1
|
Nowicka N, Zglejc-Waszak K, Juranek J, Korytko A, Wąsowicz K, Chmielewska-Krzesińska M, Wojtkiewicz J. Novel insights into RAGE signaling pathways during the progression of amyotrophic lateral sclerosis in RAGE-deficient SOD1 G93A mice. PLoS One 2024; 19:e0299567. [PMID: 38457412 PMCID: PMC10923448 DOI: 10.1371/journal.pone.0299567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is neurodegenerative disease characterized by a progressive loss of motor neurons resulting in paralysis and muscle atrophy. One of the most prospective hypothesis on the ALS pathogenesis suggests that excessive inflammation and advanced glycation end-products (AGEs) accumulation play a crucial role in the development of ALS in patients and SOD1 G93A mice. Hence, we may speculate that RAGE, receptor for advanced glycation end-products and its proinflammatory ligands such as: HMGB1, S100B and CML contribute to ALS pathogenesis. The aim of our studies was to decipher the role of RAGE as well as provide insight into RAGE signaling pathways during the progression of ALS in SOD1 G93A and RAGE-deficient SOD1 G93A mice. In our study, we observed alternations in molecular pattern of proinflammatory RAGE ligands during progression of disease in RAGE KO SOD1 G93A mice compared to SOD1 G93A mice. Moreover, we observed that the amount of beta actin (ACTB) as well as Glial fibrillary acidic protein (GFAP) was elevated in SOD1 G93A mice when compared to mice with deletion of RAGE. These data contributes to our understanding of implications of RAGE and its ligands in pathogenesis of ALS and highlight potential targeted therapeutic interventions at the early stage of this devastating disease. Moreover, inhibition of the molecular cross-talk between RAGE and its proinflammatory ligands may abolish neuroinflammation, gliosis and motor neuron damage in SOD1 G93A mice. Hence, we hypothesize that attenuated interaction of RAGE with its proinflammatory ligands may improve well-being and health status during ALS in SOD1 G93A mice. Therefore, we emphasize that the inhibition of RAGE signaling pathway may be a therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Natalia Nowicka
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Judyta Juranek
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Agnieszka Korytko
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Krzysztof Wąsowicz
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Małgorzata Chmielewska-Krzesińska
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medium, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
2
|
Palanissami G, Paul SF. AGEs and RAGE: metabolic and molecular signatures of the glycation-inflammation axis in malignant or metastatic cancers. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:812-849. [PMID: 37970208 PMCID: PMC10645465 DOI: 10.37349/etat.2023.00170] [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: 03/28/2023] [Accepted: 06/12/2023] [Indexed: 11/17/2023] Open
Abstract
From attributing mutations to cancers with the advent of cutting-edge genetic technology in recent decades, to re-searching the age-old theory of intrinsic metabolic shift of cancers (Warburg's glycolysis), the quest for a precise panacea for mainly the metastatic cancers, remains incessant. This review delineates the advanced glycation end product (AGE)-receptor for AGE (RAGE) pathway driven intricate oncogenic cues, budding from the metabolic (glycolytic) reliance of tumour cells, branching into metastatic emergence of malignancies. Strong AGE-RAGE concomitance in metastasis, chemo-resistance and cancer resurgence adversely incite disease progression and patient mortality. At the conjunction of metabolic and metastatic shift of cancers, are the "glycolytically" generated AGEs and AGE-activated RAGE, instigating aberrant molecular pathways, culminating in aggressive malignancies. AGEs as by-products of metabolic insurgence, modify the metabolome, epigenome and microbiome, besides coercing the inter-, intra- and extra-cellular micro-milieu conducive for oncogenic events like epithelial-mesenchymal transition (EMT). AGE-RAGE synergistically elicit ATP surge for surplus energy, autophagy for apoptotic evasion and chemo-resistance, insulin-like growth factor 1 (IGF-1) for meta-inflammation and angiogenesis, high mobility group box-1 (HMGB1) for immune tolerance, S100 proteins for metastasis, and p53 protein attenuation for tumour suppression. AGEs are pronouncedly reported in invasive forms of breast, prostate, colon and pancreatic cancers, higher in patients with cancer than healthy counterparts, and higher in advanced stage than localized phase. Hence, the investigation of person-specific presence of AGEs, soluble RAGE and AGE-activated RAGE can be advocated as impending bio-markers for diagnostic, prognostic and therapeutic purposes, to predict cancer risk in patients with diabetes, obesity, metabolic syndrome as well as general population, to monitor prognosis and metastasis in patients with cancer, and to reckon complications in cancer survivors. Furthermore, clinical reports of exogenous (dietary) and endogenous (internally formed) AGEs in cancer patients, and contemporary clinical trials involving AGE-RAGE axis in cancer are underlined with theranostic implications.
