1
|
Haratizadeh S, Nemati M, Basiri M, Nozari M. Erythropoietin and glial cells in central and peripheral nervous systems. Mol Biol Rep 2024; 51:1065. [PMID: 39422776 DOI: 10.1007/s11033-024-09997-2] [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: 09/04/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Erythropoietin (EPO) is the main hematopoietic growth factor prescribed to overcome anemia. It is also a neuroprotective agent. EPO binds to the erythropoietin receptor (EPOR), expressed on neurons and glial cells in the central nervous system (CNS), and exerts its neuroprotective potencies through the EPO-EPOR complex. The mechanism of the signal transduction pathways of EPO on glial cells is defined. EPO-EPOR complex can affect neurological disorders, such as Alzheimer's disease, Parkinson's disease, ischemia, retinal injury, stroke, hypoxia, trauma, and demyelinating diseases, through acting downstream signaling pathways. This review focuses on the roles of EPO in different types of glial cells (astrocytes, microglia, oligodendrocytes, and Schwann cells) and their relationships with signaling pathways. Information on the non-erythropoietic action of EPO and related signaling systems in connection with glial cells could enhance EPO treatment to restore different CNS disorders and propose new perspectives on the neuroprotective potential of EPO.
Collapse
Affiliation(s)
- Sara Haratizadeh
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mozhdeh Nemati
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohsen Basiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoumeh Nozari
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
2
|
Davoody S, Asgari Taei A, Khodabakhsh P, Dargahi L. mTOR signaling and Alzheimer's disease: What we know and where we are? CNS Neurosci Ther 2024; 30:e14463. [PMID: 37721413 PMCID: PMC11017461 DOI: 10.1111/cns.14463] [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: 07/07/2022] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
Despite the great body of research done on Alzheimer's disease, the underlying mechanisms have not been vividly investigated. To date, the accumulation of amyloid-beta plaques and tau tangles constitutes the hallmark of the disease; however, dysregulation of the mammalian target of rapamycin (mTOR) seems to be significantly involved in the pathogenesis of the disease as well. mTOR, as a serine-threonine protein kinase, was previously known for controlling many cellular functions such as cell size, autophagy, and metabolism. In this regard, mammalian target of rapamycin complex 1 (mTORC1) may leave anti-aging impacts by robustly inhibiting autophagy, a mechanism that inhibits the accumulation of damaged protein aggregate and dysfunctional organelles. Formation and aggregation of neurofibrillary tangles and amyloid-beta plaques seem to be significantly regulated by mTOR signaling. Understanding the underlying mechanisms and connection between mTOR signaling and AD may suggest conducting clinical trials assessing the efficacy of rapamycin, as an mTOR inhibitor drug, in managing AD or may help develop other medications. In this literature review, we aim to elaborate mTOR signaling network mainly in the brain, point to gaps of knowledge, and define how and in which ways mTOR signaling can be connected with AD pathogenesis and symptoms.
Collapse
Affiliation(s)
- Samin Davoody
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Afsaneh Asgari Taei
- Neuroscience Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Pariya Khodabakhsh
- Department of NeurophysiologyInstitute of Physiology, Eberhard Karls University of TübingenTübingenGermany
| | - Leila Dargahi
- Neurobiology Research CenterShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
3
|
Fussing F, Christensson J, Wörtwein G. Expression of erythropoietin receptor protein in the mouse hippocampus in response to normobaric hypoxia. Heliyon 2024; 10:e25051. [PMID: 38322970 PMCID: PMC10844123 DOI: 10.1016/j.heliyon.2024.e25051] [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: 05/07/2023] [Revised: 12/17/2023] [Accepted: 01/18/2024] [Indexed: 02/08/2024] Open
Abstract
Background Over the past decades, accumulating research on erythropoietin (EPO) and its receptor (EPOR) has revealed various neuroprotective actions and upregulation in hypoxic conditions. To our knowledge, EPOR protein levels in the hippocampus and isocortex have never been measured. Therefore, the aim of this study was to measure EPOR protein in the hippocampus (HPC) and prefrontal cortex (PFC). Further objectives were to examine the effects of exposure to normobaric hypoxia of various degrees and durations on EPOR protein and to explore how long-lasting these effects were. Method Adult C57BL/6 mice were randomized into a control group (N = 12) or various hypoxia groups (N = 5-11). Mice were exposed to three different O2 concentrations (10 %, 12 %, or 18 %) for 8 h a day for 5 days and sacrificed immediately after the last exposure. The effect of exposure to 12 % O2 for 1 day and 4 weeks (8 h per day) at this survival time was also examined. Additionally, groups of mice were exposed to 12 % O2 for 1 or 5 days (8 h per day) and euthanized at various times (up to 3 weeks) thereafter to examine the duration of EPOR protein regulation in the HPC and the PFC. EPOR protein was detected with a sandwich-ELISA method. Results EPOR protein was present in the HPC and PFC, at 206.64 ± 43.98 pg/mg and 184.25 ± 48.21 pg/mg, respectively. The highest increase in EPOR protein was observed in the HPC after 5 days of 8 h exposure to 12 % O2 and was most pronounced 24 h after last exposure. The effect of hypoxia normalized within one week after the last exposure. Conclusion This study successfully measured hippocampal EPOR protein and showed a significant association between normobaric hypoxia and acute EPOR elevation. It is our hope that this study can provide guidance to future research on the neuroprotective effects of EPO.
Collapse
Affiliation(s)
- F. Fussing
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services, Capital Region of Copenhagen and University Hospital of Copenhagen, Denmark
| | - J. Christensson
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services, Capital Region of Copenhagen and University Hospital of Copenhagen, Denmark
| | - G. Wörtwein
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Mental Health Services, Capital Region of Copenhagen and University Hospital of Copenhagen, Denmark
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Øster Farimagsgade 5, DK-1014, Copenhagen, Denmark
| |
Collapse
|
4
|
Maiese K. Microglia: Formidable Players in Alzheimer's Disease and Other Neurodegenerative Disorders. Curr Neurovasc Res 2024; 20:515-518. [PMID: 37888824 DOI: 10.2174/1567202620999231027155308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Indexed: 10/28/2023]
|
5
|
Sosa S, Bringas G, Urrutia N, Peñalver AI, López D, González E, Fernández A, Hernández ZM, Viña A, Peña Y, Batista JF, Valenzuela C, León K, Crombet T, Rodríguez T, Pérez L. NeuroEPO plus (NeuralCIM ®) in mild-to-moderate Alzheimer's clinical syndrome: the ATHENEA randomized clinical trial. Alzheimers Res Ther 2023; 15:215. [PMID: 38093366 PMCID: PMC10716956 DOI: 10.1186/s13195-023-01356-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/19/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND NeuroEPO plus is a recombinant human erythropoietin without erythropoietic activity and shorter plasma half-life due to its low sialic acid content. NeuroEPO plus prevents oxidative damage, neuroinflammation, apoptosis and cognitive deficit in an Alzheimer's disease (AD) models. The aim of this study was to assess efficacy and safety of neuroEPO plus. METHODS This was a double-blind, randomized, placebo-controlled, phase 2-3 trial involving participants ≥ 50 years of age with mild-to-moderate AD clinical syndrome. Participants were randomized in a 1:1:1 ratio to receive 0.5 or 1.0 mg of neuroEPO plus or placebo intranasally 3 times/week for 48 weeks. The primary outcome was change in the 11-item cognitive subscale of the AD Assessment Scale (ADAS-Cog11) score from baseline to 48 weeks (range, 0 to 70; higher scores indicate greater impairment). Secondary outcomes included CIBIC+, GDS, MoCA, NPI, Activities of Daily Living Scales, cerebral perfusion, and hippocampal volume. RESULTS A total of 174 participants were enrolled and 170 were treated (57 in neuroEPO plus 0.5 mg, 56 in neuroEPO plus 1.0 mg and 57 in placebo group). Mean age, 74.0 years; 121 (71.2%) women and 85% completed the trial. The median change in ADAS-Cog11 score at 48 weeks was -3.0 (95% CI, -4.3 to -1.7) in the 0.5 mg neuroEPO plus group, -4.0 (95% CI, -5.9 to -2.1) in the 1.0 mg neuroEPO plus group and 4.0 (95% CI, 1.9 to 6.1) in the placebo group. The difference of neuroEPO plus 0.5 mg vs. placebo was 7.0 points (95% CI, 4.5-9.5) P = 0.000 and between the neuroEPO plus 1.0 mg vs. placebo was 8.0 points (95% CI, 5.2-10.8) P = 0.000. NeuroEPO plus treatment induced a statistically significant improvement in some of clinical secondary outcomes vs. placebo including CIBIC+, GDS, MoCA, NPI, and the brain perfusion. CONCLUSIONS Among participants with mild-moderate Alzheimer's disease clinical syndrome, neuroEPO plus improved the cognitive evaluation at 48 weeks, with a very good safety profile. Larger trials are warranted to determine the efficacy and safety of neuroEPO plus in Alzheimer's disease. TRIAL REGISTRATION https://rpcec.sld.cu Identifier: RPCEC00000232.
Collapse
Affiliation(s)
- Saily Sosa
- Hospital Iván Portuondo, Calle 78 e/ Ave. 33 y 37, San Antonio de los Baños, Artemisa, CP 32 500, Cuba
| | - Giosmany Bringas
- National Institute of Neurology (INN), Calle 29 esquina D, Vedado, Havana, CP 10 400, Cuba
| | - Nelky Urrutia
- Hospital Iván Portuondo, Calle 78 e/ Ave. 33 y 37, San Antonio de los Baños, Artemisa, CP 32 500, Cuba
| | - Ana Ivis Peñalver
- National Institute of Neurology (INN), Calle 29 esquina D, Vedado, Havana, CP 10 400, Cuba
| | - Danay López
- Hospital Iván Portuondo, Calle 78 e/ Ave. 33 y 37, San Antonio de los Baños, Artemisa, CP 32 500, Cuba
| | - Evelio González
- Cuban Neurosciences Center (CNEURO), Avenida 25, No. 15 007, Cubanacán, Havana, CP 11 600, Cuba
| | - Ana Fernández
- Cuban Neurosciences Center (CNEURO), Avenida 25, No. 15 007, Cubanacán, Havana, CP 11 600, Cuba
| | - Zenaida Milagros Hernández
- Center of Neurological Restoration (CIREN), Calle 216 esquina 13, Siboney, Playa, Havana, CP 11 600, Cuba
| | - Ariel Viña
- Cuban Neurosciences Center (CNEURO), Avenida 25, No. 15 007, Cubanacán, Havana, CP 11 600, Cuba
| | - Yamile Peña
- Center for Clinical Investigation, CENTIS, Calle 45 No. 4501, esquina a 34, Reparto Kolhy, Havana, CP 11 300, Cuba
| | - Juan Felipe Batista
- Center for Clinical Investigation, CENTIS, Calle 45 No. 4501, esquina a 34, Reparto Kolhy, Havana, CP 11 300, Cuba
| | - Carmen Valenzuela
- Institute of Cybernetics, Mathematics and Physics (ICIMAF), Calle 15 #551 entre C y D, Plaza de la Revolución, Vedado, Havana, CP 10 400, Cuba
| | - Kalet León
- Center of Molecular Immunology (CIM), Calle 216 esquina 15, Siboney, Playa , Havana, CP 11 600, Cuba
| | - Tania Crombet
- Center of Molecular Immunology (CIM), Calle 216 esquina 15, Siboney, Playa , Havana, CP 11 600, Cuba
| | - Teresita Rodríguez
- Center of Molecular Immunology (CIM), Calle 216 esquina 15, Siboney, Playa , Havana, CP 11 600, Cuba
| | - Leslie Pérez
- Center of Molecular Immunology (CIM), Calle 216 esquina 15, Siboney, Playa , Havana, CP 11 600, Cuba.
