JK, India
| | - Ramesh K Goyal
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Sayed M Mumtaz
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Rajiv K Tonk
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Rahul Gupta
- Delhi Pharmaceutical Sciences and Research University, Mehrauli- Badarpur Rd, Sector 3, PushpVihar, New Delhi, India
| | - Faheem H Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University P.O.BOX 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
27
|
Gamache J, Yun Y, Chiba-Falek O. Sex-dependent effect of APOE on Alzheimer's disease and other age-related neurodegenerative disorders. Dis Model Mech 2020; 13:dmm045211. [PMID: 32859588 PMCID: PMC7473656 DOI: 10.1242/dmm.045211] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The importance of apolipoprotein E (APOE) in late-onset Alzheimer's disease (LOAD) has been firmly established, but the mechanisms through which it exerts its pathogenic effects remain elusive. In addition, the sex-dependent effects of APOE on LOAD risk and endophenotypes have yet to be explained. In this Review, we revisit the different aspects of APOE involvement in neurodegeneration and neurological diseases, with particular attention to sex differences in the contribution of APOE to LOAD susceptibility. We discuss the role of APOE in a broader range of age-related neurodegenerative diseases, and summarize the biological factors linking APOE to sex hormones, drawing on supportive findings from rodent models to identify major mechanistic themes underlying the exacerbation of LOAD-associated neurodegeneration and pathology in the female brain. Additionally, we list sex-by-genotype interactions identified across neurodegenerative diseases, proposing APOE variants as a shared etiology for sex differences in the manifestation of these diseases. Finally, we present recent advancements in 'omics' technologies, which provide a new platform for more in-depth investigations of how dysregulation of this gene affects the development and progression of neurodegenerative diseases. Collectively, the evidence summarized in this Review highlights the interplay between APOE and sex as a key factor in the etiology of LOAD and other age-related neurodegenerative diseases. We emphasize the importance of careful examination of sex as a contributing factor in studying the underpinning genetics of neurodegenerative diseases in general, but particularly for LOAD.
Collapse
Affiliation(s)
- Julia Gamache
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Young Yun
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
28
|
Liao Y, Cheng J, Kong X, Li S, Li X, Zhang M, Zhang H, Yang T, Dong Y, Li J, Xu Y, Yuan Z. HDAC3 inhibition ameliorates ischemia/reperfusion-induced brain injury by regulating the microglial cGAS-STING pathway. Am J Cancer Res 2020; 10:9644-9662. [PMID: 32863951 PMCID: PMC7449914 DOI: 10.7150/thno.47651] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/21/2020] [Indexed: 12/22/2022] Open
Abstract
Rationale: It is known that neuroinflammation plays a critical and detrimental role in the development of cerebral ischemia/reperfusion (I/R), but the regulation of the cyclic GMP-AMP synthase (cGAS)-mediated innate immune response in I/R-induced neuroinflammation is largely unexplored. This study aimed to investigate the function and regulatory mechanism of cGAS in I/R-induced neuroinflammation and brain injury, and to identify possible strategies for the treatment of ischemic stroke. Methods: To demonstrate that microglial histone deacetylase 3 (HDAC3) regulates the microglial cGAS-stimulator of interferon genes (cGAS-STING) pathway and is involved in I/R-induced neuroinflammation and brain injury, a series of cell biological, molecular, and biochemical approaches were utilized. These approaches include transient middle cerebral artery occlusion (tMCAO), real-time polymerase chain reaction (PCR), RNA sequencing, western blot, co-immunoprecipitation, chromosome-immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), dual-luciferase reporter assay, immunohistochemistry, and confocal imaging. Results: The microglial cGAS- STING pathway was activated by mitochondrial DNA, which promoted the formation of a pro-inflammatory microenvironment. In addition, we revealed that HDAC3 transcriptionally promoted the expression of cGAS and potentiated the activation of the cGAS-STING pathway by regulating the acetylation and nuclear localization of p65 in microglia. Our in vivo results indicated that deletion of cGAS or HDAC3 in microglia attenuated I/R-induced neuroinflammation and brain injury. Conclusion: Collectively, we elucidated that the HDAC3-p65-cGAS-STING pathway is involved in the development of I/R-induced neuroinflammation, identifying a new therapeutic avenue for the treatment of ischemic stroke.
Collapse
|
29
|
Yoon S, Kim M, Min HK, Lee YU, Kwon DH, Lee M, Lee S, Kook T, Joung H, Nam KI, Ahn Y, Kim YK, Kim J, Park WJ, McMullen JR, Eom GH, Kook H. Inhibition of heat shock protein 70 blocks the development of cardiac hypertrophy by modulating the phosphorylation of histone deacetylase 2. Cardiovasc Res 2020; 115:1850-1860. [PMID: 30596969 DOI: 10.1093/cvr/cvy317] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/22/2018] [Accepted: 12/21/2018] [Indexed: 11/14/2022] Open
Abstract
AIMS Previously, we reported that phosphorylation of histone deacetylase 2 (HDAC2) and the resulting activation causes cardiac hypertrophy. Through further study of the specific binding partners of phosphorylated HDAC2 and their mechanism of regulation, we can better understand how cardiac hypertrophy develops. Thus, in the present study, we aimed to elucidate the function of one such binding partner, heat shock protein 70 (HSP70). METHODS AND RESULTS Primary cultures of rat neonatal ventricular cardiomyocytes and H9c2 cardiomyoblasts were used for in vitro cellular experiments. HSP70 knockout (KO) mice and transgenic (Tg) mice that overexpress HSP70 in the heart were used for in vivo analysis. Peptide-precipitation and immunoprecipitation assay revealed that HSP70 preferentially binds to phosphorylated HDAC2 S394. Forced expression of HSP70 increased phosphorylation of HDAC2 S394 and its activation, but not that of S422/424, whereas knocking down of HSP70 reduced it. However, HSP70 failed to phosphorylate HDAC2 in the cell-free condition. Phosphorylation of HDAC2 S394 by casein kinase 2α1 enhanced the binding of HSP70 to HDAC2, whereas dephosphorylation induced by the catalytic subunit of protein phosphatase 2A (PP2CA) had the opposite effect. HSP70 prevented HDAC2 dephosphorylation by reducing the binding of HDAC2 to PP2CA. HSP70 KO mouse hearts failed to phosphorylate S394 HDAC2 in response to isoproterenol infusion, whereas Tg overexpression of HSP70 increased the phosphorylation and activation of HDAC2. 2-Phenylethynesulfonamide (PES), an HSP70 inhibitor, attenuated cardiac hypertrophy induced either by phenylephrine in neonatal ventricular cardiomyocytes or by aortic banding in mice. PES reduced HDAC2 S394 phosphorylation and its activation by interfering with the binding of HSP70 to HDAC2. CONCLUSION These results demonstrate that HSP70 specifically binds to S394-phosphorylated HDAC2 and maintains its phosphorylation status, which results in HDAC2 activation and the development of cardiac hypertrophy. Inhibition of HSP70 has possible application as a therapeutic.