Collapse
Affiliation(s)
- Gowri Palanissami
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai 600 116, Tamil Nadu, India
| | - Solomon F.D. Paul
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai 600 116, Tamil Nadu, India
| |
Collapse
|
3
|
Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 2023; 8:267. [PMID: 37433768 PMCID: PMC10336149 DOI: 10.1038/s41392-023-01486-5] [Citation(s) in RCA: 410] [Impact Index Per Article: 205.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 07/13/2023] Open
Abstract
Studies in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Amyotrophic lateral sclerosis, Huntington's disease, and so on, have suggested that inflammation is not only a result of neurodegeneration but also a crucial player in this process. Protein aggregates which are very common pathological phenomenon in neurodegeneration can induce neuroinflammation which further aggravates protein aggregation and neurodegeneration. Actually, inflammation even happens earlier than protein aggregation. Neuroinflammation induced by genetic variations in CNS cells or by peripheral immune cells may induce protein deposition in some susceptible population. Numerous signaling pathways and a range of CNS cells have been suggested to be involved in the pathogenesis of neurodegeneration, although they are still far from being completely understood. Due to the limited success of traditional treatment methods, blocking or enhancing inflammatory signaling pathways involved in neurodegeneration are considered to be promising strategies for the therapy of neurodegenerative diseases, and many of them have got exciting results in animal models or clinical trials. Some of them, although very few, have been approved by FDA for clinical usage. Here we comprehensively review the factors affecting neuroinflammation and the major inflammatory signaling pathways involved in the pathogenicity of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic lateral sclerosis. We also summarize the current strategies, both in animal models and in the clinic, for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Weifeng Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China
| | - Dan Xiao
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, P.R. China
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China.
| |
Collapse
|
4
|
Chen L, Wang N, Zhang Y, Li D, He C, Li Z, Zhang J, Guo Y. Proteomics analysis indicates the involvement of immunity and inflammation in the onset stage of SOD1-G93A mouse model of ALS. J Proteomics 2023; 272:104776. [PMID: 36423857 DOI: 10.1016/j.jprot.2022.104776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron degenerative disease, and the pathogenic mechanism that underlies ALS is still unclear. We analyzed the differentially expressed proteins (DEPs) in the spinal cord between SOD1-G93A transgenic mice at the onset stage and non-transgenic (NTG) littermates based on 4D label-free quantitative proteomics (4D-LFQ) with liquid chromatography-tandem mass spectrometry (LC-MS/MS). In our study, 189 DEPs were screened, of which 166 were up-regulated and 23 down-regulated. Clusters of Orthologous Groups (COG)/ EuKaryotic Orthologous Groups (KOG) classification, subcellular localization annotation, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, clustering analysis and protein-protein interaction (PPI) network analyses were performed. Parallel reaction monitoring (PRM) analysis validated 48 proteins from immunity and inflammation-related pathways of KEGG. We described the function and distribution of DEPs, most of which were involved in the following pathways: complement and coagulation cascades, antigen processing and presentation, NF-kappa B signaling pathway, Retinoic acid-inducible gene I (RIG) -I-like receptor signaling pathway, the extracellular matrix-receptor (ECM-receptor) interaction, focal adhesion, phagosome and lysosome. PPI network analysis identified Fn1, Fga, Serpina1e and Serpina3n as potential biomarkers. Our discoveries broaden the view and expand our understanding of immunity and inflammation in ALS. SIGNIFICANCE: This study gives a comprehensive description of DEPs in the spinal cord proteomics of SOD1-G93A mice at the onset period. Compared with a previous study focusing on progressive stage, we showed that immunity and inflammation play an important role at the onset stage of ALS. Several pathways validated by PRM bring new insight to the pathological mechanisms of ALS. The participation of RIG-I-like signaling pathway in ALS and potential biomarkers Fga, Fn1, Serpina1e and Serpina3n are supplements to existing knowledge.