| |
Collapse
|
6
|
Maiese K. Cornerstone Cellular Pathways for Metabolic Disorders and Diabetes Mellitus: Non-Coding RNAs, Wnt Signaling, and AMPK. Cells 2023; 12:2595. [PMID: 37998330 PMCID: PMC10670256 DOI: 10.3390/cells12222595] [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: 09/21/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Metabolic disorders and diabetes (DM) impact more than five hundred million individuals throughout the world and are insidious in onset, chronic in nature, and yield significant disability and death. Current therapies that address nutritional status, weight management, and pharmacological options may delay disability but cannot alter disease course or functional organ loss, such as dementia and degeneration of systemic bodily functions. Underlying these challenges are the onset of aging disorders associated with increased lifespan, telomere dysfunction, and oxidative stress generation that lead to multi-system dysfunction. These significant hurdles point to the urgent need to address underlying disease mechanisms with innovative applications. New treatment strategies involve non-coding RNA pathways with microRNAs (miRNAs) and circular ribonucleic acids (circRNAs), Wnt signaling, and Wnt1 inducible signaling pathway protein 1 (WISP1) that are dependent upon programmed cell death pathways, cellular metabolic pathways with AMP-activated protein kinase (AMPK) and nicotinamide, and growth factor applications. Non-coding RNAs, Wnt signaling, and AMPK are cornerstone mechanisms for overseeing complex metabolic pathways that offer innovative treatment avenues for metabolic disease and DM but will necessitate continued appreciation of the ability of each of these cellular mechanisms to independently and in unison influence clinical outcome.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
7
|
Maiese K. The impact of aging and oxidative stress in metabolic and nervous system disorders: programmed cell death and molecular signal transduction crosstalk. Front Immunol 2023; 14:1273570. [PMID: 38022638 PMCID: PMC10663950 DOI: 10.3389/fimmu.2023.1273570] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Life expectancy is increasing throughout the world and coincides with a rise in non-communicable diseases (NCDs), especially for metabolic disease that includes diabetes mellitus (DM) and neurodegenerative disorders. The debilitating effects of metabolic disorders influence the entire body and significantly affect the nervous system impacting greater than one billion people with disability in the peripheral nervous system as well as with cognitive loss, now the seventh leading cause of death worldwide. Metabolic disorders, such as DM, and neurologic disease remain a significant challenge for the treatment and care of individuals since present therapies may limit symptoms but do not halt overall disease progression. These clinical challenges to address the interplay between metabolic and neurodegenerative disorders warrant innovative strategies that can focus upon the underlying mechanisms of aging-related disorders, oxidative stress, cell senescence, and cell death. Programmed cell death pathways that involve autophagy, apoptosis, ferroptosis, and pyroptosis can play a critical role in metabolic and neurodegenerative disorders and oversee processes that include insulin resistance, β-cell function, mitochondrial integrity, reactive oxygen species release, and inflammatory cell activation. The silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), AMP activated protein kinase (AMPK), and Wnt1 inducible signaling pathway protein 1 (WISP1) are novel targets that can oversee programmed cell death pathways tied to β-nicotinamide adenine dinucleotide (NAD+), nicotinamide, apolipoprotein E (APOE), severe acute respiratory syndrome (SARS-CoV-2) exposure with coronavirus disease 2019 (COVID-19), and trophic factors, such as erythropoietin (EPO). The pathways of programmed cell death, SIRT1, AMPK, and WISP1 offer exciting prospects for maintaining metabolic homeostasis and nervous system function that can be compromised during aging-related disorders and lead to cognitive impairment, but these pathways have dual roles in determining the ultimate fate of cells and organ systems that warrant thoughtful insight into complex autofeedback mechanisms.
Collapse
Affiliation(s)
- Kenneth Maiese
- Innovation and Commercialization, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
8
|
Maiese K. Innovative therapeutic strategies for cardiovascular disease. EXCLI JOURNAL 2023; 22:690-715. [PMID: 37593239 PMCID: PMC10427777 DOI: 10.17179/excli2023-6306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023]
Abstract
As a significant non-communicable disease, cardiovascular disease is the leading cause of death for both men and women, comprises almost twenty percent of deaths in most racial and ethnic groups, can affect greater than twenty-five million individuals worldwide over the age of twenty, and impacts global economies with far-reaching financial challenges. Multiple factors can affect the onset of cardiovascular disease that include high serum cholesterol levels, elevated blood pressure, tobacco consumption and secondhand smoke exposure, poor nutrition, physical inactivity, obesity, and concurrent diabetes mellitus. Yet, addressing any of these factors cannot completely eliminate the onset or progression of cardiovascular disorders. Novel strategies are necessary to target underlying cardiovascular disease mechanisms. The silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), a histone deacetylase, can limit cardiovascular injury, assist with stem cell development, oversee metabolic homeostasis through nicotinamide adenine dinucleotide (NAD+) pathways, foster trophic factor protection, and control cell senescence through the modulation of telomere function. Intimately tied to SIRT1 pathways are mammalian forkhead transcription factors (FoxOs) which can modulate cardiac disease to reduce oxidative stress, repair microcirculation disturbances, and reduce atherogenesis through pathways of autophagy, apoptosis, and ferroptosis. AMP activated protein kinase (AMPK) also is critical among these pathways for the oversight of cardiac cellular metabolism, insulin sensitivity, mitochondrial function, inflammation, and the susceptibility to viral infections such as severe acute respiratory syndrome coronavirus that can impact cardiovascular disease. Yet, the relationship among these pathways is both intricate and complex and requires detailed insight to successfully translate these pathways into clinical care for cardiovascular disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022
| |
Collapse
|
9
|
Maiese K. Cognitive Impairment in Multiple Sclerosis. Bioengineering (Basel) 2023; 10:871. [PMID: 37508898 PMCID: PMC10376413 DOI: 10.3390/bioengineering10070871] [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: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Almost three million individuals suffer from multiple sclerosis (MS) throughout the world, a demyelinating disease in the nervous system with increased prevalence over the last five decades, and is now being recognized as one significant etiology of cognitive loss and dementia. Presently, disease modifying therapies can limit the rate of relapse and potentially reduce brain volume loss in patients with MS, but unfortunately cannot prevent disease progression or the onset of cognitive disability. Innovative strategies are therefore required to address areas of inflammation, immune cell activation, and cell survival that involve novel pathways of programmed cell death, mammalian forkhead transcription factors (FoxOs), the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), and associated pathways with the apolipoprotein E (APOE-ε4) gene and severe acute respiratory syndrome coronavirus (SARS-CoV-2). These pathways are intertwined at multiple levels and can involve metabolic oversight with cellular metabolism dependent upon nicotinamide adenine dinucleotide (NAD+). Insight into the mechanisms of these pathways can provide new avenues of discovery for the therapeutic treatment of dementia and loss in cognition that occurs during MS.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
10
|
Maiese K. Cellular Metabolism: A Fundamental Component of Degeneration in the Nervous System. Biomolecules 2023; 13:816. [PMID: 37238686 PMCID: PMC10216724 DOI: 10.3390/biom13050816] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
It is estimated that, at minimum, 500 million individuals suffer from cellular metabolic dysfunction, such as diabetes mellitus (DM), throughout the world. Even more concerning is the knowledge that metabolic disease is intimately tied to neurodegenerative disorders, affecting both the central and peripheral nervous systems as well as leading to dementia, the seventh leading cause of death. New and innovative therapeutic strategies that address cellular metabolism, apoptosis, autophagy, and pyroptosis, the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), growth factor signaling with erythropoietin (EPO), and risk factors such as the apolipoprotein E (APOE-ε4) gene and coronavirus disease 2019 (COVID-19) can offer valuable insights for the clinical care and treatment of neurodegenerative disorders impacted by cellular metabolic disease. Critical insight into and modulation of these complex pathways are required since mTOR signaling pathways, such as AMPK activation, can improve memory retention in Alzheimer's disease (AD) and DM, promote healthy aging, facilitate clearance of β-amyloid (Aß) and tau in the brain, and control inflammation, but also may lead to cognitive loss and long-COVID syndrome through mechanisms that can include oxidative stress, mitochondrial dysfunction, cytokine release, and APOE-ε4 if pathways such as autophagy and other mechanisms of programmed cell death are left unchecked.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
11
|
Maiese K. The Metabolic Basis for Nervous System Dysfunction in Alzheimer's Disease, Parkinson's Disease, and Huntington's Disease. Curr Neurovasc Res 2023; 20:314-333. [PMID: 37488757 PMCID: PMC10528135 DOI: 10.2174/1567202620666230721122957] [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: 05/04/2023] [Revised: 06/10/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Disorders of metabolism affect multiple systems throughout the body but may have the greatest impact on both central and peripheral nervous systems. Currently available treatments and behavior changes for disorders that include diabetes mellitus (DM) and nervous system diseases are limited and cannot reverse the disease burden. Greater access to healthcare and a longer lifespan have led to an increased prevalence of metabolic and neurodegenerative disorders. In light of these challenges, innovative studies into the underlying disease pathways offer new treatment perspectives for Alzheimer's Disease, Parkinson's Disease, and Huntington's Disease. Metabolic disorders are intimately tied to neurodegenerative diseases and can lead to debilitating outcomes, such as multi-nervous system disease, susceptibility to viral pathogens, and long-term cognitive disability. Novel strategies that can robustly address metabolic disease and neurodegenerative disorders involve a careful consideration of cellular metabolism, programmed cell death pathways, the mechanistic target of rapamycin (mTOR) and its associated pathways of mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), AMP-activated protein kinase (AMPK), growth factor signaling, and underlying risk factors such as the apolipoprotein E (APOE-ε4) gene. Yet, these complex pathways necessitate comprehensive understanding to achieve clinical outcomes that target disease susceptibility, onset, and progression.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022
| |
Collapse
|
12
|
Xu L, Li L, Pan C, Song J, Zhang C, Wu X, Hu F, Liu X, Zhang Z, Zhang Z. Erythropoietin signaling in peripheral macrophages is required for systemic β-amyloid clearance. EMBO J 2022; 41:e111038. [PMID: 36215698 PMCID: PMC9670197 DOI: 10.15252/embj.2022111038] [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: 02/28/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 01/13/2023] Open
Abstract
Impaired clearance of beta-amyloid (Aβ) is a primary cause of sporadic Alzheimer's disease (AD). Aβ clearance in the periphery contributes to reducing brain Aβ levels and preventing Alzheimer's disease pathogenesis. We show here that erythropoietin (EPO) increases phagocytic activity, levels of Aβ-degrading enzymes, and Aβ clearance in peripheral macrophages via PPARγ. Erythropoietin is also shown to suppress Aβ-induced inflammatory responses. Deletion of EPO receptor in peripheral macrophages leads to increased peripheral and brain Aβ levels and exacerbates Alzheimer's-associated brain pathologies and behavioral deficits in AD-model mice. Moreover, erythropoietin signaling is impaired in peripheral macrophages of old AD-model mice. Exogenous erythropoietin normalizes impaired EPO signaling and dysregulated functions of peripheral macrophages in old AD-model mice, promotes systemic Aβ clearance, and alleviates disease progression. Erythropoietin treatment may represent a potential therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Lu Xu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
- Department of Neurology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Lei Li
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Cai‐Long Pan
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
| | - Jing‐Jing Song
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Chen‐Yang Zhang
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Xiang‐Hui Wu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Fan Hu
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Xue Liu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Zhiren Zhang
- Institute of ImmunologyArmy Medical UniversityChongqingChina
| | - Zhi‐Yuan Zhang
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
- Department of Neurology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
- Key Laboratory of Human Functional Genomics of Jiangsu ProvinceNanjing Medical UniversityNanjingChina
| |
Collapse
|
13
|
Maiese K. Pyroptosis, Apoptosis, and Autophagy: Critical Players of Inflammation and Cell Demise in the Nervous System. Curr Neurovasc Res 2022; 19:241-244. [PMID: 35909267 DOI: 10.2174/1567202619666220729093449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
14
|
An Association between Insulin Resistance and Neurodegeneration in Zebrafish Larval Model ( Danio rerio). Int J Mol Sci 2022; 23:ijms23158290. [PMID: 35955446 PMCID: PMC9368350 DOI: 10.3390/ijms23158290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Type 2 diabetes mellitus has recently been identified as a mediator of neurodegeneration. However, the molecular mechanisms have not been clearly elucidated. We aimed to investigate insulin resistance associated with neurodegenerative events in zebrafish larvae. Methods: Larvae aged 72 h-post-fertilization (hpf) were induced to insulin resistance by immersion in 250 nM insulin and were then reinduced with 100 nM insulin at 96 hpf. This model was validated by a glucose levels assay, qPCR analysis of selected genes (akt, pepck, zglut3 and claudin-5a) and Oil Red-O (ORO) staining of the yolk sac for lipid distribution. The association of insulin resistance and neurodegeneration was validated by malondialdehyde (MDA), glutathione (GSH) assays, and by integrating next-generation sequencing with database for annotation, visualization and integrated discovery (DAVID). Results: There was a significant increase in glucose levels at 180 min in the insulin-resistant group. However, it decreased at 400 min after the re-challenge. Insulin-signaling mediators, akt and pepck, were showed significantly downregulated up to 400 min after insulin immersion (p < 0.05). Meanwhile, claudin-5a assessed blood−brain barrier (BBB) integrity and showed significant deterioration after 400 min of post-insulin immersion. ORO staining remarked the increase in yolk sac size in the insulin-resistant group. After the confirmation of insulin resistance, MDA levels increased significantly in the insulin-resistant group compared to the control group in the following parameters. Furthermore, dysregulated MAPK- and Wnt/Ca2+-signaling pathways were observed in the insulin-resistant group, disrupting energy metabolism and causing BBB injury. Conclusions: We conclude that the insulin-resistant zebrafish larvae alter the metabolic physiology associated with neurodegeneration.
