1
|
Chen Z, Lu Y, Wang Y, Wang Q, Yu L, Liu J. Natural Products Targeting Tau Protein Phosphorylation: A Promising Therapeutic Avenue for Alzheimer's Disease. PLANTA MEDICA 2025. [PMID: 40086889 DOI: 10.1055/a-2536-8919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder characterized by tau protein hyperphosphorylation and neurofibrillary tangle formation, which are central to its pathogenesis. This review focuses on the therapeutic potential of natural products in targeting tau phosphorylation, a key factor in Alzheimer's disease progression. It comprehensively summarizes current research on various natural compounds, including flavonoids, alkaloids, saponins, polysaccharides, phenols, phenylpropanoids, and terpenoids, highlighting their multitarget mechanisms, such as modulating kinases and phosphatases. The ability of these compounds to mitigate oxidative stress, inflammation, and tau pathology while enhancing cognitive function underscores their value as potential anti-Alzheimer's disease therapeutics. By integrating recent advances in extraction methods, pharmacological studies, and artificial intelligence-driven screening technologies, this review provides a valuable reference for future research and development of natural product-based interventions for Alzheimer's disease.
Collapse
Affiliation(s)
- Ziying Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan Lu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiyun Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangwen Yu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, China
| |
Collapse
|
2
|
Yu C, Xiang Y, Zhang M, Wen J, Duan X, Wang L, Deng G, Fang P. Glycyrrhizic Acid Alleviates Semen Strychni-Induced Neurotoxicity Through the Inhibition of HMGB1 Phosphorylation and Inflammatory Responses. J Neuroimmune Pharmacol 2024; 19:21. [PMID: 38771510 PMCID: PMC11108907 DOI: 10.1007/s11481-024-10128-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
The neurotoxicity of Semen Strychni has been reported recently in several clinical cases. Therefore, this study was conducted to investigate the role of HMGB1 in a model of neurotoxicity induced by Semen Strychni and to assess the potential alleviating effects of glycyrrhizic acid (GA), which is associated with the regulation of HMGB1 release. Forty-eight SD rats were intraperitoneally injected with Semen Strychni extract (175 mg/kg), followed by oral administration of GA (50 mg/kg) for four days. After treatment of SS and GA, neuronal degeneration, apoptosis, and necrosis were observed via histopathological examination. Inflammatory cytokines (TNF-α and IL-1β), neurotransmitter associated enzymes (MAO and AChE), serum HMGB1, nuclear and cytoplasmic HMGB1/ph-HMGB1, and the interaction between PP2A, PKC, and HMGB1 were evaluated. The influence of the MAPK pathway was also examined. As a result, this neurotoxicity was characterized by neuronal degeneration and apoptosis, the induction of pro-inflammatory cytokines, and a reduction in neurotransmitter-metabolizing enzymes. In contrast, GA treatment significantly ameliorated the abovementioned effects and alleviated nerve injury. Furthermore, Semen Strychni promoted HMGB1 phosphorylation and its translocation between the nucleus and cytoplasm, thereby activating the NF-κB and MAPK pathways, initiating various inflammatory responses. Our experiments demonstrated that GA could partially reverse these effects. In summary, GA acid alleviated Semen Strychni-induced neurotoxicity, possibly by inhibiting HMGB1 phosphorylation and preventing its release from the cell.
Collapse
Affiliation(s)
- Changwei Yu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, China
| | - Yalan Xiang
- Department of Pharmacy, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Min Zhang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, China
| | - Jing Wen
- Department of Pharmacy, the Third Hospital of Changsha, Changsha, 410015, China
| | - Xiaoyu Duan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, China
| | - Lu Wang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, China
| | - Gongying Deng
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, China
| | - Pingfei Fang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, China.
| |
Collapse
|
3
|
Dulski J, Soto-Beasley AI, Uitti RJ, Wszolek ZK, Ross OA. PTPA variants are rare in early-onset and familial Parkinson's disease. Brain 2023; 146:e125-e127. [PMID: 37448355 DOI: 10.1093/brain/awad244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/23/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023] Open
Affiliation(s)
- Jarosław Dulski
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
- Division of Neurological and Psychiatric Nursing, Faculty of Health Sciences, Medical University of Gdansk, 80-211 Gdansk, Poland
- Neurology Department, St Adalbert Hospital, Copernicus PL Ltd., 80-462 Gdansk, Poland
| | | | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
4
|
Nematullah M, Rashid F, Nimker S, Khan F. Protein Phosphatase 2A Regulates Phenotypic and Metabolic Alteration of Microglia Cells in HFD-Associated Vascular Dementia Mice via TNF-α/Arg-1 Axis. Mol Neurobiol 2023; 60:4049-4063. [PMID: 37017907 DOI: 10.1007/s12035-023-03324-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/20/2023] [Indexed: 04/06/2023]
Abstract
Protein phosphatase 2A (PP2A), the activity of which is dictated by the composition of its regulatory subunit, is strongly related to the progression of neurodegenerative disease. The potential role of PP2A on the phenotypic transition of microglial cells under obese conditions is poorly explored. An understanding of the role of PP2A and identification of regulatory subunits contributing to microglial phenotypic transitions in obese condition may serve as a therapeutic target for obesity-associated neurodegeneration. C57BL/6 mice were exposed to obese-associated vascular dementia conditions by performing unilateral common carotid artery occlusion on obese mice of microglial polarization and PP2A activity using flow cytometry, real-time PCR, western blotting, and immunoprecipitation enzymatic assay, followed identifications of PP2A regulatory subunits using LCMS and RT-PCR. Chronic HFD feeding significantly increased the populations of infiltrated macrophages, showing a high percentage of CD86+ in VaD mice, and the expression of pro-inflammatory cytokines, and we observed that PP2A modulates metabolic reprogramming of microglia by regulating OXPHOS/ECAR activity. Using Co-IP and LCMS, we identified the six specific regulatory subunits, namely PPP2R2A, PPP2R2D, PPP2R5B, PPP2R5C, PPP2R5D, and PPP2R5E, that are associated with microglial-activation during obesity-associated-VaD. Interestingly, pharmacological up-regulation of PP2A more significantly suppressed the expression of TNF-alpha than other pro-inflammatory-cytokines and increased the expression of Arginase-1, suggesting that PP2A modulates microglial-phenotypic transitions through TNF-α/Arg-1 axis. Our present findings demonstrate microglial polarization in HFD associated with VaD, and point towards a therapeutic target by providing specific PP2A regulatory-subunits implicated in microglial activation during obesity-related-vascular-dementia.