Collapse
Affiliation(s)
- Lin Chen
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, China; Beijing Geriatric Healthcare Center, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Ningyuan Wang
- Xiangya School of Medicine, Central South University, No.172 Tongzipo Road, Changsha, Hunan 410013, China
| | - Yingzhen Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Dongxiao Li
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Caili He
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Zhongzhong Li
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Jian Zhang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Yansu Guo
- Beijing Geriatric Healthcare Center, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China; Beijing Municipal Geriatric Medical Research Center, No. 45 Changchun Street, Xicheng District, Beijing 100053, China.
| |
Collapse
|
5
|
Rossi R, Douglas A, Gil SM, Jabrah D, Pandit A, Gilvarry M, McCarthy R, Prendergast J, Jood K, Redfors P, Nordanstig A, Ceder E, Dunker D, Carlqvist J, Szikora I, Thornton J, Tsivgoulis G, Psychogios K, Tatlisumak T, Rentzos A, Doyle KM. S100b in acute ischemic stroke clots is a biomarker for post-thrombectomy intracranial hemorrhages. Front Neurol 2023; 13:1067215. [PMID: 36756347 PMCID: PMC9900124 DOI: 10.3389/fneur.2022.1067215] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/01/2022] [Indexed: 01/24/2023] Open
Abstract
Background and purpose Post-thrombectomy intracranial hemorrhages (PTIH) are dangerous complications of acute ischemic stroke (AIS) following mechanical thrombectomy. We aimed to investigate if S100b levels in AIS clots removed by mechanical thrombectomy correlated to increased risk of PTIH. Methods We analyzed 122 thrombi from 80 AIS patients in the RESTORE Registry of AIS clots, selecting an equal number of patients having been pre-treated or not with rtPA (40 each group). Within each subgroup, 20 patients had developed PTIH and 20 patients showed no signs of hemorrhage. Gross photos of each clot were taken and extracted clot area (ECA) was measured using ImageJ. Immunohistochemistry for S100b was performed and Orbit Image Analysis was used for quantification. Immunofluorescence was performed to investigate co-localization between S100b and T-lymphocytes, neutrophils and macrophages. Chi-square or Kruskal-Wallis test were used for statistical analysis. Results PTIH was associated with higher S100b levels in clots (0.33 [0.08-0.85] vs. 0.07 [0.02-0.27] mm2, H1 = 6.021, P = 0.014*), but S100b levels were not significantly affected by acute thrombolytic treatment (P = 0.386). PTIH was also associated with patients having higher NIHSS at admission (20.0 [17.0-23.0] vs. 14.0 [10.5-19.0], H1 = 8.006, P = 0.005) and higher number of passes during thrombectomy (2 [1-4] vs. 1 [1-2.5], H1 = 5.995, P = 0.014*). S100b co-localized with neutrophils, macrophages and with T-lymphocytes in the clots. Conclusions Higher S100b expression in AIS clots, higher NIHSS at admission and higher number of passes during thrombectomy are all associated with PTIH. Further investigation of S100b expression in AIS clots by neutrophils, macrophages and T-lymphocytes could provide insight into the role of S100b in thromboinflammation.
Collapse
Affiliation(s)
- Rosanna Rossi
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland,CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland,*Correspondence: Rosanna Rossi ✉
| | - Andrew Douglas
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland,CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Sara Molina Gil
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland,CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Duaa Jabrah
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | | | - James Prendergast
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Katarina Jood
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Redfors
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Annika Nordanstig
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Erik Ceder
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Dennis Dunker
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Jeanette Carlqvist
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - István Szikora
- Department of Neurointerventions, National Institute of Clinical Neurosciences, Budapest, Hungary
| | - John Thornton
- Department of Radiology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Turgut Tatlisumak
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Alexandros Rentzos
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Karen M. Doyle
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland,CÚRAM–SFI Research Centre in Medical Devices, National University of Ireland Galway, Galway, Ireland,Karen M. Doyle ✉
| |
Collapse
|
6
|
Koerich S, Parreira GM, de Almeida DL, Vieira RP, de Oliveira ACP. Receptors for Advanced Glycation End Products (RAGE): Promising Targets Aiming at the Treatment of Neurodegenerative Conditions. Curr Neuropharmacol 2023; 21:219-234. [PMID: 36154605 PMCID: PMC10190138 DOI: 10.2174/1570159x20666220922153903] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 11/22/2022] Open
Abstract
Advanced glycation end products (AGEs) are compounds formed after the non-enzymatic addition of reducing sugars to lipids, proteins, and nucleic acids. They are associated with the development of various clinical complications observed in diabetes and cardiovascular diseases, such as retinopathy, nephropathy, diabetic neuropathy, and others. In addition, compelling evidence indicates that these molecules participate in the progression of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Multiple cellular and molecular alterations triggered by AGEs that could alter homeostasis have been identified. One of the main targets for AGE signaling is the receptor for advanced glycation end-products (RAGE). Importantly, this receptor is the target of not only AGEs, but also amyloid β peptides, HMGB1 (high-mobility group box-1), members of the S100 protein family, and glycosaminoglycans. The activation of this receptor induces intracellular signaling cascades that are involved in pathological processes and cell death. Therefore, RAGE represents a key target for pharmacological interventions in neurodegenerative diseases. This review will discuss the various effects of AGEs and RAGE activation in the pathophysiology of neurodegenerative diseases, as well as the currently available pharmacological tools and promising drug candidates.