Collapse
MESH Headings
- Animals
- Binding Sites
- Cell Line
- Disease Models, Animal
- Enzyme Activation
- HSP70 Heat-Shock Proteins/antagonists & inhibitors
- HSP70 Heat-Shock Proteins/deficiency
- HSP70 Heat-Shock Proteins/genetics
- HSP70 Heat-Shock Proteins/metabolism
- Histone Deacetylase 2/metabolism
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Phosphorylation
- Protein Binding
- Protein Phosphatase 2/metabolism
- Rats
- Rats, Sprague-Dawley
- Signal Transduction
- Sulfonamides/pharmacology
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Somy Yoon
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Mira Kim
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Hyun-Ki Min
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Yeong-Un Lee
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Cardiac Remodeling Research Center, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Duk-Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Cardiac Remodeling Research Center, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Miyoung Lee
- Cardiac Remodeling Research Center, Chonnam National University Medical School, Hwasun, Republic of Korea
- College of Life Science, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Sumin Lee
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Taewon Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Hosouk Joung
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Cardiac Remodeling Research Center, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Kwang-Il Nam
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Young-Kook Kim
- Cardiac Remodeling Research Center, Chonnam National University Medical School, Hwasun, Republic of Korea
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Jaetaek Kim
- Cardiac Remodeling Research Center, Chonnam National University Medical School, Hwasun, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Woo Jin Park
- Cardiac Remodeling Research Center, Chonnam National University Medical School, Hwasun, Republic of Korea
- College of Life Science, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gwang Hyeon Eom
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Republic of Korea
- Cardiac Remodeling Research Center, Chonnam National University Medical School, Hwasun, Republic of Korea
| |
Collapse
|
30
|
PCAF Involvement in Lamin A/C-HDAC2 Interplay during the Early Phase of Muscle Differentiation. Cells 2020; 9:cells9071735. [PMID: 32698523 PMCID: PMC7409167 DOI: 10.3390/cells9071735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Lamin A/C has been implicated in the epigenetic regulation of muscle gene expression through dynamic interaction with chromatin domains and epigenetic enzymes. We previously showed that lamin A/C interacts with histone deacetylase 2 (HDAC2). In this study, we deepened the relevance and regulation of lamin A/C-HDAC2 interaction in human muscle cells. We present evidence that HDAC2 binding to lamina A/C is related to HDAC2 acetylation on lysine 75 and expression of p300-CBP associated factor (PCAF), an acetyltransferase known to acetylate HDAC2. Our findings show that lamin A and farnesylated prelamin A promote PCAF recruitment to the nuclear lamina and lamin A/C binding in human myoblasts committed to myogenic differentiation, while protein interaction is decreased in differentiating myotubes. Interestingly, PCAF translocation to the nuclear envelope, as well as lamin A/C-PCAF interaction, are reduced by transient expression of lamin A mutated forms causing Emery Dreifuss muscular dystrophy. Consistent with this observation, lamin A/C interaction with both PCAF and HDAC2 is significantly reduced in Emery-Dreifuss muscular dystrophy myoblasts. Overall, these results support the view that, by recruiting PCAF and HDAC2 in a molecular platform, lamin A/C might contribute to regulate their epigenetic activity required in the early phase of muscle differentiation.
Collapse
|
31
|
Regulation of histone deacetylase activities and functions by phosphorylation and its physiological relevance. Cell Mol Life Sci 2020; 78:427-445. [PMID: 32683534 DOI: 10.1007/s00018-020-03599-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022]
Abstract
Histone deacetylases (HDACs) are conserved enzymes that regulate many cellular processes by catalyzing the removal of acetyl groups from lysine residues on histones and non-histone proteins. As appropriate for proteins that occupy such an essential biological role, HDAC activities and functions are in turn highly regulated. Overwhelming evidence suggests that the dysregulation of HDACs plays a major role in many human diseases. The regulation of HDACs is achieved by multiple different mechanisms, including posttranslational modifications. One of the most common posttranslational modifications on HDACs is reversible phosphorylation. Many HDAC phosphorylations are context-dependent, occurring in specific tissues or as a consequence of certain stimuli. Additionally, whereas phosphorylation can regulate some HDACs in a non-specific manner, many HDAC phosphorylations result in specific consequences. Although some of these modifications support normal HDAC function, aberrations can contribute to disease development. Here we review and critically evaluate how reversible phosphorylation activates or deactivates HDACs and, thereby, regulates their many functions under various cellular and physiological contexts.