Collapse
Affiliation(s)
- Md Nematullah
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Faraz Rashid
- Department of Neurology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Shwetanjali Nimker
- Application Scientist, BD Biosciences India Pvt. Ltd, Jamia Hamdard, New Delhi, 110062, India
| | - Farah Khan
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
5
|
Khan H, Kaur Grewal A, Gurjeet Singh T. Mitochondrial dynamics related neurovascular approaches in cerebral ischemic injury. Mitochondrion 2022; 66:54-66. [DOI: 10.1016/j.mito.2022.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/14/2022] [Accepted: 08/02/2022] [Indexed: 12/30/2022]
|
6
|
Eun JD, Jimenez H, Adrien L, Wolin A, Marambaud P, Davies P, Koppel JL. Anesthesia promotes acute expression of genes related to Alzheimer's disease and latent tau aggregation in transgenic mouse models of tauopathy. Mol Med 2022; 28:83. [PMID: 35858831 PMCID: PMC9297560 DOI: 10.1186/s10020-022-00506-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
Background Exposure to anesthesia in the elderly might increase the risk of dementia. Although the mechanism underlying the association is uncertain, anesthesia has been shown to induce acute tau hyperphosphorylation in preclinical models. We sought to investigate the impact of anesthesia on gene expression and on acute and long-term changes in tau biochemistry in transgenic models of tauopathy in order to better understand how anesthesia influences the pathophysiology of dementia. Methods We exposed mice with over-expressed human mutant tau (P301L and hyperdopaminergic COMTKO/P301L) to two hours of isoflurane and compared anesthetized mice to controls at several time points. We evaluated tau hyperphosphorylation with quantitative high-sensitivity enzyme-linked immunosorbent assay and performed differential expression and functional transcriptome analyses following bulk mRNA-sequencing. Results Anesthesia induced acute hyperphosphorylation of tau at epitopes related to Alzheimer’s disease (AD) in both P301L-based models. Anesthesia was associated with differential expression of genes in the neurodegenerative pathways (e.g., AD-risk genes ApoE and Trem2) and thermogenesis pathway, which is related to both mammalian hibernation and tau phosphorylation. One and three months after anesthesia, hyperphosphorylated tau aggregates were increased in the anesthetized mice. Conclusions Anesthesia may influence the expression of AD-risk genes and induce biochemical changes in tau that promote aggregation even after single exposure. Further preclinical and human studies are necessary to establish the relevance of our transcriptomic and biochemical findings in these preclinical models to the pathogenesis of dementia following anesthesia. Trial registration: Not applicable. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00506-4.
Collapse
Affiliation(s)
- John David Eun
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.,Litwin-Zucker Research Center for the Study of Alzheimer's Disease, Feinstein Institutes for Medical Research, 350 Community Drive, 4th floor, Manhasset, NY, 11030, USA
| | - Heidy Jimenez
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, Feinstein Institutes for Medical Research, 350 Community Drive, 4th floor, Manhasset, NY, 11030, USA
| | - Leslie Adrien
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, Feinstein Institutes for Medical Research, 350 Community Drive, 4th floor, Manhasset, NY, 11030, USA
| | - Adam Wolin
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, Feinstein Institutes for Medical Research, 350 Community Drive, 4th floor, Manhasset, NY, 11030, USA
| | - Philippe Marambaud
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, Feinstein Institutes for Medical Research, 350 Community Drive, 4th floor, Manhasset, NY, 11030, USA
| | - Peter Davies
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, Feinstein Institutes for Medical Research, 350 Community Drive, 4th floor, Manhasset, NY, 11030, USA
| | - Jeremy L Koppel
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, Feinstein Institutes for Medical Research, 350 Community Drive, 4th floor, Manhasset, NY, 11030, USA. .,Zucker Hillside Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA.
| |
Collapse
|
7
|
Rroji M, Figurek A, Viggiano D, Capasso G, Spasovski G. Phosphate in the Context of Cognitive Impairment and Other Neurological Disorders Occurrence in Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms23137362. [PMID: 35806367 PMCID: PMC9266940 DOI: 10.3390/ijms23137362] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 02/01/2023] Open
Abstract
The nervous system and the kidneys are linked under physiological states to maintain normal body homeostasis. In chronic kidney disease (CKD), damaged kidneys can impair the central nervous system, including cerebrovascular disease and cognitive impairment (CI). Recently, kidney disease has been proposed as a new modifiable risk factor for dementia. It is reported that uremic toxins may have direct neurotoxic (astrocyte activation and neuronal death) and/or indirect action through vascular effects (cerebral endothelial dysfunction, calcification, and inflammation). This review summarizes the evidence from research investigating the pathophysiological effects of phosphate toxicity in the nervous system, raising the question of whether the control of hyperphosphatemia in CKD would lower patients’ risk of developing cognitive impairment and dementia.
Collapse
Affiliation(s)
- Merita Rroji
- Department of Nephrology, Faculty of Medicine, University of Medicine Tirana, 1001 Tirana, Albania
- Correspondence:
| | - Andreja Figurek
- Department of Internal Medicine, Medical Faculty, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina;
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland
| | - Davide Viggiano
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (D.V.); (G.C.)