Collapse
Affiliation(s)
- Suélyn Koerich
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Gabriela Machado Parreira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Rafael Pinto Vieira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | |
Collapse
|
7
|
Ramasubbu K, Devi Rajeswari V. Impairment of insulin signaling pathway PI3K/Akt/mTOR and insulin resistance induced AGEs on diabetes mellitus and neurodegenerative diseases: a perspective review. Mol Cell Biochem 2022; 478:1307-1324. [PMID: 36308670 DOI: 10.1007/s11010-022-04587-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/12/2022] [Indexed: 12/01/2022]
Abstract
Insulin resistance is common in type 2 diabetes mellitus (T2DM), neurodegenerative diseases, cardiovascular diseases, kidney diseases, and polycystic ovary syndrome. Impairment in insulin signaling pathways, such as the PI3K/Akt/mTOR pathway, would lead to insulin resistance. It might induce the synthesis and deposition of advanced glycation end products (AGEs), reactive oxygen species, and reactive nitrogen species, resulting in stress, protein misfolding, protein accumulation, mitochondrial dysfunction, reticulum function, and metabolic syndrome dysregulation, inflammation, and apoptosis. It plays a huge role in various neurodegenerative diseases like Parkinson's disease, Alzheimer's disease, Huntington's disease, and Amyloid lateral sclerosis. In this review, we intend to focus on the possible effect of insulin resistance in the progression of neurodegeneration via the impaired P13K/Akt/mTOR signaling pathway, AGEs, and receptors for AGEs.
Collapse
Affiliation(s)
- Kanagavalli Ramasubbu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632014, India
| | - V Devi Rajeswari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632014, India.
| |
Collapse
|
8
|
Ikram FZ, Arulsamy A, Retinasamy T, Shaikh MF. The Role of High Mobility Group Box 1 (HMGB1) in Neurodegeneration: A Systematic Review. Curr Neuropharmacol 2022; 20:2221-2245. [PMID: 35034598 PMCID: PMC9886836 DOI: 10.2174/1570159x20666220114153308] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/18/2021] [Accepted: 12/29/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND High mobility group box 1 (HMGB1) protein is a damage-associated molecular pattern (DAMP) that plays an important role in the repair and regeneration of tissue injury. It also acts as a pro-inflammatory cytokine through the activation of toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE), to elicit the neuroinflammatory response. HMGB1 may aggravate several cellular responses, which may lead to pathological inflammation and cellular death. Thus, there have been a considerable amount of research into the pathological role of HMGB1 in diseases. However, whether the mechanism of action of HMGB1 is similar in all neurodegenerative disease pathology remains to be determined. OBJECTIVE Therefore, this systematic review aimed to critically evaluate and elucidate the role of HMGB1 in the pathology of neurodegeneration based on the available literature. METHODS A comprehensive literature search was performed on four databases; EMBASE, PubMed, Scopus, and CINAHL Plus. RESULTS A total of 85 articles were selected for critical appraisal, after subjecting to the inclusion and exclusion criteria in this study. The selected articles revealed that HMGB1 levels were found elevated in most neurodegeneration except in Huntington's disease and Spinocerebellar ataxia, where the levels were found decreased. This review also showcased that HMGB1 may act on distinctive pathways to elicit its pathological response leading to the various neurodegeneration processes/ diseases. CONCLUSION While there have been promising findings in HMGB1 intervention research, further studies may still be required before any HMGB1 intervention may be recommended as a therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Fathimath Zaha Ikram
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia;
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia,Address correspondence to this author at the Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia; Tel/Fax: +60 3 5514 4483; E-mail:
| |
Collapse
|
9
|
Advanced Glycation End Products in Health and Disease. Microorganisms 2022; 10:microorganisms10091848. [PMID: 36144449 PMCID: PMC9501837 DOI: 10.3390/microorganisms10091848] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022] Open
Abstract
Advanced glycation end products (AGEs), formed through the nonenzymatic reaction of reducing sugars with the side-chain amino groups of lysine or arginine of proteins, followed by further glycoxidation reactions under oxidative stress conditions, are involved in the onset and exacerbation of a variety of diseases, including diabetes, atherosclerosis, and Alzheimer’s disease (AD) as well as in the secondary stages of traumatic brain injury (TBI). AGEs, in the form of intra- and interprotein crosslinks, deactivate various enzymes, exacerbating disease progression. The interactions of AGEs with the receptors for the AGEs (RAGE) also result in further downstream inflammatory cascade events. The overexpression of RAGE and the AGE-RAGE interactions are especially involved in cases of Alzheimer’s disease and other neurodegenerative diseases, including TBI and amyotrophic lateral sclerosis (ALS). Maillard reactions are also observed in the gut bacterial species. The protein aggregates found in the bacterial species resemble those of AD and Parkinson’s disease (PD), and AGE inhibitors increase the life span of the bacteria. Dietary AGEs alter the gut microbiota composition and elevate plasma glycosylation, thereby leading to systemic proinflammatory effects and endothelial dysfunction. There is emerging interest in developing AGE inhibitor and AGE breaker compounds to treat AGE-mediated pathologies, including diabetes and neurodegenerative diseases. Gut-microbiota-derived enzymes may also function as AGE-breaker biocatalysts. Thus, AGEs have a prominent role in the pathogenesis of various diseases, and the AGE inhibitor and AGE breaker approach may lead to novel therapeutic candidates.