Collapse
|
32
|
Sun X, Wang T, Wang Y, Ai K, Pan G, Li Y, Zhou C, He S, Cong H. Downregulation of lncRNA-11496 in the Brain Contributes to Microglia Apoptosis via Regulation of Mef2c in Chronic T. gondii Infection Mice. Front Mol Neurosci 2020; 13:77. [PMID: 32499679 PMCID: PMC7243434 DOI: 10.3389/fnmol.2020.00077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/20/2020] [Indexed: 01/02/2023] Open
Abstract
Though it is well known that chronic infections of Toxoplasma gondii (T. gondii) can induce mental and behavioral disorders in the host, little is known about the role of long non-coding RNAs (lncRNAs) in this pathological process. In this study, we employed an advanced lncRNAs and mRNAs integration chip (Affymetrix HTA 2.0) to detect the expression of both lncRNAs and mRNAs in T. gondii Chinese 1 strain infected mouse brain. As a result, for the first time, the downregulation of lncRNA-11496 (NONMMUGO11496) was identified as the responsible factor for this pathological process. We showed that dysregulation of lncRNA-11496 affected proliferation, differentiation and apoptosis of mouse microglia. Furthermore, we proved that Mef2c (Myocyte-specific enhancer factor 2C), a member of the MEF2 subfamily, is the target gene of lncRNA-11496. In a more detailed study, we confirmed that lncRNA-11496 positively regulated the expression of Mef2c by binding to histone deacetylase 2 (HDAC2). Importantly, Mef2c itself could coordinate neuronal differentiation, survival, as well as synapse formation. Thus, our current study provides the first evidence in terms of the modulatory action of lncRNAs in chronic toxoplasmosis in T. gondii infected mouse brain, providing a solid scientific basis for using lncRNA-11496 as a therapeutic target to treat T. gondii induced neurological disorder.
Collapse
Affiliation(s)
- Xiahui Sun
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ting Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yongliang Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Kang Ai
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ge Pan
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Li
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunxue Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shenyi He
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hua Cong
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
33
|
Affiliation(s)
- Antonino Tuttolomondo
- Department of Promoting Health, Maternal Infant, Excellence and Internal & Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Palermo, Sicily, Italy
- Internal Medicine and Stroke Care Ward, Policlinico ‘P Giaccone’, University of Palermo, Palermo, Sicily, Italy
| | - Antonio Pinto
- Department of Promoting Health, Maternal Infant, Excellence and Internal & Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Palermo, Sicily, Italy
- Internal Medicine and Stroke Care Ward, Policlinico ‘P Giaccone’, University of Palermo, Palermo, Sicily, Italy
| |
Collapse
|
34
|
Cheng J, Wang S, Dong Y, Yuan Z. The Role and Regulatory Mechanism of Hippo Signaling Components in the Neuronal System. Front Immunol 2020; 11:281. [PMID: 32140159 PMCID: PMC7042394 DOI: 10.3389/fimmu.2020.00281] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/04/2020] [Indexed: 01/07/2023] Open
Abstract
The Hippo signaling pathway, an evolutionarily conserved protein kinase cascade, plays a critical role in controlling organ size, cancer development, and tissue regeneration. Recently, mounting evidence has suggested that Hippo signaling also has an important role in regulating immunity, including innate and adaptive immune activation. In the neuronal system, Our laboratory results, together with those from other studies, demonstrate that the Hippo signaling pathway is involved in neuroinflammation, neuronal cell differentiation, and neuronal death. In the present review, we summarize the recent findings pertaining to the function and regulatory mechanism of Hippo signaling components in the neuronal system, implicating the potential of Hippo signaling as a therapeutic target for the treatment of neuronal system diseases.
Collapse
Affiliation(s)
- Jinbo Cheng
- Center on Translational Neuroscience, College of Life and Environmental Science, Minzu University of China, Beijing, China.,The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shukun Wang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yuan Dong
- Department of Biochemistry, Medical College, Qingdao University, Qingdao, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
35
|
Dhir N, Medhi B, Prakash A, Goyal MK, Modi M, Mohindra S. Pre-clinical to Clinical Translational Failures and Current Status of Clinical Trials in Stroke Therapy: A Brief Review. Curr Neuropharmacol 2020; 18:596-612. [PMID: 31934841 PMCID: PMC7457423 DOI: 10.2174/1570159x18666200114160844] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/31/2019] [Accepted: 12/28/2019] [Indexed: 12/16/2022] Open
Abstract
In stroke (cerebral ischemia), despite continuous efforts both at the experimental and clinical level, the only approved pharmacological treatment has been restricted to tissue plasminogen activator (tPA). Stroke is the leading cause of functional disability and mortality throughout worldwide. Its pathophysiology starts with energy pump failure, followed by complex signaling cascade that ultimately ends in neuronal cell death. Ischemic cascade involves excessive glutamate release followed by raised intracellular sodium and calcium influx along with free radicals' generation, activation of inflammatory cytokines, NO synthases, lipases, endonucleases and other apoptotic pathways leading to cell edema and death. At the pre-clinical stage, several agents have been tried and proven as an effective neuroprotectant in animal models of ischemia. However, these agents failed to show convincing results in terms of efficacy and safety when the trials were conducted in humans following stroke. This article highlights the various agents which have been tried in the past but failed to translate into stroke therapy along with key points that are responsible for the lagging of experimental success to translational failure in stroke treatment.
Collapse
Affiliation(s)
| | - Bikash Medhi
- Address correspondence to this author at the Department of Pharmacology, Research Block B, 4th Floor, Room no 4043, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India; E-mail:
| | | | | | | | | |
Collapse
|
36
|
Macelignan inhibits the inflammatory response of microglia and regulates neuronal survival. J Neuroimmunol 2019; 339:577123. [PMID: 31838278 DOI: 10.1016/j.jneuroim.2019.577123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/24/2019] [Accepted: 12/05/2019] [Indexed: 01/19/2023]
Abstract
Neuroinflammation is an important pathological process of neurodegenerative diseases, and microglial contributes to chronic inflammation and neuronal loss in progressive neurodegenerative. Therefore, regulating the inflammatory response of microglia could lead to the discovery of promising treatments for neurodegenerative diseases. In this study, we investigated the effects of the nutmeg plant seed extract, macelignan, on the inflammatory response of microglia and neuronal cell survival. We detected NO and iNOS using the Griess test and Western blotting. We measured phosphoinositide 3 kinase (PI3K)/Akt expression by Western blotting. The release of NO and inflammatory cytokines and the expression of iNOS decreased in a concentration-dependent manner, with an increase in macelignan concentration. PI3K/Akt phosphorylation levels decreased in a dose-dependent manner in lipopolysaccharide (LPS)-activated microglial cells after exposure to macelignan. We also demonstrated that macelignan improved HT22 cell viability, following exposure to a microglial-conditioned medium. Furthermore, macelignan inhibited microglial cell near neurons treated with a hypoxic conditioned medium. Finally, macelignan treatment reduced the expression of p27 and cyclin D1 in neurons cultured in an LPS-activated microglia-conditioned medium. Therefore, these results imply that macelignan can inhibit the inflammatory response of microglia and regulate neuronal survival through the PI3K/Akt pathway.
