1
|
Cai H, Hou F, Wang Y, Wu L, Wang Z, Wu M, Wang X, Hölscher C. Mitochondrial Calcium Uniporter knockdown improves the viability of HT22 hippocampal neurons in Alzheimer's disease. Eur J Pharmacol 2025; 991:177347. [PMID: 39914782 DOI: 10.1016/j.ejphar.2025.177347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Alzheimer's disease (AD) is a degenerative disorder that poses a serious threat because it has no cure. Recently, mitochondrial dysfunction has been shown to directly promote the development of AD. An imbalance in mitochondrial calcium (mCa2+) homeostasis is an important cause of mitochondrial dysfunction. Abnormal expression of mitochondrial calcium uniporter (MCU), a key channel responsible for mCa2+ uptake, can induce an imbalance in mCa2+ homeostasis, ultimately leading to mitochondrial dysfunction. Importantly, we observed a much higher expression level of MCU in the hippocampus of amyloid precursor protein (APP)/presenilin 1 (PS1)/tau transgenic mice than that in the hippocampus of control mice (C57), suggesting that MCU may be a target for the development of treatments for AD. Our recent study revealed the neuroprotective effect of MCU knockdown on hippocampal neurons in APP/PS1/tau mice. In the present study, we used MCU knockdown to investigate the cellular mechanisms involved in amyloid-β (Aβ)1-42 and okadaic acid (OA) cell models of AD. We found that MCU knockdown could increase the survival and decrease the apoptosis of these two cell models by lowering mCa2+ overload, further increasing the mitochondrial membrane potential (MMP) and ultimately reducing the overproduction of reactive oxygen species (ROS). This study showed that MCU knockdown could increase the proliferation and viability of HT22 hippocampal neurons, which might explain the neuroprotective effect of MCU knockdown on AD, potentially leading to the development of novel and effective therapies for AD.
Collapse
Affiliation(s)
- Hongyan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Taiyuan, China.
| | - Fei Hou
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China.
| | - Yu Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China.
| | - Linhong Wu
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China.
| | - Zhaojun Wang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Taiyuan, China; Department of Physiology, Shanxi Medical University, Taiyuan, China.
| | - Meina Wu
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Taiyuan, China; Department of Physiology, Shanxi Medical University, Taiyuan, China.
| | - Xiao Wang
- Department of Psychiatry, First Clinical Medical College of Shanxi Medical University, Taiyuan, China.
| | - Christian Hölscher
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, China.
| |
Collapse
|
2
|
Liu Z, Zhang T, Ergashev A, Bo Z, Wang J, Shi F, Pan Z, Xie H, Chen G, Ma F, Kong L. CIP2A inhibitors TD52 and Ethoxysanguinarine promote macrophage autophagy and alleviates acute pancreatitis by modulating the AKT-mTOR pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156263. [PMID: 39615212 DOI: 10.1016/j.phymed.2024.156263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/02/2024] [Accepted: 11/13/2024] [Indexed: 01/30/2025]
Abstract
BACKGROUND Acute pancreatitis (AP) is a prevalent and serious condition within the digestive system, with approximately 20 % to 30 % of cases advancing to severe acute pancreatitis (SAP). During the initial phases of SAP, macrophages are activated in response to the substantial amounts of acinar cell contents and damage-associated molecular patterns (DAMPs) resulting from acinar cell destruction. Subsequently, activated macrophages release a significant array of pro-inflammatory factors that exacerbate the progression of SAP. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncogenic protein that is intimately linked to immune regulation. While the role of CIP2A in T-cell-mediated specific immune responses has been reported, its function and mechanism in macrophages, a component of non-specific immunity, have not been widely studied. This research fills this knowledge gap by elucidating the critical role of CIP2A in regulating macrophage autophagy and inflammation. This finding not only expands our understanding of CIP2A in immune modulation but also provides a new scientific basis and potential application prospects for targeting CIP2A in the treatment of AP. METHODS We established AP using a combination of palmitoleic acid with anhydrous ethanol or using caerulein alone. The effects of TD52 and Ethoxysanguinarine (Etho) on SAP were evaluated through serological, histopathological, and tissue inflammation observations. The effect of TD52 on macrophage activation in vitro was examined using primary macrophages (PMs) and RAW264.7 cells. RESULTS We found that TD52 and Etho inhibit CIP2A expression while reducing the levels of serum amylase, lipase, and inflammatory cytokines, thereby alleviating the pathological symptoms of SAP. Additionally, TD52 could reduce the infiltration of macrophages into pancreatic tissue. Therefore, we established a model of macrophage inflammatory response mimicking the pathophysiological process of AP and detected changes in inflammation, apoptosis, and autophagy through pre-treatment of macrophages with TD52. The results show that inhibiting CIP2A expression decreases the release of inflammatory cytokines and reduces apoptosis in macrophages. Further exploration revealed that TD52 promoted macrophage autophagy regulation and inhibited the AKT-mTOR pathway to modulate macrophage activation. CONCLUSION In summary, our findings indicate that TD52 and Etho can alleviate the severity of SAP. TD52 can block the AKT-mTOR pathway to promote macrophage autophagy, thereby improving SAP. Thus, CIP2A may serve as one of the molecular targets in SAP, highlighting its potential as a therapeutic option.
Collapse
Affiliation(s)
- Zhu Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China; Department of Hepatobiliary Surgery, Pingyang People's Hospital Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Tan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China
| | - Akmal Ergashev
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China
| | - Zhiyuan Bo
- Department of Surgery, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Jinhui Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China
| | - Fengyu Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China
| | - Zhenyan Pan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China
| | - Haonan Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China.
| | - Feng Ma
- National Key Laboratory of Immunity and Inflammation, and CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China.
| | - Lingming Kong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Zhejiang Key Laboratory of intelligent Cancer Biomarker Discovery & Translation, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, China; Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
3
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
4
|
Mayo P, Pascual J, Crisman E, Domínguez C, López MG, León R. Innovative pathological network-based multitarget approaches for Alzheimer's disease treatment. Med Res Rev 2024; 44:2367-2419. [PMID: 38678582 DOI: 10.1002/med.22045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/02/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is a major health threat globally. Its prevalence is forecasted to exponentially increase during the next 30 years due to the global aging population. Currently, approved drugs are merely symptomatic, being ineffective in delaying or blocking the relentless disease advance. Intensive AD research describes this disease as a highly complex multifactorial disease. Disclosure of novel pathological pathways and their interconnections has had a major impact on medicinal chemistry drug development for AD over the last two decades. The complex network of pathological events involved in the onset of the disease has prompted the development of multitarget drugs. These chemical entities combine pharmacological activities toward two or more drug targets of interest. These multitarget-directed ligands are proposed to modify different nodes in the pathological network aiming to delay or even stop disease progression. Here, we review the multitarget drug development strategy for AD during the last decade.
Collapse
Affiliation(s)
- Paloma Mayo
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Jorge Pascual
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Enrique Crisman
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Cristina Domínguez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| |
Collapse
|
5
|
Johnson H, Singh A, Raza A, Sha CM, Wang J, Gowda K, Shen Z, Nair H, Li C, Dokholyan NV, Narayan S, Sharma AK. Identification of a Novel Protein Phosphatase 2A Activator, PPA24, as a Potential Therapeutic for FOLFOX-Resistant Colorectal Cancer. J Med Chem 2024; 67:18070-18089. [PMID: 39004939 DOI: 10.1021/acs.jmedchem.4c01077] [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: 07/16/2024]
Abstract
A series of compounds were designed utilizing molecular modeling and fragment-based design based upon the known protein phosphatase 2A (PP2A) activators, NSC49L and iHAP1, and evaluated for their ability to inhibit the viability of colorectal cancer (CRC) and folinic acid, 5-fluorouracil, and oxaliplatin (FOLFOX)-resistant CRC cells. PPA24 (19a) was identified as the most cytotoxic compound with IC50 values in the range of 2.36-6.75 μM in CRC and FOLFOX-resistant CRC cell lines. It stimulated PP2A activity to a greater extent, displayed lower binding energies through molecular docking, and showed higher binding affinity through surface plasmon resonance for PP2A catalytic subunit α than the known PP2A activators. PPA24 dose-dependently induced apoptosis and oxidative stress, decreased the level of c-Myc expression, and synergistically potentiated cytotoxicity when combined with gemcitabine and cisplatin. Furthermore, a PPA24-encapsulated nanoformulation significantly inhibited the growth of CRC xenografts without systemic toxicities. Together, these results signify the potential of PPA24 as a novel PP2A activator and a prospective therapeutic for CRC and FOLFOX-resistant CRC.
Collapse
Affiliation(s)
- Hannah Johnson
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Amandeep Singh
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Asif Raza
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Congzhou M Sha
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Jian Wang
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Krishne Gowda
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Zhihang Shen
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Haritha Nair
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, Florida 32610, United States
| | - Chenglong Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Satya Narayan
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, Florida 32610, United States
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
6
|
Guo J, Yue L, Ning L, Han A, Wang J. Phosphopeptide-bridged NH 2-TiO 2-mediated carbon dots self-enhancing and electrochemiluminescence microsensors for label-free protein kinase A detection. Mikrochim Acta 2024; 191:622. [PMID: 39320530 DOI: 10.1007/s00604-024-06711-8] [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: 06/18/2024] [Accepted: 09/14/2024] [Indexed: 09/26/2024]
Abstract
A novel electrochemiluminescence (ECL) method was developed for determination of protein kinase A (PKA) ultra-sensitively based on amidated nano-titanium (NH2-TiO2) embellished carbon dots (Mg@N-CDs) fluorescent probe, which integrated the target recognition and ECL signal enhancement. The Cys-labeled kemptides were employed to build a serine-rich synthetic substrate-heptapeptide (Cys-kemptide) on the Au-electrode surface. Then, the PKA-induced biosensor was triggered as a signal switch to introduce the large amounts of TiO2 decorated Mg@N-CD nanohybrid (Ti@NMg-CDs) into AuE/Cys-phosphopeptides for signal output. In particular, the presence of PKA could induce the formation of Cys-phosphopeptides by the catalytic reaction between specific substrate (kemptide) and PKA, which acts as an initiator to link the Ti@NMg-CDs according to the bridge interactions Ti-O-P. In this way, multiple Cys-phosphopeptides were adsorbed onto a single Ti@NMg-CDs, and the Ti@NMg-CDs not only provided high specific selectivity but also large surface area, as well as unprecedented high ECL efficiency. Using this PKA-induced enhanced sensor, the limit of detection of the PKA was 4.89 × 10-4 U/mL (S/N = 3). The proposed ECL biosensor was also universally applicable for the screening of PKA inhibitors and determining of other kinases activity. Our sensing system has excellent performance of specificity and the screening of kinase inhibitors, as well as it will inspire future effort in clinical diagnostics and new drug discovery.