- BioGeM, Institute of Molecular Biology and Genetics, 83031 Ariano Irpino, Italy
| | - Giovambattista Capasso
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (D.V.); (G.C.)
- BioGeM, Institute of Molecular Biology and Genetics, 83031 Ariano Irpino, Italy
| | - Goce Spasovski
- University Clinic for Nephrology, Medical Faculty, University St. Cyril and Methodius, 1000 Skopje, North Macedonia;
| |
Collapse
|
8
|
Mahamane Salissou MT, Razak MYA, Wang X, Magaji RA. The role of protein phosphatase 2A tau axis in traumatic brain injury therapy. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00223-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Traumatic brain injury (TBI) is a debilitating disorder due to trauma caused by an external mechanical force eventually leading to disruption in the normal function of the brain, with possible outcomes including permanent or temporary dysfunction of cognitive, physical, and psychosocial abilities. There have been several studies focusing on the search and innovation of neuroprotective agents that could have therapeutic relevance in TBI management. Due to its complexity, TBI is divided into two major components. The first initial event is known as the primary injury; it is a result of the mechanical insult itself and is known to be irreversible and resistant to a vast variety of therapeutics. The secondary event or secondary brain injury is viewed as a cellular injury that does not manifest immediately after the trauma but evolved after a delay period of hours or several days. This category of injury is known to respond favorably to different pharmacological treatment approaches.
Main body
Due to the complexity in the pathophysiology of the secondary injury, the therapeutic strategy needs to be in a multi-facets model and to have the ability to simultaneously regulate different cellular changes. Several studies have investigated in deep the possible approaches relying on natural compounds as an alternative therapeutic strategy for the management of TBI. In addition, many natural compounds have the potential to target numerous different components of the secondary injury including neuroinflammation, apoptosis, PP2A, tau, and Aβ among others. Here, we review past and current strategies in the therapeutic management of TBI, focusing on the PP2A-tau axis both in animal and human subjects. This review uncovers, in addition, a variety of compounds used in TBI therapy.
Conclusion
Despite beneficial therapeutic effects observed in animals for many compounds, studies are still needed to be conducted on human subjects to validate their therapeutic virtues. Furthermore, potential therapeutic virtues observed among studies might likely be dependent on the TBI animal model used and the type of induced injury. In addition, specificity and side effects are challenges in TBI therapy specifically which site of PP2A dysfunction to be targeted.
Collapse
|
9
|
Phosphate and Endothelial Function: How Sensing of Elevated Inorganic Phosphate Concentration Generates Signals in Endothelial Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:85-98. [DOI: 10.1007/978-3-030-91623-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
10
|
Viengkhou B, White MY, Cordwell SJ, Campbell IL, Hofer MJ. A novel phosphoproteomic landscape evoked in response to type I interferon in the brain and in glial cells. J Neuroinflammation 2021; 18:237. [PMID: 34656141 PMCID: PMC8520650 DOI: 10.1186/s12974-021-02277-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/16/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Type I interferons (IFN-I) are key responders to central nervous system infection and injury and are also increased in common neurodegenerative diseases. Their effects are primarily mediated via transcriptional regulation of several hundred interferon-regulated genes. In addition, IFN-I activate several kinases including members of the MAPK and PI3K families. Yet, how changes to the global protein phosphoproteome contribute to the cellular response to IFN-I is unknown. METHODS The cerebral phosphoproteome of mice with brain-targeted chronic production of the IFN-I, IFN-α, was obtained. Changes in phosphorylation were analyzed by ontology and pathway analysis and kinase enrichment predictions. These were verified by phenotypic analysis, immunohistochemistry and immunoblots. In addition, primary murine microglia and astrocytes, the brain's primary IFN-I-responding cells, were acutely treated with IFN-α and the global phosphoproteome was similarly analyzed. RESULTS We identified widespread protein phosphorylation as a novel mechanism by which IFN-I mediate their effects. In our mouse model for IFN-I-induced neurodegeneration, protein phosphorylation, rather than the proteome, aligned with the clinical hallmarks and pathological outcome, including impaired development, motor dysfunction and seizures. In vitro experiments revealed extensive and rapid IFN-I-induced protein phosphorylation in microglia and astrocytes. Response to acute IFN-I stimulation was independent of gene expression and mediated by a small number of kinase families. The changes in the phosphoproteome affected a diverse range of cellular processes and functional analysis suggested that this response induced an immediate reactive state and prepared cells for subsequent transcriptional responses. CONCLUSIONS Our studies reveal a hitherto unappreciated role for changes in the protein phosphorylation landscape in cellular responses to IFN-I and thus provide insights for novel diagnostic and therapeutic strategies for neurological diseases caused by IFN-I.
