1
|
Chatterjee S, Verma A, Thakkar H, Shah RP, Khairnar A. Glycated α-Synuclein Renders Glial Cell Activation and Induces Degeneration of Dopaminergic Neurons: A Potential Implication for the Development of Parkinson's Disease. ACS Chem Biol 2025. [PMID: 39982250 DOI: 10.1021/acschembio.4c00777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Accumulation of misfolded α-synuclein (α-Syn) leads to the formation of Lewy bodies and is a major hallmark of Parkinson's disease (PD). The accumulation of α-Syn involves several post-translational modifications. Recently, though, glycation of α-Syn (advanced glycation end products) and activation of the receptor for advanced glycation end products (RAGE) have been linked to neuroinflammation, which leads to oxidative stress and accumulation of α-Syn. The present study aims to detect the effect of glycated α-Syn (gly-α-Syn)-induced synucleinopathy and loss of dopaminergic (DAergic) neurons in the development of PD. We isolated, purified, and prepared glycated recombinant human α-Syn using d-ribose. Gly-α-Syn was characterized by SDS-PAGE, intact mass analysis, and bottom-up peptide sequence through LC-HRMS/MS. The aggregation propensity of gly-α-Syn has been verified by morphological and shape analysis through Bio-AFM. The gly-α-Syn (2 μg/μL) was injected stereotaxically in the substantia nigra (SN) of ICR mice (3-4 months) and compared with the normal α-Syn, d ribose, and Tris-HCl/artificial CSF groups. 56 days postsurgery (DPS), an immunohistochemical examination was conducted to investigate gly-α-Syn-induced α-Syn accumulation, neuroinflammation, and neurodegeneration. The glycation of α-Syn led to the expression of transglutaminase 2 (TGM2), an enzyme that cross-linked with AGEs and may have caused the accumulation of α-Syn. Significant RAGE activation was also observed in gly-α-Syn, which might have induced glial cell activation, resulting in oxidative stress and, ultimately, apoptosis of dopaminergic neurons. It is important to note that TGM2, phosphorylated α-Syn, RAGE expression, and glial cell activation were only found in the gly-α-Syn group and not in the other groups. This suggests that gly-α-Syn plays a major role in synucleinopathy, neuroinflammation, and neurodegeneration. Overall, the present study demonstrated glycation of α-Syn as one of the important age-associated post-translational modifications that are involved in the degeneration of dopaminergic neurons, at least in a subset of the diabetic patients susceptible to developing PD.
Collapse
Affiliation(s)
- Sayan Chatterjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ahmedabad, Gujarat 382355, India
| | - Arvind Verma
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ahmedabad, Gujarat 382355, India
| | - Harsh Thakkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ahmedabad, Gujarat 382355, India
| | - Ravi P Shah
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ahmedabad, Gujarat 382355, India
| | - Amit Khairnar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ahmedabad, Gujarat 382355, India
- International Clinical Research Centre, St. Anne's University Hospital Brno, Brno 60200, Czech Republic
- ICRC, FNUSA, Brno 65691, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic
- International Clinical Research Centre, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic
| |
Collapse
|
2
|
Peixoto DO, Bittencourt RR, Gasparotto J, Kessler FGC, Brum PO, Somensi N, Girardi CS, Dos Santos da Silva L, Outeiro TF, Moreira JCF, Gelain DP. Increased alpha-synuclein and neuroinflammation in the substantia nigra triggered by systemic inflammation are reversed by targeted inhibition of the receptor for advanced glycation end products (RAGE). J Neurochem 2024; 168:1587-1607. [PMID: 37661637 DOI: 10.1111/jnc.15956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/18/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a protein of the immunoglobulin superfamily capable of regulating inflammation. Considering the role of this receptor in the initiation and establishment of neuroinflammation, and the limited understanding of the function of RAGE in the maintenance of this condition, this study describes the effects of RAGE inhibition in the brain, through an intranasal treatment with the antagonist FPS-ZM1, in an animal model of chronic neuroinflammation induced by acute intraperitoneal injection of lipopolysaccharide (LPS). Seventy days after LPS administration (2 mg/kg, i.p.), Wistar rats received, intranasally, 1.2 mg of FPS-ZM1 over 14 days. On days 88 and 89, the animals were submitted to the open-field test and were killed on day 90 after the intraperitoneal injection of LPS. Our results indicate that blockade of encephalic RAGE attenuates LPS-induced chronic neuroinflammation in different brain regions. Furthermore, we found that intranasal FPS-ZM1 administration reduced levels of gliosis markers, RAGE ligands, and α-synuclein in the substantia nigra pars compacta. Additionally, the treatment also reversed the increase in S100 calcium-binding protein B (RAGE ligand) in the cerebrospinal fluid and the cognitive-behavioral deficits promoted by LPS-less time spent in the central zone of the open-field arena (more time in the lateral zones), decreased total distance traveled, and increased number of freezing episodes. In summary, our study demonstrates the prominent role of RAGE in the maintenance of a chronic neuroinflammatory state triggered by a single episode of systemic inflammation and also points to possible future RAGE-based therapeutic approaches to treat conditions in which chronic neuroinflammation and increased α-synuclein levels could play a relevant role, such as in Parkinson's disease.
Collapse
Affiliation(s)
- Daniel Oppermann Peixoto
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Alicante, Spain
| | - Reykla Ramon Bittencourt
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Juciano Gasparotto
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (ICB-UNIFAL), Alfenas, Brazil
| | - Flávio Gabriel Carazza Kessler
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | | | - Nauana Somensi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Carolina Saibro Girardi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Lucas Dos Santos da Silva
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Natural Sciences, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| |
Collapse
|
3
|
Gasparotto J, Somensi N, Girardi CS, Bittencourt RR, de Oliveira LM, Hoefel LP, Scheibel IM, Peixoto DO, Moreira JCF, Outeiro TF, Gelain DP. Is it all the RAGE? Defining the role of the receptor for advanced glycation end products in Parkinson's disease. J Neurochem 2024; 168:1608-1624. [PMID: 37381043 DOI: 10.1111/jnc.15890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/09/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a transmembrane receptor that belongs to the immunoglobulin superfamily and is extensively associated with chronic inflammation in non-transmissible diseases. As chronic inflammation is consistently present in neurodegenerative diseases, it was largely assumed that RAGE could act as a critical modulator of neuroinflammation in Parkinson's disease (PD), similar to what was reported for Alzheimer's disease (AD), where RAGE is postulated to mediate pro-inflammatory signaling in microglia by binding to amyloid-β peptide. However, accumulating evidence from studies of RAGE in PD models suggests a less obvious scenario. Here, we review physiological aspects of RAGE and address the current questions about the potential involvement of this receptor in the cellular events that may be critical for the development and progression of PD, exploring possible mechanisms beyond the classical view of the microglial activation/neuroinflammation/neurodegeneration axis that is widely assumed to be the general mechanism of RAGE action in the adult brain.
Collapse
Affiliation(s)
- Juciano Gasparotto
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, MG, Brazil
| | - Nauana Somensi
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carolina Saibro Girardi
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Reykla Ramon Bittencourt
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Laura Martinewski de Oliveira
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Laura Piloneto Hoefel
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ingrid Matsubara Scheibel
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniel Oppermann Peixoto
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - José Claudio Fonseca Moreira
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Daniel Pens Gelain
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
4
|
Jarosławska J, Kordas B, Miłowski T, Juranek JK. Mammalian Diaphanous1 signalling in neurovascular complications of diabetes. Eur J Neurosci 2024; 59:2628-2645. [PMID: 38491850 DOI: 10.1111/ejn.16310] [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: 12/20/2023] [Accepted: 02/18/2024] [Indexed: 03/18/2024]
Abstract
Over the past few decades, diabetes gradually has become one of the top non-communicable disorders, affecting 476.0 million in 2017 and is predicted to reach 570.9 million people in 2025. It is estimated that 70 to 100% of all diabetic patients will develop some if not all, diabetic complications over the course of the disease. Despite different symptoms, mechanisms underlying the development of diabetic complications are similar, likely stemming from deficits in both neuronal and vascular components supplying hyperglycaemia-susceptible tissues and organs. Diaph1, protein diaphanous homolog 1, although mainly known for its regulatory role in structural modification of actin and related cytoskeleton proteins, in recent years attracted research attention as a cytoplasmic partner of the receptor of advanced glycation end-products (RAGE) a signal transduction receptor, whose activation triggers an increase in proinflammatory molecules, oxidative stressors and cytokines in diabetes and its related complications. Both Diaph1 and RAGE are also a part of the RhoA signalling cascade, playing a significant role in the development of neurovascular disturbances underlying diabetes-related complications. In this review, based on the existing knowledge as well as compelling findings from our past and present studies, we address the role of Diaph1 signalling in metabolic stress and neurovascular degeneration in diabetic complications. In light of the most recent developments in biochemical, genomic and transcriptomic research, we describe current theories on the aetiology of diabetes complications, highlighting the function of the Diaph1 signalling system and its role in diabetes pathophysiology.