Collapse
|
10
|
Shen T, Li Y, Wang D, Su Y, Li G, Shang Z, Niu Y, Tan X. YAP1-TEAD1 mediates the perineural invasion of prostate cancer cells induced by cancer-associated fibroblasts. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166540. [PMID: 36100154 DOI: 10.1016/j.bbadis.2022.166540] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/10/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022]
Abstract
Perineural invasion (PNI) driven by the tumor microenvironment (TME) has emerged as a key pattern of metastasis of prostate cancer (PCa), while its underlying mechanism is still elusive. Here, we identified increased CAFs and YAP1 expression levels in patients with metastatic PCa. In the cultured PCa cell line LNCaP, co-culture with cancer-associated fibroblasts (CAFs) could upregulate YAP1 protein expression. Either ectopic overexpression of YAP1 or co-culture with CAFs could promote the infiltration of LNCaPs towards dorsal root ganglia (DRG). This effect could be blocked using an YAP1 inhibitor. In vivo, overexpression of YAP1 could increase PNI in a mouse model of sciatic nerve tumor invasion. Mechanistically, TEAD1 binds to the NGF promotor and YAP1/TEAD1 activates its transcription and consequently increases NGF secretion. In turn, PCa cells treated with CM from CAFs or stable YAP1 overexpression can stimulate DRG to secrete CCL2. The epithelial-to-mesenchymal transition (EMT) of PCa cells is thus activated via CCL2/CCR2. Overall, our data demonstrate that CAFs can activate YAP1/TEAD1 signaling and increase the secretion of NGF, therefore promoting PCa PNI. In addition, EMT induced by PNI suggests a feedback loop is present between neurons and PCa cells.
Collapse
Affiliation(s)
- Tianyu Shen
- School of Medicine, Nankai University, Tianjin, China
| | - Yang Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Dekun Wang
- School of Medicine, Nankai University, Tianjin, China
| | - Yu Su
- School of Medicine, Nankai University, Tianjin, China
| | - Gang Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiqun Shang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuanjie Niu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Xiaoyue Tan
- School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
11
|
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
|
12
|
RNA Molecular Signature Profiling in PBMCs of Sporadic ALS Patients: HSP70 Overexpression Is Associated with Nuclear SOD1. Cells 2022; 11:cells11020293. [PMID: 35053410 PMCID: PMC8774074 DOI: 10.3390/cells11020293] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Superoxide dismutase 1 (SOD1) is one of the causative genes associated with amyotrophic lateral sclerosis (ALS), a neurodegenerative disorder. SOD1 aggregation contributes to ALS pathogenesis. A fraction of the protein is localized in the nucleus (nSOD1), where it seems to be involved in the regulation of genes participating in the oxidative stress response and DNA repair. Peripheral blood mononuclear cells (PBMCs) were collected from sporadic ALS (sALS) patients (n = 18) and healthy controls (n = 12) to perform RNA-sequencing experiments and differential expression analysis. Patients were stratified into groups with “high” and “low” levels of nSOD1. We obtained different gene expression patterns for high- and low-nSOD1 patients. Differentially expressed genes in high nSOD1 form a cluster similar to controls compared to the low-nSOD1 group. The pathways activated in high-nSOD1 patients are related to the upregulation of HSP70 molecular chaperones. We demonstrated that, in this condition, the DNA damage is reduced, even under oxidative stress conditions. Our findings highlight the importance of the nuclear localization of SOD1 as a protective mechanism in sALS patients.