Collapse
Affiliation(s)
- Barney Viengkhou
- School of Life and Environmental Sciences, Charles Perkins Centre and Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Melanie Y White
- School of Life and Environmental Sciences, School of Medical Sciences, Charles Perkins Centre and Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Stuart J Cordwell
- School of Life and Environmental Sciences, School of Medical Sciences, Charles Perkins Centre and Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Iain L Campbell
- School of Life and Environmental Sciences, Charles Perkins Centre and Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Markus J Hofer
- School of Life and Environmental Sciences, Charles Perkins Centre and Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
11
|
Bryant JP, Levy A, Heiss J, Banasavadi-Siddegowda YK. Review of PP2A Tumor Biology and Antitumor Effects of PP2A Inhibitor LB100 in the Nervous System. Cancers (Basel) 2021; 13:cancers13123087. [PMID: 34205611 PMCID: PMC8235527 DOI: 10.3390/cancers13123087] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Central and peripheral nervous system tumors represent a heterogenous group of neoplasms which often demonstrate resistance to treatment. Given that these tumors are often refractory to conventional therapy, novel pharmaceutical regimens are needed for successfully treating this pathology. One such therapeutic is the serine/threonine phosphatase inhibitor, LB100. LB100 is a water-soluble competitive protein phosphtase inhibitor that has demonstrated antitumor effects in preclinical and clinical trials. In this review, we aim to summarize current evidence demonstrating the efficacy of LB100 as an inhibitor of nervous system tumors. Furthermore, we review the involvement of the well-studied phosphatase, protein phosphatase 2A, in oncogenic cell signaling pathways, neurophysiology, and neurodevelopment. Abstract Protein phosphatase 2A (PP2A) is a ubiquitous serine/threonine phosphatase implicated in a wide variety of regulatory cellular functions. PP2A is abundant in the mammalian nervous system, and dysregulation of its cellular functions is associated with myriad neurodegenerative disorders. Additionally, PP2A has oncologic implications, recently garnering attention and emerging as a therapeutic target because of the antitumor effects of a potent PP2A inhibitor, LB100. LB100 abrogation of PP2A is believed to exert its inhibitory effects on tumor progression through cellular chemo- and radiosensitization to adjuvant agents. An updated and unifying review of PP2A biology and inhibition with LB100 as a therapeutic strategy for targeting cancers of the nervous system is needed, as other reviews have mainly covered broader applications of LB100. In this review, we discuss the role of PP2A in normal cells and tumor cells of the nervous system. Furthermore, we summarize current evidence regarding the therapeutic potential of LB100 for treating solid tumors of the nervous system.
Collapse
Affiliation(s)
- Jean-Paul Bryant
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (J.-P.B.); (J.H.)
| | - Adam Levy
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - John Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (J.-P.B.); (J.H.)
| | - Yeshavanth Kumar Banasavadi-Siddegowda
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (J.-P.B.); (J.H.)
- Correspondence: ; Tel.: +1-301-451-0970
| |
Collapse
|
12
|
Seumen CHT, Grimm TM, Hauck CR. Protein phosphatases in TLR signaling. Cell Commun Signal 2021; 19:45. [PMID: 33882943 PMCID: PMC8058998 DOI: 10.1186/s12964-021-00722-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are critical sensors for the detection of potentially harmful microbes. They are instrumental in initiating innate and adaptive immune responses against pathogenic organisms. However, exaggerated activation of TLR receptor signaling can also be responsible for the onset of autoimmune and inflammatory diseases. While positive regulators of TLR signaling, such as protein serine/threonine kinases, have been studied intensively, only little is known about phosphatases, which counterbalance and limit TLR signaling. In this review, we summarize protein phosphorylation events and their roles in the TLR pathway and highlight the involvement of protein phosphatases as negative regulators at specific steps along the TLR-initiated signaling cascade. Then, we focus on individual phosphatase families, specify the function of individual enzymes in TLR signaling in more detail and give perspectives for future research. A better understanding of phosphatase-mediated regulation of TLR signaling could provide novel access points to mitigate excessive immune activation and to modulate innate immune signaling.![]() Video Abstract
Collapse
Affiliation(s)
- Clovis H T Seumen
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany
| | - Tanja M Grimm
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology, Universität Konstanz, 78457, Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457, Konstanz, Germany. .,Konstanz Research School Chemical Biology, Universität Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
13
|
Rivard RS, Morris JM, Youngman MJ. The PP2A/4/6 subfamily of phosphoprotein phosphatases regulates DAF-16 and confers resistance to environmental stress in postreproductive adult C. elegans. PLoS One 2020; 15:e0229812. [PMID: 33315870 PMCID: PMC7735605 DOI: 10.1371/journal.pone.0229812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 11/13/2020] [Indexed: 11/28/2022] Open
Abstract
Insulin and insulin-like growth factors are longevity determinants that negatively regulate Forkhead box class O (FoxO) transcription factors. In C. elegans mutations that constitutively activate DAF-16, the ortholog of mammalian FoxO3a, extend lifespan by two-fold. While environmental insults induce DAF-16 activity in younger animals, it also becomes activated in an age-dependent manner in the absence of stress, modulating gene expression well into late adulthood. The mechanism by which DAF-16 activity is regulated during aging has not been defined. Since phosphorylation of DAF-16 generally leads to its inhibition, we asked whether phosphatases might be necessary for its increased transcriptional activity in adult C. elegans. We focused on the PP2A/4/6 subfamily of phosphoprotein phosphatases, members of which had been implicated to regulate DAF-16 under low insulin signaling conditions but had not been investigated during aging in wildtype animals. Using reverse genetics, we functionally characterized all C. elegans orthologs of human catalytic, regulatory, and scaffolding subunits of PP2A/4/6 holoenzymes in postreproductive adults. We found that PP2A complex constituents PAA-1 and PPTR-1 regulate DAF-16 transcriptional activity during aging and that they cooperate with the catalytic subunit LET-92 to protect adult animals from ultraviolet radiation. PP4 complex members PPH-4.1/4.2, and SMK-1 also appear to regulate DAF-16 in an age-dependent manner, and together with PPFR-2 they contribute to innate immunity. Interestingly, SUR-6 but no other subunit of the PP2A complex was necessary for the survival of pathogen-infected animals. Finally, we found that PP6 complex constituents PPH-6 and SAPS-1 contribute to host defense during aging, apparently without affecting DAF-16 transcriptional activity. Our studies indicate that a set of PP2A/4/6 complexes protect adult C. elegans from environmental stress, thus preserving healthspan. Therefore, along with their functions in cell division and development, the PP2A/4/6 phosphatases also appear to play critical roles later in life.