Collapse
|
37
|
Wu X, Fan Z, Chen M, Chen Y, Rong D, Cui Z, Yuan Y, Zhuo L, Xu Y. Forkhead transcription factor FOXO3a mediates interferon-γ-induced MHC II transcription in macrophages. Immunology 2019; 158:304-313. [PMID: 31509237 DOI: 10.1111/imm.13116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/23/2019] [Accepted: 09/03/2019] [Indexed: 12/23/2022] Open
Abstract
Macrophages are professional antigen-presenting cells relying on the expression of class II major histocompatibility complex (MHC II) genes. Interferon-γ (IFN-γ) activates MHC II transcription via the assembly of an enhanceosome centred on class II trans-activator (CIITA). In the present study, we investigated the role of the forkhead transcription factor FOXO3a in IFN- γ-induced MHC II transcription in macrophages. Knockdown of FOXO3a, but not FOXO1 or FOXO4, diminished IFN-γ-induced MHC II expression in RAW cells. On the contrary, over-expression of FOXO3a, but neither FOXO1 nor FOXO4, enhanced CIITA-mediated trans-activation of the MHC II promoter. IFN-γ treatment promoted the recruitment of FOXO3a to the MHC II promoter. Co-immunoprecipitation and RE-ChIP assays showed that FOXO3a was a component of the MHC II enhanceosome forming interactions with CIITA, RFX5, RFXB and RFXAP. FOXO3a contributed to MHC II transcription by altering histone modifications surrounding the MHC II promoter. Of interest, FOXO3a was recruited to the type IV CIITA promoter and directly activated CIITA transcription by interacting with signal transducer of activation and transcription 1 in response to IFN-γ stimulation. In conclusion, our data unveil a novel role for FOXO3a in the regulation of MHC II transcription in macrophages.
Collapse
Affiliation(s)
- Xiaoyan Wu
- The Laboratory Centre for Basic Medical Sciences, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China.,Department of Pathophysiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zhiwen Fan
- Department of Pathology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Ming Chen
- The Laboratory Centre for Basic Medical Sciences, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yi Chen
- The Laboratory Centre for Basic Medical Sciences, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Danyan Rong
- The Laboratory Centre for Basic Medical Sciences, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zhiwei Cui
- The Laboratory Centre for Basic Medical Sciences, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yibiao Yuan
- The Laboratory Centre for Basic Medical Sciences, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lili Zhuo
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Xu
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China.,Department of Pathophysiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
38
|
Demyanenko SV, Dzreyan VA, Neginskaya MA, Uzdensky AB. Expression of Histone Deacetylases HDAC1 and HDAC2 and Their Role in Apoptosis in the Penumbra Induced by Photothrombotic Stroke. Mol Neurobiol 2019; 57:226-238. [PMID: 31493239 DOI: 10.1007/s12035-019-01772-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/18/2022]
Abstract
In ischemic stroke, vascular occlusion rapidly induces tissue infarct. Over the ensuing hours, damage spreads to adjacent tissue and forms transition zone (penumbra), which is potentially salvageable. Epigenetic regulation of chromatin structure controls gene expression and protein synthesis. We studied the expression of histone deacetylases HDAC1 and HDAC2 in the penumbra at 4 or 24 h after photothrombotic stroke (PTS) in the rat brain cortex. PTS increased the expression of HDAC1 and HDAC2 in penumbra and caused the redistribution of HDAC1 but not HDAC2 from the neuronal nuclei to cytoplasm. In astrocytes, HDAC1 expression and localization did not change. In neurons, HDAC2 localized exclusively in nuclei, but in astrocytes, it was also observed in processes. PTS induced neuronal apoptosis in the penumbra. TUNEL-stained apoptotic neurons co-localized with HDAC2 but not HDAC1. These data suggest that HDAC2 may represent the potential target for anti-stroke therapy and its selective inhibition may be a promising strategy for the protection of the penumbra tissue after ischemic stroke.
Collapse
Affiliation(s)
- S V Demyanenko
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky Ave., Rostov-on-Don, Russia, 344090
| | - V A Dzreyan
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky Ave., Rostov-on-Don, Russia, 344090
| | - M A Neginskaya
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky Ave., Rostov-on-Don, Russia, 344090
| | - A B Uzdensky
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky Ave., Rostov-on-Don, Russia, 344090.
| |
Collapse
|
39
|
FOXO3 Is Expressed in Ovarian Tissues and Acts as an Apoptosis Initiator in Granulosa Cells of Chickens. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6902906. [PMID: 31380433 PMCID: PMC6657628 DOI: 10.1155/2019/6902906] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/21/2019] [Accepted: 07/02/2019] [Indexed: 11/18/2022]
Abstract
FOXO3, which encodes the transcription factor forkhead box O-3 (FoxO3), is a member of the FOXO subfamily of the forkhead box (FOX) family. FOXO3 can be negatively regulated by its phosphorylation by the PI3K/Akt signaling pathway and ultimately drives apoptosis when activated. In mammalian ovaries, the FOXO3 protein regulates atresia and follicle growth by promoting apoptosis of ovarian granulosa cells. Nonetheless, the specific effects of the FOXO3 protein on granulosa apoptosis of avian ovaries have not been elucidated. Therefore, we studied FOXO3 expression in follicles with different organization and at all hierarchical levels of chicken follicles. Via an immunofluorescence assay, the chicken follicular theca at all hierarchical levels were found to be strongly stained with an anti-FOXO3 antibody. In chicken primary ovarian granulosa cells, mRNA levels of proapoptotic factors BNIP3 and BCL2L11 decreased in the absence of FOXO3, and so did PARP-1 and cleaved caspase 3 protein levels. After treatment with a recombinant FOXO3 protein, PARP-1 and caspase 3 protein levels increased, along with mRNA levels of Bnip3 and BCL2L11 (significantly, p<0.05). In addition, FOXO3 was downregulated in chicken granulosa cells when different estradiol or FSH concentrations were applied. In conclusion, FOXO3 is expressed in chicken reproductive tissues, including follicles and ovarian granulosa cells, and promotes apoptosis of chicken ovarian granulosa cells.