Collapse
Affiliation(s)
- Jianping Guo
- School of Food Science, Shanxi Engineering Research Center of Microbial Application Technologies, Shanxi Normal University, Taiyuan, 030031, Shanxi, China.
| | - Lele Yue
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Lingya Ning
- State Key Laboratory of Food Nutrition and Safety, Tianjin Economy and Technology Development Area, Tianjin University of Science & Technology, 29 The Thirteenth Road, Tianjin, 300457, P.R. China
| | - Ailing Han
- School of Food Science, Shanxi Engineering Research Center of Microbial Application Technologies, Shanxi Normal University, Taiyuan, 030031, Shanxi, China.
| | - Junping Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin Economy and Technology Development Area, Tianjin University of Science & Technology, 29 The Thirteenth Road, Tianjin, 300457, P.R. China.
| |
Collapse
|
7
|
Vlahopoulos SA. Divergent Processing of Cell Stress Signals as the Basis of Cancer Progression: Licensing NFκB on Chromatin. Int J Mol Sci 2024; 25:8621. [PMID: 39201306 PMCID: PMC11354898 DOI: 10.3390/ijms25168621] [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: 07/10/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Inflammation is activated by diverse triggers that induce the expression of cytokines and adhesion molecules, which permit a succession of molecules and cells to deliver stimuli and functions that help the immune system clear the primary cause of tissue damage, whether this is an infection, a tumor, or a trauma. During inflammation, short-term changes in the expression and secretion of strong mediators of inflammation occur, while long-term changes occur to specific groups of cells. Long-term changes include cellular transdifferentiation for some types of cells that need to regenerate damaged tissue, as well as death for specific immune cells that can be detrimental to tissue integrity if they remain active beyond the boundaries of essential function. The transcriptional regulator NFκB enables some of the fundamental gene expression changes during inflammation, as well as during tissue development. During recurrence of malignant disease, cell stress-induced alterations enable the growth of cancer cell clones that are substantially resistant to therapeutic intervention and to the immune system. A number of those alterations occur due to significant defects in feedback signal cascades that control the activity of NFκB. Specifically, cell stress contributes to feedback defects as it overrides modules that otherwise control inflammation to protect host tissue. NFκB is involved in both the suppression and promotion of cancer, and the key distinctive feature that determines its net effect remains unclear. This paper aims to provide a clear answer to at least one aspect of this question, namely the mechanism that enables a divergent response of cancer cells to critical inflammatory stimuli and to cell stress in general.
Collapse
|
8
|
Wang M, Zhu M, Jia X, Wu J, Yuan Q, Xu T, Wang Z, Huang M, Ji N, Zhang M. LincR-PPP2R5C regulates IL-1β ubiquitination in macrophages and promotes airway inflammation and emphysema in a murine model of COPD. Int Immunopharmacol 2024; 139:112680. [PMID: 39018689 DOI: 10.1016/j.intimp.2024.112680] [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: 11/17/2023] [Revised: 06/15/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common disease with high global morbidity and mortality. Macrophages release IL-1β and orchestrate airway inflammation in COPD. Previously, we explored the role of a new lncRNA, LincR-PPP2R5C, in regulating Th2 cells in asthma. Here, we established a murine model of COPD and explored the roles and mechanisms by which LincR-PPP2R5C regulates IL-1β in macrophages. LincR-PPP2R5C was highly expressed in pulmonary macrophages from COPD-like mice. LincR-PPP2R5C deficiency ameliorated emphysema and pulmonary inflammation, as characterized by reduced IL-1β in macrophages. Unexpectedly, in both lung tissues and macrophages, LincR-PPP2R5C deficiency decreased the expression of the IL-1β protein but not the IL-1β mRNA. Furthermore, we found that LincR-PPP2R5C deficiency increased the level of ubiquitinated IL-1β in macrophages, which was mediated by PP2A activity. Targeting PP2A with FTY720 decreased IL-1β and improved COPD. In conclusion, LincR-PPP2R5C regulates IL-1β ubiquitination by affecting PP2A activity in macrophages, contributing to the airway inflammation and emphysema in a murine model of COPD. PP2A and IL-1β ubiquitination in macrophages might be new therapeutic avenues for COPD therapy.
Collapse
Affiliation(s)
- Min Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Manni Zhu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyu Jia
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Wu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Yuan
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Xu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Ningfei Ji
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Mingshun Zhang
- Jiangsu Province Engineering Research Center of Antibody Drug, NHC Key Laboratory of Antibody Technique, Department of Immunology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
9
|
Cappelli E, Ravera S, Bertola N, Grilli F, Squillario M, Regis S, Degan P. Advanced Analysis and Validation of a microRNA Signature for Fanconi Anemia. Genes (Basel) 2024; 15:820. [PMID: 39062599 PMCID: PMC11276059 DOI: 10.3390/genes15070820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Some years ago, we reported the generation of a Fanconi anemia (FA) microRNA signature. This study aims to develop an analytical strategy to select a smaller and more reliable set of molecules that could be tested for potential benefits for the FA phenotype, elucidate its biochemical and molecular mechanisms, address experimental activity, and evaluate its possible impact on FA therapy. In silico analyses of the data obtained in the original study were thoroughly processed and anenrichment analysis was employed to identify the classes of genes that are over-represented in the FA-miRNA population under study. Primary bone marrow mononuclear cells (MNCs) from sixFA patients and sixhealthy donors as control samples were employed in the study. RNAs containing the small RNA fractions were reverse-transcribed and real-time PCR was performed in triplicate using the specific primers. Experiments were performed in triplicate.The in-silico analysis reported six miRNAs as likely contributors to the complex pathological spectrum of FA. Among these, three miRNAs were validated by real-time PCR. Primary bone marrow mononuclear cells (MNCs) reported a significant reduction in the expression level of miRNA-1246 and miRNA-206 in the FA samples in comparison to controls.This study highlights several biochemical pathways as culprits in the phenotypic manifestations and the pathophysiological mechanisms acting in FA. A relatively low number of miRNAs appear involved in all these different phenotypes, demonstrating the extreme plasticity of the gene expression modulation. This study further highlights miR-206 as a pivotal player in regulatory functions and signaling in the bone marrow mesenchymal stem cell (BMSC) process in FA. Due to this evidence, the activity of miR-206 in FA deserves specific experimental scrutiny. The results, here presented, might be relevant in the management of FA.
Collapse
Affiliation(s)
- Enrico Cappelli
- Haematology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16148 Genova, Italy;
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Via De Toni 14, 16132 Genova, Italy;
| | - Nadia Bertola
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, L. go R. Benzi 10, 16132 Genoa, Italy;
| | - Federica Grilli
- Haematology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16148 Genova, Italy;
| | | | - Stefano Regis
- Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16148 Genova, Italy;
| | - Paolo Degan
- Mutagenesi e Prevenzione Oncologica Unit—IRCCS Ospedale Policlinico San Martino, L. go R. Benzi 10, 16132 Genoa, Italy;
| |
Collapse
|
10
|
Chuang YT, Yen CY, Tang JY, Chang FR, Tsai YH, Wu KC, Chien TM, Chang HW. Protein phosphatase 2A modulation and connection with miRNAs and natural products. ENVIRONMENTAL TOXICOLOGY 2024; 39:3612-3627. [PMID: 38491812 DOI: 10.1002/tox.24199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/28/2024] [Accepted: 02/10/2024] [Indexed: 03/18/2024]
Abstract
Protein phosphatase 2A (PP2A), a heterotrimeric holoenzyme (scaffolding, catalytic, and regulatory subunits), regulates dephosphorylation for more than half of serine/threonine phosphosites and exhibits diverse cellular functions. Although several studies on natural products and miRNAs have emphasized their impacts on PP2A regulation, their connections lack systemic organization. Moreover, only part of the PP2A family has been investigated. This review focuses on the PP2A-modulating effects of natural products and miRNAs' interactions with potential PP2A targets in cancer and non-cancer cells. PP2A-modulating natural products and miRNAs were retrieved through a literature search. Utilizing the miRDB database, potential PP2A targets of these PP2A-modulating miRNAs for the whole set (17 members) of the PP2A family were retrieved. Finally, PP2A-modulating natural products and miRNAs were linked via a literature search. This review provides systemic directions for assessing natural products and miRNAs relating to the PP2A-modulating functions in cancer and disease treatments.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei, Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - Kuo-Chuan Wu
- Department of Computer Science and Information Engineering, National Pingtung University, Pingtung, Taiwan
| | - Tsu-Ming Chien
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
11
|
Kang JB, Son HK, Park DJ, Jin YB, Koh PO. Chlorogenic acid regulates the expression of protein phosphatase 2A subunit B in the cerebral cortex of a rat stroke model and glutamate-exposed neurons. Lab Anim Res 2024; 40:8. [PMID: 38429854 PMCID: PMC10905799 DOI: 10.1186/s42826-024-00196-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Ischemic stroke is a serious neurological disorder caused by blockages in cerebral artery. Protein phosphatase 2A (PP2A) is a phosphatase that performs a critical role in cell signaling and growth. PP2A subunit B acts as a neuroprotective agent in the nerve system. Chlorogenic acid, which is mainly found in roasted coffee, has antioxidant, anti-inflammatory, and anti-apoptotic effects. We hypothesized that chlorogenic acid modulates PP2A subunit B expression in ischemic stroke models and glutamate-mediated neurons. Middle artery occlusion (MCAO) surgery was operated and chlorogenic acid (30 mg/kg) or phosphate buffer saline was treated 2 h after MCAO. The cerebral cortex was collected 24 h after surgery and the change of PP2A subunit B expression was analyzed. Glutamate and/or chlorogenic acid were treated in cultured neurons, further study was performed. RESULTS A decrease in PP2A subunit B expression in MCAO animals was identified. Chlorogenic acid alleviated this decrease due to ischemic injury. Moreover, the number of PP2A subunit B-positive cells in the ischemic cerebral cortex was significantly decreased, chlorogenic acid alleviated this decrease. We also found protective effects of chlorogenic acid in neurons exposed to glutamate. Glutamate decreased the expression of PP2A subunit B and chlorogenic acid mitigated this decrease. Our results elucidated that chlorogenic acid performs neuroprotective functions and attenuates the reduction of PP2A subunit B by brain damage and glutamate-mediated excitotoxicity. CONCLUSIONS We showed that chlorogenic acid attenuated the decrease of PP2A subunit B in ischemic injury and neurons exposed to glutamate. Since PP2A subunit B contributes to the protection of brain tissue, we can suggest that chlorogenic acid preserves neurons by modulating PP2A subunit B during ischemic damage.