Collapse
|
15
|
Arik E, Heinisch O, Bienert M, Gubeljak L, Slowik A, Reich A, Schulz JB, Wilhelm T, Huber M, Habib P. Erythropoietin Enhances Post-ischemic Migration and Phagocytosis and Alleviates the Activation of Inflammasomes in Human Microglial Cells. Front Cell Neurosci 2022; 16:915348. [PMID: 35813499 PMCID: PMC9263298 DOI: 10.3389/fncel.2022.915348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022] Open
Abstract
Recombinant human erythropoietin (rhEPO) has been shown to exert anti-apoptotic and anti-inflammatory effects after cerebral ischemia. Inflammatory cytokines interleukin-1β and -18 (IL-1β and IL-18) are crucial mediators of apoptosis and are maturated by multiprotein complexes termed inflammasomes. Microglia are the first responders to post-ischemic brain damage and are a main source of inflammasomes. However, the impact of rhEPO on microglial activation and the subsequent induction of inflammasomes after ischemia remains elusive. To address this, we subjected human microglial clone 3 (HMC-3) cells to various durations of oxygen-glucose-deprivation/reperfusion (OGD/R) to assess the impact of rhEPO on cell viability, metabolic activity, oxidative stress, phagocytosis, migration, as well as on the regulation and activation of the NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes. Administration of rhEPO mitigated OGD/R-induced oxidative stress and cell death. Additionally, it enhanced metabolic activity, migration and phagocytosis of HMC-3. Moreover, rhEPO attenuated post-ischemic activation and regulation of the NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes as well as their downstream effectors CASPASE1 and IL-1β. Pharmacological inhibition of NLRP3 via MCC950 had no effect on the activation of CASPASE1 and maturation of IL-1β after OGD/R, but increased protein levels of NLRP1, NLRC4, and AIM2, suggesting compensatory activities among inflammasomes. We provide evidence that EPO-conveyed anti-inflammatory actions might be mediated via the regulation of the inflammasomes.
Collapse
Affiliation(s)
- Eren Arik
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ole Heinisch
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michaela Bienert
- Institute of Molecular and Cellular Anatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Lara Gubeljak
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Department of Anatomy and Cell Biology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jörg B. Schulz
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-BRAIN Institute of Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-BRAIN Institute of Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
- *Correspondence: Pardes Habib, ; orcid.org/0000-0002-5771-216X
| |
Collapse
|
16
|
Maiese K. A Common Link in Neurovascular Regenerative Pathways: Protein Kinase B (Akt). Curr Neurovasc Res 2022; 19:1-4. [PMID: 35139797 DOI: 10.2174/1567202619666220209111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Al-Onaizi MA, Thériault P, Lecordier S, Prefontaine P, Rivest S, ElAli A. Early monocyte modulation by the non-erythropoietic peptide ARA 290 decelerates AD-like pathology progression. Brain Behav Immun 2022; 99:363-382. [PMID: 34343617 DOI: 10.1016/j.bbi.2021.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/06/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) pathology is characterized by amyloid-β (Aβ) deposition and tau hyper-phosphorylation, accompanied by a progressive cognitive decline. Monocytes have been recently shown to play a major role in modulating Aβ pathology, and thereby have been pointed as potential therapeutic targets. However, the main challenge remains in identifying clinically relevant interventions that could modulate monocyte immune functions in absence of undesired off-target effects. Erythropoietin (EPO), a key regulator of erythrocyte production, has been shown to possess immunomodulatory potential and to provide beneficial effects in preclinical models of AD. However, the transition to use recombinant human EPO in clinical trials was hindered by unwanted erythropoietic effects that could lead to thrombosis. Here, we used a recently identified non-erythropoietic analogue of EPO, ARA 290, to evaluate its therapeutic potential in AD therapy. We first evaluated the effects of early systemic ARA 290 administration on AD-like pathology in an early-onset model, represented by young APP/PS1 mice. Our findings indicate that ARA 290 early treatment decelerated Aβ pathology progression in APP/PS1 mice while improving cognitive functions. ARA 290 potently increased the levels of total monocytes by specifically stimulating the generation of Ly6CLow patrolling subset, which are implicated in clearing Aβ from the cerebral vasculature, and subsequently reducing overall Aβ burden in the brain. Moreover, ARA 290 increased the levels of monocyte progenitors in the bone marrow. Using chimeric APP/PS1 mice in which Ly6CLow patrolling subset are selectively depleted, ARA 290 was inefficient in attenuating Aβ pathology and ameliorating cognitive functions in young animals. Interestingly, ARA 290 effects were compromised when delivered in a late-onset model, represented by aged APP1/PS1. In aged APP/PS1 mice in which AD-like pathology is at advanced stages, ARA 290 failed to reverse Aβ pathology and to increase the levels of circulating monocytes. Our study suggests that ARA 290 early systemic treatment could prevent AD-like progression via modulation of monocyte functions by specifically increasing the ratio of patrolling monocytes.
Collapse
Affiliation(s)
- Mohammed A Al-Onaizi
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Peter Thériault
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Paul Prefontaine
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Serge Rivest
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
18
|
Kaur D, Behl T, Sehgal A, Singh S, Sharma N, Badavath VN, Ul Hassan SS, Hasan MM, Bhatia S, Al-Harassi A, Khan H, Bungau S. Unravelling the potential neuroprotective facets of erythropoietin for the treatment of Alzheimer's disease. Metab Brain Dis 2022; 37:1-16. [PMID: 34436747 DOI: 10.1007/s11011-021-00820-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023]
Abstract
During the last three decades, recombinant DNA technology has produced a wide range of hematopoietic and neurotrophic growth factors, including erythropoietin (EPO), which has emerged as a promising protein drug in the treatment of several diseases. Cumulative studies have recently indicated the neuroprotective role of EPO in preclinical models of acute and chronic neurodegenerative disorders, including Alzheimer's disease (AD). AD is one of the most prevalent neurodegenerative illnesses in the elderly, characterized by the accumulation of extracellular amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles (NFTs), which serve as the disease's two hallmarks. Unfortunately, AD lacks a successful treatment strategy due to its multifaceted and complex pathology. Various clinical studies, both in vitro and in vivo, have been conducted to identify the various mechanisms by which erythropoietin exerts its neuroprotective effects. The results of clinical trials in patients with AD are also promising. Herein, it is summarized and reviews all such studies demonstrating erythropoietin's potential therapeutic benefits as a pleiotropic neuroprotective agent in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Syed Shams Ul Hassan
- School of Medicine and Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mohammad Mehedi Hasan
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
- Amity Institute of Pharmacy, Amity University, Noida, Haryana, India
| | - Ahmed Al-Harassi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
19
|
Maiese K. Neurodegeneration, memory loss, and dementia: the impact of biological clocks and circadian rhythm. FRONT BIOSCI-LANDMRK 2021; 26:614-627. [PMID: 34590471 PMCID: PMC8756734 DOI: 10.52586/4971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022]
Abstract
Introduction: Dementia and cognitive loss impact a significant proportion of the global population and present almost insurmountable challenges for treatment since they stem from multifactorial etiologies. Innovative avenues for treatment are highly warranted. Methods and results: Novel work with biological clock genes that oversee circadian rhythm may meet this critical need by focusing upon the pathways of the mechanistic target of rapamycin (mTOR), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), mammalian forkhead transcription factors (FoxOs), the growth factor erythropoietin (EPO), and the wingless Wnt pathway. These pathways are complex in nature, intimately associated with autophagy that can maintain circadian rhythm, and have an intricate relationship that can lead to beneficial outcomes that may offer neuroprotection, metabolic homeostasis, and prevention of cognitive loss. However, biological clocks and alterations in circadian rhythm also have the potential to lead to devastating effects involving tumorigenesis in conjunction with pathways involving Wnt that oversee angiogenesis and stem cell proliferation. Conclusions: Current work with biological clocks and circadian rhythm pathways provide exciting possibilities for the treating dementia and cognitive loss, but also provide powerful arguments to further comprehend the intimate and complex relationship among these pathways to fully potentiate desired clinical outcomes.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
20
|
Zhang L, Qian Y, Li J, Zhou X, Xu H, Yan J, Xiang J, Yuan X, Sun B, Sisodia SS, Jiang YH, Cao X, Jing N, Lin A. BAD-mediated neuronal apoptosis and neuroinflammation contribute to Alzheimer's disease pathology. iScience 2021; 24:102942. [PMID: 34430820 PMCID: PMC8369003 DOI: 10.1016/j.isci.2021.102942] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/07/2021] [Accepted: 07/30/2021] [Indexed: 11/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most common progressive neurodegenerative disease. However, the underlying molecular mechanism is incompletely understood. Here we report that the pro-apoptotic protein BAD as a key regulator for neuronal apoptosis, neuroinflammation and Aβ clearance in AD. BAD pro-apoptotic activity is significantly increased in neurons of AD patients and 5XFAD mice. Conversely, genetic disruption of Bad alleles restores spatial learning and memory deficits in 5XFAD mice. Mechanistically, phosphorylation and inactivation of BAD by neurotropic factor-activated Akt is abrogated in neurons under AD condition. Through reactive oxygen species (ROS)-oxidized mitochondrial DNA (mtDNA) axis, BAD also promotes microglial NLRP3 inflammasome activation, thereby skewing microglia toward neuroinflammatory microglia to inhibit microglial phagocytosis of Aβ in AD mice. Our results support a model in which BAD contributes to AD pathologies by driving neuronal apoptosis and neuroinflammation but suppressing microglial phagocytosis of Aβ, suggesting that BAD is a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Liansheng Zhang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Modern Biology, Nanjing University, Nanjing 210023, China
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Yun Qian
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jie Li
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuan Zhou
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - He Xu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jie Yan
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allery & Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Jialing Xiang
- Department of Biological and Chemical Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Xiang Yuan
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Modern Biology, Nanjing University, Nanjing 210023, China
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Sangram S. Sisodia
- Department of Neurobiology, The University of Chicago, Chicago, IL 60637, USA
- The Microbiome Center, The University of Chicago, Chicago, IL 60637, USA
| | - Yong-Hui Jiang
- Department of Pediatrics and Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaohua Cao
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Naihe Jing
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Center of Cell Lineage and Atlas, Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510005, China
- Corresponding author
| | - Anning Lin
- Institute of Modern Biology, Nanjing University, Nanjing 210023, China
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
- Department of Neurobiology, The University of Chicago, Chicago, IL 60637, USA
- Grossman Institute for Neuroscience, Quantitative Biology, and Haman Behavior, The University of Chicago, Chicago, IL 60637, USA
- Corresponding author
| |
Collapse
|
21
|
Vittori DC, Chamorro ME, Hernández YV, Maltaneri RE, Nesse AB. Erythropoietin and derivatives: Potential beneficial effects on the brain. J Neurochem 2021; 158:1032-1057. [PMID: 34278579 DOI: 10.1111/jnc.15475] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/24/2021] [Accepted: 07/14/2021] [Indexed: 12/16/2022]
Abstract
Erythropoietin (Epo), the main erythropoiesis-stimulating factor widely prescribed to overcome anemia, is also known nowadays for its cytoprotective action on non-hematopoietic tissues. In this context, Epo showed not only its ability to cross the blood-brain barrier, but also its expression in the brain of mammals. In clinical trials, recombinant Epo treatment has been shown to stimulate neurogenesis; improve cognition; and activate antiapoptotic, antioxidant, and anti-inflammatory signaling pathways. These mechanisms, proposed to characterize a neuroprotective property, opened new perspectives on the Epo pharmacological potencies. However, many questions arise about a possible physiological role of Epo in the central nervous system (CNS) and the factors or environmental conditions that induce its expression. Although Epo may be considered a strong candidate to be used against neuronal damage, long-term treatments, particularly when high Epo doses are needed, may induce thromboembolic complications associated with increases in hematocrit and blood viscosity. To avoid these adverse effects, different Epo analogs without erythropoietic activity but maintaining neuroprotection ability are currently being investigated. Carbamylated erythropoietin, as well as alternative molecules like Epo fusion proteins and partial peptides of Epo, seems to match this profile. This review will focus on the discussion of experimental evidence reported in recent years linking erythropoietin and CNS function through investigations aimed at finding benefits in the treatment of neurodegenerative diseases. In addition, it will review the proposed mechanisms for novel derivatives which may clarify and, eventually, improve the neuroprotective action of Epo.