Collapse
|
13
|
Kinscherf NA, Pehar M. Role and Therapeutic Potential of RAGE Signaling in Neurodegeneration. Curr Drug Targets 2022; 23:1191-1209. [PMID: 35702767 PMCID: PMC9589927 DOI: 10.2174/1389450123666220610171005] [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: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/29/2022] [Indexed: 01/03/2023]
Abstract
Activation of the receptor for advanced glycation end products (RAGE) has been shown to play an active role in the development of multiple neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis. Although originally identified as a receptor for advanced glycation end products, RAGE is a pattern recognition receptor able to bind multiple ligands. The final outcome of RAGE signaling is defined in a context and cell type specific manner and can exert both neurotoxic and neuroprotective functions. Contributing to the complexity of the RAGE signaling network, different RAGE isoforms with distinctive signaling capabilities have been described. Moreover, multiple RAGE ligands bind other receptors and RAGE antagonism can significantly affect their signaling. Here, we discuss the outcome of celltype specific RAGE signaling in neurodegenerative pathologies. In addition, we will review the different approaches that have been developed to target RAGE signaling and their therapeutic potential. A clear understanding of the outcome of RAGE signaling in a cell type- and disease-specific manner would contribute to advancing the development of new therapies targeting RAGE. The ability to counteract RAGE neurotoxic signaling while preserving its neuroprotective effects would be critical for the success of novel therapies targeting RAGE signaling.
Collapse
Affiliation(s)
- Noah Alexander Kinscherf
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Mariana Pehar
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI, USA
| |
Collapse
|
14
|
Pucci M, Aria F, Premoli M, Maccarinelli G, Mastinu A, Bonini S, Memo M, Uberti D, Abate G. Methylglyoxal affects cognitive behaviour and modulates RAGE and Presenilin-1 expression in hippocampus of aged mice. Food Chem Toxicol 2021; 158:112608. [PMID: 34656697 DOI: 10.1016/j.fct.2021.112608] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 12/12/2022]
Abstract
Methylglyoxal (MG), a potent glycotoxin that can be found in the diet, is one of the main precursors of Advanced glycation end products (AGEs). It is well known that modifications in lifestyle such as nutritional interventions can be of great value for preventing brain deterioration. This study aimed to evaluate in vivo how an oral MG treatment, that mimics a high MG dietary intake, could affect brain health. From our results, we demonstrated that MG administration affected working memory, and induced neuroinflammation and oxidative stress by modulating the Receptor for Advanced glycation end products (RAGE). The gene and protein expressions of RAGE were increased in the hippocampus of MG mice, an area where the activity of glyoxalase 1, one of the main enzymes involved in MG detoxification, was found reduced. Furthermore, at hippocampus level, MG mice showed increased expression of proinflammatory cytokines and increased activities of NADPH oxidase and catalase. MG administration also increased the gene and protein expressions of Presenilin-1, a subunit of the gamma-secretase protein complex linked to Alzheimer's disease. These findings suggest that high MG oral intake induces alteration directly in the brain and might establish an environment predisposing to AD-like pathological conditions.
Collapse
Affiliation(s)
- M Pucci
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - F Aria
- Department of Molecular and Translational Medicine, University of Brescia, Italy; Center for Neural Science, New York University, New York, United States
| | - M Premoli
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - G Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - A Mastinu
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - S Bonini
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - M Memo
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - D Uberti
- Department of Molecular and Translational Medicine, University of Brescia, Italy; Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - G Abate
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| |
Collapse
|
15
|
Gerou M, Hall B, Woof R, Allsop J, Kolb SJ, Meyer K, Shaw PJ, Allen SP. Amyotrophic lateral sclerosis alters the metabolic aging profile in patient derived fibroblasts. Neurobiol Aging 2021; 105:64-77. [PMID: 34044197 PMCID: PMC8346650 DOI: 10.1016/j.neurobiolaging.2021.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022]
Abstract
Aging is a major risk factor for neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). As metabolic alterations are a hallmark of aging and have previously been observed in ALS, it is important to examine the effect of aging in the context of ALS metabolic function. Here, using a newly established phenotypic metabolic approach, we examined the effect of aging on the metabolic profile of fibroblasts derived from ALS cases compared to controls. We found that ALS fibroblasts have an altered metabolic profile, which is influenced by age. In control cases, we found significant increases with age in NADH metabolism in the presence of several metabolites including lactic acid, trehalose, uridine and fructose, which was not recapitulated in ALS cases. Conversely, we found a reduction of NADH metabolism with age of biopsy, age of onset and age of death in the presence of glycogen in the ALS cohort. Furthermore, we found that NADH production correlated with disease progression rates in relation to a number of metabolites including inosine and α-ketoglutaric acid. Inosine or α-ketoglutaric acid supplementation in ALS fibroblasts was bioenergetically favourable. Overall, we found aging related defects in energy substrates that feed carbon into glycolysis at various points as well as the tricarboxylic acid (TCA) cycle in ALS fibroblasts, which was validated in induced neuronal progenitor cell derived iAstrocytes. Our results suggest that supplementing those pathways may protect against age related metabolic dysfunction in ALS.