Collapse
Affiliation(s)
- Julia Jarosławska
- Department of Biological Functions of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Bernard Kordas
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Tadeusz Miłowski
- Department of Emergency Medicine, School of Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Judyta K Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
5
|
Deepu V, Rai V, Agrawal DK. Quantitative Assessment of Intracellular Effectors and Cellular Response in RAGE Activation. ARCHIVES OF INTERNAL MEDICINE RESEARCH 2024; 7:80-103. [PMID: 38784044 PMCID: PMC11113086 DOI: 10.26502/aimr.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
The review delves into the methods for the quantitative assessment of intracellular effectors and cellular response of Receptor for Advanced Glycation End products (RAGE), a vital transmembrane receptor involved in a range of physiological and pathological processes. RAGE bind to Advanced Glycation End products (AGEs) and other ligands, which in turn activate diverse downstream signaling pathways that impact cellular responses such as inflammation, oxidative stress, and immune reactions. The review article discusses the intracellular signaling pathways activated by RAGE followed by differential activation of RAGE signaling across various diseases. This will ultimately guide researchers in developing targeted and effective interventions for diseases associated with RAGE activation. Further, we have discussed how PCR, western blotting, and microscopic examination of various molecules involved in downstream signaling can be leveraged to monitor, diagnose, and explore diseases involving proteins with unique post-translational modifications. This review article underscores the pressing need for advancements in molecular approaches for disease detection and management involving RAGE.
Collapse
Affiliation(s)
- Vinitha Deepu
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| |
Collapse
|
6
|
Wong W, Sari Y. Effects of Hydrocodone Overdose and Ceftriaxone on Astrocytic Glutamate Transporters and Glutamate Receptors, and Associated Signaling in Nucleus Accumbens as well as Locomotor Activity in C57/BL Mice. Brain Sci 2024; 14:361. [PMID: 38672013 PMCID: PMC11048659 DOI: 10.3390/brainsci14040361] [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: 02/28/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic opioid treatments dysregulate the glutamatergic system, inducing a hyperglutamatergic state in mesocorticolimbic brain regions. This study investigated the effects of exposure to hydrocodone overdose on locomotor activity, expression of target proteins related to the glutamatergic system, signaling kinases, and neuroinflammatory factors in the nucleus accumbens. The locomotor activity of mice was measured using the Comprehensive Laboratory Animal Monitoring System (CLAMS). CLAMS data showed that exposure to hydrocodone overdose increased locomotion activity in mice. This study tested ceftriaxone, known to upregulate major glutamate transporter 1 (GLT-1), in mice exposed to an overdose of hydrocodone. Thus, ceftriaxone normalized hydrocodone-induced hyperlocomotion activity in mice. Furthermore, exposure to hydrocodone overdose downregulated GLT-1, cystine/glutamate antiporter (xCT), and extracellular signal-regulated kinase activity (p-ERK/ERK) expression in the nucleus accumbens. However, exposure to an overdose of hydrocodone increased metabotropic glutamate receptor 5 (mGluR5), neuronal nitric oxide synthase activity (p-nNOS/nNOS), and receptor for advanced glycation end products (RAGE) expression in the nucleus accumbens. Importantly, ceftriaxone treatment attenuated hydrocodone-induced upregulation of mGluR5, p-nNOS/nNOS, and RAGE, as well as hydrocodone-induced downregulation of GLT-1, xCT, and p-ERK/ERK expression. These data demonstrated that exposure to hydrocodone overdose can cause dysregulation of the glutamatergic system, neuroinflammation, hyperlocomotion activity, and the potential therapeutic role of ceftriaxone in attenuating these effects.
Collapse
Affiliation(s)
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA;
| |
Collapse
|
7
|
Nunes MJ, Carvalho AN, Rosa AI, Videira PA, Gama MJ, Rodrigues E, Castro-Caldas M. Altered expression of Sialyl Lewis X in experimental models of Parkinson's disease. J Mol Med (Berl) 2024; 102:365-377. [PMID: 38197965 PMCID: PMC10879467 DOI: 10.1007/s00109-023-02415-3] [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: 05/16/2023] [Revised: 12/07/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
The mechanisms underlying neurodegeneration in Parkinson's disease (PD) are still not fully understood. Glycosylation is an important post-translational modification that affects protein function, cell-cell contacts and inflammation and can be modified in pathologic conditions. Although the involvement of aberrant glycosylation has been proposed for PD, the knowledge of the diversity of glycans and their role in PD is still minimal. Sialyl Lewis X (sLeX) is a sialylated and fucosylated tetrasaccharide with essential roles in cell-to-cell recognition processes. Pathological conditions and pro-inflammatory mediators can up-regulate sLeX expression on cell surfaces, which has important consequences in intracellular signalling and immune function. Here, we investigated the expression of this glycan using in vivo and in vitro models of PD. We show the activation of deleterious glycation-related pathways in mouse striatum upon treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a toxin-based model of PD. Importantly, our results show that MPTP triggers the presentation of more proteins decorated with sLeX in mouse cortex and striatum in a time-dependent manner, as well as increased mRNA expression of its rate-limiting enzyme fucosyltransferase 7. sLeX is expressed in neurons, including dopaminergic neurons, and microglia. Although the underlying mechanism that drives increased sLeX epitopes, the nature of the protein scaffolds and their functional importance in PD remain unknown, our data suggest for the first time that sLeX in the brain may have a role in neuronal signalling and immunomodulation in pathological conditions. KEY MESSAGES: MPTP triggers the presentation of proteins decorated with sLeX in mouse brain. MPTP triggers the expression of sLeX rate-limiting enzyme FUT 7 in striatum. sLeX is expressed in neurons, including dopaminergic neurons, and microglia. sLeX in the brain may have a role in neuronal signalling and immunomodulation.
Collapse
Affiliation(s)
- Maria João Nunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Andreia Neves Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Alexandra I Rosa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Paula A Videira
- Department of Life Sciences, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| | - Maria João Gama
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Elsa Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Margarida Castro-Caldas
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
- Department of Life Sciences, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| |
Collapse
|
8
|
Wal P, Wal A, Vig H, Mahmood D, Khan MMU. Potential Applications of Mitochondrial Therapy with a Focus on Parkinson's Disease and Mitochondrial Transplantation. Adv Pharm Bull 2024; 14:147-160. [PMID: 38585467 PMCID: PMC10997929 DOI: 10.34172/apb.2024.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/28/2023] [Accepted: 10/08/2023] [Indexed: 04/09/2024] Open
Abstract
Purpose Both aging and neurodegenerative illnesses are thought to be influenced by mitochondrial malfunction and free radical formation. Deformities of the energy metabolism, mitochondrial genome polymorphisms, nuclear DNA genetic abnormalities associated with mitochondria, modifications of mitochondrial fusion or fission, variations in shape and size, variations in transit, modified mobility of mitochondria, transcription defects, and the emergence of misfolded proteins associated with mitochondria are all linked to Parkinson's disease. Methods This review is a condensed compilation of data from research that has been published between the years of 2014 and 2022, using search engines like Google Scholar, PubMed, and Scopus. Results Mitochondrial transplantation is a one-of-a-kind treatment for mitochondrial diseases and deficits in mitochondrial biogenesis. The replacement of malfunctioning mitochondria with transplanted viable mitochondria using innovative methodologies has shown promising outcomes as a cure for Parkinson's, involving tissue sparing coupled with enhanced energy generation and lower oxidative damage. Numerous mitochondria-targeted therapies, including mitochondrial gene therapy, redox therapy, and others, have been investigated for their effectiveness and potency. Conclusion The development of innovative therapeutics for mitochondria-directed treatments in Parkinson's disease may be aided by optimizing mitochondrial dynamics. Many neurological diseases have been studied in animal and cellular models, and it has been found that mitochondrial maintenance can slow the death of neuronal cells. It has been hypothesized that drug therapies for neurodegenerative diseases that focus on mitochondrial dysfunction will help to delay the onset of neuronal dysfunction.
Collapse
Affiliation(s)
- Pranay Wal
- Pharmacy Department, PSIT- Pranveer Singh Institute of Technology, (PHARMACY) Kanpur-Agra-Delhi National Highway (NH-2), Bhauti-Kanpur-209305
| | - Ankita Wal
- Pharmacy Department, PSIT- Pranveer Singh Institute of Technology, (PHARMACY) Kanpur-Agra-Delhi National Highway (NH-2), Bhauti-Kanpur-209305
| | - Himangi Vig
- Pharmacy Department, PSIT- Pranveer Singh Institute of Technology, (PHARMACY) Kanpur-Agra-Delhi National Highway (NH-2), Bhauti-Kanpur-209305
| | - Danish Mahmood
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Unaizah 51911, Saudi Arabia
| | - Mohd Masih Uzzaman Khan
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Unaizah 51911, Saudi Arabia
| |
Collapse
|
9
|
Xue J, Tao K, Wang W, Wang X. What Can Inflammation Tell Us about Therapeutic Strategies for Parkinson's Disease? Int J Mol Sci 2024; 25:1641. [PMID: 38338925 PMCID: PMC10855787 DOI: 10.3390/ijms25031641] [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: 01/01/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder with a complicated etiology and pathogenesis. α-Synuclein aggregation, dopaminergic (DA) neuron loss, mitochondrial injury, oxidative stress, and inflammation are involved in the process of PD. Neuroinflammation has been recognized as a key element in the initiation and progression of PD. In this review, we summarize the inflammatory response and pathogenic mechanisms of PD. Additionally, we describe the potential anti-inflammatory therapies, including nod-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome inhibition, nuclear factor κB (NF-κB) inhibition, microglia inhibition, astrocyte inhibition, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibition, the peroxisome proliferator-activated receptor γ (PPARγ) agonist, targeting the mitogen-activated protein kinase (MAPK) pathway, targeting the adenosine monophosphate-activated protein kinase (AMPK)-dependent pathway, targeting α-synuclein, targeting miRNA, acupuncture, and exercise. The review focuses on inflammation and will help in designing new prevention strategies for PD.