Collapse
Affiliation(s)
- Daniela C Vittori
- Department of Biological Chemistry, National Scientific and Technical Research Council, Institute of Biological Chemistry (IQUIBICEN), School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - María E Chamorro
- Department of Biological Chemistry, National Scientific and Technical Research Council, Institute of Biological Chemistry (IQUIBICEN), School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Yender V Hernández
- Department of Biological Chemistry, National Scientific and Technical Research Council, Institute of Biological Chemistry (IQUIBICEN), School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Romina E Maltaneri
- Department of Biological Chemistry, National Scientific and Technical Research Council, Institute of Biological Chemistry (IQUIBICEN), School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Alcira B Nesse
- Department of Biological Chemistry, National Scientific and Technical Research Council, Institute of Biological Chemistry (IQUIBICEN), School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
22
|
Xu JX, Fang K, Gao XR, Liu S, Ge JF. Resveratrol Protects SH-SY5Y Cells Against Oleic Acid-Induced Glucolipid Metabolic Dysfunction and Cell Injuries Via the Wnt/β-Catenin Signalling Pathway. Neurochem Res 2021; 46:2936-2947. [PMID: 34260003 DOI: 10.1007/s11064-021-03398-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/23/2022]
Abstract
Resveratrol (RES) is a polyphenol with diverse beneficial biological and pharmacological activities, and our previous results have demonstrated its neuroprotective effects in several metabolic diseases, including non-alcoholic fatty liver disease. The aim of the present study is to investigate the potential effect of RES against oleic acid (OA)-induced cell injuries in SH-SY5Y cells and explore the possible mechanism. Based on the dose- and time-dependent effects of OA on cell proliferation and LDH release, SH-SY5Y cells were challenged with OA and incubated with or without RES (10-5-10-9 mM) or sitagliptin (STG, 10-7 mM). Lipid accumulation, SREBP1 and PPARα protein expression, glucose consumption and IRS1, AKT, ERK phosphorylation under insulin stimulation, and ROS production were detected. The protein expression of brain-derived neurotrophic factor (BDNF), Copine 6, and key molecules in the Wnt/β-catenin signalling pathway were measured via western blot. The expression of Wnt 1 was also measured via immunofluorescence staining. The results showed that RES treatment could alleviate the neurotoxicity induced by OA, as indicated by the increased cell proliferation and the decreased concentration of LDH in the supernatant. The increased lipid deposition and protein expression of SREBP1 and PPARα induced by OA was also reversed by treatment with RES. Moreover, RES could upregulate glucose consumption and the protein expression of phosphorylated IRS1, AKT, ERK and reduced ROS production in OA-induced SH-SY5Y cells. Furthermore, RES treatment reversed the imbalanced protein expression of BDNF, Copine 6, p-β-catenin, and Wnt 1 in SH-SY5Y cells induced by OA and decreased the hyperexpression of p-GSK3β. However, these effects were suppressed by DKK1, which is a specific antagonist of the Wnt signalling pathway. These results suggested that RES has a neuroprotective effect against OA-induced cell injury and dysfunctional glucolipid metabolism, and the mechanism might involve its ability to regulate oxidative stress and insulin resistance via the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Jing-Xian Xu
- School of Pharmacy, Anhui Medical University, 81 Mei-Shan Road, Hefei, 230032, Anhui, China.,Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Ke Fang
- School of Pharmacy, Anhui Medical University, 81 Mei-Shan Road, Hefei, 230032, Anhui, China.,Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xin-Ran Gao
- School of Pharmacy, Anhui Medical University, 81 Mei-Shan Road, Hefei, 230032, Anhui, China.,Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Sen Liu
- School of Pharmacy, Anhui Medical University, 81 Mei-Shan Road, Hefei, 230032, Anhui, China.,Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jin-Fang Ge
- School of Pharmacy, Anhui Medical University, 81 Mei-Shan Road, Hefei, 230032, Anhui, China. .,Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China. .,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| |
Collapse
|
23
|
Maiese K. Cognitive Impairment and Dementia: Gaining Insight through Circadian Clock Gene Pathways. Biomolecules 2021; 11:1002. [PMID: 34356626 PMCID: PMC8301848 DOI: 10.3390/biom11071002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 01/18/2023] Open
Abstract
Neurodegenerative disorders affect fifteen percent of the world's population and pose a significant financial burden to all nations. Cognitive impairment is the seventh leading cause of death throughout the globe. Given the enormous challenges to treat cognitive disorders, such as Alzheimer's disease, and the inability to markedly limit disease progression, circadian clock gene pathways offer an exciting strategy to address cognitive loss. Alterations in circadian clock genes can result in age-related motor deficits, affect treatment regimens with neurodegenerative disorders, and lead to the onset and progression of dementia. Interestingly, circadian pathways hold an intricate relationship with autophagy, the mechanistic target of rapamycin (mTOR), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), mammalian forkhead transcription factors (FoxOs), and the trophic factor erythropoietin. Autophagy induction is necessary to maintain circadian rhythm homeostasis and limit cortical neurodegenerative disease, but requires a fine balance in biological activity to foster proper circadian clock gene regulation that is intimately dependent upon mTOR, SIRT1, FoxOs, and growth factor expression. Circadian rhythm mechanisms offer innovative prospects for the development of new avenues to comprehend the underlying mechanisms of cognitive loss and forge ahead with new therapeutics for dementia that can offer effective clinical treatments.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
24
|
Querfurth H, Lee HK. Mammalian/mechanistic target of rapamycin (mTOR) complexes in neurodegeneration. Mol Neurodegener 2021; 16:44. [PMID: 34215308 PMCID: PMC8252260 DOI: 10.1186/s13024-021-00428-5] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Novel targets to arrest neurodegeneration in several dementing conditions involving misfolded protein accumulations may be found in the diverse signaling pathways of the Mammalian/mechanistic target of rapamycin (mTOR). As a nutrient sensor, mTOR has important homeostatic functions to regulate energy metabolism and support neuronal growth and plasticity. However, in Alzheimer's disease (AD), mTOR alternately plays important pathogenic roles by inhibiting both insulin signaling and autophagic removal of β-amyloid (Aβ) and phospho-tau (ptau) aggregates. It also plays a role in the cerebrovascular dysfunction of AD. mTOR is a serine/threonine kinase residing at the core in either of two multiprotein complexes termed mTORC1 and mTORC2. Recent data suggest that their balanced actions also have implications for Parkinson's disease (PD) and Huntington's disease (HD), Frontotemporal dementia (FTD) and Amyotrophic Lateral Sclerosis (ALS). Beyond rapamycin; an mTOR inhibitor, there are rapalogs having greater tolerability and micro delivery modes, that hold promise in arresting these age dependent conditions.
Collapse
Affiliation(s)
- Henry Querfurth
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA.
| | - Han-Kyu Lee
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Akhter R, Shao Y, Formica S, Khrestian M, Bekris LM. TREM2 alters the phagocytic, apoptotic and inflammatory response to Aβ 42 in HMC3 cells. Mol Immunol 2021; 131:171-179. [PMID: 33461764 DOI: 10.1016/j.molimm.2020.12.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/20/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation in the brain of extracellular amyloid β (Aβ) plaques as well as intraneuronal inclusions (neurofibrillary tangles) consisting of total tau and phosphorylated tau. Also present are dystrophic neurites, loss of synapses, neuronal death, and gliosis. AD genetic studies have highlighted the importance of inflammation in this disease by identifying several risk associated immune response genes, including TREM2. TREM2 has been strongly implicated in basic microglia function including, phagocytosis, apoptosis, and the inflammatory response to Aβ in mouse brain and primary cells. These studies show that microglia are key players in the response to Aβ and in the accumulation of AD pathology. However, details are still missing about which apoptotic or inflammatory factors rely on TREM2 in their response to Aβ, especially in human cell lines. Given these previous findings our hypothesis is that TREM2 influences the response to Aβ toxicity by enhancing phagocytosis and inhibiting both the BCL-2 family of apoptotic proteins and pro-inflammatory cytokines. Aβ42 treatment of the human microglial cell line, HMC3 cells, was performed and TREM2 was overexpressed or silenced and the phagocytosis, apoptosis and inflammatory response were evaluated. Results indicate that a robust phagocytic response to Aβ after 24 h requires TREM2 in HMC3 cells. Also, TREM2 inhibits Aβ induced apoptosis by activating the Mcl-1/Bim complex. TREM2 is involved in activation of IP-10, MIP-1a, and IL-8, while it inhibits FGF-2, VEGF and GRO. Taken together, TREM2 plays a role in enhancing the microglial functional response to Aβ toxicity in HMC3 cells. This novel information suggests that therapeutic strategies that seek to activate TREM2 may not only enhance phagocytosis and inhibit apoptosis, but may also inhibit beneficial inflammatory factors, emphasizing the need to define TREM2-related inflammatory activity in not only mouse models of AD, but also in human AD.