Collapse
Affiliation(s)
- Rebecca S. Rivard
- Department of Biology, Villanova University, Villanova, PA, United States of America
| | - Julia M. Morris
- Department of Biology, Villanova University, Villanova, PA, United States of America
| | - Matthew J. Youngman
- Department of Biology, Villanova University, Villanova, PA, United States of America
- * E-mail:
| |
Collapse
|
14
|
Abbasian N, Bevington A, Burton JO, Herbert KE, Goodall AH, Brunskill NJ. Inorganic Phosphate (Pi) Signaling in Endothelial Cells: A Molecular Basis for Generation of Endothelial Microvesicles in Uraemic Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21196993. [PMID: 32977471 PMCID: PMC7583816 DOI: 10.3390/ijms21196993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Hyperphosphataemia increases cardiovascular mortality in patients with kidney disease. Direct effects of high inorganic phosphate (Pi) concentrations have previously been demonstrated on endothelial cells (ECs), including generation of procoagulant endothelial microvesicles (MVs). However, no mechanism directly sensing elevated intracellular Pi has ever been described in mammalian cells. Here, we investigated the hypothesis that direct inhibition by Pi of the phosphoprotein phosphatase PP2A fulfils this sensing role in ECs, culminating in cytoskeleton disruption and MV generation. ECs were treated with control (1 mM [Pi]) vs. high (2.5 mM [Pi]), a condition that drives actin stress fibre depletion and MV generation demonstrated by confocal microscopy of F-actin and NanoSight Nanoparticle tracking, respectively. Immuno-blotting demonstrated that high Pi increased p-Src, p-PP2A-C and p-DAPK-1 and decreased p-TPM-3. Pi at 100 μM directly inhibited PP2A catalytic activity. Inhibition of PP2A enhanced inhibitory phosphorylation of DAPK-1, leading to hypophosphorylation of Tropomyosin-3 at S284 and MV generation. p-Src is known to perform inhibitory phosphorylation on DAPK-1 but also on PP2A-C. However, PP2A-C can itself dephosphorylate (and therefore inhibit) p-Src. The direct inhibition of PP2A-C by Pi is, therefore, amplified by the feedback loop between PP2A-C and p-Src, resulting in further PP2A-C inhibition. These data demonstrated that PP2A/Src acts as a potent sensor and amplifier of Pi signals which can further signal through DAPK-1/Tropomyosin-3 to generate cytoskeleton disruption and generation of potentially pathological MVs.
Collapse
Affiliation(s)
- Nima Abbasian
- Department of Cardiovascular Sciences, University of Leicester, and Leicester NIHR Cardiovascular Biomedical Research Unit, Leicester LE3 9QP, UK; (J.O.B.); (K.E.H.); (A.H.G.); (N.J.B.)
- Correspondence: (N.A.); (A.B.); Tel.: +44-(0)116-246-0951 (A.B.)
| | - Alan Bevington
- Department of Cardiovascular Sciences, University of Leicester, and Leicester NIHR Cardiovascular Biomedical Research Unit, Leicester LE3 9QP, UK; (J.O.B.); (K.E.H.); (A.H.G.); (N.J.B.)
- Correspondence: (N.A.); (A.B.); Tel.: +44-(0)116-246-0951 (A.B.)
| | - James O. Burton
- Department of Cardiovascular Sciences, University of Leicester, and Leicester NIHR Cardiovascular Biomedical Research Unit, Leicester LE3 9QP, UK; (J.O.B.); (K.E.H.); (A.H.G.); (N.J.B.)
- Department of Nephrology, Leicester General Hospital, Leicester LE5 4PW, UK
| | - Karl E. Herbert
- Department of Cardiovascular Sciences, University of Leicester, and Leicester NIHR Cardiovascular Biomedical Research Unit, Leicester LE3 9QP, UK; (J.O.B.); (K.E.H.); (A.H.G.); (N.J.B.)
| | - Alison H. Goodall
- Department of Cardiovascular Sciences, University of Leicester, and Leicester NIHR Cardiovascular Biomedical Research Unit, Leicester LE3 9QP, UK; (J.O.B.); (K.E.H.); (A.H.G.); (N.J.B.)
| | - Nigel J. Brunskill
- Department of Cardiovascular Sciences, University of Leicester, and Leicester NIHR Cardiovascular Biomedical Research Unit, Leicester LE3 9QP, UK; (J.O.B.); (K.E.H.); (A.H.G.); (N.J.B.)
- Department of Nephrology, Leicester General Hospital, Leicester LE5 4PW, UK
| |
Collapse
|
15
|
Frohner IE, Mudrak I, Kronlachner S, Schüchner S, Ogris E. Antibodies recognizing the C terminus of PP2A catalytic subunit are unsuitable for evaluating PP2A activity and holoenzyme composition. Sci Signal 2020; 13:13/616/eaax6490. [PMID: 31992581 DOI: 10.1126/scisignal.aax6490] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The methyl-esterification of the C-terminal leucine of the protein phosphatase 2A (PP2A) catalytic (C) subunit is essential for the assembly of specific trimeric PP2A holoenzymes, and this region of the C subunit also contains two threonine and tyrosine phosphorylation sites. Most commercial antibodies-including the monoclonal antibody 1D6 that is part of a frequently used, commercial phosphatase assay kit-are directed toward the C terminus of the C subunit, raising questions as to their ability to recognize methylated and phosphorylated forms of the enzyme. Here, we tested several PP2A C antibodies, including monoclonal antibodies 1D6, 7A6, G-4, and 52F8 and the polyclonal antibody 2038 for their ability to specifically detect PP2A in its various modified forms, as well as to coprecipitate regulatory subunits. The tested antibodies preferentially recognized the nonmethylated form of the enzyme, and they did not coimmunoprecipitate trimeric holoenzymes containing the regulatory subunits B or B', an issue that precludes their use to monitor PP2A holoenzyme activity. Furthermore, some of the antibodies also recognized the phosphatase PP4, demonstrating a lack of specificity for PP2A. Together, these findings suggest that reinterpretation of the data generated by using these reagents is required.