Collapse
Affiliation(s)
- Jinsong Xue
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| | | | | | - Xiaofei Wang
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| |
Collapse
|
10
|
Chegão A, Vicente Miranda H. Unveiling new secrets in Parkinson's disease: The glycatome. Behav Brain Res 2023; 442:114309. [PMID: 36706808 DOI: 10.1016/j.bbr.2023.114309] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/04/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
We are witnessing a considerable increase in the incidence of Parkinson's disease (PD), which may be due to the general ageing of the population. While there is a plethora of therapeutic strategies for this disease, they still fail to arrest disease progression as they do not target and prevent the neurodegenerative process. The identification of disease-causing mutations allowed researchers to better dissect the underlying causes of this disease, highlighting, for example, the pathogenic role of alpha-synuclein. However, most PD cases are sporadic, which is making it hard to unveil the major causative mechanisms of this disease. In the recent years, epidemiological evidence suggest that type-2 diabetes mellitus (T2DM) individuals have higher risk and worst outcomes of PD, allowing to raise the hypothesis that some dysregulated processes in T2DM may contribute or even trigger the neurodegenerative process in PD. One major consequence of T2DM is the unprogrammed reaction between sugars, increased in T2DM, and proteins, a reaction named glycation. Pre-clinical reports show that alpha-synuclein is a target of glycation, and glycation potentiates its pathogenicity which contributes for the neurodegenerative process. Moreover, it triggers, anticipates, or aggravates several PD-like motor and non-motor complications. A given profile of proteins are differently glycated in diseased conditions, altering the brain proteome and leading to brain dysfunction and neurodegeneration. Herein we coin the term Glycatome as the profile of glycated proteins. In this review we report on the mechanisms underlying the association between T2DM and PD, with particular focus on the impact of protein glycation.
Collapse
Affiliation(s)
- Ana Chegão
- iNOVA4Health, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Hugo Vicente Miranda
- iNOVA4Health, NOVA Medical School, NMS, Universidade NOVA de Lisboa, Lisboa, Portugal.
| |
Collapse
|
11
|
Wang Q, Zheng J, Pettersson S, Reynolds R, Tan EK. The link between neuroinflammation and the neurovascular unit in synucleinopathies. SCIENCE ADVANCES 2023; 9:eabq1141. [PMID: 36791205 PMCID: PMC9931221 DOI: 10.1126/sciadv.abq1141] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 01/19/2023] [Indexed: 05/28/2023]
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glial cells, and neurons. As a fundamental functional module in the central nervous system, the NVU maintains homeostasis in the microenvironment and the integrity of the blood-brain barrier. Disruption of the NVU and interactions among its components are involved in the pathophysiology of synucleinopathies, which are characterized by the pathological accumulation of α-synuclein. Neuroinflammation contributes to the pathophysiology of synucleinopathies, including Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. This review aims to summarize the neuroinflammatory response of glial cells and vascular cells in the NVU. We also review neuroinflammation in the context of the cross-talk between glial cells and vascular cells, between glial cells and pericytes, and between microglia and astroglia. Last, we discuss how α-synuclein affects neuroinflammation and how neuroinflammation influences the aggregation and spread of α-synuclein and analyze different properties of α-synuclein in synucleinopathies.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Sven Pettersson
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
- Karolinska Institutet, Department of Odontology, 171 77 Solna, Sweden
- Faculty of Medical Sciences, Sunway University, Subang Jaya, 47500 Selangor, Malaysia
- Department of Microbiology and Immunology, National University Singapore, Singapore 117545, Singapore
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London W12 0NN, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
12
|
Chen YY, Chen SY, Lin JA, Yen GC. Preventive Effect of Indian Gooseberry (Phyllanthus emblica L.) Fruit Extract on Cognitive Decline in High-Fat Diet (HFD)-Fed Rats. Mol Nutr Food Res 2023; 67:e2200791. [PMID: 36738163 DOI: 10.1002/mnfr.202200791] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/27/2022] [Indexed: 02/05/2023]
Abstract
SCOPE Methylglyoxal (MG)-derived advanced glycation end products (AGEs) directly bind to the receptor for advanced glycation end products (RAGE), subsequently exacerbating obesity and obesity-induced cognitive decline. Indian gooseberry (Phyllanthus emblica L.) fruit has antiobesity properties. However, the underlying mechanism by which Indian gooseberry fruit prevents obesity-induced cognitive decline remains unclear. METHODS AND RESULTS This study aims to investigate the preventive effect of a water extract of Indian gooseberry fruit (WEIG) and its bioactive compound gallic acid (GA) on the obesity-induced cognitive decline through MG suppression and gut microbiota modulation in high-fat diet (HFD)-fed rats. Trapping MG, WEIG, and GA significantly ameliorate fat accumulation in adipose tissue and learning and memory deficits. Mechanistically, WEIG and GA administration effectively reduces brain MG and AGE levels and subsequently reduces insulin resistance, inflammatory cytokines, MDA production, and Alzheimer's disease-related proteins, but increases both antioxidant enzyme activities and anti-inflammatory cytokine with inhibiting RAGE, MAPK, and NF-κB levels in HFD-fed rats. Additionally, WEIG and GA supplementation increases the relative abundances of Bacteroidetes, Gammaproteobacteria, and Parasutterella, which negatively correlate with MG, inflammatory cytokine, and Alzheimer's disease-related protein expressions. CONCLUSION This novel finding provides a possible mechanism by which WEIG prevents obesity-induced cognitive decline through the gut-brain axis.
Collapse
Affiliation(s)
- Ying-Yin Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Sheng-Yi Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Jer-An Lin
- Graduate Institute of Food Safety, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Road, Taichung, 40227, Taiwan
| |
Collapse
|
13
|
Moradi S, Zamani A, Mazdeh M, Ramezani M, Komaki A, Talebi-Ghane E, Mahdi Eftekharian M. An inclusive study on cytokine gene expression in Parkinson's disease: Advanced analysis using Bayesian regression model. Hum Immunol 2023; 84:123-129. [PMID: 36400640 DOI: 10.1016/j.humimm.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 10/22/2022] [Accepted: 11/01/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is the second most prevalent neurodegenerative disease throughout the globe whose specific pathophysiology is unknown. Researchers believe that inflammation and oxidative stress contribute to PD development. Also, alterations in cytokines production appear to have a key role in the pathogenesis of PD. The aim of the current study was to evaluate gene expression levels of nine cytokines in the peripheral blood of PD patients compared to a healthy control group. METHODS Real-time PCR was used to analyze cytokines gene expression followed by advanced statistical analysis performed using Bayesian regression model in R (version 4.1.0) statistical software. RESULTS TNF-α, IL-1β, IL-2, IL-4, IFN-γ, IL-17 and IL-6 transcript levels were upregulated in patients compared to healthy controls. However, CXCL8 expression was downregulated in patients compared to controls and IFN-β expression was not statistically different between the two groups. While we found no significant difference between the groups based on gender and age regarding TNF-α, IL-1β, CXCL8, IL-2, IL-4, IFN-γ and IFN-β gene expression, IL-6 and IL-17 transcript levels showed significant upregulations in older subjects and in females, respectively. In addition, we found that the interaction effects between gender and group on gene expression levels were not significant. In this way, the subgroup analysis within gender revealed that in each gender, expression levels of TNF-α, IL-2, IL-4, IL-6, IFN-γ and IL-17 were significantly higher in patients than controls. However, IFN-β expression level did not show any significant difference between groups and subgroups. CONCLUSION The present study provides evidence on significant alterations in cytokine expression with different patterns and points to immune system dysregulation in PD.