Collapse
Affiliation(s)
- Rumana Akhter
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Yvonne Shao
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Shane Formica
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Maria Khrestian
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Lynn M Bekris
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
26
|
Abstract
The global increase in lifespan noted not only in developed nations, but also in large developing countries parallels an observed increase in a significant number of non-communicable diseases, most notable neurodegenerative disorders. Neurodegenerative disorders present a number of challenges for treatment options that do not resolve disease progression. Furthermore, it is believed by the year 2030, the services required to treat cognitive disorders in the United States alone will exceed $2 trillion annually. Mammalian forkhead transcription factors, silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae), the mechanistic target of rapamycin, and the pathways of autophagy and apoptosis offer exciting avenues to address these challenges by focusing upon core cellular mechanisms that may significantly impact nervous system disease. These pathways are intimately linked such as through cell signaling pathways involving protein kinase B and can foster, sometimes in conjunction with trophic factors, enhanced neuronal survival, reduction in toxic intracellular accumulations, and mitochondrial stability. Feedback mechanisms among these pathways also exist that can oversee reparative processes in the nervous system. However, mammalian forkhead transcription factors, silent mating type information regulation 2 homolog 1, mechanistic target of rapamycin, and autophagy can lead to cellular demise under some scenarios that may be dependent upon the precise cellular environment, warranting future studies to effectively translate these core pathways into successful clinical treatment strategies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling New York, New York, NY, USA
| |
Collapse
|
27
|
Gaindh D, Choi YB, Marchese M, Dowling P, Cook S, Blumberg B, Park JH, Lu W. Prolonged Beneficial Effect of Brief Erythropoietin Peptide JM4 Therapy on Chronic Relapsing EAE. Neurotherapeutics 2021; 18:401-411. [PMID: 32959273 PMCID: PMC8116362 DOI: 10.1007/s13311-020-00923-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2020] [Indexed: 12/28/2022] Open
Abstract
Potent beneficial immunomodulatory and anti-inflammatory effects of whole-molecule erythropoietin have been demonstrated in a variety of animal disease models including experimental autoimmune encephalomyelitis (EAE); however, excessive hematopoiesis limits its use in clinical applications. Our group previously generated an Epo-derived small peptide JM4 that is side-effect free and has strong neuroprotective activity without hematologic effects. Here, we investigated the long-term clinical effects of brief treatment with JM4 in chronic relapsing EAE using bioluminescence imaging (BLI) in transgenic mice containing the luciferase gene driven by the murine GFAP promoter. EAE mice treated with JM4 exhibited marked improvement in clinical scores and showed fewer disease flareups than control animals. JM4 therapy concomitantly led to markedly decreased GFAP bioluminescence in the brain and spinal cord in both acute and chronic relapsing EAE mouse models. We found a marker for toxic A1 astrocytes, complement component C3, that is upregulated in the brain and cord of EAE mice and sharply reduced in JM4-treated animals. In addition, an abnormally leaky neurovascular unit permeability was rapidly normalized within 5 days by JM4 therapy. The prolonged therapeutic benefit seen following brief JM4 treatment in EAE mice closely resemble that recently described in humans receiving pulsed immune reconstitution therapy with the disease-modifying compounds, alemtuzumab and cladribine. Our study suggests that JM4 therapy may have widespread clinical applicability for long-term treatment of inflammatory demyelinating diseases and that BLI is a useful noninvasive means of monitoring murine disease activity of the central nervous system.
Collapse
Affiliation(s)
- Deeya Gaindh
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA.
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA.
| | - Yun-Beom Choi
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Michelle Marchese
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
| | - Peter Dowling
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Stuart Cook
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Benjamin Blumberg
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
| | - James H Park
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
| | - Wei Lu
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
| |
Collapse
|
28
|
Huang C, Wen C, Yang M, Li A, Fan C, Gan D, Li Q, Zhao J, Zhu L, Lu D. Astaxanthin Improved the Cognitive Deficits in APP/PS1 Transgenic Mice Via Selective Activation of mTOR. J Neuroimmune Pharmacol 2020; 16:609-619. [PMID: 32944864 DOI: 10.1007/s11481-020-09953-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/26/2020] [Indexed: 01/20/2023]
Abstract
Astaxanthin (Ast) is an effective neuroprotective and antioxidant compound used to treat Alzheimer's disease (AD); however, the underlying in vivo molecular mechanisms remain unknown. In this study, we report that Ast can activate the mammalian target of rapamycin (mTOR) pathway in the 8-month-old APP/PS1 transgenic mouse model of AD. Our results suggest that Ast could ameliorate the cognitive defects in APP/PS1 mice by activating the mTOR pathway. Moreover, mTOR activation perturbed the mitochondrial dynamics, increased the synaptic plasticity after 21 days of treatment with Ast (10 mg/kg/day), and increased the expression of Aβ-degrading enzymes, mitochondrial fusion, and synapse-associated proteins and decreased the expression of mitochondrial fission proteins. Intraperitoneal injection of the mTOR inhibitor, rapamycin, abolished the effects of Ast. In conclusion, Ast activates the mTOR pathway, which is necessary for mitochondrial dynamics and synaptic plasticity, leading to improved learning and memory. Our results support the use of Ast for the treatment of cognitive deficits. Graphical abstract In summary, Ast ameliorates cognitive deficits via facilitating the mTOR-dependent mitochondrial dynamics and synaptic damage, and reducing Aβ accumulation. This model supports the use of Ast for the treatment of cognitive deficits.
Collapse
Affiliation(s)
- Cuiqin Huang
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Caiyan Wen
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Mei Yang
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - An Li
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Chongzhu Fan
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Danhui Gan
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Qin Li
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Jiayi Zhao
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Lihong Zhu
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Daxiang Lu
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China.
| |
Collapse
|
29
|
Maiese K. Dysregulation of metabolic flexibility: The impact of mTOR on autophagy in neurodegenerative disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:1-35. [PMID: 32854851 DOI: 10.1016/bs.irn.2020.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Non-communicable diseases (NCDs) that involve neurodegenerative disorders and metabolic disease impact over 400 million individuals globally. Interestingly, metabolic disorders, such as diabetes mellitus, are significant risk factors for the development of neurodegenerative diseases. Given that current therapies for these NCDs address symptomatic care, new avenues of discovery are required to offer treatments that affect disease progression. Innovative strategies that fill this void involve the mechanistic target of rapamycin (mTOR) and its associated pathways of mTOR complex 1 (mTORC1), mTOR complex 2 (mTORC2), AMP activated protein kinase (AMPK), trophic factors that include erythropoietin (EPO), and the programmed cell death pathways of autophagy and apoptosis. These pathways are intriguing in their potential to provide effective care for metabolic and neurodegenerative disorders. Yet, future work is necessary to fully comprehend the entire breadth of the mTOR pathways that can effectively and safely translate treatments to clinical medicine without the development of unexpected clinical disabilities.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY, United States.
| |
Collapse
|
30
|
Abstract
Metabolic disorders, such as diabetes mellitus (DM), are increasingly becoming significant risk factors for the health of the global population and consume substantial portions of the gross domestic product of all nations. Although conventional therapies that include early diagnosis, nutritional modification of diet, and pharmacological treatments may limit disease progression, tight serum glucose control cannot prevent the onset of future disease complications. With these concerns, novel strategies for the treatment of metabolic disorders that involve the vitamin nicotinamide, the mechanistic target of rapamycin (mTOR), mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), AMP activated protein kinase (AMPK), and the cellular pathways of autophagy and apoptosis offer exceptional promise to provide new avenues of treatment. Oversight of these pathways can promote cellular energy homeostasis, maintain mitochondrial function, improve glucose utilization, and preserve pancreatic beta-cell function. Yet, the interplay among mTOR, AMPK, and autophagy pathways can be complex and affect desired clinical outcomes, necessitating further investigations to provide efficacious treatment strategies for metabolic dysfunction and DM.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022,
| |
Collapse
|
31
|
Rey F, Balsari A, Giallongo T, Ottolenghi S, Di Giulio AM, Samaja M, Carelli S. Erythropoietin as a Neuroprotective Molecule: An Overview of Its Therapeutic Potential in Neurodegenerative Diseases. ASN Neuro 2020; 11:1759091419871420. [PMID: 31450955 PMCID: PMC6712762 DOI: 10.1177/1759091419871420] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Erythropoietin (EPO) is a cytokine mainly induced in hypoxia conditions. Its major production site is the kidney. EPO primarily acts on the erythroid progenitor cells in the bone marrow. More and more studies are highlighting its secondary functions, with a crucial focus on its role in the central nervous system. Here, EPO may interact with up to four distinct isoforms of its receptor (erythropoietin receptor [EPOR]), activating different signaling cascades with roles in neuroprotection and neurogenesis. Indeed, the EPO/EPOR axis has been widely studied in the neurodegenerative diseases field. Its potential therapeutic effects have been evaluated in multiple disorders, such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, spinal cord injury, as well as brain ischemia, hypoxia, and hyperoxia. EPO is showing great promise by counteracting secondary neuroinflammatory processes, reactive oxygen species imbalance, and cell death in these diseases. Multiple studies have been performed both in vitro and in vivo, characterizing the mechanisms through which EPO exerts its neurotrophic action. In some cases, clinical trials involving EPO have been performed, highlighting its therapeutic potential. Together, all these works indicate the potential beneficial effects of EPO.
Collapse
Affiliation(s)
- Federica Rey
- 1 Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Italy
| | - Alice Balsari
- 1 Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Italy
| | - Toniella Giallongo
- 1 Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Italy
| | - Sara Ottolenghi
- 2 Laboratory of Biochemistry, Department of Health Sciences, University of Milan, Italy
| | - Anna M Di Giulio
- 1 Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Italy.,3 Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Italy
| | - Michele Samaja
- 2 Laboratory of Biochemistry, Department of Health Sciences, University of Milan, Italy
| | - Stephana Carelli
- 1 Laboratory of Pharmacology, Department of Health Sciences, University of Milan, Italy.,3 Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Italy
| |
Collapse
|
32
|
Pan YR, Song JY, Fan B, Wang Y, Che L, Zhang SM, Chang YX, He C, Li GY. mTOR may interact with PARP-1 to regulate visible light-induced parthanatos in photoreceptors. Cell Commun Signal 2020; 18:27. [PMID: 32066462 PMCID: PMC7025415 DOI: 10.1186/s12964-019-0498-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Excessive light exposure is a detrimental environmental factor that plays a critical role in the pathogenesis of retinal degeneration. However, the mechanism of light-induced death of retina/photoreceptor cells remains unclear. The mammalian/mechanistic target of rapamycin (mTOR) and Poly (ADP-ribose) polymerase-1 (PARP-1) have become the primary targets for treating many neurodegenerative disorders. The aim of this study was to elucidate the mechanisms underlying light-induced photoreceptor cell death and whether the neuroprotective effects of mTOR and PARP-1 inhibition against death are mediated through apoptosis-inducing factor (AIF). METHODS Propidium iodide (PI)/Hoechst staining, lentiviral-mediated short hairpin RNA (shRNA), Western blot analysis, cellular fraction separation, plasmid transient transfection, laser confocal microscopy, a mice model, electroretinography (ERG), and hematoxylin-eosin (H & E) staining were employed to explore the mechanisms by which rapamycin/3-Aminobenzamide (3AB) exert neuroprotective effects of mTOR/PARP-1 inhibition in light-injured retinas. RESULTS A parthanatos-like death mechanism was evaluated in light-injured 661 W cells that are an immortalized photoreceptor-like cell line that exhibit cellular and biochemical feature characteristics of cone photoreceptor cells. The death process featured over-activation of PARP-1 and AIF nuclear translocation. Either PARP-1 or AIF knockdown played a significantly protective role for light-damaged photoreceptors. More importantly, crosstalk was observed between mTOR and PARP-1 signaling and mTOR could have regulated parthanatos via the intermediate factor sirtuin 1 (SIRT1). The parthanatos-like injury was also verified in vivo, wherein either PARP-1 or mTOR inhibition provided significant neuroprotection against light-induced injury, which is evinced by both structural and functional retinal analysis. Overall, these results elucidate the mTOR-regulated parthanatos death mechanism in light-injured photoreceptors/retinas and may facilitate the development of novel neuroprotective therapies for retinal degeneration diseases. CONCLUSIONS Our results demonstrate that inhibition of the mTOR/PARP-1 axis exerts protective effects on photoreceptors against visible-light-induced parthanatos. These protective effects are conducted by regulating the downstream factors of AIF, while mTOR possibly interacts with PARP-1 via SIRT1 to regulate parthanatos. Video Abstract Schematic diagram of mTOR interacting with PARP-1 to regulate visible light-induced parthanatos. Increased ROS caused by light exposure penetrates the nuclear membrane and causes nuclear DNA strand breaks. PARP-1 detects DNA breaks and synthesizes PAR polymers to initiate the DNA repair system that consumes a large amount of cellular NAD+. Over-production of PAR polymers prompts the release of AIF from the mitochondria and translocation to the nucleus, which leads to parthanatos. Activated mTOR may interact with PARP-1 via SIRT1 to regulate visible light-induced parthanatos.