Collapse
|
40
|
FoxO3a inhibiting expression of EPS8 to prevent progression of NSCLC: A new negative loop of EGFR signaling. EBioMedicine 2019; 40:198-209. [PMID: 30738830 PMCID: PMC6413682 DOI: 10.1016/j.ebiom.2019.01.053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 01/25/2019] [Accepted: 01/25/2019] [Indexed: 12/21/2022] Open
Abstract
Background The resistance to EGF receptor (EGFR) tyrosine kinase inhibitors (TKI) is a major challenge in the treatment of non-small cell lung cancer (NSCLC). Understanding the molecular mechanisms behind resistance is therefore an important issue. Here we assessed the role of EGFR pathway substrate 8 (EPS8) and Forkhead box O 3a (FoxO3a) as potentially valuable targets in the resistance of NSCLC . Methods The expression levels of EPS8 and FoxO3a in patients with NSCLC (n = 75) were examined by immunohistochemistry staining, while in cells were detected by qPCR and western blot. The effects of EPS8 and FoxO3a on resistance, migration and invasion, cell cycle arrest were detected by MTT, transwell and flow cytometry, respectively. Chromatin immunoprecipitation and luciferase reporter assays were performed to determine the mechanisms of EPS8 expression and FoxO3a regulation. Findings We observed that the expression of EPS8 inversely correlated with FoxO3a in NSCLC cell lines and NSCLC patients. FoxO3a levels were significantly decreased in tumor tissues compared with para-carcinoma tissues, while EPS8 is opposite. Besides, they play reverse roles in the resistance to gefitinib, the migration and invasion abilities, the cell cycle arrest in vitro and the tumor growth in vivo. Mechanistically, FoxO3a inhibits EPS8 levels by directly binding its gene promoter and they form a negative loop in EGFR pathway. Interpretation Targeting FoxO3a and EPS8 in EGFR signaling pathway prevents the progression of NSCLC, which implied that the negative loop they formed could served as a therapeutic target for overcoming resistance in NSCLC. Funds National Natural Science Foundation of China, Science and Technology Project of Henan, Outstanding Young Talent Research Fund of Zhengzhou University and the National Scholarship Fund.
Collapse
|
41
|
Lucia D, Burgess D, Cullen CL, Dorey ES, Rawashdeh O, Moritz KM. Periconceptional maternal alcohol consumption leads to behavioural changes in adult and aged offspring and alters the expression of hippocampal genes associated with learning and memory and regulators of the epigenome. Behav Brain Res 2019; 362:249-257. [PMID: 30633938 DOI: 10.1016/j.bbr.2019.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Maternal alcohol consumption throughout pregnancy can result in long term behavioural deficits in offspring. However, less is known about the impact of alcohol during the periconceptional period (PC). The aim of this study was to examine the effect of PC ethanol (PC:EtOH) exposure on long term cognitive function; including memory and anxiety. Rats were exposed to a liquid diet containing ethanol (EtOH) (12.5% vol;vol) or a control diet from 4 days prior to mating until day 4 of pregnancy. Separate cohorts of animals were tested at 6 months (adult) or 15-18 months of age (aged). Offspring underwent a series of behavioural tests to assess anxiety, spatial and recognition memory. The hippocampus was collected, and mRNA expression of epigenetic modifiers and genes implicated in learning and memory were examined. PC:EtOH exposure resulted in a subtle anxiety like behaviour in adult female offspring with a significant reduction in directed exploring/head dipping behaviour during holeboard testing. In aged male offspring, PC:EtOH exposure resulted in a tendency for increased directed exploring/head dipping behaviour during holeboard testing. No differences between treatments were observed in the elevated plus maze. Aged female offspring exposed to PC:EtOH demonstrated short term spatial memory impairment (P < 0.05). PC:EtOH resulted in an upregulation of hippocampal mRNA expression of bdnf, grin2a and grin2b at 18 months of age along with increased expression of epigenetic modifiers (dnmt1, dnmt3a and hdac2). In conclusion, PC:EtOH can lead to sex specific anxiety-like behaviour and impairments in spatial memory and altered hippocampal gene expression.
Collapse
Affiliation(s)
- D Lucia
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - D Burgess
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - C L Cullen
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland 4072, Australia; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - E S Dorey
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - O Rawashdeh
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - K M Moritz
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland 4072, Australia; Child Health Research Centre, The University of Queensland, 4072, Australia.
| |
Collapse
|
42
|
Li D, Ni H, Rui Q, Gao R, Chen G. Mst1: Function and Mechanism in Brain and Myocardial Ischemia Reperfusion Injury. Curr Neuropharmacol 2018; 16:1358-1364. [PMID: 29766810 PMCID: PMC6251045 DOI: 10.2174/1570159x16666180516095949] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/14/2017] [Accepted: 02/28/2018] [Indexed: 01/09/2023] Open
Abstract
Mammalian STE20-like kinase-1 (Mst1) is a generally expressed apoptosis-promoting kinase and a key bridgebuilder of apoptotic signaling in the etiology of tissue injury. Despite the fact that the biological function of Mst1 and its role in the cell's signalling network have yet to be determined, however, there is a lot of evidence that Mst1 plays an important role in cell death which results from tissue injury. Previous studies have shown that Mst1 is not only a target for some apoptosis- related molecules such as caspase 3 and P53, but also act as an activator of these proteinases to magnify apoptosis signal pathways. This article reviews the role of Mst1 in the signaling pathways which is related with the neuronal cell apoptosis or microglia activation following myocardial and brain injury. Therefore, this work contributes to better understanding of the pathological process of myocardial and brain injury.