Collapse
Affiliation(s)
- Ingrid E Frohner
- Center for Medical Biochemistry, Max Perutz Labs, Vienna BioCenter, Medical University of Vienna, Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Ingrid Mudrak
- Center for Medical Biochemistry, Max Perutz Labs, Vienna BioCenter, Medical University of Vienna, Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Stephanie Kronlachner
- Center for Medical Biochemistry, Max Perutz Labs, Vienna BioCenter, Medical University of Vienna, Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Stefan Schüchner
- Center for Medical Biochemistry, Max Perutz Labs, Vienna BioCenter, Medical University of Vienna, Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Egon Ogris
- Center for Medical Biochemistry, Max Perutz Labs, Vienna BioCenter, Medical University of Vienna, Dr. Bohr-Gasse 9, A-1030 Vienna, Austria.
| |
Collapse
|
16
|
Xu J, Zhang L, Xu Y, Zhang H, Gao J, Wang Q, Tian Z, Xuan L, Chen H, Wang Y. PP2A Facilitates Porcine Reproductive and Respiratory Syndrome Virus Replication by Deactivating irf3 and Limiting Type I Interferon Production. Viruses 2019; 11:v11100948. [PMID: 31618847 PMCID: PMC6832233 DOI: 10.3390/v11100948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Protein phosphatase 2A (PP2A), a major serine/threonine phosphatase in mammalian cells, is known to regulate the kinase-driven intracellular signaling pathways. Emerging evidences have shown that the PP2A phosphatase functions as a bona-fide therapeutic target for anticancer therapy, but it is unclear whether PP2A affects a porcine reproductive and respiratory syndrome virus infection. In the present study, we demonstrated for the first time that inhibition of PP2A activity by either inhibitor or small interfering RNA duplexes in target cells significantly reduced their susceptibility to porcine reproductive and respiratory syndrome virus (PRRSV) infection. Further analysis revealed that inhibition of PP2A function resulted in augmented production of type I interferon (IFN). The mechanism is that inhibition of PP2A activity enhances the levels of phosphorylated interferon regulatory factor 3, which activates the transcription of IFN-stimulated genes. Moreover, inhibition of PP2A activity mainly blocked PRRSV replication in the early stage of viral life cycle, after virus entry but before virus release. Using type I IFN receptor 2 specific siRNA in combination with PP2A inhibitor, we confirmed that the effect of PP2A on viral replication within target cells was an interferon-dependent manner. Taken together, these findings demonstrate that PP2A serves as a negative regulator of host cells antiviral responses and provides a novel therapeutic target for virus infection.
Collapse
Affiliation(s)
- Jiayu Xu
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Lu Zhang
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yunfei Xu
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - He Zhang
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Junxin Gao
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Qian Wang
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Zhijun Tian
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Lv Xuan
- Department of public health policy, University of California, Irvine, CA 92697, USA
| | - Hongyan Chen
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yue Wang
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| |
Collapse
|
17
|
Javadpour P, Dargahi L, Ahmadiani A, Ghasemi R. To be or not to be: PP2A as a dual player in CNS functions, its role in neurodegeneration, and its interaction with brain insulin signaling. Cell Mol Life Sci 2019; 76:2277-2297. [PMID: 30874837 PMCID: PMC11105459 DOI: 10.1007/s00018-019-03063-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/16/2019] [Accepted: 03/07/2019] [Indexed: 12/26/2022]
Abstract
Accumulating evidence has reached the consensus that the balance of phosphorylation state of signaling molecules is a pivotal point in the regulation of cell signaling. Therefore, characterizing elements (kinases-phosphatases) in the phosphorylation balance are at great importance. However, the role of phosphatase enzymes is less investigated than kinase enzymes. PP2A is a member of serine/threonine protein phosphatase that its imbalance has been reported in neurodegenerative diseases. Therefore, we reviewed the superfamily of phosphatases and more specifically PP2A, its regulation, and physiological functions participate in CNS. Thereafter, we discussed the latest findings about PP2A dysregulation in Alzheimer and Parkinson diseases and possible interplay between this phosphatase and insulin signaling pathways. Finally, activating/inhibitory modulators for PP2A activity as well as experimental methods for PP2A study have been reviewed.
Collapse
Affiliation(s)
- Pegah Javadpour
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Wang J, Xie R, Kou X, Liu Y, Qi C, Liu R, You W, Gao J, Gao X. A protein phosphatase 2A deficit in the hippocampal CA1 area impairs memory extinction. Mol Brain 2019; 12:51. [PMID: 31113458 PMCID: PMC6528246 DOI: 10.1186/s13041-019-0469-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/30/2019] [Indexed: 01/17/2023] Open
Abstract
Protein phosphorylation plays an important role in learning and memory. Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase involved in the regulation of neural synaptic plasticity. Here, to determine if PP2A is necessary for successful learning and memory, we have utilized a Tg (Camk2a-cre) T29–2Stl mice to specific knock down the expression of hippocampal PP2A in mice. By analysing behavioural, we observed that loss of PP2A in the hippocampal CA1 area did not affect the formation of memory but impaired contextual fear memory extinction. We use the electrophysiological recording to find the synaptic mechanisms. The results showed that the basic synapse transmission and synaptic plasticity of PP2A conditional knockout (CKO) mice were impaired. Moreover, PP2A CKO mice exhibited a saturating long-term potentiation inducted by strong theta burst stimulation but no depotentiation after low-frequency stimulation. Taken together, our results provide the evidence that PP2A is involved in synaptic transmission and hippocampus-dependent memory extinction.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ran Xie
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xiaolin Kou
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yu Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Cui Qi
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Rui Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Weiyan You
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jun Gao
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Xiang Gao
- Model Animal Research Center and MOE Key Laboratory of Model Animals for Disease Study, Nanjing University, Nanjing, 210093, Jiangsu, China.