Collapse
Affiliation(s)
- Yi-Ran Pan
- Department of Ophthalmology, Second Hospital of JiLin University, No.218 Zi-Qiang St, ChangChun, 130041 China
| | - Jing-Yao Song
- Department of Ophthalmology, Second Hospital of JiLin University, No.218 Zi-Qiang St, ChangChun, 130041 China
| | - Bin Fan
- Department of Ophthalmology, Second Hospital of JiLin University, No.218 Zi-Qiang St, ChangChun, 130041 China
| | - Ying Wang
- Department of Hemooncolog, Second Hospital of JiLin University, ChangChun, 130041 China
| | - Lin Che
- Department of Ophthalmology, Second Hospital of JiLin University, No.218 Zi-Qiang St, ChangChun, 130041 China
| | - Si-Ming Zhang
- Department of Ophthalmology, Second Hospital of JiLin University, No.218 Zi-Qiang St, ChangChun, 130041 China
| | - Yu-Xin Chang
- Department of Orthopedics, Second Hospital of JiLin University, ChangChun, 130041 China
| | - Chang He
- Department of Genetics,Basic, Medical College of Jilin University, ChangChun, 130041 China
| | - Guang-Yu Li
- Department of Ophthalmology, Second Hospital of JiLin University, No.218 Zi-Qiang St, ChangChun, 130041 China
| |
Collapse
|
33
|
Maiese K. Cognitive impairment with diabetes mellitus and metabolic disease: innovative insights with the mechanistic target of rapamycin and circadian clock gene pathways. Expert Rev Clin Pharmacol 2020; 13:23-34. [PMID: 31794280 PMCID: PMC6959472 DOI: 10.1080/17512433.2020.1698288] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022]
Abstract
Introduction: Dementia is the 7th leading cause of death that imposes a significant financial and service burden on the global population. Presently, only symptomatic care exists for cognitive loss, such as Alzheimer's disease.Areas covered: Given the advancing age of the global population, it becomes imperative to develop innovative therapeutic strategies for cognitive loss. New studies provide insight to the association of cognitive loss with metabolic disorders, such as diabetes mellitus.Expert opinion: Diabetes mellitus is increasing in incidence throughout the world and affects 350 million individuals. Treatment strategies identifying novel pathways that oversee metabolic and neurodegenerative disorders offer exciting prospects to treat dementia. The mechanistic target of rapamycin (mTOR) and circadian clock gene pathways that include AMP activated protein kinase (AMPK), Wnt1 inducible signaling pathway protein 1 (WISP1), erythropoietin (EPO), and silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) provide novel strategies to treat cognitive loss that has its basis in metabolic cellular dysfunction. However, these pathways are complex and require precise regulation to maximize treatment efficacy and minimize any potential clinical disability. Further investigations hold great promise to treat both the onset and progression of cognitive loss that is associated with metabolic disease.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022
| |
Collapse
|
34
|
Maiese K. Nicotinamide: Oversight of Metabolic Dysfunction Through SIRT1, mTOR, and Clock Genes. Curr Neurovasc Res 2020; 17:765-783. [PMID: 33183203 PMCID: PMC7914159 DOI: 10.2174/1567202617999201111195232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
Metabolic disorders that include diabetes mellitus present significant challenges for maintaining the welfare of the global population. Metabolic diseases impact all systems of the body and despite current therapies that offer some protection through tight serum glucose control, ultimately such treatments cannot block the progression of disability and death realized with metabolic disorders. As a result, novel therapeutic avenues are critical for further development to address these concerns. An innovative strategy involves the vitamin nicotinamide and the pathways associated with the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), the mechanistic target of rapamycin (mTOR), mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), AMP activated protein kinase (AMPK), and clock genes. Nicotinamide maintains an intimate relationship with these pathways to oversee metabolic disease and improve glucose utilization, limit mitochondrial dysfunction, block oxidative stress, potentially function as antiviral therapy, and foster cellular survival through mechanisms involving autophagy. However, the pathways of nicotinamide, SIRT1, mTOR, AMPK, and clock genes are complex and involve feedback pathways as well as trophic factors such as erythropoietin that require a careful balance to ensure metabolic homeostasis. Future work is warranted to gain additional insight into these vital pathways that can oversee both normal metabolic physiology and metabolic disease.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022
| |
Collapse
|
35
|
Dmytriyeva O, Belmeguenai A, Bezin L, Soud K, Drucker Woldbye DP, Gøtzsche CR, Pankratova S. Short erythropoietin-derived peptide enhances memory, improves long-term potentiation, and counteracts amyloid beta–induced pathology. Neurobiol Aging 2019; 81:88-101. [DOI: 10.1016/j.neurobiolaging.2019.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 03/27/2019] [Accepted: 05/06/2019] [Indexed: 12/23/2022]
|
36
|
Zhao R, He XW, Shi YH, Liu YS, Liu FD, Hu Y, Zhuang MT, Feng XY, Zhao L, Zhao BQ, Liu HQ, Shi GP, Liu JR. Cathepsin K Knockout Exacerbates Haemorrhagic Transformation Induced by Recombinant Tissue Plasminogen Activator After Focal Cerebral Ischaemia in Mice. Cell Mol Neurobiol 2019; 39:823-831. [PMID: 31065924 DOI: 10.1007/s10571-019-00682-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/29/2019] [Indexed: 12/19/2022]
Abstract
Severe haemorrhagic transformation (HT), a common complication of recombinant tissue plasminogen activator (rtPA) treatment, predicts poor clinical outcomes in acute ischaemic stroke. The search for agents to mitigate this effect includes investigating biomolecules involved in neovascularization. This study examines the role of Cathepsin K (Ctsk) in rtPA-induced HT after focal cerebral ischaemia in mice. After knockout of Ctsk, the gene encoding Ctsk, the outcomes of Ctsk+/+ and Ctsk-/- mice were compared 24 h after rtPA-treated cerebral ischaemia with respect to HT severity, neurological deficits, brain oedema, infarct volume, number of apoptotic neurons and activated microglia/macrophage, blood-brain barrier integrity, vascular endothelial growth factor (VEGF) expression and Akt-mTOR pathway activation. We observed that haemoglobin levels, brain oedema and infarct volume were significantly greater and resulted in more severe neurological deficits in Ctsk-/- than in Ctsk+/+ mice. Consistent with our hypothesis, the number of NeuN-positive neurons was lower and the number of TUNEL-positive apoptotic neurons and activated microglia/macrophage was higher in Ctsk-/- than in Ctsk+/+ mice. Ctsk knockout mice exhibited more severe blood-brain barrier (BBB) disruption, with microvascular endothelial cells exhibiting greater VEGF expression and lower ratios of phospo-Akt/Akt and phospo-mTOR/mTOR than in Ctsk+/+ mice. This study is the first to provide molecular insights into Ctsk-regulated HT after cerebral ischaemia, suggesting that Ctsk deficiency may disrupt the BBB via Akt/mTOR/VEGF signalling, resulting in neurological deficits and neuron apoptosis. Ctsk administration has the potential as a novel modality for improving the safety of rtPA treatment following stroke.
Collapse
Affiliation(s)
- Rong Zhao
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Xin-Wei He
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Yan-Hui Shi
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Yi-Sheng Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Feng-Di Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Yue Hu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Mei-Ting Zhuang
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Xiao-Yan Feng
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Lei Zhao
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China
| | - Bing-Qiao Zhao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Collaborative Innovation Center for Brain Science and School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Hui-Qin Liu
- Department of Neurology, Henan Provincial People's Hospital, Henan University, Zhengzhou, People's Republic of China
| | - Guo-Ping Shi
- Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, NRB-7, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Jian-Ren Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Huangpu District, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
37
|
Guo HT, Yang SD, Zhang F, Liu S, Yang DL, Ma L, Wang H, Ding WY. 17β‑Estradiol protects against interleukin‑1β‑induced apoptosis in rat nucleus pulposus cells via the mTOR/caspase‑3 pathway. Mol Med Rep 2019; 20:1523-1530. [PMID: 31257459 PMCID: PMC6625415 DOI: 10.3892/mmr.2019.10384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 04/04/2019] [Indexed: 12/31/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is the main pathological basis of spinal degenerative diseases, and aberrant apoptosis of nucleus pulposus cells (NPCs) is the main cellular process that causes IVDD. In our previous studies, 17β-estradiol (E2) was demonstrated to protect rat NPCs from interleukin-1β (IL-1β)-induced apoptosis via the PI3K/Akt signaling pathway. However, the downstream signaling pathway of PI3K/Akt is currently unclear. The present study aimed to explore the signaling pathways that are downstream of the PI3K/Akt pathway, including mTOR, NF-κB and glycogen synthase kinase-3β (GSK-3β). Annexin V/propidium iodide double staining was used to determine the incidence of apoptosis. Cell Counting kit-8 and MTS assays were used to determine the proliferation and viability of NPCs, respectively. Cellular binding was evaluated using a cell-collagen binding assay. Western blotting was used to determine the protein expression levels of mTOR, NF-κB and GSK-3β, and their phosphorylation levels, as well as the expression levels of active caspase-3. The results revealed that IL-1β induced NPC apoptosis and increased the early apoptotic rate of NPCs. However, E2 reduced the early apoptosis of NPCs induced by IL-1β. In addition, E2 suppressed the decrease in cell viability and binding ability caused by IL-1β cytotoxicity. Western blotting revealed that E2 also reduced the expression of activated caspase-3, and increased the expression of activated mTOR. As a specific inhibitor of mTOR, rapamycin effectively attenuated the effects of E2. These findings indicated that E2 protected NPCs against apoptosis via activation of the mTOR/caspase-3 pathway.
Collapse
Affiliation(s)
- Hong-Tao Guo
- Department of Spinal Surgery, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Si-Dong Yang
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Sen Liu
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Da-Long Yang
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Lei Ma
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Hui Wang
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Wen-Yuan Ding
- Department of Spinal Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
38
|
Sun J, Martin JM, Vanderpoel V, Sumbria RK. The Promises and Challenges of Erythropoietin for Treatment of Alzheimer's Disease. Neuromolecular Med 2019; 21:12-24. [PMID: 30656553 DOI: 10.1007/s12017-019-08524-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder in the world, and intracellular neurofibrillary tangles and extracellular amyloid-beta protein deposits represent the major pathological hallmarks of the disease. Currently available treatments provide some symptomatic relief but fail to modify primary pathological processes that underlie the disease. Erythropoietin (EPO), a hematopoietic growth factor, acts primarily to stimulate erythroid cell production, and is clinically used to treat anemia. EPO has evolved as a therapeutic agent for neurodegeneration and has improved neurological outcomes and AD pathology in rodents. However, penetration of the blood-brain barrier (BBB) and negative hematopoietic effects are the two major challenges for the therapeutic development of EPO for chronic neurodegenerative diseases like AD. The transferrin receptors at the BBB, which are responsible for transporting transferrin-bound iron from the blood into the brain parenchyma, can be used to shuttle therapeutic molecules across the BBB. In this review, we discuss the role of EPO as a potential neurotherapeutic for AD, challenges associated with EPO development for AD, and targeting the BBB transferrin receptor for EPO brain delivery.