Collapse
Affiliation(s)
- Di Li
- Department of Neurosurgery and Translational Medicine Center, The First People `s Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Haibo Ni
- Department of Neurosurgery, The First People `s Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Qin Rui
- Clinical laboratory,The First People`s Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Rong Gao
- Department of Neurosurgery, The First People `s Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
43
|
Mahady L, Nadeem M, Malek-Ahmadi M, Chen K, Perez SE, Mufson EJ. HDAC2 dysregulation in the nucleus basalis of Meynert during the progression of Alzheimer's disease. Neuropathol Appl Neurobiol 2018; 45:380-397. [PMID: 30252960 DOI: 10.1111/nan.12518] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 08/23/2018] [Indexed: 02/06/2023]
Abstract
AIMS Alzheimer's disease (AD) is characterized by degeneration of cholinergic basal forebrain (CBF) neurons in the nucleus basalis of Meynert (nbM), which provides the major cholinergic input to the cortical mantle and is related to cognitive decline in patients with AD. Cortical histone deacetylase (HDAC) dysregulation has been associated with neuronal degeneration during AD progression. However, whether HDAC alterations play a role in CBF degeneration during AD onset is unknown. We investigated global HDAC protein levels and nuclear HDAC2 immunoreactivity in tissue containing the nbM, changes and their association with neurofibrillary tangles (NFTs) during the progression of AD. METHODS We used semi-quantitative western blotting and immunohistochemistry to evaluate HDAC and sirtuin (SIRT) levels in individuals that died with a premortem clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), mild/moderate AD (mAD) or severe AD (sAD). Quantitative immunohistochemistry was used to identify HDAC2 protein levels in individual cholinergic nbM nuclei and their colocalization with the early phosphorylated tau marker AT8, the late-stage apoptotic tau marker TauC3 and Thioflavin-S, a marker of β-pleated sheet structures in NFTs. RESULTS In AD patients, HDAC2 protein levels were dysregulated in the basal forebrain region containing cholinergic neurons of the nbM. HDAC2 nuclear immunoreactivity was reduced in individual cholinergic nbM neurons across disease stages. HDAC2 nuclear reactivity correlated with multiple cognitive domains and with NFT formation. CONCLUSIONS These findings suggest that HDAC2 dysregulation contributes to cholinergic nbM neuronal dysfunction, NFT pathology, and cognitive decline during clinical progression of AD.
Collapse
Affiliation(s)
- L Mahady
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA.,Arizona State University Interdisciplinary Graduate Program in Neuroscience, Tempe, Arizona, USA
| | - M Nadeem
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | | | - K Chen
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | - S E Perez
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - E J Mufson
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
44
|
Hong-Qiang H, Mang-Qiao S, Fen X, Shan-Shan L, Hui-Juan C, Wu-Gang H, Wen-Jun Y, Zheng-Wu P. Sirt1 mediates improvement of isoflurane-induced memory impairment following hyperbaric oxygen preconditioning in middle-aged mice. Physiol Behav 2018; 195:1-8. [PMID: 30040951 DOI: 10.1016/j.physbeh.2018.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/20/2018] [Accepted: 07/20/2018] [Indexed: 01/04/2023]
Abstract
Hyperbaric oxygen (HBO) preconditioning (PC) has been suggested as a feasible method to provide neuroprotection from postoperative cognitive dysfunction (POCD). However, whether HBO-PC can ameliorate cognitive deficits induced by isoflurane, and the possible mechanism by which it may exert its effect, has not yet been clarified. In the present study, middle-aged mice were exposed to isoflurane anesthesia (1.5 minimal alveolar concentration [MAC]) for 2 h to establish a POCD model. After HBO preconditioning, cognitive function and expression of hippocampal sirtuin 1 (Sirt1), nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) were evaluated 24 h following isoflurane treatment, in the presence or absence of Sirt1 knockdown by short hairpin RNA (shRNA). HBO preconditioning increased the expression of Sirt1, Nrf2, and HO-1 and ameliorated memory dysfunction. Meanwhile, Sirt1 knockdown inhibited the expression of Nrf2 and HO-1 and attenuated the HBO preconditioning-associated memory improvement. Our results suggest that the application of HBO preconditioning is a useful treatment for POCD, and that Sirt1 may be a potential molecular target for POCD therapy.
Collapse
Affiliation(s)
- Hu Hong-Qiang
- Department of Anesthesiology, PLA No. 174 Hospital, Chenggong Hospital Affiliated to Xiamen University, Xiamen, Fujian 361003, China
| | - Shu Mang-Qiao
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Department of Psychiatry, Changan Hospital, Xi'an 710016, China
| | - Xue Fen
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Liu Shan-Shan
- Department of Anesthesiology, PLA No. 174 Hospital, Chenggong Hospital Affiliated to Xiamen University, Xiamen, Fujian 361003, China
| | - Cao Hui-Juan
- Department of Anesthesiology, PLA No. 174 Hospital, Chenggong Hospital Affiliated to Xiamen University, Xiamen, Fujian 361003, China
| | - Hou Wu-Gang
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yan Wen-Jun
- Department of Anesthesiology, Gansu Provincial Hospital, Lanzhou 730000, China.
| | - Peng Zheng-Wu
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
45
|
Mattioli E, Andrenacci D, Garofalo C, Prencipe S, Scotlandi K, Remondini D, Gentilini D, Di Blasio AM, Valente S, Scarano E, Cicchilitti L, Piaggio G, Mai A, Lattanzi G. Altered modulation of lamin A/C-HDAC2 interaction and p21 expression during oxidative stress response in HGPS. Aging Cell 2018; 17:e12824. [PMID: 30109767 PMCID: PMC6156291 DOI: 10.1111/acel.12824] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/22/2018] [Accepted: 06/26/2018] [Indexed: 02/01/2023] Open
Abstract
Defects in stress response are main determinants of cellular senescence and organism aging. In fibroblasts from patients affected by Hutchinson-Gilford progeria, a severe LMNA-linked syndrome associated with bone resorption, cardiovascular disorders, and premature aging, we found altered modulation of CDKN1A, encoding p21, upon oxidative stress induction, and accumulation of senescence markers during stress recovery. In this context, we unraveled a dynamic interaction of lamin A/C with HDAC2, an histone deacetylase that regulates CDKN1A expression. In control skin fibroblasts, lamin A/C is part of a protein complex including HDAC2 and its histone substrates; protein interaction is reduced at the onset of DNA damage response and recovered after completion of DNA repair. This interplay parallels modulation of p21 expression and global histone acetylation, and it is disrupted by LMNAmutations leading to progeroid phenotypes. In fact, HGPS cells show impaired lamin A/C-HDAC2 interplay and accumulation of p21 upon stress recovery. Collectively, these results link altered physical interaction between lamin A/C and HDAC2 to cellular and organism aging. The lamin A/C-HDAC2 complex may be a novel therapeutic target to slow down progression of progeria symptoms.