Collapse
Affiliation(s)
- Margarita Gerou
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Benjamin Hall
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Ryan Woof
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Jessica Allsop
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Stephen J Kolb
- Department of Neurology, The Ohio State University Wexner Medical Centre, Columbus, OH, USA
| | - Kathrin Meyer
- Centre for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
| | - Pamela J Shaw
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Scott P Allen
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK.
| |
Collapse
|
16
|
Zglejc-Waszak K, Mukherjee K, Juranek JK. The cross-talk between RAGE and DIAPH1 in neurological complications of diabetes: A review. Eur J Neurosci 2021; 54:5982-5999. [PMID: 34449932 DOI: 10.1111/ejn.15433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 01/10/2023]
Abstract
Neuropathy, or dysfunction of peripheral nerve, is one of the most common neurological manifestation in patients with diabetes mellitus (DM). DM is typically associated with a hyperglycaemic milieu, which promotes non-enzymatic glycation of proteins. Proteins with advanced glycation are known to engage a cell-surface receptor called the receptor for advanced glycation end products (RAGE). Thus, it is reasonable to assume that RAGE and its associated molecule-mediated cellular signalling may contribute to DM-induced symmetrical axonal (length-dependent) neuropathy. Of particular interest is diaphanous related formin 1 (DIAPH1), a cytoskeletal organizing molecule, which interacts with the cytosolic domain of RAGE and whose dysfunction may precipitate axonopathy/neuropathy. Indeed, it has been demonstrated that both RAGE and DIAPH1 are expressed in the motor and sensory fibres of nerve harvested from DM animal models. Although the detailed molecular role of RAGE and DIAPH1 in diabetic neurological complications remains unclear, here we will discuss available evidence of their involvement in peripheral diabetic neuropathy. Specifically, we will discuss how a hyperglycaemic environment is not only likely to elevate advanced glycation end products (ligands of RAGE) and induce a pro-inflammatory environment but also alter signalling via RAGE and DIAPH1. Further, hyperglycaemia may regulate epigenetic mechanisms that interacts with RAGE signalling. We suggest the cumulative effect of hyperglycaemia on RAGE-DIAPH1-mediated signalling may be disruptive to axonal cytoskeletal organization and transport and is therefore likely to play a key role in pathogenesis of diabetic symmetrical axonal neuropathy.
Collapse
Affiliation(s)
- Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Virginia Tech Roanoke, Roanoke, Virginia, USA
| | - Judyta Karolina Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
17
|
MacLean M, Juranek J, Cuddapah S, López-Díez R, Ruiz HH, Hu J, Frye L, Li H, Gugger PF, Schmidt AM. Microglia RAGE exacerbates the progression of neurodegeneration within the SOD1 G93A murine model of amyotrophic lateral sclerosis in a sex-dependent manner. J Neuroinflammation 2021; 18:139. [PMID: 34130712 PMCID: PMC8207569 DOI: 10.1186/s12974-021-02191-2] [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: 02/10/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Background Burgeoning evidence highlights seminal roles for microglia in the pathogenesis of neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). The receptor for advanced glycation end products (RAGE) binds ligands relevant to ALS that accumulate in the diseased spinal cord and RAGE has been previously implicated in the progression of ALS pathology. Methods We generated a novel mouse model to temporally delete Ager from microglia in the murine SOD1G93A model of ALS. Microglia Ager deficient SOD1G93A mice and controls were examined for changes in survival, motor function, gliosis, motor neuron numbers, and transcriptomic analyses of lumbar spinal cord. Furthermore, we examined bulk-RNA-sequencing transcriptomic analyses of human ALS cervical spinal cord. Results Transcriptomic analysis of human cervical spinal cord reveals a range of AGER expression in ALS patients, which was negatively correlated with age at disease onset and death or tracheostomy. The degree of AGER expression related to differential expression of pathways involved in extracellular matrix, lipid metabolism, and intercellular communication. Microglia display increased RAGE immunoreactivity in the spinal cords of high AGER expressing patients and in the SOD1G93A murine model of ALS vs. respective controls. We demonstrate that microglia Ager deletion at the age of symptomatic onset, day 90, in SOD1G93A mice extends survival in male but not female mice. Critically, many of the pathways identified in human ALS patients that accompanied increased AGER expression were significantly ameliorated by microglia Ager deletion in male SOD1G93A mice. Conclusions Our results indicate that microglia RAGE disrupts communications with cell types including astrocytes and neurons, intercellular communication pathways that divert microglia from a homeostatic to an inflammatory and tissue-injurious program. In totality, microglia RAGE contributes to the progression of SOD1G93A murine pathology in male mice and may be relevant in human disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02191-2.