| |
Collapse
|
19
|
Adhikari B, De Silva B, Molina JA, Allen A, Peck SH, Lee SY. Neuronal ceroid lipofuscinosis related ER membrane protein CLN8 regulates PP2A activity and ceramide levels. Biochim Biophys Acta Mol Basis Dis 2019; 1865:322-328. [DOI: 10.1016/j.bbadis.2018.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/09/2018] [Accepted: 11/15/2018] [Indexed: 01/02/2023]
|
20
|
Singh S, Jangid RK, Crowder A, Groves AK. Foxi3 transcription factor activity is mediated by a C-terminal transactivation domain and regulated by the Protein Phosphatase 2A (PP2A) complex. Sci Rep 2018; 8:17249. [PMID: 30467319 PMCID: PMC6250667 DOI: 10.1038/s41598-018-35390-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/02/2018] [Indexed: 01/20/2023] Open
Abstract
The Forkhead box (FOX) family consists of at least 19 subgroups of transcription factors which are characterized by the presence of an evolutionary conserved ‘forkhead’ or ‘winged-helix’ DNA-binding domain. Despite having a conserved core DNA binding domain, FOX proteins display remarkable functional diversity and are involved in many developmental and cell specific processes. In the present study, we focus on a poorly characterized member of the Forkhead family, Foxi3, which plays a critical role in the development of the inner ear and jaw. We show that Foxi3 contains at least two important functional domains, a nuclear localization sequence (NLS) and a C-terminal transactivation domain (TAD), and that it directly binds its targets in a sequence specific manner. We also show that the transcriptional activity of Foxi3 is regulated by phosphorylation, and that the activity of Foxi3 can be attenuated by its physical interaction with the protein phosphatase 2A (PP2A) complex.
Collapse
Affiliation(s)
- Sunita Singh
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Rahul K Jangid
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Alyssa Crowder
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA. .,Program in Developmental Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
Murcia Pienkowski V, Kucharczyk M, Młynek M, Szczałuba K, Rydzanicz M, Poszewiecka B, Skórka A, Sykulski M, Biernacka A, Koppolu AA, Posmyk R, Walczak A, Kosińska J, Krajewski P, Castaneda J, Obersztyn E, Jurkiewicz E, Śmigiel R, Gambin A, Chrzanowska K, Krajewska-Walasek M, Płoski R. Mapping of breakpoints in balanced chromosomal translocations by shallow whole-genome sequencing points to EFNA5, BAHD1 and PPP2R5E as novel candidates for genes causing human Mendelian disorders. J Med Genet 2018; 56:104-112. [PMID: 30352868 DOI: 10.1136/jmedgenet-2018-105527] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/21/2018] [Accepted: 09/22/2018] [Indexed: 11/04/2022]
Abstract
BACKGROUND Mapping the breakpoints in de novo balanced chromosomal translocations (BCT) in symptomatic individuals provides a unique opportunity to identify in an unbiased way the likely causative genetic defect and thus find novel human disease candidate genes. Our aim was to fine-map breakpoints of de novo BCTs in a case series of nine patients. METHODS Shallow whole-genome mate pair sequencing (SGMPS) together with long-range PCR and Sanger sequencing. In one case (BCT disrupting BAHD1 and RET) cDNA analysis was used to verify expression of a fusion transcript in cultured fibroblasts. RESULTS In all nine probands 11 disrupted genes were found, that is, EFNA5, EBF3, LARGE, PPP2R5E, TXNDC5, ZNF423, NIPBL, BAHD1, RET, TRPS1 and SLC4A10. Five subjects had translocations that disrupted genes with so far unknown (EFNA5, BAHD1, PPP2R5E, TXNDC5) or poorly delineated impact on the phenotype (SLC4A10, two previous reports of BCT disrupting the gene). The four genes with no previous disease associations (EFNA5, BAHD1, PPP2R5E, TXNDC5), when compared with all human genes by a bootstrap test, had significantly higher pLI (p<0.017) and DOMINO (p<0.02) scores indicating enrichment in genes likely to be intolerant to single copy damage. Inspection of individual pLI and DOMINO scores, and local topologically associating domain structure suggested that EFNA5, BAHD1 and PPP2R5E were particularly good candidates for novel disease loci. The pathomechanism for BAHD1 may involve deregulation of expression due to fusion with RET promoter. CONCLUSION SGMPS in symptomatic carriers of BCTs is a powerful approach to delineate novel human gene-disease associations.
Collapse
Affiliation(s)
- Victor Murcia Pienkowski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marzena Kucharczyk
- Department of Medical Genetics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Marlena Młynek
- Department of Medical Genetics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Krzysztof Szczałuba
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | | | - Barbara Poszewiecka
- Faculty of Mathematics, Informatics and Mechanics, Institute of Informatics, University of Warsaw, Warsaw, Poland
| | - Agata Skórka
- Department of Medical Genetics, The Children's Memorial Health Institute, Warsaw, Poland.,Department of Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Sykulski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland.,genXone, Poznan, Poland
| | - Anna Biernacka
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Anna Koppolu
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Renata Posmyk
- Department of Clinical Genetics, Podlaskie Medical Center, Bialystok, Poland.,Department of Perinatology, Medical University of Bialystok, Bialystok, Poland
| | - Anna Walczak
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Kosińska
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Paweł Krajewski
- Department of Forensic Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jennifer Castaneda
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Ewa Obersztyn
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Elżbieta Jurkiewicz
- Department of Diagnostic Imaging, The Children's Memorial Health Institute, Warsaw, Poland
| | - Robert Śmigiel
- Department of Pediatrics and Rare Disorder, Wroclaw Medical University, Wroclaw, Poland
| | - Anna Gambin
- Faculty of Mathematics, Informatics and Mechanics, Institute of Informatics, University of Warsaw, Warsaw, Poland
| | - Krystyna Chrzanowska
- Department of Medical Genetics, The Children's Memorial Health Institute, Warsaw, Poland
| | | | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Guo S. Cancer driver mutations in endometriosis: Variations on the major theme of fibrogenesis. Reprod Med Biol 2018; 17:369-397. [PMID: 30377392 PMCID: PMC6194252 DOI: 10.1002/rmb2.12221] [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: 04/18/2018] [Revised: 06/03/2018] [Accepted: 06/24/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND One recent study reports cancer driver mutations in deep endometriosis, but its biological/clinical significance remains unclear. Since the natural history of endometriosis is essentially gradual progression toward fibrosis, it is thus hypothesized that the six driver genes reported to be mutated in endometriosis (the RP set) may play important roles in fibrogenesis but not necessarily malignant transformation. METHODS Extensive PubMed search to see whether RP and another set of driver genes not yet reported (NR) to be mutated in endometriosis have any roles in fibrogenesis. All studies reporting on the role of fibrogenesis of the genes in both RP and NR sets were retrieved and evaluated in this review. RESULTS All six RP genes were involved in various aspects of fibrogenesis as compared with only three NR genes. These nine genes can be anchored in networks linking with their upstream and downstream genes that are known to be aberrantly expressed in endometriosis, piecing together seemingly unrelated findings. CONCLUSIONS Given that somatic driver mutations can and do occur frequently in physiologically normal tissues, it is argued that these mutations in endometriosis are not necessarily synonymous with malignancy or premalignancy, but the result of enormous pressure for fibrogenesis.