Collapse
Affiliation(s)
- Elisabetta Mattioli
- CNR Institute of Molecular Genetics, Unit of Bologna; Bologna Italy
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
| | - Davide Andrenacci
- CNR Institute of Molecular Genetics, Unit of Bologna; Bologna Italy
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
| | - Cecilia Garofalo
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
- CRS Development of Biomolecular Therapies, Experimental Oncology Lab; Rizzoli Institute; Bologna Italy
| | - Sabino Prencipe
- CNR Institute of Molecular Genetics, Unit of Bologna; Bologna Italy
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
| | - Katia Scotlandi
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
- CRS Development of Biomolecular Therapies, Experimental Oncology Lab; Rizzoli Institute; Bologna Italy
| | - Daniel Remondini
- Department of Physics and Astronomy; University of Bologna; Bologna Italy
| | - Davide Gentilini
- Centre for Biomedical Research and Technologies; Italian Auxologic Institute, IRCCS; Milan Italy
| | - Anna Maria Di Blasio
- Centre for Biomedical Research and Technologies; Italian Auxologic Institute, IRCCS; Milan Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies; Pasteur Institute Italy; Cenci-Bolognetti Foundation; Sapienza University of Rome; Rome Italy
| | - Emanuela Scarano
- Pediatric Endocrinology and Rare Diseases Unit; University of Bologna; Bologna Italy
| | - Lucia Cicchilitti
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies; IRCCS - Regina Elena National Cancer Institute; Rome Italy
| | - Giulia Piaggio
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies; IRCCS - Regina Elena National Cancer Institute; Rome Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies; Pasteur Institute Italy; Cenci-Bolognetti Foundation; Sapienza University of Rome; Rome Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics, Unit of Bologna; Bologna Italy
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
| |
Collapse
|
46
|
Yoon S, Kook T, Min HK, Kwon DH, Cho YK, Kim M, Shin S, Joung H, Jeong SH, Lee S, Kang G, Park Y, Kim YS, Ahn Y, McMullen JR, Gergs U, Neumann J, Kim KK, Kim J, Nam KI, Kim YK, Kook H, Eom GH. PP2A negatively regulates the hypertrophic response by dephosphorylating HDAC2 S394 in the heart. Exp Mol Med 2018; 50:1-14. [PMID: 30050113 PMCID: PMC6062565 DOI: 10.1038/s12276-018-0121-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/20/2018] [Accepted: 03/27/2018] [Indexed: 12/23/2022] Open
Abstract
Cardiac hypertrophy occurs in response to increased hemodynamic demand and can progress to heart failure. Identifying the key regulators of this process is clinically important. Though it is thought that the phosphorylation of histone deacetylase (HDAC) 2 plays a crucial role in the development of pathological cardiac hypertrophy, the detailed mechanism by which this occurs remains unclear. Here, we performed immunoprecipitation and peptide pull-down assays to characterize the functional complex of HDAC2. Protein phosphatase (PP) 2 A was confirmed as a binding partner of HDAC2. PPP2CA, the catalytic subunit of PP2A, bound to HDAC2 and prevented its phosphorylation. Transient overexpression of PPP2CA specifically regulated both the phosphorylation of HDAC2 S394 and hypertrophy-associated HDAC2 activation. HDAC2 S394 phosphorylation was increased in a dose-dependent manner by PP2A inhibitors. Hypertrophic stresses, such as phenylephrine in vitro or pressure overload in vivo, caused PPP2CA to dissociate from HDAC2. Forced expression of PPP2CA negatively regulated the hypertrophic response, but PP2A inhibitors provoked hypertrophy. Adenoviral delivery of a phosphomimic HDAC2 mutant, adenovirus HDAC2 S394E, successfully blocked the anti-hypertrophic effect of adenovirus-PPP2CA, implicating HDAC2 S394 phosphorylation as a critical event for the anti-hypertrophic response. PPP2CA transgenic mice were protected against isoproterenol-induced cardiac hypertrophy and subsequent cardiac fibrosis, whereas simultaneous expression of HDAC2 S394E in the heart did induce hypertrophy. Taken together, our results suggest that PP2A is a critical regulator of HDAC2 activity and pathological cardiac hypertrophy and is a promising target for future therapeutic interventions.