Collapse
Affiliation(s)
- Ju-Bin Kang
- Department of Anatomy and Histology, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea
| | - Hyun-Kyoung Son
- Department of Anatomy and Histology, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea
| | - Dong-Ju Park
- Department of Anatomy and Histology, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea
| | - Yeung-Bae Jin
- Department of Anatomy and Histology, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea
| | - Phil-Ok Koh
- Department of Anatomy and Histology, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju, 52828, South Korea.
| |
Collapse
|
12
|
Yang F, Zhang XL, Liu HH, Qian LL, Wang RX. Post translational modifications of connexin 43 in ventricular arrhythmias after myocardial infarction. Mol Biol Rep 2024; 51:329. [PMID: 38393658 DOI: 10.1007/s11033-024-09290-2] [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: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
Ventricular arrhythmias are the leading cause of sudden cardiac death in patients after myocardial infarction (MI). Connexin43 (Cx43) is the most important gap junction channel-forming protein in cardiomyocytes. Dysfunction of Cx43 contributes to impaired myocardial conduction and the development of ventricular arrhythmias. Following an MI, Cx43 undergoes structural remodeling, including expression abnormalities, and redistribution. These alterations detrimentally affect intercellular communication and electrical conduction within the myocardium, thereby increasing the susceptibility to post-infarction ventricular arrhythmias. Emerging evidence suggests that post-translational modifications play essential roles in Cx43 regulation after MI. Therefore, Cx43-targeted management has the potential to be a promising protective strategy for the prevention and treatment of post infarction ventricular arrhythmias. In this article, we primarily reviewed the regulatory mechanisms of Cx43 mediated post-translational modifications on post-infarction ventricular arrhythmias. Furthermore, Cx43-targeted therapy have also been discussed, providing insights into an innovative treatment strategy for ventricular arrhythmias after MI.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cardiology, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
| | - Xiao-Lu Zhang
- Department of Cardiology, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
| | - Huan-Huan Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ling-Ling Qian
- Department of Cardiology, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China.
| | - Ru-Xing Wang
- Department of Cardiology, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
13
|
Dhapola R, Beura SK, Sharma P, Singh SK, HariKrishnaReddy D. Oxidative stress in Alzheimer's disease: current knowledge of signaling pathways and therapeutics. Mol Biol Rep 2024; 51:48. [PMID: 38165499 DOI: 10.1007/s11033-023-09021-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024]
Abstract
Alzheimer's disease's pathophysiology is still a conundrum. Growing number of evidences have elucidated the involvement of oxidative stress in the pathology of AD rendering it a major target for therapeutic development. Reactive oxygen species (ROS) generated by altered mitochondrial function, dysregulated electron transport chain and other sources elevate aggregated Aβ and neurofibrillary tangles which further stimulating the production of ROS. Oxidative stress induced damage to lipids, proteins and DNA result in neuronal death which leads to AD. In addition, oxidative stress induces apoptosis that is triggered by the modulation of ERK1/2 and Nrf2 pathway followed by increased GSK-3β expression and decreased PP2A activity. Oxidative stress exaggerates disease condition by interfering with various signaling pathways like RCAN1, CREB/ERK, Nrf2, PP2A, NFκB and PI3K/Akt. Studies have reported the role of TNF-α in oxidative stress stimulation that has been regulated by drugs like etanercept increasing the level of anti-oxidants. Other drugs like pramipexole, memantine, carvedilol, and melatonin have been reported to activate CREB/RCAN1 and Nrf2 pathways. In line with this, epigallocatechin gallate and genistein also target Nrf2 and CREB pathway leading to activation of downstream pathways like ARE and Keap1 which ameliorate oxidative stress condition. Donepezil and resveratrol reduce oxidative stress and activate AMPK pathway along with PP2A activation thus promoting tau dephosphorylation and neuronal survival. This study describes in detail the role of oxidative stress in AD, major signaling pathways involving oxidative stress induced AD and drugs under development targeting these pathways which may aid in therapeutic advances for AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratoty, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Samir K Beura
- Department of Zoology, School of Basic and Applied Science, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratoty, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Sunil K Singh
- Department of Zoology, School of Basic and Applied Science, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratoty, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
14
|
Dave BP, Shah YB, Maheshwari KG, Mansuri KA, Prajapati BS, Postwala HI, Chorawala MR. Pathophysiological Aspects and Therapeutic Armamentarium of Alzheimer's Disease: Recent Trends and Future Development. Cell Mol Neurobiol 2023; 43:3847-3884. [PMID: 37725199 PMCID: PMC11407742 DOI: 10.1007/s10571-023-01408-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023]
Abstract
Alzheimer's disease (AD) is the primary cause of dementia and is characterized by the death of brain cells due to the accumulation of insoluble amyloid plaques, hyperphosphorylation of tau protein, and the formation of neurofibrillary tangles within the cells. AD is also associated with other pathologies such as neuroinflammation, dysfunction of synaptic connections and circuits, disorders in mitochondrial function and energy production, epigenetic changes, and abnormalities in the vascular system. Despite extensive research conducted over the last hundred years, little is established about what causes AD or how to effectively treat it. Given the severity of the disease and the increasing number of affected individuals, there is a critical need to discover effective medications for AD. The US Food and Drug Administration (FDA) has approved several new drug molecules for AD management since 2003, but these drugs only provide temporary relief of symptoms and do not address the underlying causes of the disease. Currently, available medications focus on correcting the neurotransmitter disruption observed in AD, including cholinesterase inhibitors and an antagonist of the N-methyl-D-aspartate (NMDA) receptor, which temporarily alleviates the signs of dementia but does not prevent or reverse the course of AD. Research towards disease-modifying AD treatments is currently underway, including gene therapy, lipid nanoparticles, and dendrimer-based therapy. These innovative approaches aim to target the underlying pathological processes of AD rather than just managing the symptoms. This review discusses the novel aspects of pathogenesis involved in the causation of AD of AD and in recent developments in the therapeutic armamentarium for the treatment of AD such as gene therapy, lipid nanoparticles, and dendrimer-based therapy, and many more.
Collapse
Affiliation(s)
- Bhavarth P Dave
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Yesha B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Kunal G Maheshwari
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Kaif A Mansuri
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Bhadrawati S Prajapati
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Humzah I Postwala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| |
Collapse
|
15
|
Li Y, Fan H, Ni M, Zhang W, Fang F, Sun J, Lyu P, Ma P. Targeting lncRNA NEAT1 Hampers Alzheimer's Disease Progression. Neuroscience 2023; 529:88-98. [PMID: 37286157 DOI: 10.1016/j.neuroscience.2023.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 06/09/2023]
Abstract
Long noncoding RNA nuclear enriched abundant transcript 1 (lnc-NEAT1) is closely implicated in neurological diseases, while its implication in Alzheimer's disease (AD) is rarely reported. This study aimed to investigate the effect of lnc-NEAT1 knockdown on neuron injury, inflammation, and oxidative stress in AD, as well as its interaction with downstream targets and pathways. APPswe/PS1dE9 transgenic mice were injected with negative control or lnc-NEAT1 interference lentivirus. Besides, AD cellular model was constructed by amyloid β treatment in mice primary neuron cells; then, knockdown of lnc-NEAT1 and microRNA-193a was performed alone or in combination. In vivo experiments revealed that Lnc-NEAT1 knockdown improved cognition in AD mice reflected by Morrison water maze and Y-maze assays. Besides, lnc-NEAT1 knockdown reduced injury and apoptosis, decreased inflammatory cytokine levels, repressed oxidative stress level, and activated adenosine cyclophosphate response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF) and nuclear factor erythroid 2-related factor 2 (NRF2)/nicotinamide adenine dinucleotide phosphate dehydrogenase 1 (NQO1) pathways in hippocampi of AD mice. Notably, lnc-NEAT1 down-regulated microRNA-193a both in vitro and in vivo and acted as a decoy of microRNA-193a. In vitro experiments showed that lnc-NEAT1 knockdown decreased apoptosis and oxidative stress, improved cell viability, also activated CREB/BDNF and NRF2/NQO1 pathways in AD cellular model. Meanwhile, microRNA-193a knockdown showed the opposite effects, which also attenuated lnc-NEAT1 knockdown-mediated reduction in injury, oxidative stress, and CREB/BDNF and NRF2/NQO1 pathways of AD cellular model. In conclusion, lnc-NEAT1 knockdown reduces neuron injury, inflammation, and oxidative stress through activating microRNA-193a mediated CREB/BDNF and NRF2/NQO1 pathways in AD.
Collapse
Affiliation(s)
- Yuanlong Li
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Hua Fan
- School of Clinical Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China
| | - Ming Ni
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Clinical Pharmacy, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Wei Zhang
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Fengqin Fang
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Jun Sun
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Pin Lyu
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Peizhi Ma
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China.
| |
Collapse
|
16
|
Fujiki H, Sueoka E, Watanabe T, Komori A, Suganuma M. Cancer progression by the okadaic acid class of tumor promoters and endogenous protein inhibitors of PP2A, SET and CIP2A. J Cancer Res Clin Oncol 2023; 149:9425-9433. [PMID: 37097392 PMCID: PMC10374699 DOI: 10.1007/s00432-023-04800-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/15/2023] [Indexed: 04/26/2023]
Abstract
PURPOSE Okadaic acid class of tumor promoters are transformed into endogenous protein inhibitors of PP2A, SET, and CIP2A in human cancers. This indicates that inhibition of PP2A activity is a common mechanism of cancer progression in humans. It is important to study the roles of SET and CIP2A vis-à-vis their clinical significance on the basis of new information gathered from a search of PubMed. RESULTS AND DISCUSSION The first part of this review introduces the carcinogenic roles of TNF-α and IL-1, which are induced by the okadaic acid class of compounds. The second part describes unique features of SET and CIP2A in cancer progression for several types of human cancer: (1) SET-expressing circulating tumor cells (SET-CTCs) in breast cancer, (2) knockdown of CIP2A and increased PP2A activity in chronic myeloid leukemia, (3) CIP2A and epidermal growth factor receptor (EGFR) activity in erlotinib sensitive- and resistant-non-small cell lung cancer, (4) SET antagonist EMQA plus radiation therapy against hepatocellular carcinoma, (5) PP2A inactivation as a common event in colorectal cancer, (6) prostate cancer susceptibility variants, homeobox transcription factor (HOXB13 T) and CIP2A T, and (7) SET inhibitor OP449 for pre-clinical investigation of pancreatic cancer. In the Discussion, the binding complex of SET is briefly introduced, and overexpression of SET and CIP2A proteins is discussed in relation to age-associated chronic inflammation (inflammaging). CONCLUSION This review establishes the concept that inhibition of PP2A activity is a common mechanism of human cancer progression and activation of PP2A activity leads to effective anticancer therapy.