Collapse
Affiliation(s)
- Shadi Moradi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Zamani
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehrdokht Mazdeh
- Department of Neurology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahdi Ramezani
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Elahe Talebi-Ghane
- Modeling of Noncommunicable Diseases Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Mahdi Eftekharian
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
14
|
Gou T, Jin X, Xia J. Idebenone reduces sepsis-induced oxidative stress and apoptosis in hepatocytes via RAGE/p38 signaling. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1363. [PMID: 36660726 PMCID: PMC9843342 DOI: 10.21037/atm-22-5758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022]
Abstract
Background Sepsis-induced liver dysfunction is believed to be an independent risk factor for multiple organ dysfunction and death. Idebenone (IDE), a synthetic analog of coenzyme Q10 (CoQ10), possesses an antioxidizing property. The present study aimed to investigate the efficacy of IDE on sepsis-induced liver injury and discuss its reaction mechanism in vivo and in vitro. Methods To establish an in vivo model of sepsis-induced liver injury, rats were treated with high-grade cecal ligation and puncture (CLP). Hematoxylin-eosin staining was applied to observe the liver pathological changes, and liver function was examined using alanine aminotransferase (ALT) and aspartate aminotransferase (AST) assay kits. Enzyme-linked immunosorbent assay (ELISA) kits were employed to assess the levels of inflammatory cytokines in serum and tissues. The activities of malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were analyzed using MDA, SOD, and GSH-Px assay kits, respectively. The apoptosis of liver tissues was measured by terminal deoxynucleotidyl transferase-mediated nick-end labeling (TUNEL) staining, and western blot was employed to estimate apoptosis-related proteins. In vitro, 0.5 μg/mL lipopolysaccharide (LPS) was adopted to administrate primary hepatocytes. The expressions of receptor for advanced glycation end products (RAGE)/p38-related proteins were evaluated by western blot. Cell counting kit-8 (CCK-8) and dichloro-dihydro-fluorescein diacetate (DCFH-DA) assays were utilized to estimate cell viability and reactive oxygen species (ROS) content. Moreover, the transfection efficacy of overexpression (Ov)-RAGE in primary hepatocytes was tested by real-time quantitative polymerase chain reaction (RT-qPCR) and western blot. Results IDE could improve liver function and reduce sepsis-induced pathological damage. The levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in the serum and liver tissue of sepsis rats were suppressed by IDE. Additionally, IDE repressed the oxidative stress and apoptosis of liver tissues in sepsis-induced rats. IDE also inhibited RAGE/p38 signaling. Furthermore, IDE revived the decreased viability in LPS-induced hepatocytes concentration-dependently. After overexpressing RAGE, RAGE expression in hepatocytes was significantly elevated. Further functional experiments revealed that IDE attenuated cell viability injury, apoptosis, oxidative stress, and inflammatory damage in LPS-induced hepatocytes via RAGE/p38 signaling. Conclusions IDE helped to protect against sepsis-induced liver injury via the regulation of RAGE/p38 signaling.
Collapse
Affiliation(s)
- Tao Gou
- Department of Emergency, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xiaoyu Jin
- Department of Emergency, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Jinming Xia
- Department of Emergency, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
15
|
Juranek J, Mukherjee K, Kordas B, Załęcki M, Korytko A, Zglejc-Waszak K, Szuszkiewicz J, Banach M. Role of RAGE in the Pathogenesis of Neurological Disorders. Neurosci Bull 2022; 38:1248-1262. [PMID: 35729453 PMCID: PMC9554177 DOI: 10.1007/s12264-022-00878-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/03/2022] [Indexed: 11/30/2022] Open
Abstract
This review reflects upon our own as well as other investigators' studies on the role of receptor for advanced glycation end-products (RAGE), bringing up the latest information on RAGE in physiology and pathology of the nervous system. Over the last ten years, major progress has been made in uncovering many of RAGE-ligand interactions and signaling pathways in nervous tissue; however, the translation of these discoveries into clinical practice has not come to fruition yet. This is likely, in part to be the result of our incomplete understanding of this crucial signaling pathway. Clinical trials examining the therapeutic efficacy of blocking RAGE-external ligand interactions by genetically engineered soluble RAGE or an endogenous RAGE antagonist, has not stood up to its promise; however, other trials with different blocking agents are being considered with hope for therapeutic success in diseases of the nervous system.
Collapse
Affiliation(s)
- Judyta Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-085, Olsztyn, Poland.
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Blacksburg, VA, 24016, USA
| | - Bernard Kordas
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-085, Olsztyn, Poland
| | - Michał Załęcki
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719, Olsztyn, Poland
| | - Agnieszka Korytko
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-085, Olsztyn, Poland
| | - Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-085, Olsztyn, Poland
| | - Jarosław Szuszkiewicz
- Department of Materials and Machines Technology, Faculty of Technical Sciences, University of Warmia and Mazury, 10-719, Olsztyn, Poland
| | - Marta Banach
- Department of Neurology, Collegium Medicum, Jagiellonian University, 31-008, Kraków, Poland.
| |
Collapse
|
16
|
Long H, Zhang S, Zeng S, Tong Y, Liu J, Liu C, Li D. Interaction of RAGE with α-synuclein fibrils mediates inflammatory response of microglia. Cell Rep 2022; 40:111401. [PMID: 36130498 DOI: 10.1016/j.celrep.2022.111401] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 08/03/2022] [Accepted: 08/31/2022] [Indexed: 11/03/2022] Open
Abstract
Microglia-mediated neuroinflammation and α-synuclein (α-syn) aggregation, both as pathological hallmarks of Parkinson's disease (PD), crosstalk to exacerbate degeneration of dopaminergic neurons and PD progression. However, the mechanism underlying their interaction is poorly understood, which obstructs effective therapeutic inhibition of α-syn-induced neuroinflammation. Here, we initiate from structure-based interaction predictions and find that receptor for advanced glycation end products (RAGE) serves as a receptor of α-syn fibrils on microglia. Results of nuclear magnetic resonance (NMR) spectroscopy and mutagenesis validate that the V domain of RAGE that contains an alkaline surface can bind with acidic C-terminal residues of α-syn. Furthermore, the binding of α-syn fibrils with RAGE induces neuroinflammation, which is blocked by both genetic depletion of RAGE and inhibitor FPS-ZM1. Our work shows the important role, as well as the structural mechanism, of RAGE in mediating the inflammatory response of microglia to α-syn fibrils, which may help to establish effective therapeutic strategies to alleviate α-syn-induced neuroinflammation and neuronal damage.
Collapse
Affiliation(s)
- Houfang Long
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyi Zeng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yilun Tong
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China; WLA Laboratories, World Laureates Association, Shanghai 201203, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
17
|
Al-Ghraiybah NF, Wang J, Alkhalifa AE, Roberts AB, Raj R, Yang E, Kaddoumi A. Glial Cell-Mediated Neuroinflammation in Alzheimer's Disease. Int J Mol Sci 2022; 23:10572. [PMID: 36142483 PMCID: PMC9502483 DOI: 10.3390/ijms231810572] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder; it is the most common cause of dementia and has no treatment. It is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of Neurofibrillary tangles (NFTs). Yet, those two hallmarks do not explain the full pathology seen with AD, suggesting the involvement of other mechanisms. Neuroinflammation could offer another explanation for the progression of the disease. This review provides an overview of recent advances on the role of the immune cells' microglia and astrocytes in neuroinflammation. In AD, microglia and astrocytes become reactive by several mechanisms leading to the release of proinflammatory cytokines that cause further neuronal damage. We then provide updates on neuroinflammation diagnostic markers and investigational therapeutics currently in clinical trials to target neuroinflammation.
Collapse
Affiliation(s)
- Nour F. Al-Ghraiybah
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Junwei Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amer E. Alkhalifa
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Andrew B. Roberts
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Euitaek Yang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| |
Collapse
|
18
|
Dong H, Zhang Y, Huang Y, Deng H. Pathophysiology of RAGE in inflammatory diseases. Front Immunol 2022; 13:931473. [PMID: 35967420 PMCID: PMC9373849 DOI: 10.3389/fimmu.2022.931473] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a non-specific multi-ligand pattern recognition receptor capable of binding to a range of structurally diverse ligands, expressed on a variety of cell types, and performing different functions. The ligand-RAGE axis can trigger a range of signaling events that are associated with diabetes and its complications, neurological disorders, cancer, inflammation and other diseases. Since RAGE is involved in the pathophysiological processes of many diseases, targeting RAGE may be an effective strategy to block RAGE signaling.
Collapse
|
19
|
The "Cerebrospinal Fluid Sink Therapeutic Strategy" in Alzheimer's Disease-From Theory to Design of Applied Systems. Biomedicines 2022; 10:biomedicines10071509. [PMID: 35884814 PMCID: PMC9313192 DOI: 10.3390/biomedicines10071509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a global health problem, with incidence and prevalence considered to increase during the next decades. However, no currently available effective treatment exists despite numerous clinical trials in progress. Moreover, although many hypotheses are accepted regarding the pathophysiological mechanisms of AD onset and evolution, there are still many unknowns about the disorder. A relatively new approach, based on the amyloid-beta dynamics among different biological compartments, is currently intensely discussed, as it seems to offer a promising solution with significant therapeutic impact. Known as the “cerebrospinal-fluid-sink therapeutic strategy”, part of the “three-sink therapeutic strategy”, this theoretical model focuses on the dynamics of amyloid-beta among the three main liquid compartments of the human body, namely blood, cerebrospinal fluid, and the (brain) interstitial fluid. In this context, this article aims to describe in detail the abovementioned hypothesis, by reviewing in the first part the most relevant anatomical and physiological aspects of amyloid-beta dynamics. Subsequently, explored therapeutic strategies based on the clearance of amyloid-beta from the cerebrospinal fluid level are presented, additionally highlighting their limitations. Finally, the originality and novelty of this work rely on the research experience of the authors, who focus on implantable devices and their utility in AD treatment.