Collapse
Affiliation(s)
- Sun‐Wei Guo
- Shanghai Obstetrics and Gynecology HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Female Reproductive Endocrine‐Related DiseasesShanghaiChina
| |
Collapse
|
23
|
Yin J, Li R, Liu W, Chen Y, Zhang X, Li X, He X, Duan C. Neuroprotective Effect of Protein Phosphatase 2A/Tristetraprolin Following Subarachnoid Hemorrhage in Rats. Front Neurosci 2018; 12:96. [PMID: 29535596 PMCID: PMC5835096 DOI: 10.3389/fnins.2018.00096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/06/2018] [Indexed: 11/25/2022] Open
Abstract
Early brain injury (EBI) following subarachnoid hemorrhage (SAH) can lead to inflammation and neuronal dysfunction. There is a need for effective strategies to mitigate these effects and improve the outcome of patients who experience SAH. The mRNA-destabilizing protein tristetraprolin (TTP) is an anti-inflammatory factor that induces the decay of cytokine transcripts and has been implicated in diseases such as glioma. However, the mechanism of action of TTP in EBI after SAH is unclear. The present study investigated the effects of TTP regulation via phosphorylation in a rat model of SAH by protein phosphatase (PP)2A, which is a pleiotropic enzyme complex with multiple substrate phospho-proteins. We hypothesized that inhibitory phosphorylation of TTP by PP2A would reduce neuroinflammation and apoptosis. To evaluate the function of each factor, the PP2A agonist FTY720, short interfering (si)RNAs targeting TTP and PP2A were administered to rats by intracerebroventricular injection 24 h before SAH. Rats were evaluated with SAH grade, neurological score, brain water content and by western blotting, and terminal deoxynucleotidyltransferase dUTP nick-end labeling. We found that endogenous PP2A and TTP levels were increased after SAH. FTY720 induced PP2A activation would lead to dephosphorylation and activation of TTP and decreased production of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-8. SiRNA-mediated TTP knockdown abolished anti-inflammatory effects of FTY720 treatment, indicating that PP2A was associated with TTP activation in vivo. Decreased TNF-α, IL-6, and IL-8 levels were associated with improvement of neurological function, reduction of brain edema, suppression of caspase-3, and up-regulation of B cell lymphoma-2. These results demonstrated that PP2A activation could enhance the anti-inflammatory and anti-apoptotic effects of TTP, by which it might shed light on the development of an effective therapeutic strategy against EBI following SAH.
Collapse
Affiliation(s)
- Jian Yin
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Departments of Neurosurgery, Hanghzou Red Cross Hospital, Hangzhou, China
| | - Ran Li
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenchao Liu
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yunchang Chen
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Zhang
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xifeng Li
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuying He
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chuanzhi Duan
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
Pajarillo EAB, Kim SH, Valeriano VD, Lee JY, Kang DK. Proteomic View of the Crosstalk between Lactobacillus mucosae and Intestinal Epithelial Cells in Co-culture Revealed by Q Exactive-Based Quantitative Proteomics. Front Microbiol 2017; 8:2459. [PMID: 29312173 PMCID: PMC5732961 DOI: 10.3389/fmicb.2017.02459] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/27/2017] [Indexed: 01/28/2023] Open
Abstract
Lactobacilli are bacteria that are beneficial to host health, but information on communication between Lactobacilli and host cells in the intestine is lacking. In this study, we examined the proteomes of the Lactobacillus mucosae strain LM1, as a model of beneficial bacteria, and the intestinal porcine epithelial cell line (IPEC-J2) after co-culture. Label-free proteomics demonstrated the high-throughput capability of the technique, and robust characterization of the functional profiles and changes in the bacteria and intestinal cells was achieved in pure and mixed cultures. After co-culture, we identified totals of 376 and 653 differentially expressed proteins in the LM1 and IPEC-J2 proteomes, respectively. The major proteomic changes in the LM1 strain occurred in the functional categories of transcription, general function, and translation, whereas those in IPEC-J2 cells involved metabolic and cellular processes, and cellular component organization/biogenesis. Among them, elongation factor Tu, glyceraldehyde 3-phosphate dehydrogenase, and phosphocarrier protein HPr, which are known to be involved in bacterial adhesion, were upregulated in LM1. In contrast, proteins involved in tight junction assembly, actin organization, and genetic information processing (i.e., histones and signaling pathways) were significantly upregulated in IPEC-J2 cells. Furthermore, we identified functional pathways that are possibly involved in host–microbe crosstalk and response. These findings will provide novel insights into host–bacteria communication and the molecular mechanism of probiotic establishment in the intestine.
Collapse
Affiliation(s)
| | - Sang Hoon Kim
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | | | - Ji Yoon Lee
- National Instrumentation Center for Environmental Management, Seoul National University, Seoul, South Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| |
Collapse
|