Collapse
Affiliation(s)
- Hirota Fujiki
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, Nabeshima, Saga 849-8501 Japan
| | - Eisaburo Sueoka
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, Nabeshima, Saga 849-8501 Japan
| | - Tatsuro Watanabe
- Department of Drug Discovery and Biomedical Sciences, Faculty of Medicine, Saga University, Nabeshima, Saga 849-8501 Japan
| | - Atsumasa Komori
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center and Department of Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Omura, Nagasaki 856-8562 Japan
| | - Masami Suganuma
- Department of Strategic Research, Graduate School of Science and Engineering, Saitama University, Saitama, 338-8570 Japan
| |
Collapse
|
17
|
Guffens L, Derua R, Janssens V. PME-1 sensitizes glioblastoma cells to oxidative stress-induced cell death by attenuating PP2A-B55α-mediated inactivation of MAPKAPK2-RIPK1 signaling. Cell Death Discov 2023; 9:265. [PMID: 37500619 PMCID: PMC10374899 DOI: 10.1038/s41420-023-01572-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor in adults. Current standard therapy is surgery followed by radiotherapy, with concurrent and adjuvant temozolomide chemotherapy. GBM is characterized by almost uniformly fatal outcomes, highlighting the unmet clinical need for more efficient, biomarker-guided treatments. Protein phosphatase methylesterase-1 (PME-1), a regulator of the tumor suppressive phosphatase PP2A, promotes PP2A demethylation and inactivation, and is overexpressed in 44% of GBM, associated with increased tumor grade and cellular proliferation. Here, we aimed to investigate how reactive oxygen species (ROS), a frequent by-product of radiotherapy and temozolomide chemotherapy, regulate PP2A function via its methylesterase PME-1, and how PME-1 overexpression impacts the response of GBM cells to oxidative stress. We found that in two glioblastoma cell lines, U87MG and U251MG, expression of PME-1 is positively correlated with the sensitivity of the cells to H2O2 or t-BHP-induced oxidative stress. Experiments using the irreversible pharmacologic PME-1 inhibitor, AMZ30, and different PME-1 mutants, revealed that the methylesterase function, the PP2A binding capacity, and the nuclear localization of PME-1 are all important for the sensitizing effect of PME-1 expression. Furthermore, we identified increased nuclear localization of the PP2A-B55α subunit, increased binding of PP2A-B55α to PME-1, and increased B55α-bound PP2A-C demethylation upon oxidative stress. Lastly, we uncovered increased stress-induced phosphorylation and activity of MAPKAPK2 and RIPK1 in PME-1 overexpressing U87MG cells, which caused the observed sensitization to t-BHP treatment. Our data reveal a novel role for PME-1 in oxidative stress-induced GBM cell death, regulating nuclear PP2A-B55α activity and MAPKAPK2-RIPK1 signaling. Patients with GBM tumors overexpressing PME-1, although having a worse prognosis due to increased cellular proliferation of the tumor, could actually be more responsive to oxidative stress-inducing therapies.
Collapse
Affiliation(s)
- Liesbeth Guffens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. Cellular & Molecular Medicine, KU Leuven, B-3000, Leuven, Belgium
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation & Proteomics, Dept. Cellular & Molecular Medicine, KU Leuven, B-3000, Leuven, Belgium
- SyBioMa, KU Leuven, B-3000, Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. Cellular & Molecular Medicine, KU Leuven, B-3000, Leuven, Belgium.
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium.
| |
Collapse
|
18
|
Vertyshev AY, Akberdin IR, Kolpakov FA. Numerous Trigger-like Interactions of Kinases/Protein Phosphatases in Human Skeletal Muscles Can Underlie Transient Processes in Activation of Signaling Pathways during Exercise. Int J Mol Sci 2023; 24:11223. [PMID: 37446402 DOI: 10.3390/ijms241311223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Optimizing physical training regimens to increase muscle aerobic capacity requires an understanding of the internal processes that occur during exercise that initiate subsequent adaptation. During exercise, muscle cells undergo a series of metabolic events that trigger downstream signaling pathways and induce the expression of many genes in working muscle fibers. There are a number of studies that show the dependence of changes in the activity of AMP-activated protein kinase (AMPK), one of the mediators of cellular signaling pathways, on the duration and intensity of single exercises. The activity of various AMPK isoforms can change in different directions, increasing for some isoforms and decreasing for others, depending on the intensity and duration of the load. This review summarizes research data on changes in the activity of AMPK, Ca2+/calmodulin-dependent protein kinase II (CaMKII), and other components of the signaling pathways in skeletal muscles during exercise. Based on these data, we hypothesize that the observed changes in AMPK activity may be largely related to metabolic and signaling transients rather than exercise intensity per se. Probably, the main events associated with these transients occur at the beginning of the exercise in a time window of about 1-10 min. We hypothesize that these transients may be partly due to putative trigger-like kinase/protein phosphatase interactions regulated by feedback loops. In addition, numerous dynamically changing factors, such as [Ca2+], metabolite concentration, and reactive oxygen and nitrogen species (RONS), can shift the switching thresholds and change the states of these triggers, thereby affecting the activity of kinases (in particular, AMPK and CaMKII) and phosphatases. The review considers the putative molecular mechanisms underlying trigger-like interactions. The proposed hypothesis allows for a reinterpretation of the experimental data available in the literature as well as the generation of ideas to optimize future training regimens.
Collapse
Affiliation(s)
| | - Ilya R Akberdin
- Department of Computational Biology, Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- Biosoft.Ru, Ltd., 630058 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Fedor A Kolpakov
- Department of Computational Biology, Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- Biosoft.Ru, Ltd., 630058 Novosibirsk, Russia
- Federal Research Center for Information and Computational Technologies, 630090 Novosibirsk, Russia
| |
Collapse
|
19
|
Adeyi OE, Somade OT, James AS, Adeyi AO, Ogbonna-Eze SN, Salako OQ, Makinde TV, Ajadi OM, Nosiru SA. Ferulic acid mitigates 2-methoxyethanol-induced testicular oxidative stress via combined downregulation of FoxO1, PTEN, and modulation of Nrf2-Hmox1-NQO1 signaling pathway in rats. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2023; 7:100257. [DOI: 10.1016/j.prmcm.2023.100257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
20
|
Sarkar P, Kumar A, Behera PS, Thirumurugan K. Phytotherapeutic targeting of the mitochondria in neurodegenerative disorders. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:415-455. [PMID: 37437986 DOI: 10.1016/bs.apcsb.2023.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Neurodegenerative diseases are characterized by degeneration or cellular atrophy within specific structures of the brain. Neurons are the major target of neurodegeneration. Neurons utilize 75-80% of the energy produced in the brain. This energy is either formed by utilizing the glucose provided by the cerebrovascular blood flow or by the in-house energy producers, mitochondria. Mitochondrial dysfunction has been associated with neurodegenerative diseases. But recently it has been noticed that neurodegenerative diseases are often associated with cerebrovascular diseases. Cerebral blood flow requires vasodilation which to an extent regulated by mitochondria. We hypothesize that when mitochondrial functioning is disrupted, it is not able to supply energy to the neurons. This disruption also affects cerebral blood flow, further reducing the possibilities of energy supply. Loss of sufficient energy leads to neuronal dysfunction, atrophy, and degeneration. In this chapter, we will discuss the metabolic modifications of mitochondria in aging-related neurological disorders and the potential of phytocompounds targeting them.
Collapse
Affiliation(s)
- Priyanka Sarkar
- Structural Biology Lab, Pearl Research Park, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Ashish Kumar
- Structural Biology Lab, Pearl Research Park, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Partha Sarathi Behera
- Structural Biology Lab, Pearl Research Park, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Kavitha Thirumurugan
- Structural Biology Lab, Pearl Research Park, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
21
|
Paciello F, Ripoli C, Fetoni AR, Grassi C. Redox Imbalance as a Common Pathogenic Factor Linking Hearing Loss and Cognitive Decline. Antioxidants (Basel) 2023; 12:antiox12020332. [PMID: 36829891 PMCID: PMC9952092 DOI: 10.3390/antiox12020332] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Experimental and clinical data suggest a tight link between hearing and cognitive functions under both physiological and pathological conditions. Indeed, hearing perception requires high-level cognitive processes, and its alterations have been considered a risk factor for cognitive decline. Thus, identifying common pathogenic determinants of hearing loss and neurodegenerative disease is challenging. Here, we focused on redox status imbalance as a possible common pathological mechanism linking hearing and cognitive dysfunctions. Oxidative stress plays a critical role in cochlear damage occurring during aging, as well as in that induced by exogenous factors, including noise. At the same time, increased oxidative stress in medio-temporal brain regions, including the hippocampus, is a hallmark of neurodegenerative disorders like Alzheimer's disease. As such, antioxidant therapy seems to be a promising approach to prevent and/or counteract both sensory and cognitive neurodegeneration. Here, we review experimental evidence suggesting that redox imbalance is a key pathogenetic factor underlying the association between sensorineural hearing loss and neurodegenerative diseases. A greater understanding of the pathophysiological mechanisms shared by these two diseased conditions will hopefully provide relevant information to develop innovative and effective therapeutic strategies.