Collapse
|
20
|
Li Y, Peng Y, Shen Y, Zhang Y, Liu L, Yang X. Dietary polyphenols: regulate the advanced glycation end products-RAGE axis and the microbiota-gut-brain axis to prevent neurodegenerative diseases. Crit Rev Food Sci Nutr 2022; 63:9816-9842. [PMID: 35587161 DOI: 10.1080/10408398.2022.2076064] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Advanced glycation end products (AGEs) are formed in non-enzymatic reaction, oxidation, rearrangement and cross-linking between the active carbonyl groups of reducing sugars and the free amines of amino acids. The Maillard reaction is related to sensory characteristics in thermal processed food, while AGEs are formed in food matrix in this process. AGEs are a key link between carbonyl stress and neurodegenerative disease. AGEs can interact with receptors for AGEs (RAGE), causing oxidative stress, inflammation response and signal pathways activation related to neurodegenerative diseases. Neurodegenerative diseases are closely related to gut microbiota imbalance and intestinal inflammation. Polyphenols with multiple hydroxyl groups showed a powerful ability to scavenge ROS and capture α-dicarbonyl species, which led to the formation of mono- and di- adducts, thereby inhibiting AGEs formation. Neurodegenerative diseases can be effectively prevented by inhibiting AGEs production, and interaction with RAGEs, or regulating the microbiota-gut-brain axis. These strategies include polyphenols multifunctional effects on AGEs inhibition, RAGE-ligand interactions blocking, and regulating the abundance and diversity of gut microbiota, and intestinal inflammation alleviation to delay or prevent neurodegenerative diseases progress. It is a wise and promising strategy to supplement dietary polyphenols for preventing neurodegenerative diseases via AGEs-RAGE axis and microbiota-gut-brain axis regulation.
Collapse
Affiliation(s)
- Yueqin Li
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang, PR China
| | - Yao Peng
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, Guangdong, PR China
| | - Yingbin Shen
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong, PR China
| | - Yunzhen Zhang
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang, PR China
| | - Lianliang Liu
- College of Food and Pharmaceutical Sciences, Deep Processing Technology Key Laboratory of Zhejiang Province Animal Protein Food, Ningbo University, Ningbo, Zhejiang, PR China
| | - Xinquan Yang
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, Guangdong, PR China
| |
Collapse
|
21
|
Abdallah IM, Al-Shami KM, Yang E, Wang J, Guillaume C, Kaddoumi A. Oleuropein-Rich Olive Leaf Extract Attenuates Neuroinflammation in the Alzheimer's Disease Mouse Model. ACS Chem Neurosci 2022; 13:1002-1013. [PMID: 35263086 DOI: 10.1021/acschemneuro.2c00005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia among several neurodegenerative disorders afflicting the elderly. AD is characterized by the deposition of extracellular amyloid-β (Aβ) plaques, disrupted blood-brain barrier (BBB), and neuroinflammation. Several studies have demonstrated the health benefits of olive oil and olive leaf extract (OLE) due to their polyphenolic content. The main phenolic compound in OLE is glycosylated oleuropein (OLG), while the aglycon form of oleuropein (OLA) exists in much lower amounts. This work aimed to evaluate the effect of a low dose of OLG-rich OLE and the mechanism(s) that contributed to the observed beneficial effects against Aβ pathology in the homozygous 5xFAD mouse model. Mice were fed with OLE-enriched diet (695 μg/kg body weight/day) for 3 months, starting at 3 months old. Overall findings demonstrated that OLE reduced neuroinflammation by inhibiting the NF-κB pathway and suppressing the activation of NLRP3 inflammasomes and RAGE/HMGB1 pathways. In addition, OLE reduced total Aβ brain levels due to increased clearance and reduced production of Aβ and enhanced BBB integrity and function, which collectively improved the memory function. Thus, the consumption of OLE as a dietary supplement is expected to stop and/or slow the progression of AD.
Collapse
Affiliation(s)
- Ihab M. Abdallah
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, Alabama 36849, United States
| | - Kamal M. Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, Alabama 36849, United States
| | - Euitaek Yang
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, Alabama 36849, United States
| | - Junwei Wang
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, Alabama 36849, United States
| | | | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, Alabama 36849, United States
| |
Collapse
|
22
|
Schmidt S, Vogt Weisenhorn DM, Wurst W. Chapter 5 – “Parkinson's disease – A role of non-enzymatic posttranslational modifications in disease onset and progression?”. Mol Aspects Med 2022; 86:101096. [PMID: 35370007 DOI: 10.1016/j.mam.2022.101096] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 12/14/2022]
|
23
|
Reis PA, Castro-Faria-Neto HC. Systemic Response to Infection Induces Long-Term Cognitive Decline: Neuroinflammation and Oxidative Stress as Therapeutical Targets. Front Neurosci 2022; 15:742158. [PMID: 35250433 PMCID: PMC8895724 DOI: 10.3389/fnins.2021.742158] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/31/2021] [Indexed: 12/29/2022] Open
Abstract
In response to pathogens or damage signs, the immune system is activated in order to eliminate the noxious stimuli. The inflammatory response to infectious diseases induces systemic events, including cytokine storm phenomenon, vascular dysfunction, and coagulopathy, that can lead to multiple-organ dysfunction. The central nervous system (CNS) is one of the major organs affected, and symptoms such as sickness behavior (depression and fever, among others), or even delirium, can be observed due to activation of endothelial and glial cells, leading to neuroinflammation. Several reports have been shown that, due to CNS alterations caused by neuroinflammation, some sequels can be developed in special cognitive decline. There is still no any treatment to avoid cognitive impairment, especially those developed due to systemic infectious diseases, but preclinical and clinical trials have pointed out controlling neuroinflammatory events to avoid the development of this sequel. In this minireview, we point to the possible mechanisms that triggers long-term cognitive decline, proposing the acute neuroinflammatory events as a potential therapeutical target to treat this sequel that has been associated to several infectious diseases, such as malaria, sepsis, and, more recently, the new SARS-Cov2 infection.
Collapse
Affiliation(s)
- Patricia Alves Reis
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- Biochemistry Department, Roberto Alcântara Gomes Biology Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Alves Reis,
| | | |
Collapse
|
24
|
Zhou X, Ying C, Hu B, Zhang Y, Gan T, Zhu Y, Wang N, Li A, Song Y. Receptor for advanced glycation end products aggravates cognitive deficits in type 2 diabetes through binding of C-terminal AAs 2-5 to mitogen-activated protein kinase kinase 3 (MKK3) and facilitation of MEKK3-MKK3-p38 module assembly. Aging Cell 2022; 21:e13543. [PMID: 35080104 PMCID: PMC8844116 DOI: 10.1111/acel.13543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/28/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022] Open
Abstract
In this study, we explored the precise mechanisms underlying the receptor for advanced glycation end products (RAGE)‐mediated neuronal loss and behavioral dysfunction induced by hyperglycemia. We used immunoprecipitation (IP) and GST pull‐down assays to assess the interaction between RAGE and mitogen‐activated protein kinase kinase 3 (MKK3). Then, we investigated the effect of specific mutation of RAGE on plasticity at hippocampal synapses and behavioral deficits in db/db mice through electrophysiological recordings, morphological assays, and behavioral tests. We discovered that RAGE binds MKK3 and that this binding is required for assembly of the MEKK3‐MKK3‐p38 signaling module. Mechanistically, we found that activation of p38 mitogen‐activated protein kinase (MAPK)/NF‐κB signaling depends on mediation of the RAGE‐MKK3 interaction by C‐terminal RAGE (ctRAGE) amino acids (AAs) 2‐5. We found that ctRAGE R2A‐K3A‐R4A‐Q5A mutation suppressed neuronal damage, improved synaptic plasticity, and alleviated behavioral deficits in diabetic mice by disrupting the RAGE‐MKK3 conjugation. High glucose induces direct binding of RAGE and MKK3 via ctRAGE AAs 2‐5, which leads to assembly of the MEKK3‐MKK3‐p38 signaling module and subsequent activation of the p38MAPK/NF‐κB pathway, and ultimately results in diabetic encephalopathy (DE).