Collapse
Affiliation(s)
- Jiahong Sun
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, 535 Watson Dr, Claremont, CA, 91711, USA
| | - Jan Michelle Martin
- College of Medicine, California Northstate University, Elk Grove, CA, 95757, USA
| | | | - Rachita K Sumbria
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, 535 Watson Dr, Claremont, CA, 91711, USA. .,Department of Neurology, University of California, Irvine, CA, 92868, USA.
| |
Collapse
|
39
|
L'Episcopo F, Tirolo C, Peruzzotti-Jametti L, Serapide MF, Testa N, Caniglia S, Balzarotti B, Pluchino S, Marchetti B. Neural Stem Cell Grafts Promote Astroglia-Driven Neurorestoration in the Aged Parkinsonian Brain via Wnt/β-Catenin Signaling. Stem Cells 2018; 36:1179-1197. [PMID: 29575325 DOI: 10.1002/stem.2827] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 02/19/2018] [Accepted: 03/06/2018] [Indexed: 12/16/2022]
Abstract
neuronal phenotype. Wnt/β-catenin signaling antagonism abolished mDA neurorestoration and immune modulatory effects of NSC grafts. Our work implicates an unprecedented therapeutic potential for somatic NSC grafts in the restoration of mDA neuronal function in the aged Parkinsonian brain. Stem Cells 2018;36:1179-1197.
Collapse
Affiliation(s)
| | | | - Luca Peruzzotti-Jametti
- Dept of Clinical Neurosciences, Clifford Allbutt Building - Cambridge Biosciences Campus and NIHR Biomedical Research,Centre, University of Cambridge, Hills Road, CB2 0HA Cambridge, UK
| | - Maria F Serapide
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology and Physiology Sections, University of Catania Medical School, Catania, Italy
| | | | | | - Beatrice Balzarotti
- Dept of Clinical Neurosciences, Clifford Allbutt Building - Cambridge Biosciences Campus and NIHR Biomedical Research,Centre, University of Cambridge, Hills Road, CB2 0HA Cambridge, UK
| | - Stefano Pluchino
- Dept of Clinical Neurosciences, Clifford Allbutt Building - Cambridge Biosciences Campus and NIHR Biomedical Research,Centre, University of Cambridge, Hills Road, CB2 0HA Cambridge, UK
| | - Bianca Marchetti
- Oasi Research Institute-IRCCS, Troina, Italy.,Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology and Physiology Sections, University of Catania Medical School, Catania, Italy
| |
Collapse
|
40
|
Castillo C, Zaror S, Gonzalez M, Hidalgo A, Burgos CF, Cabezas OI, Hugues F, Jiménez SP, González-Horta E, González-Chavarría I, Gavilán J, Montesino R, Sánchez O, Lopez MG, Fuentealba J, Toledo JR. Neuroprotective effect of a new variant of Epo nonhematopoietic against oxidative stress. Redox Biol 2018; 14:285-294. [PMID: 28987867 PMCID: PMC5975214 DOI: 10.1016/j.redox.2017.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 12/19/2022] Open
Abstract
Human erythropoietin is mainly recognized for its hematopoietic function; however, by binding to its receptor (EpoR), it can activate different signaling pathways as STAT, PI3K, MAPK and RAS to increase cellular differentiation or provide neuroprotective effects, among others. A recombinant human erythropoietin variant with low glycosylation and without hematopoietic effect (EpoL) was purified from skimmed goat milk. Recombinant human erythropoietin (Epo) was obtained from CHO cell line and used as control to compare EpoL effects. Neuroprotection studies were performed in PC12 cells and rat hippocampal slices. Cells were pretreated during 1h with EpoL or Epo and exposed to oxidative agents (H2O2 or FCCP); cell viability was assayed at the end of the experiment by the MTT method. Hippocampal slices were exposed to 15min of oxygen and glucose deprivation (OGD) and the neuroprotective drugs EpoL or Epo were incubated for 2h post-OGD in re-oxygenated medium. Cell cultures stressed with oxidative agents, and pretreated with EpoL, showed neuroprotective effects of 30% at a concentration 10 times lower than that of Epo. Moreover, similar differences were observed in OGD ex vivo assays. Neuroprotection elicited by EpoL was lost when an antibody against EpoR was present, indicating that its effect is EpoR-dependent. In conclusion, our results suggest that EpoL has a more potent neuroprotective profile than Epo against oxidative stress, mediated by activation of EpoR, thus EpoL represents an important target to develop a potential biopharmaceutical to treat different central nervous system pathologies related to oxidative stress such as stroke or neurodegenerative diseases.
Collapse
Affiliation(s)
- C Castillo
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - S Zaror
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - M Gonzalez
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - A Hidalgo
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - C F Burgos
- Laboratory of Screening of Neuroactive Compound, Physiology Department. School of Biological Sciences, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - O I Cabezas
- Clinical Sciences Department, School of Veterinary Sciences, Universidad de Concepción, Avenida Vicente Méndez 595, Chillan, Chile
| | - F Hugues
- Clinical Sciences Department, School of Veterinary Sciences, Universidad de Concepción, Avenida Vicente Méndez 595, Chillan, Chile
| | - S P Jiménez
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - E González-Horta
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - I González-Chavarría
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - J Gavilán
- Laboratory of Screening of Neuroactive Compound, Physiology Department. School of Biological Sciences, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - R Montesino
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - O Sánchez
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - Manuela G Lopez
- Department of Pharmacology and Therapeutics, "Instituo Teófilo Hernando", Universidad Autónoma de Madrid, Spain
| | - J Fuentealba
- Laboratory of Screening of Neuroactive Compound, Physiology Department. School of Biological Sciences, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - J R Toledo
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| |
Collapse
|
41
|
Maiese K. Warming Up to New Possibilities with the Capsaicin Receptor TRPV1: mTOR, AMPK, and Erythropoietin. Curr Neurovasc Res 2018; 14:184-189. [PMID: 28294062 DOI: 10.2174/1567202614666170313105337] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 02/26/2017] [Accepted: 03/03/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Transient receptor potential (TRP) channels are a superfamily of ion channels termed after the trp gene in Drosophila that are diverse in structure and control a wide range of biological functions including cell development and growth, thermal regulation, and vascular physiology. Of significant interest is the transient receptor potential cation channel subfamily V member 1 (TRPV1) receptor, also known as the capsaicin receptor and the vanilloid receptor 1, that is a non-selective cation channel sensitive to a host of external stimuli including capsaicin and camphor, venoms, acid/basic pH changes, and temperature. METHODS Given the multiple modalities that TRPV1 receptors impact in the body, we examined and discussed the role of these receptors in vasomotor control, metabolic disorders, cellular injury, oxidative stress, apoptosis, autophagy, and neurodegenerative disorders and their overlap with other signal transduction pathways that impact trophic factors. RESULTS Surprisingly, TRPV1 receptors do not rely entirely upon calcium signaling to affect cellular biology, but also have a close relationship with the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), and protein kinase B (Akt) that have roles in pain sensitivity, stem cell development, cellular survival, and cellular metabolism. These pathways with TRPV1 converge in the signaling of growth factors with recent work highlighting a relationship with erythropoietin (EPO). Angiogenesis and endothelial tube formation controlled by EPO requires, in part, the activation of TRPV1 receptors in conjunction with Akt and AMPK pathways. CONCLUSION TRPV1 receptors could prove to become vital to target disorders of vascular origin and neurodegeneration. Broader and currently unrealized implementations for both EPO and TRPV1 receptors can be envisioned for for the development of novel therapeutic strategies in multiple systems of the body.
Collapse
|
42
|
The mechanistic target of rapamycin (mTOR) and the silent mating-type information regulation 2 homolog 1 (SIRT1): oversight for neurodegenerative disorders. Biochem Soc Trans 2018. [PMID: 29523769 DOI: 10.1042/bst20170121] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a result of the advancing age of the global population and the progressive increase in lifespan, neurodegenerative disorders continue to increase in incidence throughout the world. New strategies for neurodegenerative disorders involve the novel pathways of the mechanistic target of rapamycin (mTOR) and the silent mating-type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) that can modulate pathways of apoptosis and autophagy. The pathways of mTOR and SIRT1 are closely integrated. mTOR forms the complexes mTOR Complex 1 and mTOR Complex 2 and can impact multiple neurodegenerative disorders that include Alzheimer's disease, Huntington's disease, and Parkinson's disease. SIRT1 can control stem cell proliferation, block neuronal injury through limiting programmed cell death, drive vascular cell survival, and control clinical disorders that include dementia and retinopathy. It is important to recognize that oversight of programmed cell death by mTOR and SIRT1 requires a fine degree of precision to prevent the progression of neurodegenerative disorders. Additional investigations and insights into these pathways should offer effective and safe treatments for neurodegenerative disorders.
Collapse
|
43
|
Maiese K. Novel Treatment Strategies for the Nervous System: Circadian Clock Genes, Non-coding RNAs, and Forkhead Transcription Factors. Curr Neurovasc Res 2018; 15:81-91. [PMID: 29557749 PMCID: PMC6021214 DOI: 10.2174/1567202615666180319151244] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/23/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND With the global increase in lifespan expectancy, neurodegenerative disorders continue to affect an ever-increasing number of individuals throughout the world. New treatment strategies for neurodegenerative diseases are desperately required given the lack of current treatment modalities. METHODS Here, we examine novel strategies for neurodegenerative disorders that include circadian clock genes, non-coding Ribonucleic Acids (RNAs), and the mammalian forkhead transcription factors of the O class (FoxOs). RESULTS Circadian clock genes, non-coding RNAs, and FoxOs offer exciting prospects to potentially limit or remove the significant disability and death associated with neurodegenerative disorders. Each of these pathways has an intimate relationship with the programmed death pathways of autophagy and apoptosis and share a common link to the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) and the mechanistic target of rapamycin (mTOR). Circadian clock genes are necessary to modulate autophagy, limit cognitive loss, and prevent neuronal injury. Non-coding RNAs can control neuronal stem cell development and neuronal differentiation and offer protection against vascular disease such as atherosclerosis. FoxOs provide exciting prospects to block neuronal apoptotic death and to activate pathways of autophagy to remove toxic accumulations in neurons that can lead to neurodegenerative disorders. CONCLUSION Continued work with circadian clock genes, non-coding RNAs, and FoxOs can offer new prospects and hope for the development of vital strategies for the treatment of neurodegenerative diseases. These innovative investigative avenues have the potential to significantly limit disability and death from these devastating disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
44
|
Wang SP, Wang LH. Disease implication of hyper-Hippo signalling. Open Biol 2017; 6:rsob.160119. [PMID: 27805903 PMCID: PMC5090056 DOI: 10.1098/rsob.160119] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022] Open
Abstract
The Hippo signalling pathway regulates cellular proliferation, apoptosis and differentiation, thus exerting profound effects on cellular homeostasis. Inhibition of Hippo signalling has been frequently implicated in human cancers, indicating a well-known tumour suppressor function of the Hippo pathway. However, it is less certain whether and how hyperactivation of the Hippo pathway affects biological outcome in living cells. This review describes current knowledge of the regulatory mechanisms of the Hippo pathway, mainly focusing on hyperactivation of the Hippo signalling nexus. The disease implications of hyperactivated Hippo signalling have also been discussed, including arrhythmogenic cardiomyopathy, Sveinsson's chorioretinal atrophy, Alzheimer's disease, amyotrophic lateral sclerosis and diabetes. By highlighting the significance of disease-relevant Hippo signalling activation, this review can offer exciting prospects to address the onset and potential reversal of Hippo-related disorders.