Collapse
Affiliation(s)
- Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-0630154966
| | - Anna Rita Fetoni
- Unit of Audiology, Department of Neuroscience, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
22
|
Creighton MT, Nemie-Feyissa D, Zaman N, Johansen SS, Dysjaland H, Heidari B, Lillo C. Loss of LEUCINE CARBOXYL METHYLTRANSFERASE 1 interferes with metal homeostasis in Arabidopsis and enhances susceptibility to environmental stresses. JOURNAL OF PLANT PHYSIOLOGY 2022; 279:153843. [PMID: 36265226 DOI: 10.1016/j.jplph.2022.153843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/30/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
The biochemical function of LEUCINE CARBOXYL METHYLTRANSFERASE 1 (LCMT1) is to transfer a methyl group from the methyl donor S-adenosylmethionine (SAM) to the catalytic subunits of PROTEIN PHOSPHATASE 2A (PP2Ac), PP4 and PP6. This post-translational modification by LCMT1 is found throughout eukaryotes from yeast to animals and plants, indicating that its function is essential. However, Arabidopsis with knocked out LCMT1 still grows and develops almost normally, at least under optimal growth conditions. We therefore proposed that the presence of LCMT1 would be important under non-optimal growth conditions and favoured plant survival during evolution. To shed light on the physiological functions of plant LCMT1, phenotypes of the lcmt1 mutant and wild type Arabidopsis were compared under various conditions including exposure to heavy metals, variable chelator concentrations, and increased temperature. The lcmt1 mutant was found to be more susceptible to these environmental changes than wild type and resulted in poor growth of seedlings and rosette stage plants. Element analysis of rosette stage plants mainly showed a difference between the lcmt1 mutant and wild type regarding concentrations of sodium and boron, two-fold up or halved, respectively. In both lcmt1 and wild type, lack of EDTA in the growth medium resulted in enhanced concentration of copper, manganese, zinc and sulphur, and especially lcmt1 growth was hampered by these conditions. The altered phenotype in response to stress, the element and mRNA transcript analysis substantiate that LCMT1 has an important role in metal homeostasis and show that functional LCMT1 is necessary to prevent damages from heat, heavy metals or lack of chelator.
Collapse
Affiliation(s)
- Maria T Creighton
- IKBM, Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036, Stavanger, Norway
| | - Dugassa Nemie-Feyissa
- IKBM, Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036, Stavanger, Norway
| | - Nabeela Zaman
- IKBM, Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036, Stavanger, Norway
| | - Sverre S Johansen
- IKBM, Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036, Stavanger, Norway
| | - Hege Dysjaland
- IKBM, Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036, Stavanger, Norway
| | - Behzad Heidari
- IKBM, Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036, Stavanger, Norway
| | - Cathrine Lillo
- IKBM, Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036, Stavanger, Norway.
| |
Collapse
|
23
|
Manipulating PP2Acα-ASK-JNK signaling to favor apoptotic over necroptotic hepatocyte fate reduces the extent of necrosis and fibrosis upon acute liver injury. Cell Death Dis 2022; 13:985. [PMID: 36418313 PMCID: PMC9684557 DOI: 10.1038/s41419-022-05353-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022]
Abstract
In the widely used Carbon tetrachloride (CCl4)-induced acute liver injury (ALI) mouse model, hepatocytes are known to die from programmed cell death (PCD) processes including apoptosis and necroptosis. Both in vivo and in vitro experiments showed that CCl4 treatment could induce both apoptosis and necroptosis. Treatment of mice with the apoptosis inducer SMAC mimetic reduced necroptosis, led to less pronounced liver damage, and improved overall liver function. By LC-MS/MS, we found that PP2Acα expression was increased in ALI mice liver, and we confirmed its high expression in subacute hepatitis patients. We observed that ALI severity (including aggravated fibrogenesis) was significantly alleviated in hepatocyte-specific PP2Acα conditional knockout (PP2Acα cKO) mice. Furthermore, the relative extent of apoptosis over necroptosis was increased in the PP2Acα cKO ALI mice. Pursuing the idea that biasing the type of PCD towards apoptosis may reduce liver damage, we found that treatment of PP2Acα cKO ALI mice with the apoptosis inhibitor z-Vad-fmk increased the extent of necroptosis and caused severer damage. Mechanistically, disruption of PP2Acα prevents the dephosphorylation of pASK1(Ser967), thereby preventing the sustained activation of JNK. Inhibition of PP2Acα prevents CCl4-induced liver injury and fibrogenesis by disrupting ASK/JNK pathway mediated PCD signaling, ultimately improving liver function by biasing hepatocytes towards an apoptotic rather than necroptotic cell fate. Thus, targeting PP2A and/or ASK1 to favor apoptotic over necroptotic hepatocyte fate may represent an attractive therapeutic strategy for treating ALI.
Collapse
|
24
|
Jurcău MC, Andronie-Cioara FL, Jurcău A, Marcu F, Ţiț DM, Pașcalău N, Nistor-Cseppentö DC. The Link between Oxidative Stress, Mitochondrial Dysfunction and Neuroinflammation in the Pathophysiology of Alzheimer's Disease: Therapeutic Implications and Future Perspectives. Antioxidants (Basel) 2022; 11:2167. [PMID: 36358538 PMCID: PMC9686795 DOI: 10.3390/antiox11112167] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 08/26/2023] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, has increasing incidence, increasing mortality rates, and poses a huge burden on healthcare. None of the currently approved drugs for the treatment of AD influence disease progression. Many clinical trials aiming at inhibiting amyloid plaque formation, increasing amyloid beta clearance, or inhibiting neurofibrillary tangle pathology yielded inconclusive results or failed. Meanwhile, research has identified many interlinked vicious cascades implicating oxidative stress, mitochondrial dysfunction, and chronic neuroinflammation, and has pointed to novel therapeutic targets such as improving mitochondrial bioenergetics and quality control, diminishing oxidative stress, or modulating the neuroinflammatory pathways. Many novel molecules tested in vitro or in animal models have proven efficient, but their translation into clinic needs further research regarding appropriate doses, delivery routes, and possible side effects. Cell-based therapies and extracellular vesicle-mediated delivery of messenger RNAs and microRNAs seem also promising strategies allowing to target specific signaling pathways, but need further research regarding the most appropriate harvesting and culture methods as well as control of the possible tumorigenic side effects. The rapidly developing area of nanotechnology could improve drug delivery and also be used in early diagnosis.
Collapse
Affiliation(s)
| | - Felicia Liana Andronie-Cioara
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Anamaria Jurcău
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Florin Marcu
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Delia Mirela Ţiț
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Nicoleta Pașcalău
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Delia Carmen Nistor-Cseppentö
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
25
|
Chen GS, Chen SY, Liu ST, Hsieh CC, Lee SP, Huang SM. Stabilization of the c-Myc Protein via the Modulation of Threonine 58 and Serine 62 Phosphorylation by the Disulfiram/Copper Complex in Oral Cancer Cells. Int J Mol Sci 2022; 23:ijms23169137. [PMID: 36012403 PMCID: PMC9409128 DOI: 10.3390/ijms23169137] [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: 07/14/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
MYC has a short half-life that is tightly regulated through phosphorylation and proteasomal degradation. Many studies have claimed that treatment with disulfiram (DSF) with or without copper ions can cause cancer cell death in a reactive oxygen species (ROS)-dependent manner in cancer cells. Our previous study showed that the levels of c-Myc protein and the phosphorylation of threonine 58 (T58) and serine 62 (S62) increased in DSF-Cu-complex-treated oral epidermoid carcinoma Meng-1 (OECM-1) cells. These abovementioned patterns were suppressed by pretreatment with an ROS scavenger, N-acetyl cysteine. The overexpression of c-Myc failed to induce hypoxia-inducible factor 1α protein expression, which was stabilized by the DSF-Cu complex. In this study, we further examined the regulatory mechanism behind the induction of the c-Myc of the DSF-Cu complex in an OECM-1 cell compared with a Smulow–Glickman (SG) human normal gingival epithelial cell. Our data showed that the downregulation of c-Myc truncated nick and p62 and the induction of the ratio of H3P/H3 and p-ERK/ERK might not be involved in the increase in the amount of c-Myc via the DSF/copper complexes in OECM-1 cells. Combined with the inhibitors for various signaling pathways and cycloheximde treatment, the increase in the amount of c-Myc with the DSF/copper complexes might be mediated through the increase in the stabilities of c-Myc (T58) and c-Myc (S62) proteins in OECM-1 cells. In SG cells, only the c-Myc (T58) protein was stabilized by the DSF-Cu (I and II) complexes. Hence, our findings could provide novel regulatory insights into the phosphorylation-dependent stability of c-Myc in DSF/copper-complex-treated oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Gunng-Shinng Chen
- School of Dentistry, Department of Dentistry of Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan
| | - Ssu-Yu Chen
- Department of Biochemistry, National Defense Medical Center, Taipei City 114, Taiwan
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taipei City 114, Taiwan
| | - Cheng-Chih Hsieh
- School of Pharmacy and Institute of Pharmacy, National Defense Medical Center, Taipei City 114, Taiwan
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Shiao-Pieng Lee
- School of Dentistry, Department of Dentistry of Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan
- Correspondence: (S.-P.L.); (S.-M.H.); Tel.: +886-2-87923100 (ext. 18790) (S.-M.H.)
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei City 114, Taiwan
- Correspondence: (S.-P.L.); (S.-M.H.); Tel.: +886-2-87923100 (ext. 18790) (S.-M.H.)
| |
Collapse
|
26
|
Bartolome F, Carro E, Alquezar C. Oxidative Stress in Tauopathies: From Cause to Therapy. Antioxidants (Basel) 2022; 11:antiox11081421. [PMID: 35892623 PMCID: PMC9332496 DOI: 10.3390/antiox11081421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress (OS) is the result of an imbalance between the production of reactive oxygen species (ROS) and the antioxidant capacity of cells. Due to its high oxygen demand, the human brain is highly susceptible to OS and, thus, it is not a surprise that OS has emerged as an essential component of the pathophysiology of several neurodegenerative diseases, including tauopathies. Tauopathies are a heterogeneous group of age-related neurodegenerative disorders characterized by the deposition of abnormal tau protein in the affected neurons. With the worldwide population aging, the prevalence of tauopathies is increasing, but effective therapies have not yet been developed. Since OS seems to play a key role in tauopathies, it has been proposed that the use of antioxidants might be beneficial for tau-related neurodegenerative diseases. Although antioxidant therapies looked promising in preclinical studies performed in cellular and animal models, the antioxidant clinical trials performed in tauopathy patients have been disappointing. To develop effective antioxidant therapies, the molecular mechanisms underlying OS in tauopathies should be completely understood. Here, we review the link between OS and tauopathies, emphasizing the causes of OS in these diseases and the role of OS in tau pathogenesis. We also summarize the antioxidant therapies proposed as a potential treatment for tauopathies and discuss why they have not been completely translated to clinical trials. This review aims to provide an integrated perspective of the role of OS and antioxidant therapies in tauopathies. In doing so, we hope to enable a more comprehensive understanding of OS in tauopathies that will positively impact future studies.