Collapse
Affiliation(s)
- Xiao‐Yan Zhou
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology Xuzhou Medical University Xuzhou China
- Department of Genetics, Xuzhou Engineering Research Center of Medical Genetics and Transformation Xuzhou Medical University Xuzhou China
| | - Chang‐Jiang Ying
- Department of Endocrinology Affiliated Hospital of Xuzhou Medical University Xuzhou China
| | - Bin Hu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology Xuzhou Medical University Xuzhou China
| | - Yu‐Sheng Zhang
- The Graduate School Xuzhou Medical University Xuzhou China
| | - Tian Gan
- The Graduate School Xuzhou Medical University Xuzhou China
| | - Yan‐Dong Zhu
- The Graduate School Xuzhou Medical University Xuzhou China
| | - Nan Wang
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology Xuzhou Medical University Xuzhou China
| | - An‐An Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology Xuzhou Medical University Xuzhou China
| | - Yuan‐Jian Song
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology Xuzhou Medical University Xuzhou China
- Department of Genetics, Xuzhou Engineering Research Center of Medical Genetics and Transformation Xuzhou Medical University Xuzhou China
| |
Collapse
|
25
|
Chunduri A, Crusio WE, Delprato A. Narcolepsy in Parkinson's disease with insulin resistance. F1000Res 2022; 9:1361. [PMID: 34745571 PMCID: PMC8543173 DOI: 10.12688/f1000research.27413.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Parkinson’s disease (PD) is characterized by its progression of motor-related symptoms such as tremors, rigidity, slowness of movement, and difficulty with walking and balance. Comorbid conditions in PD individuals include insulin resistance (IR) and narcolepsy-like sleep patterns. The intersecting sleep symptoms of both conditions include excessive daytime sleepiness, hallucinations, insomnia, and falling into REM sleep more quickly than an average person. Understanding of the biological basis and relationship of these comorbid disorders with PD may help with early detection and intervention strategies to improve quality of life. Methods: In this study, an integrative genomics and systems biology approach was used to analyze gene expression patterns associated with PD, IR, and narcolepsy in order to identify genes and pathways that may shed light on how these disorders are interrelated. A correlation analysis with known genes associated with these disorders (LRRK2, HLA-DQB1, and HCRT) was used to query microarray data corresponding to brain regions known to be involved in PD and narcolepsy. This includes the hypothalamus, dorsal thalamus, pons, and subcoeruleus nucleus. Risk factor genes for PD, IR, and narcolepsy were also incorporated into the analysis. Results: The PD and narcolepsy signaling networks are connected through insulin and immune system pathways. Important genes and pathways that link PD, narcolepsy, and IR are CACNA1C, CAMK1D, BHLHE41, HMGB1, and AGE-RAGE. Conclusions: We have identified the genetic signatures that link PD with its comorbid disorders, narcolepsy and insulin resistance, from the convergence and intersection of dopaminergic, insulin, and immune system related signaling pathways. These findings may aid in the design of early intervention strategies and treatment regimes for non-motor symptoms in PD patients as well as individuals with diabetes and narcolepsy.
Collapse
Affiliation(s)
- Alisha Chunduri
- Department of Biotechnology, Chaitanya Bharathi Institute of Technology, Hyderabad, 500075, India
- Department of Research and Education, BioScience Project, Wakefield, MA, 01880, USA
| | - Wim E. Crusio
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Pessac, 33615, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287 University of Bordeaux, Pessac, 33615, France
| | - Anna Delprato
- Department of Research and Education, BioScience Project, Wakefield, MA, 01880, USA
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Pessac, 33615, France
| |
Collapse
|
26
|
Lin KH, Ali A, Kuo CH, Yang PC, Kumar VB, Padma VV, Lo JF, Huang CY, Kuo WW. Carboxyl terminus of HSP70-interacting protein attenuates advanced glycation end products-induced cardiac injuries by promoting NFκB proteasomal degradation. J Cell Physiol 2021; 237:1888-1901. [PMID: 34958118 DOI: 10.1002/jcp.30660] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 11/06/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022]
Abstract
Advanced glycation end products (AGEs), which are highly reactive molecules resulting from persistent high-glucose levels, can lead to the generation of oxidative stress and cardiac complications. The carboxyl terminus of HSP70 interacting protein (CHIP) has been demonstrated to have a protective role in several diseases, including cardiac complications; however, the role in preventing AGE-induced cardiac damages remains poorly understood. Here, we found that elevated AGE levels impaired cardiac CHIP expression in streptozotocin-induced diabetes and high-fat diet-administered animals, representing AGE exposure models. We used the TUNEL assay, hematoxylin and eosin, Masson's trichrome staining, and western blotting to prove that cardiac injuries were induced in diabetic animals and AGE-treated cardiac cells. Interestingly, our results collectively indicated that CHIP overexpression significantly rescued the AGE-induced cardiac injuries and promoted cell survival. Moreover, CHIP knockdown-mediated stabilization of nuclear factor κB (NFκB) was attenuated by overexpressing CHIP in the cells. Furthermore, co-immunoprecipitation and immunoblot assay revealed that CHIP promotes the ubiquitination and proteasomal degradation of AGE-induced NFκB. Importantly, fluorescence microscopy, a luciferase reporter assay, electrophoretic mobility shift assay, and subcellular fractionation further demonstrated that CHIP overexpression inhibits AGE-induced NFκB nuclear translocation, reduced its binding ability with the promoter sequences of the receptor of AGE, consequently inhibiting the translocation of the receptor AGE to the cell membrane for its proper function. Overall, our current study findings suggest that CHIP can target NFκB for ubiquitin-mediated proteasomal degradation, and thereby potentially rescue AGE-induced cardiac damages.
Collapse
Affiliation(s)
- Kuan-Ho Lin
- College of Medicine, China Medical University, Taichung, Taiwan.,Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ayaz Ali
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - Pei-Chen Yang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | | | | | - Jeng-Fan Lo
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Genome Research Centre, National Yang-Ming University, Taipei, Taiwan.,Cancer Progression Centre of Excellence, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Yang Huang
- Department of Biotechnology, Asia University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Centre of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| |
Collapse
|
27
|
Zhang X, Wang J, Gao JZ, Zhang XN, Dou KX, Shi WD, Xie AM. P2X4 receptor participates in autophagy regulation in Parkinson's disease. Neural Regen Res 2021; 16:2505-2511. [PMID: 33907041 PMCID: PMC8374561 DOI: 10.4103/1673-5374.313053] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/15/2020] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
Dysfunctional autophagy often occurs during the development of neurodegenerative diseases, such as Parkinson's disease, Huntington's disease, and Alzheimer's disease. The purinergic P2X4 receptor is an ATP-gated ion channel that is widely expressed in the microglia, astrocytes, and neurons of the central and peripheral nervous systems. P2X4R is involved in the regulation of cellular excitability, synaptic transmission, and neuroinflammation. However, the role played by P2X4R in Parkinson's disease remains poorly understood. Rat models of Parkinson's disease were established by injecting 6-hydroxydopamine into the substantia nigra pars compacta. P2X4R-targeted small interfering RNA (siRNA) was injected into the same area 1 week before injury induction to inhibit the expression of the P2X4 receptor. The results showed that the inhibition of P2X4 receptor expression in Parkinson's disease model rats reduced the rotation behavior induced by apomorphine treatment, increased the latency on the rotarod test, and upregulated the expression of tyrosine hydroxylase, brain-derived neurotrophic factor, LC3-II/LC3-I, Beclin-1, and phosphorylated tropomyosin receptor kinase B (TrkB) in brain tissue, while simultaneously reducing p62 levels. These findings suggest that P2X4 receptor activation might inhibit neuronal autophagy through the regulation of the brain-derived neurotrophic factor/TrkB signaling pathway, leading to dopaminergic neuron damage in the substantia nigra and the further inhibition of P2X4 receptor-mediated autophagy. These results indicate that P2X4 receptor might serve as a potential novel target for the treatment of Parkinson's disease. This study was approved by the Animal Ethics Committee of Affiliated Hospital of Qingdao University (approval No. QYFYWZLL26119) on April 12, 2016.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jing Wang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jin-Zhao Gao
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiao-Na Zhang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Kai-Xin Dou
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Wan-Da Shi
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - An-Mu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
- Neurological Regulation Institute of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
28
|
Badner A, Reinhardt EK, Nguyen TV, Midani N, Marshall AT, Lepe CA, Echeverria K, Lepe JJ, Torrecampo V, Bertan SH, Tran SH, Anderson AJ, Cummings BJ. Freshly Thawed Cryobanked Human Neural Stem Cells Engraft within Endogenous Neurogenic Niches and Restore Cognitive Function after Chronic Traumatic Brain Injury. J Neurotrauma 2021; 38:2731-2746. [PMID: 34130484 DOI: 10.1089/neu.2021.0045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Human neural stem cells (hNSCs) have potential as a cell therapy after traumatic brain injury (TBI). While various studies have demonstrated the efficacy of NSCs from ongoing culture, there is a significant gap in our understanding of freshly thawed cells from cryobanked stocks-a more clinically relevant source. To address these shortfalls, the therapeutic potential of our previously validated Shef-6.0 human embryonic stem cell (hESC)-derived hNSC line was tested after long-term cryostorage and thawing before transplant. Immunodeficient athymic nude rats received a moderate unilateral controlled cortical impact (CCI) injury. At four weeks post-injury, 6 × 105 freshly thawed hNSCs were transplanted into six injection sites (two ipsi- and four contra-lateral) with 53.4% of cells surviving three months post-transplant. Interestingly, most hNSCs were engrafted in the meninges and the lining of lateral ventricles, associated with high CXCR4 expression and a chemotactic response to SDF1alpha (CXCL12). While some expressed markers of neuron, astrocyte, and oligodendrocyte lineages, the majority remained progenitors, identified through doublecortin expression (78.1%). Importantly, transplantation resulted in improved spatial learning and memory in Morris water maze navigation and reduced risk taking in an elevated plus maze. Investigating potential mechanisms of action, we identified an increase in ipsilateral host hippocampus cornu ammonis (CA) neuron survival, contralateral dentate gyrus (DG) volume, and DG neural progenitor morphology as well as a reduction in neuroinflammation. Together, these findings validate the potential of hNSCs to improve function after TBI and demonstrate that long-term biobanking of cells and thawing aliquots before use may be suitable for clinical deployment.