Collapse
Affiliation(s)
- Somy Yoon
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Taewon Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Hyun-Ki Min
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Basic Research Laboratory for Cardiac Remodeling Research Laboratory, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Duk-Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Basic Research Laboratory for Cardiac Remodeling Research Laboratory, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Young Kuk Cho
- Department of Pediatrics, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Mira Kim
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Sera Shin
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Basic Research Laboratory for Cardiac Remodeling Research Laboratory, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Hosouk Joung
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Basic Research Laboratory for Cardiac Remodeling Research Laboratory, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Seung Hoon Jeong
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Sumin Lee
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Gaeun Kang
- Division of Clinical Pharmacology, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Yunchul Park
- Division of Trauma Surgery, Department of Surgery, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Yong Sook Kim
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Ulrich Gergs
- Institute of Pharmacology and Toxicology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06097, Halle, Germany
| | - Joachim Neumann
- Institute of Pharmacology and Toxicology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06097, Halle, Germany
| | - Kyung Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Jungchul Kim
- Division of Trauma Surgery, Department of Surgery, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Kwang-Il Nam
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Young-Kook Kim
- Basic Research Laboratory for Cardiac Remodeling Research Laboratory, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.,Department of Biochemistry, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea. .,Basic Research Laboratory for Cardiac Remodeling Research Laboratory, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| | - Gwang Hyeon Eom
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea. .,Medical Research Center for Gene Regulation, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| |
Collapse
|
47
|
The roles of bile acids and applications of microencapsulation technology in treating Type 1 diabetes mellitus. Ther Deliv 2018; 8:401-409. [PMID: 28530150 DOI: 10.4155/tde-2017-0010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease characterized by the loss of glycemic control. Recent studies have shown significant inflammation and disturbed bile acid homeostasis, associated with T1DM. Bile acids are endogenously produced as a result of cholesterol catabolism in the liver and solely metabolized by gut microflora. This review investigates their potential oral delivery in T1DM using targeted delivery and encapsulation technologies. A sensitive and selective search was carried out using different search engines and databases. Keywords used included diabetes mellitus, bile acids and inflammation. To conclude, bile acids have a significant impact on diabetes symptoms and, when microencapsulated, may be used as an adjunct therapy to supplement T1DM treatment.
Collapse
|
48
|
Jhelum P, Karisetty BC, Kumar A, Chakravarty S. Implications of Epigenetic Mechanisms and their Targets in Cerebral Ischemia Models. Curr Neuropharmacol 2018; 15:815-830. [PMID: 27964703 PMCID: PMC5652028 DOI: 10.2174/1570159x14666161213143907] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/07/2016] [Accepted: 12/09/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Understanding the complexities associated with the ischemic condition and identifying therapeutic targets in ischemia is a continued challenge in stroke biology. Emerging evidence reveals the potential involvement of epigenetic mechanisms in the incident and outcome of stroke, suggesting novel therapeutic options of targeting different molecules related to epigenetic regulation. OBJECTIVE This review summarizes our current understanding of ischemic pathophysiology, describes various in vivo and in vitro models of ischemia, and examines epigenetic modifications associated with the ischemic condition. METHOD We focus on microRNAs, DNA methylation, and histone modifying enzymes, and present how epigenetic studies are revealing novel drug target candidates in stroke. CONCLUSION Finally, we discuss emerging approaches for the prevention and treatment of stroke and post-stroke effects using pharmacological interventions with a wide therapeutic window.
Collapse
Affiliation(s)
- Priya Jhelum
- Chemical Biology, CSIR, Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Bhanu C Karisetty
- Chemical Biology, CSIR, Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Arvind Kumar
- CSIR, Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad 500007, India
| | - Sumana Chakravarty
- Chemical Biology, CSIR-Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad-500007, India
| |
Collapse
|
49
|
Wang MY, Lin ZR, Cao Y, Zheng LS, Peng LX, Sun R, Meng DF, Xie P, Yang JP, Cao L, Xu L, Huang BJ, Qian CN. PDZ binding kinase (PBK) is a theranostic target for nasopharyngeal carcinoma: driving tumor growth via ROS signaling and correlating with patient survival. Oncotarget 2018; 7:26604-16. [PMID: 27049917 PMCID: PMC5042002 DOI: 10.18632/oncotarget.8445] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 02/19/2016] [Indexed: 12/11/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is well known as one of the most common malignancies in southern China and Southeast Asia. However, the mechanisms underlying NPC progression remain poorly understood. Herein, through overlapping the differentially expressed genes from 3 microarray data sets with the human kinome, we identified PBK, a serine-threonine kinase, is highly upregulated and has not been intensively investigated in NPC. PBK was required for malignant phenotypes of NPC, as PBK depletion by RNAi and inhibition by specific inhibitor HI-TOPK-032 obviously reduced cell proliferation and xenograft tumor growth in mice. Moreover, we determined that targeting PBK could accelerate apoptosis by inducing ROS that activates JNK/p38 signaling pathway. In NPC patients, elevated PBK expression in primary tumor positively correlated to clinical severity such as advanced T stage, high death risk and disease progression, and it could serve as an unfavorable independent indicator of overall survival and disease-free survival. Altogether, our results indicate that PBK is a novel significant regulator of NPC progression and a potential therapeutic target for NPC patients.
Collapse
Affiliation(s)
- Meng-Yao Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhi-Rui Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yun Cao
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Li-Sheng Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Li-Xia Peng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Dong-Fang Meng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ping Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jun-Ping Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Li Cao
- Department of Pharmacy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Liang Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Bi-Jun Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
50
|
Brain insulin resistance impairs hippocampal synaptic plasticity and memory by increasing GluA1 palmitoylation through FoxO3a. Nat Commun 2017; 8:2009. [PMID: 29222408 PMCID: PMC5722929 DOI: 10.1038/s41467-017-02221-9] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 11/13/2017] [Indexed: 11/08/2022] Open
Abstract
High-fat diet (HFD) and metabolic diseases cause detrimental effects on hippocampal synaptic plasticity, learning, and memory through molecular mechanisms still poorly understood. Here, we demonstrate that HFD increases palmitic acid deposition in the hippocampus and induces hippocampal insulin resistance leading to FoxO3a-mediated overexpression of the palmitoyltransferase zDHHC3. The excess of palmitic acid along with higher zDHHC3 levels causes hyper-palmitoylation of AMPA glutamate receptor subunit GluA1, hindering its activity-dependent trafficking to the plasma membrane. Accordingly, AMPAR current amplitudes and, more importantly, their potentiation underlying synaptic plasticity were inhibited, as well as hippocampal-dependent memory. Hippocampus-specific silencing of Zdhhc3 and, interestingly enough, intranasal injection of the palmitoyltransferase inhibitor, 2-bromopalmitate, counteract GluA1 hyper-palmitoylation and restore synaptic plasticity and memory in HFD mice. Our data reveal a key role of FoxO3a/Zdhhc3/GluA1 axis in the HFD-dependent impairment of cognitive function and identify a novel mechanism underlying the cross talk between metabolic and cognitive disorders.
Collapse
|