Collapse
Affiliation(s)
- Fernando Bartolome
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain;
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain;
| | - Eva Carro
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain;
- Neurobiology of Alzheimer’s Disease Unit, Chronic Disease Program, Instituto de Salud Carlos III, 28222 Madrid, Spain
| | - Carolina Alquezar
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain;
- Correspondence:
| |
Collapse
|
27
|
Changes in Tyrosine Hydroxylase Activity and Dopamine Synthesis in the Nigrostriatal System of Mice in an Acute Model of Parkinson's Disease as a Manifestation of Neurodegeneration and Neuroplasticity. Brain Sci 2022; 12:brainsci12060779. [PMID: 35741664 PMCID: PMC9221104 DOI: 10.3390/brainsci12060779] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 02/06/2023] Open
Abstract
The progressive degradation of the nigrostriatal system leads to the development of Parkinson’s disease (PD). The synthesis of dopamine, the neurotransmitter of the nigrostriatal system, depends on the rate-limiting enzyme, tyrosine hydroxylase (TH). In this study, we evaluated the synthesis of dopamine during periods of neurodegradation and neuroplasticity in the nigrostriatal system on a model of the early clinical stage of PD. It was shown that the concentration of dopamine correlated with activity of TH, while TH activity did not depend on total protein content either in the SN or in the striatum. Both during the period of neurodegeneration and neuroplasticity, TH activity in SN was determined by the content of P19-TH, and in the striatum it was determined by P31-TH and P40-TH (to a lesser extent). The data obtained indicate a difference in the regulation of dopamine synthesis between DA-neuron bodies and their axons, which must be considered for the further development of symptomatic pharmacotherapy aimed at increasing TH activity.
Collapse
|
28
|
Haslem L, Hays JM, Hays FA. p66Shc in Cardiovascular Pathology. Cells 2022; 11:cells11111855. [PMID: 35681549 PMCID: PMC9180016 DOI: 10.3390/cells11111855] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
p66Shc is a widely expressed protein that governs a variety of cardiovascular pathologies by generating, and exacerbating, pro-apoptotic ROS signals. Here, we review p66Shc’s connections to reactive oxygen species, expression, localization, and discuss p66Shc signaling and mitochondrial functions. Emphasis is placed on recent p66Shc mitochondrial function discoveries including structure/function relationships, ROS identity and regulation, mechanistic insights, and how p66Shc-cyt c interactions can influence p66Shc mitochondrial function. Based on recent findings, a new p66Shc mitochondrial function model is also put forth wherein p66Shc acts as a rheostat that can promote or antagonize apoptosis. A discussion of how the revised p66Shc model fits previous findings in p66Shc-mediated cardiovascular pathology follows.
Collapse
Affiliation(s)
- Landon Haslem
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Jennifer M. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Franklin A. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
- Stephenson Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
29
|
Gergs U, Jahn T, Schulz N, Großmann C, Rueckschloss U, Demus U, Buchwalow IB, Neumann J. Protein Phosphatase 2A Improves Cardiac Functional Response to Ischemia and Sepsis. Int J Mol Sci 2022; 23:ijms23094688. [PMID: 35563079 PMCID: PMC9101092 DOI: 10.3390/ijms23094688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022] Open
Abstract
Reversible protein phosphorylation is a posttranslational modification of regulatory proteins involved in cardiac signaling pathways. Here, we focus on the role of protein phosphatase 2A (PP2A) for cardiac gene expression and stress response using a transgenic mouse model with cardiac myocyte-specific overexpression of the catalytic subunit of PP2A (PP2A-TG). Gene and protein expression were assessed under basal conditions by gene chip analysis and Western blotting. Some cardiac genes related to the cell metabolism and to protein phosphorylation such as kinases and phosphatases were altered in PP2A-TG compared to wild type mice (WT). As cardiac stressors, a lipopolysaccharide (LPS)-induced sepsis in vivo and a global cardiac ischemia in vitro (stop-flow isolated perfused heart model) were examined. Whereas the basal cardiac function was reduced in PP2A-TG as studied by echocardiography or as studied in the isolated work-performing heart, the acute LPS- or ischemia-induced cardiac dysfunction deteriorated less in PP2A-TG compared to WT. From the data, we conclude that increased PP2A activity may influence the acute stress tolerance of cardiac myocytes.
Collapse
Affiliation(s)
- Ulrich Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097 Halle, Germany; (T.J.); (N.S.); (J.N.)
- Correspondence: ; Tel.: +49-345-557-4093
| | - Tina Jahn
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097 Halle, Germany; (T.J.); (N.S.); (J.N.)
| | - Nico Schulz
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097 Halle, Germany; (T.J.); (N.S.); (J.N.)
| | - Claudia Großmann
- Julius-Bernstein-Institut für Physiologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097 Halle, Germany;
| | - Uwe Rueckschloss
- Institut für Anatomie und Zellbiologie, Julius-Maximilians-Universität Würzburg, D-97070 Würzburg, Germany;
| | - Uta Demus
- Gesellschaft zur Förderung von Medizin-, Bio-und Umwelttechnologien e. V., D-06120 Halle, Germany;
| | - Igor B. Buchwalow
- Institut für Hämatopathologie, D-22547 Hamburg, Germany;
- Scientific and Educational Resource Center for Molecular Morphology, Peoples’ Friendship University of Russia, Moscow 117198, Russia
| | - Joachim Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, D-06097 Halle, Germany; (T.J.); (N.S.); (J.N.)
| |
Collapse
|
30
|
Li R, Zhang C, Xie F, Zhou X, Hu X, Shi J, Du X, Lin Z, Dong N. Protein Phosphatase 2A Deficiency in Macrophages Increases Foam Cell Formation and Accelerates Atherosclerotic Lesion Development. Front Cardiovasc Med 2022; 8:745009. [PMID: 35118139 PMCID: PMC8803755 DOI: 10.3389/fcvm.2021.745009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Protein phosphatase 2A (PP2A), a crucial serine/threonine phosphatase, has recently been reported to play an important role in cardiovascular disease. Previous studies have hinted that PP2A is involved in atherosclerosis formation, but the associated mechanisms remain poorly understood. In this study, we investigate the role of PP2A in the pathogenesis of atherosclerosis. In human atherosclerotic coronary arteries, we found that the expression and activity of PP2A decreased significantly when compared to non-atherosclerotic arteries. Additional experiments demonstrated that pharmacological inhibition of PP2A aggravated atherosclerosis of ApoE−/− mice. Considering the central role of macrophages in atherosclerosis, mice with conditional knockout of the PP2A-Cα subunit in myeloid cells were produced to investigate the function of PP2A in macrophages. Results showed that PP2A deficiency in myeloid cells aggravated atherosclerotic lesions in mice. in vitro experiments indicated that PP2A-deficient macrophages had an enhanced ability of lipid uptake and foam cell formation. Mechanistically, the deficiency of the PP2A in macrophages led to an increase in the phosphorylation level of p38, which contributed to the elevated expression of scavenger receptor CD36, a key factor involved in lipoprotein uptake. Our data suggest that PP2A participates in the pathophysiological process of atherosclerosis. The decrease of PP2A expression and activity in macrophages is a crucial determinant for foam cell formation and the initiation of atherosclerosis. Our study may provide a potential novel approach for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Rui Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianming Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjian Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinling Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xinling Du
| | - Zhiyong Lin
- Cardiology Division, Emory University School of Medicine, Atlanta, GA, United States
- Zhiyong Lin
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Nianguo Dong
| |
Collapse
|
31
|
Encapsulated Activated Grape Seed Extract: A Novel Formulation with Anti-Aging, Skin-Brightening, and Hydration Properties. COSMETICS 2021. [DOI: 10.3390/cosmetics9010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a master regulatory protein that plays a critical role in oxidative stress signaling. A novel, proprietary grape seed extract called Activated Grape Seed Extract (AGSE), enriched for PP2A-activating flavonoids, was recently developed and demonstrated to have antioxidant and anti-inflammatory activities. AGSE is a purple-colored powder, with limited solubility restricting its use in a broad range of formulations. Our aim was to develop a formulation that reduced the color and increased the solubility of AGSE, allowing its skin-health-enhancing properties to be utilized in a wider array of products, and to test it clinically. Encapsulation was performed utilizing a liposome and hydroxypropyl-β-cyclodextrin, (HPCD)-based approach to produce Encapsulated AGSE (E-AGSE). Human dermal fibroblasts and epidermal keratinocytes were used to determine expression levels of aging and dermal–epidermal junction (DEJ) markers. EpiDerm™ was UVB-irradiated to measure the effects against cytokine release, DNA damage, apoptosis, and skin barrier. Human melanocytes were used to determine melanin production and mushroom tyrosinase was used for inhibitory activity. A 4-week, 31-subject sensitive-skin clinical was performed with 2% E-AGSE Essence to assess its activity on human skin. We demonstrated that E-AGSE inhibits PP2A demethylation, increases key anti-aging (collagen I, III, elastin) and DEJ markers, protects against UVB-induced DNA damage, reduces inflammation, and promotes filaggrin in vitro. Moreover, E-AGSE reduces melanin production via tyrosinase inhibition. Clinical assessment of E-AGSE showed that it reduces the appearance of wrinkles, brightens the skin, and boosts hydration. E-AGSE is a novel grape seed extract formulation enriched for PP2A-activating flavonoids that is clinically effective in sensitive skin, providing several benefits.