Collapse
Affiliation(s)
- Anna Badner
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
| | - Emily K Reinhardt
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, California, USA
| | - Theodore V Nguyen
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
| | - Nicole Midani
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
| | - Andrew T Marshall
- Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Cherie A Lepe
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
| | - Karla Echeverria
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
| | - Javier J Lepe
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
| | - Vincent Torrecampo
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
| | - Sara H Bertan
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, California, USA
| | - Serinee H Tran
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, California, USA
| | - Aileen J Anderson
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, California, USA
- Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, California, USA
- Anatomy and Neurobiology, University of California, Irvine, Irvine, California, USA
| | - Brian J Cummings
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, California, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, California, USA
- Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, California, USA
- Anatomy and Neurobiology, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
29
|
Fontana L, Ghezzi L, Cross AH, Piccio L. Effects of dietary restriction on neuroinflammation in neurodegenerative diseases. J Exp Med 2021; 218:211666. [PMID: 33416892 PMCID: PMC7802371 DOI: 10.1084/jem.20190086] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Recent and accumulating work in experimental animal models and humans shows that diet has a much more pervasive and prominent role than previously thought in modulating neuroinflammatory and neurodegenerative mechanisms leading to some of the most common chronic central nervous system (CNS) diseases. Chronic or intermittent food restriction has profound effects in shaping brain and peripheral metabolism, immunity, and gut microbiome biology. Interactions among calorie intake, meal frequency, diet quality, and the gut microbiome modulate specific metabolic and molecular pathways that regulate cellular, tissue, and organ homeostasis as well as inflammation during normal brain aging and CNS neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis, among others. This review discusses these findings and their potential application to the prevention and treatment of CNS neuroinflammatory diseases and the promotion of healthy brain aging.
Collapse
Affiliation(s)
- Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia.,Department of Clinical and Experimental Sciences, Brescia University, Brescia, Italy
| | - Laura Ghezzi
- Department of Neurology, Washington University in St. Louis, St. Louis, MO.,University of Milan, Milan, Italy
| | - Anne H Cross
- Department of Neurology, Washington University in St. Louis, St. Louis, MO
| | - Laura Piccio
- Department of Neurology, Washington University in St. Louis, St. Louis, MO.,Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
30
|
Ding W, Qi M, Ma L, Xu X, Chen Y, Zhang W. ADP/ATP translocase 1 protects against an α-synuclein-associated neuronal cell damage in Parkinson's disease model. Cell Biosci 2021; 11:130. [PMID: 34246309 PMCID: PMC8272299 DOI: 10.1186/s13578-021-00645-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND ADP/ATP translocase 1 (ANT1) is involved in the exchange of cytosolic ADP and mitochondrial ATP, and its defection plays an important role in mitochondrial pathogenesis. To reveal an etiological implication of ANT1 for Parkinson's disease (PD), a neurodegenerative disorder, a mouse model treated with 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine and neuroblastoma cell model induced by 1-methyl-4-pehny1-pyridine were utilized in this study. RESULTS The tissue-specific abundance in ANT1 in mouse brains was accessed using the analysis of Western blot and immunohistochemistry. Down-regulated soluble ANT1 was found to be correlated with PD, and ANT1 was associated with PD pathogenesis via forming protein aggregates with α-synuclein. This finding was confirmed at cellular level using neuroblastoma cell models. ANT1 supplement in neuronal cells revealed the protective roles of ANT1 against cytotoxicity caused by MPP+. Protein interaction assay, coupled with the analysis of LC-MS/MS, silver-stained SDS-PAGE and Western blot against anti-ANT1 antibody respectively, illustrated the interaction of ANT1 with α-synuclein using the expressed α-synuclein as a bite. Additionally, a significant increasing ROSs was detected in the MPP+-treated cells. CONCLUSIONS This study indicated that ANT1 was a potentially causative factor of PD, and led to neuropathogenic injury via promoting the formation of protein aggregates with α-synuclein. This investigation potentially promotes an innovative understanding of ANT1 on the etiology of PD and provides valuable information on developing potential drug targets in PD treatment or reliable biomarkers in PD prognostication.
Collapse
Affiliation(s)
- Wenyong Ding
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Minghua Qi
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Li Ma
- Department of Epidemiology, Dalian Medical University, Dalian, 116044, China
| | - Xuefei Xu
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yingfei Chen
- Grade 2020, Capital Medical University, Beijing, 100069, China
| | - Wenli Zhang
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
31
|
Sesamin suppresses advanced glycation end products induced microglial reactivity using BV2 microglial cell line as a model. Brain Res Bull 2021; 172:190-202. [PMID: 33894297 DOI: 10.1016/j.brainresbull.2021.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 04/12/2021] [Accepted: 04/17/2021] [Indexed: 11/22/2022]
Abstract
Neuroinflammation-mediated microglial reactivity is a major process, which explains the increased risk of Alzheimer's disease (AD) development in patients with Type 2 diabetes mellitus (T2DM). Advanced glycation end products (AGEs), formed by hyperglycemic condition in diabetes, is characterized as an intermediary of brain injury with diabetes through induction of microglial reactivity. Here, we explored the effect of AGEs on microglial reactivity using BV2 as a model. The NF-κB, p38 and JNK pathways were found to be important mechanism in AGEs-induced BV2 microglial reactivity. NF-κB inhibitor (BAY-11-7082), p38 inhibitor (SB203580) and JNK inhibitor (SP600125) exhibited the potential inhibition of AGEs-induced NO production. We also found that the sesamin, a major lignan found in sesame seed oils, exerts an anti-inflammatory effect under AGEs-induced microglial reactivity via suppressing the phosphorylation of NF-κB, p38 and JNK pathways. Moreover, sesamin also ameliorated AGEs-induced-receptor for advanced glycation end products (RAGE) expression. Taken together, sesamin may be a promising phytochemical compound to delay inflammatory progress by AGEs microglia function. Similarly, inhibition of AGEs-induced microglial reactivity might be potential therapeutic targets of neuroinflammation-based mechanisms in T2DM link progressive AD.
Collapse
|
32
|
Zhang BP, Wu L, Wu XW, Wang F, Zhao X. Dexmedetomidine protects against degeneration of dopaminergic neurons and improves motor activity in Parkinson's disease mice model. Saudi J Biol Sci 2021; 28:3198-3203. [PMID: 34121856 PMCID: PMC8176059 DOI: 10.1016/j.sjbs.2021.04.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/04/2021] [Accepted: 04/05/2021] [Indexed: 11/18/2022] Open
Abstract
Parkinson’s disease (PD) is the result of dopaminergic (DA) neuronal death in the substantianigra pars compacta (SNc). Current treatments for PD such as L-dopa are limited in effectiveness and fail to address the cause. Targeted anti-inflammatory therapies, particularly directed at nuclear factor kappa B (NF‐κB) activity in alleviating degeneration of DA-neurons is of evolving interest. In the present study, we hypothesised that dexmedetomidine (DEX), an alpha-2 receptor adrenergic agonist, suppress the inflammatory responses associated with PD and restores dopaminergic levels by alleviating substantia nigral degeneration. Male mice (C57Bl/10, 8–11 months old and of 34–40 g of weight) were divided into: the control, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and MPTP + dexmedetomidine (MPTP + DEX) (n = 26 each group). Dex restored dopamine levels in SNpc of MPTP-induced PD mice model. Results of immunohisto staining revealed that Dex treatment post-MPTP induction restored TH-positive cells, with only 12.37% increase (##p < 0.01 vs MPTP) on the third day and a steep 55% increase (###p < 0.001 vs MPTP) following the seventh day of Dex treatment. Moreover, the expressions of proinflammatory markers regulated by NF-κB were diminished in Dex + MPTP group. In addition, cylinder test revealed that Dex treatment improved asymmetric limb usage pattern in MPTP induced mice over the course of 7 days. Hence, in this study, we provided insight on the effect of Dex in the inhibition of NF-κB1 regulated proinflammatory mediators to improve dopamine levels and reduce SNpc dopaminergic neuronal degeneration.
Collapse
Affiliation(s)
- Bao-Ping Zhang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| | - Li Wu
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| | - Xian-Wei Wu
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| | - Fang Wang
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| | - Xin Zhao
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054 Shaanxi, China
| |
Collapse
|
33
|
Atypical p38 Signaling, Activation, and Implications for Disease. Int J Mol Sci 2021; 22:ijms22084183. [PMID: 33920735 PMCID: PMC8073329 DOI: 10.3390/ijms22084183] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
The mitogen-activated protein kinase (MAPK) p38 is an essential family of kinases, regulating responses to environmental stress and inflammation. There is an ever-increasing plethora of physiological and pathophysiological conditions attributed to p38 activity, ranging from cell division and embryonic development to the control of a multitude of diseases including retinal, cardiovascular, and neurodegenerative diseases, diabetes, and cancer. Despite the decades of intense investigation, a viable therapeutic approach to disrupt p38 signaling remains elusive. A growing body of evidence supports the pathological significance of an understudied atypical p38 signaling pathway. Atypical p38 signaling is driven by a direct interaction between the adaptor protein TAB1 and p38α, driving p38 autophosphorylation independent from the classical MKK3 and MKK6 pathways. Unlike the classical MKK3/6 signaling pathway, atypical signaling is selective for just p38α, and at present has only been characterized during pathophysiological stimulation. Recent studies have linked atypical signaling to dermal and vascular inflammation, myocardial ischemia, cancer metastasis, diabetes, complications during pregnancy, and bacterial and viral infections. Additional studies are required to fully understand how, when, where, and why atypical p38 signaling is induced. Furthermore, the development of selective TAB1-p38 inhibitors represents an exciting new opportunity to selectively inhibit pathological p38 signaling in a wide array of diseases.