Collapse
Affiliation(s)
- Shu-Ping Wang
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - Lan-Hsin Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 114, Taiwan
| |
Collapse
|
45
|
Maiese K. Erythropoietin and mTOR: A "One-Two Punch" for Aging-Related Disorders Accompanied by Enhanced Life Expectancy. Curr Neurovasc Res 2017; 13:329-340. [PMID: 27488211 DOI: 10.2174/1567202613666160729164900] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/16/2022]
Abstract
Life expectancy continues to increase throughout the world, but is accompanied by a rise in the incidence of non-communicable diseases. As a result, the benefits of an increased lifespan can be limited by aging-related disorders that necessitate new directives for the development of effective and safe treatment modalities. With this objective, the mechanistic target of rapamycin (mTOR), a 289-kDa serine/threonine protein, and its related pathways of mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), proline rich Akt substrate 40 kDa (PRAS40), AMP activated protein kinase (AMPK), Wnt signaling, and silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), have generated significant excitement for furthering novel therapies applicable to multiple systems of the body. Yet, the biological and clinical outcome of these pathways can be complex especially with oversight of cell death mechanisms that involve apoptosis and autophagy. Growth factors, and in particular erythropoietin (EPO), are one avenue under consideration to implement control over cell death pathways since EPO can offer potential treatment for multiple disease entities and is intimately dependent upon mTOR signaling. In experimental and clinical studies, EPO appears to have significant efficacy in treating several disorders including those involving the developing brain. However, in mature populations that are affected by aging-related disorders, the direction for the use of EPO to treat clinical disease is less clear that may be dependent upon a number of factors including the understanding of mTOR signaling. Continued focus upon the regulatory elements that control EPO and mTOR signaling could generate critical insights for targeting a broad range of clinical maladies.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101, USA.
| |
Collapse
|
46
|
Na EJ, Nam HY, Park J, Chung MA, Woo HA, Kim HJ. PI3K-mTOR-S6K Signaling Mediates Neuronal Viability via Collapsin Response Mediator Protein-2 Expression. Front Mol Neurosci 2017; 10:288. [PMID: 28966575 PMCID: PMC5605571 DOI: 10.3389/fnmol.2017.00288] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/25/2017] [Indexed: 01/22/2023] Open
Abstract
Collapsin response mediator protein (CRMP)-2 and the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway are associated with common physiological functions such as neuronal polarity, axonal outgrowth and synaptic strength, as well as various brain disorders including epilepsy. But, their regulatory and functional links are unclear. Alterations in CRMP-2 expression that lead to its functional changes are implicated in brain disorders such as epilepsy. Here, we investigate whether changes in CRMP-2 expression, possibly regulated by mTOR-related signaling, correlates with neuronal growth and viability. Inhibition of mTOR and/or phosphoinositol-3-kinase (PI3K) led to deceased p-S6K, and p-S6 signals also reduced CRMP-2 expression. These changes corresponded to inhibition of neuronal viability and proliferation in cultured hippocampal HT-22 cells under both basal serum-free and serum- or insulin-induced mTOR pathway-activated conditions. CRMP-2 expression tended to be increased by mTOR activation, indicated by an increase in p-S6/S6 level, in pentylentetrazole (PTZ)-induced epileptic rat hippocampal tissues was also significantly reduced by mTOR inhibition. Knockdown of CRMP-2 by si-RNA reduced the neuronal viability without changes in mTOR signaling, and overexpression of CRMP-2 recovered the glutamate-induced neurotoxicity and decrease of mTOR signaling in HT-22 cells. In conclusion, CRMP-2 protein expression controlled by the PI3K-mTOR-S6K signaling axis exerts its important functional roles in neuronal growth and survival.
Collapse
Affiliation(s)
- Eun J Na
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Hye Yeon Nam
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Jiyoung Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Myung Ah Chung
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Hyun Ae Woo
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| | - Hwa-Jung Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Ewha Womans UniversitySeoul, South Korea
| |
Collapse
|
47
|
Hernández CC, Burgos CF, Gajardo AH, Silva-Grecchi T, Gavilan J, Toledo JR, Fuentealba J. Neuroprotective effects of erythropoietin on neurodegenerative and ischemic brain diseases: the role of erythropoietin receptor. Neural Regen Res 2017; 12:1381-1389. [PMID: 29089974 PMCID: PMC5649449 DOI: 10.4103/1673-5374.215240] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2017] [Indexed: 12/11/2022] Open
Abstract
Erythropoietin (Epo) is a fundamental hormone in the regulation of hematopoiesis, and other secondary roles mediated by the binding of the hormone to its specific receptor (EpoR), which leads to an activation of key signaling pathways that induce an increase in cell differentiation, apoptosis control and neuroprotection. It has been suggested that their function depends on final conformation of glycosylations, related with affinity to the receptor and its half-life. The presence of EpoR has been reported in different tissues including central nervous system, where it has been demonstrated to exert a neuroprotective function against oxidative stress conditions, such as ischemic injury and neurodegenerative diseases. There is also evidence of an increase in EpoR expression in brain cell lysates of Alzheimer's patients with respect to healthy patients. These results are related with extensive in vitro experimental data of neuroprotection obtained from cell lines, primary cell cultures and hippocampal slices. Additionally, this data is correlated with in vivo experiments (water maze test) in mouse models of Alzheimer's disease where Epo treatment improved cognitive function. These studies support the idea that receptor activation induces a neuroprotective effect in neurodegenerative disorders including dementias, and especially Alzheimer's disease. Taken together, available evidence suggests that Epo appears to be a central element for EpoR activation and neuroprotective properties in the central nervous system. In this review, we will describe the mechanisms associated with neuroprotection and its relation with the activation of EpoR in order with identify new targets to develop pharmacological strategies.
Collapse
Affiliation(s)
- Carolina Castillo Hernández
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
- Laboratory of Biotechnology and Biopharmaceutical, Department of Pathophysiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Carlos Felipe Burgos
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Angela Hidalgo Gajardo
- Laboratory of Biotechnology and Biopharmaceutical, Department of Pathophysiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Tiare Silva-Grecchi
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Javiera Gavilan
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Jorge Roberto Toledo
- Laboratory of Biotechnology and Biopharmaceutical, Department of Pathophysiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| |
Collapse
|
48
|
Joeng KS, Lee YC, Lim J, Chen Y, Jiang MM, Munivez E, Ambrose C, Lee BH. Osteocyte-specific WNT1 regulates osteoblast function during bone homeostasis. J Clin Invest 2017. [PMID: 28628032 DOI: 10.1172/jci92617] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mutations in WNT1 cause osteogenesis imperfecta (OI) and early-onset osteoporosis, identifying it as a key Wnt ligand in human bone homeostasis. However, how and where WNT1 acts in bone are unclear. To address this mechanism, we generated late-osteoblast-specific and osteocyte-specific WNT1 loss- and gain-of-function mouse models. Deletion of Wnt1 in osteocytes resulted in low bone mass with spontaneous fractures similar to that observed in OI patients. Conversely, Wnt1 overexpression from osteocytes stimulated bone formation by increasing osteoblast number and activity, which was due in part to activation of mTORC1 signaling. While antiresorptive therapy is the mainstay of OI treatment, it has limited efficacy in WNT1-related OI. In this study, anti-sclerostin antibody (Scl-Ab) treatment effectively improved bone mass and dramatically decreased fracture rate in swaying mice, a model of global Wnt1 loss. Collectively, our data suggest that WNT1-related OI and osteoporosis are caused in part by decreased mTORC1-dependent osteoblast function resulting from loss of WNT1 signaling in osteocytes. As such, this work identifies an anabolic function of osteocytes as a source of Wnt in bone development and homoeostasis, complementing their known function as targets of Wnt signaling in regulating osteoclastogenesis. Finally, this study suggests that Scl-Ab is an effective genotype-specific treatment option for WNT1-related OI and osteoporosis.
Collapse
Affiliation(s)
- Kyu Sang Joeng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Yi-Chien Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Joohyun Lim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Elda Munivez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Catherine Ambrose
- Department of Orthopedic Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
49
|
Pathipati P, Ferriero DM. The Differential Effects of Erythropoietin Exposure to Oxidative Stress on Microglia and Astrocytes in vitro. Dev Neurosci 2017; 39:310-322. [PMID: 28511187 DOI: 10.1159/000467391] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/28/2017] [Indexed: 12/15/2022] Open
Abstract
The neonatal brain is especially susceptible to oxidative stress owing to its reduced antioxidant capacity. Following hypoxic-ischemic (HI) injury, for example, there is a prolonged elevation in levels of hydrogen peroxide (H2O2) in the immature brain compared to the adult brain, resulting in lasting injury that can lead to life-long disability or morbidity. Erythropoietin (Epo) is one of few multifaceted treatment options that have been promising enough to trial in the clinic for both term and preterm brain injury. Despite this, there is a lack of clear understanding of how Epo modulates glial cell activity following oxidative injury, specifically, whether it affects microglia (Mg) and astrocytes (Ast) differently. Using an in vitro approach using primary murine Mg and Ast subjected to H2O2 injury, we studied the oxidative and inflammatory responses of Mg and Ast to recombinant murine (rm)Epo treatment. We found that Epo protects Ast from H2O2 injury (p < 0.05) and increases secreted nitric oxide levels in these cells (p < 0.05) while suppressing intracellular reactive oxygen species (p < 0.05) and superoxide ion (p < 0.05) levels only in Mg. Using a multiplex analysis, we noted that although H2O2 induced the levels of several chemokines, rmEpo did not have any significant specific effects on their levels, either with or without the presence of conditioned medium from injured neurons (NCM). Ultimately, it appears that rmEpo has pleiotropic effects based on the cell type; it has a protective effect on Ast but an antioxidative effect only on Mg without any significant modulation of chemokine and cytokine levels in either cell type. These findings highlight the importance of considering all cell types when assessing the benefits and pitfalls of Epo use.
Collapse
Affiliation(s)
- Praneeti Pathipati
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
50
|
Rosa MD, Distefano G, Gagliano C, Rusciano D, Malaguarnera L. Autophagy in Diabetic Retinopathy. Curr Neuropharmacol 2017; 14:810-825. [PMID: 26997506 PMCID: PMC5333581 DOI: 10.2174/1570159x14666160321122900] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/08/2015] [Accepted: 11/10/2015] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an important homeostatic cellular process encompassing a number of consecutive steps indispensable for degrading and recycling cytoplasmic materials. Basically autophagy is an adaptive response that under stressful conditions guarantees the physiological turnover of senescent and impaired organelles and, thus, controls cell fate by various cross-talk signals. Diabetic retinopathy (DR) is a serious microvascular complication of diabetes and accounts for 5% of all blindness. Although, various metabolic disorders have been linked with the onset of DR, due to the complex character of this multi-factorial disease, a connection between any particular defect and DR becomes speculative. Diabetes increases inflammation, advanced glycation end products (AGEs) and oxidative stress in the retina and its capillary cells. Particularly, a great number of evidences suggest a mutual connection between oxidative stress and other major metabolic abnormalities implicated in the development of DR. In addition, the intricate networks between autophagy and apoptosis establish the degree of cellular apoptosis and the progression of DR. Growing data underline the crucial role of reactive oxygen species (ROS) in the activation of autophagy. Depending on their delicate balance both redox signaling and autophagy, being detrimental or beneficial, retain opposing effects. The molecular mechanisms of autophagy are very complex and involve many signaling pathways cooperating at various steps. This review summarizes recent advances of the possible molecular mechanisms in autophagic process that are involved in pathophysiology of DR. In-depth analysis on the molecular mechanisms leading to autophagy in the retinal pigment epithelial (RPE) will be helpful to plan new therapies aimed at preventing or improving the progression of DR.
Collapse
Affiliation(s)
| | | | | | | | - Lucia Malaguarnera
- Department of Biomedical and Biotechnological Sciences, Faculty of Medicine, University of Catania, 95124 Catania, Italy
| |
Collapse
|