Collapse
|
32
|
Behl T, Arora A, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Bungau S, Mostafavi E. Molecular and Biochemical Pathways Encompassing Diabetes Mellitus and Dementia. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:542-556. [PMID: 34758720 DOI: 10.2174/1871527320666211110115257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/25/2021] [Accepted: 09/15/2021] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus is a major metabolic disorder that has now emerged as an epidemic, and it affects the brain through an array of pathways. Diabetes mellitus patients can develop pathological changes in the brain, which eventually take the shape of mild cognitive impairment progressing to Alzheimer's Disease. A number of preclinical and clinical studies demonstrate this fact, and it comes out to be those molecular pathways such as amyloidogenesis, oxidative stress, inflammation, and impaired insulin signaling are identical in diabetes mellitus and dementia. However, the critical player involved in the vicious cycle of diabetes mellitus and dementia is insulin, whose signaling, when impaired in diabetes mellitus (both type 1 and 2), leads to a decline in cognition, although other pathways are also essential contributors. Moreover, it is not only that diabetes mellitus patients indicate cognitive decline at a later stage; many Alzheimer's Disease patients also reflect symptoms of diabetes mellitus, thus creating a vicious cycle inculcating a web of complex molecular mechanisms and hence categorizing Alzheimer's Disease as 'brain diabetes'. Thus, it is practical to suggest that anti-diabetic drugs are beneficial in Alzheimer's Disease; but only smaller trials, not the larger ones, have showcased positive outcomes mainly because of the late onset of therapy. Therefore, it is extremely important to develop more of such molecules that target insulin in dementia patients along with such methods that diagnose impaired insulin signaling and the associated cognitive decline so that early therapy may be initiated and the progression of the disease be prevented.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Arpita Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Haryana. India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa. Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea. Romania
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA. United States
| |
Collapse
|
33
|
Kaur D, Behl T, Sehgal A, Singh S, Sharma N, Chigurupati S, Alhowail A, Abdeen A, Ibrahim SF, Vargas-De-La-Cruz C, Sachdeva M, Bhatia S, Al-Harrasi A, Bungau S. Decrypting the potential role of α-lipoic acid in Alzheimer's disease. Life Sci 2021; 284:119899. [PMID: 34450170 DOI: 10.1016/j.lfs.2021.119899] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases with motor disturbances, cognitive decline, and behavioral impairment. It is characterized by the extracellular aggregation of amyloid-β plaques and the intracellular accumulation of tau protein. AD patients show a cognitive decline, which has been associated with oxidative stress, as well as mitochondrial dysfunction. Alpha-lipoic acid (α-LA), a natural antioxidant present in food and used as a dietary supplement, has been considered a promising agent for the prevention or treatment of neurodegenerative disorders. Despite multiple preclinical studies indicating beneficial effects of α-LA in memory functioning, and pointing to its neuroprotective effects, to date only a few studies have examined its effects in humans. Studies performed in animal models of memory loss associated with aging and AD have shown that α-LA improves memory in a variety of behavioral paradigms. Furthermore, molecular mechanisms underlying α-LA effects have also been investigated. Accordingly, α-LA shows antioxidant, antiapoptotic, anti-inflammatory, glioprotective, metal chelating properties in both in vivo and in vitro studies. In addition, it has been shown that α-LA reverses age-associated loss of neurotransmitters and their receptors. The review article aimed at summarizing and discussing the main studies investigating the neuroprotective effects of α-LA on cognition as well as its molecular effects, to improve the understanding of the therapeutic potential of α-LA in patients suffering from neurodegenerative disorders, supporting the development of clinical trials with α-LA.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
| | - Ahmed Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt; Center of Excellence for Screening of Environmental Contaminants, Benha University, Toukh, Egypt
| | - Samah F Ibrahim
- Clinical Sciences Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia; Forensic Medicine and Clinical Toxicology Department, College of Medicine, Cairo University, Cairo, Egypt
| | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Ensenanza e Investigacion en Bacteriologia Alimentaria, Universidad Nacinol Mayor de San Marcos, Lima, Peru; E-Health Research Center, Universidad de Ciencias y Humanidades, Lima, Peru
| | - Monika Sachdeva
- Fatima College of Health Sciences, Alain, United Arab Emirates
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman; School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
34
|
Huber KL, Fernández JR, Webb C, Rouzard K, Healy J, Tamura M, Stock JB, Stock M, Pérez E. AGSE: A Novel Grape Seed Extract Enriched for PP2A Activating Flavonoids That Combats Oxidative Stress and Promotes Skin Health. Molecules 2021; 26:molecules26216351. [PMID: 34770760 PMCID: PMC8587015 DOI: 10.3390/molecules26216351] [Citation(s) in RCA: 3] [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: 10/01/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 12/01/2022] Open
Abstract
Environmental stimuli attack the skin daily resulting in the generation of reactive oxygen species (ROS) and inflammation. One pathway that regulates oxidative stress in skin involves Protein Phosphatase 2A (PP2A), a phosphatase which has been previously linked to Alzheimer’s Disease and aging. Oxidative stress decreases PP2A methylation in normal human dermal fibroblasts (NHDFs). Thus, we hypothesize agents that increase PP2A methylation and activity will promote skin health and combat aging. To discover novel inhibitors of PP2A demethylation activity, we screened a library of 32 natural botanical extracts. We discovered Grape Seed Extract (GSE), which has previously been reported to have several benefits for skin, to be the most potent PP2A demethylating extract. Via several fractionation and extraction steps we developed a novel grape seed extract called Activated Grape Seed Extract (AGSE), which is enriched for PP2A activating flavonoids that increase potency in preventing PP2A demethylation when compared to commercial GSE. We then determined that 1% AGSE and 1% commercial GSE exhibit distinct gene expression profiles when topically applied to a 3D human skin model. To begin to characterize AGSE’s activity, we investigated its antioxidant potential and demonstrate it reduces ROS levels in NHDFs and cell-free assays equal to or better than Vitamin C and E. Moreover, AGSE shows anti-inflammatory properties, dose-dependently inhibiting UVA, UVB and chemical-induced inflammation. These results demonstrate AGSE is a novel, multi-functional extract that modulates methylation levels of PP2A and supports the hypothesis of PP2A as a master regulator for oxidative stress signaling and aging in skin.
Collapse
Affiliation(s)
- Kristen L. Huber
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
| | - José R. Fernández
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
| | - Corey Webb
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
| | - Karl Rouzard
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
| | - Jason Healy
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
| | - Masanori Tamura
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
| | - Jeffry B. Stock
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
- Department of Molecular Biology, Princeton University, Princeton, NJ 08852, USA
| | - Maxwell Stock
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
| | - Eduardo Pérez
- Research and Development Department, Signum Biosciences, 11 Deer Park Drive Suite 202, Monmouth Junction, NJ 08852, USA; (K.L.H.); (J.R.F.); (C.W.); (K.R.); (J.H.); (M.T.); (J.B.S.); (M.S.)
- Correspondence: ; Tel.: +1-732-329-6344; Fax: +1-732-329-8344
| |
Collapse
|
35
|
Zeng R, Jin C, Zheng C, Li S, Qian S, Pan J, Wang L, Zhao J, Qin L. OCT4 Represses Inflammation and Cell Injury During Orchitis by Regulating CIP2A Expression. Front Cell Dev Biol 2021; 9:683209. [PMID: 34513828 PMCID: PMC8427512 DOI: 10.3389/fcell.2021.683209] [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: 04/09/2021] [Accepted: 08/06/2021] [Indexed: 11/14/2022] Open
Abstract
Octamer-binding transcription factor 4 (OCT4) and cancerous inhibitor of protein phosphatase 2A (CIP2A) are upregulated in testicular cancer and cell lines. However, its contribution to orchitis (testicular inflammation) is unclear and was thus, investigated herein. Cell-based experiments on a lipopolysaccharide (LPS)-induced orchitis mouse model revealed robust inflammation, apoptotic cell death, and redox disorder in the Leydig (interstitial), Sertoli (supporting), and, germ cells. Meanwhile, real-time quantitative PCR revealed low OCT4 and CIP2A levels in testicular tissue and LPS-stimulated cells. A gain-of-function study showed that OCT4 overexpression not only increased CIP2A expression but also repressed LPS-induced inflammation, apoptosis, and redox disorder in the aforementioned cells. Furthermore, the re-inhibition of CIP2A expression by TD-19 in OCT4-overexpressing cells counteracted the effects of OCT4 overexpression on inflammation, apoptosis, and redox equilibrium. In addition, our results indicated that the Keap1-Nrf2-HO-1 signaling pathway was mediated by OCT4 and CIP2A. These findings provide insights into the potential mechanism underlying OCT4- and CIP2A-mediated testicular inflammation.
Collapse
Affiliation(s)
- Ruifeng Zeng
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengli Jin
- Department of First Clinical Medical School, Wenzhou Medical University, Wenzhou, China
| | - Chuchu Zheng
- Department of Second Clinical Medical School, Wenzhou Medical University, Wenzhou, China
| | - Shaoqi Li
- Department of Second Clinical Medical School, Wenzhou Medical University, Wenzhou, China
| | - Siyue Qian
- Department of Second Clinical Medical School, Wenzhou Medical University, Wenzhou, China
| | - Jingsa Pan
- Department of Second Clinical Medical School, Wenzhou Medical University, Wenzhou, China
| | - Lvhe Wang
- Department of Second Clinical Medical School, Wenzhou Medical University, Wenzhou, China
| | - Junfeng Zhao
- Department of Pediatric Surgery, Ningbo Women and Children's Hospital, Wenzhou, China
| | - Le Qin
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
36
|
Voronkov M, Ataiants J, Cocchiaro B, Stock JB, Lankenau SE. A vicious cycle of neuropathological, cognitive and behavioural sequelae of repeated opioid overdose. THE INTERNATIONAL JOURNAL OF DRUG POLICY 2021; 97:103362. [PMID: 34314956 DOI: 10.1016/j.drugpo.2021.103362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/03/2021] [Accepted: 06/23/2021] [Indexed: 01/15/2023]
Abstract
In the midst of an escalating U.S. opioid crisis, the immediate focus of public health interventions is on fatal overdose prevention. Few studies, however, have sought to examine the long-term health consequences of exposure to repeated nonfatal opioid overdose. We reviewed recent literature to examine three corresponding downstream health outcomes of repeated overdose: a) neurodegenerative processes; b) cognition and memory; and c) overdose risk behaviours. We found a remarkable congruency among available biochemical and cognitive data on how nonfatal overdose precipitates various pathological feedforward and feedback loops that affect people who use opioids for years to come. We found however that downstream behavioural implications of neurodegenerative and cognitive sequelae are less studied despite being most proximal to an overdose. Findings point to a vicious cycle of nonfatal overdose leading to neurodegeneration - closely resembling Alzheimer Disease - that results in cognitive decline that in turn leads to potentially reduced adherence to safe drug use behaviours. The collected evidence not only brings into the focus the long-term health consequences of nonfatal overdose from the perspectives of biology, neuroscience, and public health, but also creates new cross-disciplinary context and awareness in the research and public health community that should benefit people at risk.