Collapse
|
34
|
Ni J, Wu Z. Inflammation Spreading: Negative Spiral Linking Systemic Inflammatory Disorders and Alzheimer's Disease. Front Cell Neurosci 2021; 15:638686. [PMID: 33716675 PMCID: PMC7947253 DOI: 10.3389/fncel.2021.638686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022] Open
Abstract
As a physiological response to injury in the internal body organs, inflammation is responsible for removing dangerous stimuli and initiating healing. However, persistent and exaggerative chronic inflammation causes undesirable negative effects in the organs. Inflammation occurring in the brain and spinal cord is known as neuroinflammation, with microglia acting as the central cellular player. There is increasing evidence suggesting that chronic neuroinflammation is the most relevant pathological feature of Alzheimer’s disease (AD), regulating other pathological features, such as the accumulation of amyloid-β (Aβ) and hyperphosphorylation of Tau. Systemic inflammatory signals caused by systemic disorders are known to strongly influence neuroinflammation as a consequence of microglial activation, inflammatory mediator production, and the recruitment of peripheral immune cells to the brain, resulting in neuronal dysfunction. However, the neuroinflammation-accelerated neuronal dysfunction in AD also influences the functions of peripheral organs. In the present review, we highlight the link between systemic inflammatory disorders and AD, with inflammation serving as the common explosion. We discuss the molecular mechanisms that govern the crosstalk between systemic inflammation and neuroinflammation. In our view, inflammation spreading indicates a negative spiral between systemic diseases and AD. Therefore, “dampening inflammation” through the inhibition of cathepsin (Cat)B or CatS may be a novel therapeutic approach for delaying the onset of and enacting early intervention for AD.
Collapse
Affiliation(s)
- Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhou Wu
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
35
|
Ghadimi M, Foroughi F, Hashemipour S, Nooshabadi MR, Ahmadi MH, Yari MG, Kavianpour M, Haghighian HK. Decreased insulin resistance in diabetic patients by influencing Sirtuin1 and Fetuin-A following supplementation with ellagic acid: a randomized controlled trial. Diabetol Metab Syndr 2021; 13:16. [PMID: 33546744 PMCID: PMC7866694 DOI: 10.1186/s13098-021-00633-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/22/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The beneficial effects of polyphenols have been reported. This study aimed to investigate the effect of oral Ellagic acid (EA) supplement on insulin resistance (IR) and Fetuin-A and serum sirtuin1 (SIRT1) in type 2 diabetics. METHODS In this double-blind, randomized clinical trial, 44 diabetic patients were selected. Patients were assigned to the intervention group (22 subjects) and placebo (22 subjects) and received a capsule containing 180 mg of EA per day or placebo for eight weeks, respectively. At the beginning and end of the study, anthropometric indices, fasting plasma glucose (FPG), plasma insulin level, IR, Fetuin-A, and SIRT1 were measured. Statistical analysis was performed using SPSS software. RESULTS At the beginning and end of the study, there was no significant difference between the two groups regarding anthropometric indices (P > 0.05). At the end of the survey, EA supplementation significantly reduced FPG, insulin, IR, and Fetuin-A and increased SIRT1 levels compared with the placebo group (P < 0.05). However, these changes were not significant in the placebo group (P > 0.05). CONCLUSION EA with antioxidant properties plays an essential role in reducing the macrovascular and microvascular complications of diabetes by reducing inflammation and insulin resistance. Trial registration The protocol of this clinical trial is registered with the Iranian Registry of Clinical Trials ( http://www.IRCT.IR , identifier: IRCT20141025019669N13).
Collapse
Affiliation(s)
- Mahnaz Ghadimi
- Department of Nutrition, School of Health, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farshad Foroughi
- Department of Immunology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sima Hashemipour
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Mohammad Hossein Ahmadi
- Department of Laboratory Sciences, School of Allied Medical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Maria Kavianpour
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Khadem Haghighian
- Department of Nutrition, School of Health, Qazvin University of Medical Sciences, Qazvin, Iran.
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
36
|
Ullah R, Ikram M, Park TJ, Ahmad R, Saeed K, Alam SI, Rehman IU, Khan A, Khan I, Jo MG, Kim MO. Vanillic Acid, a Bioactive Phenolic Compound, Counteracts LPS-Induced Neurotoxicity by Regulating c-Jun N-Terminal Kinase in Mouse Brain. Int J Mol Sci 2020; 22:ijms22010361. [PMID: 33396372 PMCID: PMC7795830 DOI: 10.3390/ijms22010361] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE), a pattern recognition receptor signaling event, has been associated with several human illnesses, including neurodegenerative diseases, particularly in Alzheimer’s disease (AD). Vanillic acid (V.A), a flavoring agent, is a benzoic acid derivative having a broad range of biological activities, including antioxidant, anti-inflammatory, and neuroprotective effects. However, the underlying molecular mechanisms of V.A in exerting neuroprotection are not well investigated. The present study aims to explore the neuroprotective effects of V.A against lipopolysaccharides (LPS)-induced neuroinflammation, amyloidogenesis, synaptic/memory dysfunction, and neurodegeneration in mice brain. Behavioral tests and biochemical and immunofluorescence assays were applied. Our results indicated increased expression of RAGE and its downstream phospho-c-Jun n-terminal kinase (p-JNK) in the LPS-alone treated group, which was significantly reduced in the V.A + LPS co-treated group. We also found that systemic administration of LPS-injection induced glial cells (microglia and astrocytes) activation and significantly increased expression level of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-KB) and secretion of proinflammatory cytokines including tumor necrosis factor alpha (TNF-α), interleukin-1 β (IL1-β), and cyclooxygenase (COX-2). However, V.A + LPS co-treatment significantly inhibited the LPS-induced activation of glial cells and neuroinflammatory mediators. Moreover, we also noted that V.A treatment significantly attenuated LPS-induced increases in the expression of AD markers, such as β-site amyloid precursor protein (APP)–cleaving enzyme 1 (BACE1) and amyloid-β (Aβ). Furthermore, V.A treatment significantly reversed LPS-induced synaptic loss via enhancing the expression level of pre- and post-synaptic markers (PSD-95 and SYP), and improved memory performance in LPS-alone treated group. Taken together; we suggest that neuroprotective effects of V.A against LPS-induced neurotoxicity might be via inhibition of LPS/RAGE mediated JNK signaling pathway; and encourage future studies that V.A would be a potential neuroprotective and neurotherapeutic candidate in various neurological disorders.
Collapse
Affiliation(s)
- Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Muhammad Ikram
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow G12OZD, UK;
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Kamran Saeed
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Sayed Ibrar Alam
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Inayat Ur Rehman
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Amjad Khan
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Ibrahim Khan
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Min Gi Jo
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
37
|
Liu Z, Bian M, Ma QQ, Zhang Z, Du HH, Wei CX. Design and Synthesis of New Benzo[d]oxazole-Based Derivatives and Their Neuroprotective Effects on β-Amyloid-Induced PC12 Cells. Molecules 2020; 25:E5391. [PMID: 33218007 PMCID: PMC7698601 DOI: 10.3390/molecules25225391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022] Open
Abstract
A series of novel synthetic substituted benzo[d]oxazole-based derivatives (5a-5v) exerted neuroprotective effects on β-amyloid (Aβ)-induced PC12 cells as a potential approach for the treatment of Alzheimer's disease (AD). In vitro studies show that most of the synthesized compounds were potent in reducing the neurotoxicity of Aβ25-35-induced PC12 cells at 5 μg/mL. We found that compound 5c was non-neurotoxic at 30 μg/mL and significantly increased the viability of Aβ25-35-induced PC12 cells at 1.25, 2.5 and 5 μg/mL. Western blot analysis showed that compound 5c promoted the phosphorylation of Akt and glycogen synthase kinase (GSK-3β) and decreased the expression of nuclear factor-κB (NF-κB) in Aβ25-35-induced PC12 cells. In addition, our findings demonstrated that compound 5c protected PC12 cells from Aβ25-35-induced apoptosis and reduced the hyperphosphorylation of tau protein, and decreased the expression of receptor for AGE (RAGE), β-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1), inducible nitric oxide synthase (iNOS) and Bcl-2-associated X protein/B-cell lymphoma 2 (Bax/Bcl-2) via Akt/GSK-3β/NF-κB signaling pathway. In vivo studies suggest that compound 5c shows less toxicity than donepezil in the heart and nervous system of zebrafish.
Collapse
Affiliation(s)
- Zheng Liu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
| | - Ming Bian
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| | - Qian-Qian Ma
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| | - Zhuo Zhang
- College of Pharmaceutical Sciences, Yanbian University, Yanji 133022, China;
| | - Huan-Huan Du
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| | - Cheng-Xi Wei
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| |
Collapse
|