Collapse
Affiliation(s)
| | - Janna Ataiants
- Dornsife School of Public Health, Drexel University, Philadelphia, PA 19104, USA.
| | - Benjamin Cocchiaro
- Center for Public Health Initiatives, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffry B Stock
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Stephen E Lankenau
- Dornsife School of Public Health, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
37
|
Shaw RL, Norton CE, Segal SS. Apoptosis in resistance arteries induced by hydrogen peroxide: greater resilience of endothelium versus smooth muscle. Am J Physiol Heart Circ Physiol 2021; 320:H1625-H1633. [PMID: 33606587 DOI: 10.1152/ajpheart.00956.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Reactive oxygen species (ROS) are implicated in cardiovascular and neurologic disorders including atherosclerosis, heart attack, stroke, and traumatic brain injury. Although oxidative stress can lead to apoptosis of vascular cells, such findings are largely based upon isolated vascular smooth muscle cells (SMCs) and endothelial cells (ECs) studied in culture. Studying intact resistance arteries, we have focused on understanding how SMCs and ECs in the blood vessel wall respond to acute oxidative stress induced by hydrogen peroxide, a ubiquitous, membrane-permeant ROS. We find that apoptosis induced by H2O2 is far greater in SMCs compared to ECs. For both cell types, apoptosis is associated with a rise in intracellular calcium concentration ([Ca2+]i) during H2O2 exposure. Consistent with their greater death, the rise in [Ca2+]i for SMCs exceeds that in ECs. Finding that disruption of the endothelium increases SMC death, we address how myoendothelial coupling and paracrine signaling attenuate apoptosis. Remarkably, conditions associated with chronic oxidative stress (advanced age, Western-style diet) protect SMCs during H2O2 exposure, as does female sex. In light of intracellular Ca2+ handling, we consider how glycolytic versus oxidative pathways for ATP production and changes in mitochondrial structure and function impact cellular resilience to H2O2-induced apoptosis. Gaining new insight into protective signaling within and between SMCs and ECs of the arterial wall can be applied to promote vascular cell survival (and recovery of blood flow) in tissues subjected to acute oxidative stress as occurs during reperfusion following myocardial infarction and thrombotic stroke.
Collapse
Affiliation(s)
- Rebecca L Shaw
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Charles E Norton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
38
|
Misrani A, Tabassum S, Yang L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer's Disease. Front Aging Neurosci 2021; 13:617588. [PMID: 33679375 PMCID: PMC7930231 DOI: 10.3389/fnagi.2021.617588] [Citation(s) in RCA: 306] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Mitochondria play a pivotal role in bioenergetics and respiratory functions, which are essential for the numerous biochemical processes underpinning cell viability. Mitochondrial morphology changes rapidly in response to external insults and changes in metabolic status via fission and fusion processes (so-called mitochondrial dynamics) that maintain mitochondrial quality and homeostasis. Damaged mitochondria are removed by a process known as mitophagy, which involves their degradation by a specific autophagosomal pathway. Over the last few years, remarkable efforts have been made to investigate the impact on the pathogenesis of Alzheimer’s disease (AD) of various forms of mitochondrial dysfunction, such as excessive reactive oxygen species (ROS) production, mitochondrial Ca2+ dyshomeostasis, loss of ATP, and defects in mitochondrial dynamics and transport, and mitophagy. Recent research suggests that restoration of mitochondrial function by physical exercise, an antioxidant diet, or therapeutic approaches can delay the onset and slow the progression of AD. In this review, we focus on recent progress that highlights the crucial role of alterations in mitochondrial function and oxidative stress in the pathogenesis of AD, emphasizing a framework of existing and potential therapeutic approaches.
Collapse
Affiliation(s)
- Afzal Misrani
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Sidra Tabassum
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| |
Collapse
|
39
|
Qadri M, ElSayed S, Elsaid KA. Fingolimod Phosphate (FTY720-P) Activates Protein Phosphatase 2A in Human Monocytes and Inhibits Monosodium Urate Crystal-Induced Interleukin-1 β Production. J Pharmacol Exp Ther 2021; 376:222-230. [PMID: 33239408 PMCID: PMC7873533 DOI: 10.1124/jpet.120.000321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/20/2020] [Indexed: 12/27/2022] Open
Abstract
Gout is a chronic inflammatory arthritis caused by monosodium urate monohydrate (MSU) crystal deposits in joints of lower limbs. Phagocytic uptake of MSU crystals by joint-resident macrophages and recruited circulating monocytes results in IL-1β expression and production. Current acute gout treatments have serious toxicities and suffer suboptimal clinical outcomes. Protein phosphatase 2A (PP2A) plays an important role in regulating signaling pathways relevant to inflammation. We hypothesized that innate immune danger signals, e.g., lipopolysaccharide (LPS) and soluble uric acid (sUA), prime human monocytes toward MSU crystal phagocytosis and that increased IL-1β production mediated by a reduction in PP2A activity and restoring PP2A activity exerts an anti-inflammatory effect in this setting. Priming monocytes with LPS + sUA increased cytosolic pro-IL-1β and mature IL-1β and enhanced MSU crystal phagocytosis and its downstream IL-1β expression (P < 0.001). A combination of LPS + sUA priming and MSU crystals reduced PP2A activity in monocytes by 60% (P = 0.013). PP2A catalytic subunit gene knockdown reduced PP2A activity and exacerbated MSU crystal-induced IL-1β expression and secretion (P < 0.0001). Fingolimod (FTY720) and its active metabolite, fingolimod phosphate (FTY720-P), were evaluated for their ability to activate PP2A in human monocytes over 24 hours. FTY720 and FTY720-P activated PP2A to a similar extent, and maximal enzyme activity occurred at 24 hours for FTY720 and at 6 hours for FTY720-P. FTY720-P (2.5 μM) reduced pro-IL-1β production and IL-1β secretion in primed and MSU crystal-stimulated monocytes (P < 0.0001) without changing the magnitude of crystal phagocytosis. We conclude that PP2A is a promising new target in acute gout. SIGNIFICANCE STATEMENT: The activity of protein phosphatase 2A (PP2A) is implicated in the enhanced expression and production of IL-1β by human monocytes in response to priming with soluble uric acid and lipopolysaccharide and phagocytosis of monosodium urate monohydrate (MSU) crystals. Fingolimod phosphate activates PP2A in human monocytes and reduces cytosolic pro-IL-1β content and its conversion to biologically active IL-1β in human monocytes exposed to MSU crystals.
Collapse
Affiliation(s)
- Marwa Qadri
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia (M.Q.) and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, Irvine, California (S.E., K.A.E.)
| | - Sandy ElSayed
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia (M.Q.) and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, Irvine, California (S.E., K.A.E.)
| | - Khaled A Elsaid
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia (M.Q.) and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Rinker Health Sciences Campus, Irvine, California (S.E., K.A.E.)
| |
Collapse
|
40
|
Kowluru A. Potential roles of PP2A-Rac1 signaling axis in pancreatic β-cell dysfunction under metabolic stress: Progress and promise. Biochem Pharmacol 2020; 180:114138. [PMID: 32634437 DOI: 10.1016/j.bcp.2020.114138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/16/2022]
Abstract
Recent estimates by the International Diabetes Federation suggest that the incidence of diabetes soared to an all-time high of 463 million in 2019, and the federation predicts that by 2045 the number of individuals afflicted with this disease will increase to 700 million. Therefore, efforts to understand the pathophysiology of diabetes are critical for moving toward the development of novel therapeutic strategies for this disease. Several contributors (oxidative stress, endoplasmic reticulum stress and others) have been proposed for the onset of metabolic dysfunction and demise of the islet β-cell leading to the pathogenesis of diabetes. Existing experimental evidence revealed sustained activation of PP2A and Rac1 in pancreatic β-cells exposed to metabolic stress (diabetogenic) conditions. Evidence in a variety of cell types implicates modulatory roles for specific signaling proteins (α4, SET, nm23-H1, Pak1) in the functional regulation of PP2A and Rac1. In this Commentary, I overviewed potential cross-talk between PP2A and Rac1 signaling modules in the onset of metabolic dysregulation of the islet β-cell leading to impaired glucose-stimulated insulin secretion (GSIS), loss of β-cell mass and the onset of diabetes. Potential knowledge gaps and future directions in this fertile area of islet biology are also highlighted. It is hoped that this Commentary will provide a basis for future studies toward a better understanding of roles of PP2A-Rac1 signaling module in pancreatic β-cell dysfunction, and identification of therapeutic targets for the treatment of islet β-cell dysfunction in diabetes.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- Biomedical Laboratory Research Service, John D. Dingell VA Medical Center and Departments of Pharmaceutical Sciences and Internal Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
41
|
Li X, Yang W, Li X, Chen M, Liu C, Li J, Yu S. Alpha-synuclein oligomerization and dopaminergic degeneration occur synchronously in the brain and colon of MPTP-intoxicated parkinsonian monkeys. Neurosci Lett 2019; 716:134640. [PMID: 31759083 DOI: 10.1016/j.neulet.2019.134640] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/01/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023]
Abstract
Dopaminergic (DAergic) degeneration and abnormal α-synuclein (α-syn) expression, phosphorylation and aggregation are observed in both the nigrostriatal system (NSS) and enteric nervous system (ENS) of patients with Parkinson's disease (PD). Whether these alterations in α-syn and DAergic neurons occur synchronously in the two nervous systems or follow a process that spreads from the gut to the brain remains a subject of debate. Here, in MPTP-intoxicated cynomolgus monkeys, we showed a parallel DAergic degeneration in the colon as well as in the substantia nigra and striatum (SN/STR), as indicated by reduced expression of tyrosine hydroxylase (TH) and dopamine transporter (DAT). In addition, we observed a simultaneous increase in the concentrations of total, phosphorylated, and oligomeric α-syn in the colon and SN/STR. Moreover, we identified that the above changes in α-syn were associated with an increase in the expression of polo-like kinase 2 (PLK2), an enzyme that promotes α-syn phosphorylation, and a decrease in the activity of protein phosphatase 2A (PP2A), an enzyme that facilitates α-syn dephosphorylation. Because the colonic ENS can be readily analyzed using routine biopsies, the shared pathological features between the colonic ENS and the brain NSS found in this study provide useful information for assessing and understanding the neuropathology in PD patients using colonic biopsies.
Collapse
Affiliation(s)
- Xuran Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Weiwei Yang
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Xin Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Min Chen
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Chengwei Liu
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jie Li
- Department of Neurology, Beijing Daxing District Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, China
| | - Shun Yu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China.
| |
Collapse
|
42
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
43
|
Xue J, Yan X, Yang Y, Chen M, Wu L, Gou Z, Sun Z, Talabieke S, Zheng Y, Luo D. Connexin 43 dephosphorylation contributes to arrhythmias and cardiomyocyte apoptosis in ischemia/reperfusion hearts. Basic Res Cardiol 2019; 114:40. [DOI: 10.1007/s00395-019-0748-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022]
|