Collapse
Affiliation(s)
- Michael MacLean
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Judyta Juranek
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA.,Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Swetha Cuddapah
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Raquel López-Díez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Henry H Ruiz
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jiyuan Hu
- Division of Biostatistics, Department of Population Health and the Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Laura Frye
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Huilin Li
- Division of Biostatistics, Department of Population Health and the Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Paul F Gugger
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
18
|
Semchyshyn H. Is carbonyl/AGE/RAGE stress a hallmark of the brain aging? Pflugers Arch 2021; 473:723-734. [PMID: 33742308 DOI: 10.1007/s00424-021-02529-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
Recent studies have linked carbonyl stress to many physiological processes. Increase in the levels of carbonyl compounds, derived from both endogenous and exogenous sources, is believed to accompany normal age-related decline as well as different pathologies. Reactive carbonyl species (RCS) are capable of damaging biomolecules via their involvement in a net of nonspecific reactions. In the advanced stages of RCS metabolism, variety of poorly degraded adducts and crosslinks, collectively named advanced glycoxidation end products (AGEs), arises. They are accumulated in an age-dependent manner in different tissues and organs and can contribute to inflammatory processes. In particular, detrimental effects of the end products are realized via activation of the specific receptor for AGEs (RAGE) and RAGE-dependent inflammatory signaling cascade. Although it is unclear, whether carbonyl stress is causal for age-associated impairments or it results from age- and disease-related cell damages, increased levels of RCS and AGEs are tightly related to inflammaging, and therefore, attenuation of the RAGE signaling is suggested as an effective approach for the treatment of inflammation and age-related disorders. The question raised in this review is whether specific metabolism in the aging brain related to carbonyl/RCS/AGE/RAGE stress.
Collapse
Affiliation(s)
- Halyna Semchyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str, Ivano-Frankivsk, 76018, Ukraine.
| |
Collapse
|
19
|
Molecular insights into the therapeutic promise of targeting HMGB1 in depression. Pharmacol Rep 2020; 73:31-42. [PMID: 33015736 DOI: 10.1007/s43440-020-00163-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/09/2020] [Accepted: 09/19/2020] [Indexed: 12/17/2022]
Abstract
Depression is a common psychiatric disorder, the exact pathogenesis of which is still elusive. Studies have proposed that immunity disproportion and enhancement in proinflammatory cytokines might be linked with the development of depression. HMGB1 (High-mobility group box (1) protein has obtained more interest as an essential factor in inherent immune reactions and a regulating factor in various inflammation-related diseases. HMGB1 is a ubiquitous chromatin protein and is constitutively expressed in nucleated mammalian cells. HMGB1 is released by glial cells and neurons upon inflammasome activation and act as a pro-inflammatory cytokine. HMGB1 is a late mediator of inflammation and has been indicated as a major mediator in various neuroinflammatory diseases. Microglia, which is the brain immune cell, is stimulated by HMGB1 and released inflammatory mediators and induces chronic neurodegeneration in the CNS (central nervous system). In the current review, we aimed to investigate the role of HMGB1 in the pathogenesis of depression. The studies found that HMGB1 functions as proinflammatory cytokines primarily via binding receptors like RAGE (receptor for advanced glycation end product), TLR2 and TLR4 (Toll-like receptor 2 and 4). Further, HMGB1 added to the preparing impacts of stress-pretreatment and assumed a major function in neurodegenerative conditions through moderating neuroinflammation. Studies demonstrated that neuroinflammation played a major role in the development of depression. The patients of depression generally exhibited an elevated amount of proinflammatory cytokines in the serum, microglia activation and neuronal deficit in the CNS.
Collapse
|
20
|
Béland LC, Markovinovic A, Jakovac H, De Marchi F, Bilic E, Mazzini L, Kriz J, Munitic I. Immunity in amyotrophic lateral sclerosis: blurred lines between excessive inflammation and inefficient immune responses. Brain Commun 2020; 2:fcaa124. [PMID: 33134918 PMCID: PMC7585698 DOI: 10.1093/braincomms/fcaa124] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Despite wide genetic, environmental and clinical heterogeneity in amyotrophic lateral sclerosis, a rapidly fatal neurodegenerative disease targeting motoneurons, neuroinflammation is a common finding. It is marked by local glial activation, T cell infiltration and systemic immune system activation. The immune system has a prominent role in the pathogenesis of various chronic diseases, hence some of them, including some types of cancer, are successfully targeted by immunotherapeutic approaches. However, various anti-inflammatory or immunosuppressive therapies in amyotrophic lateral sclerosis have failed. This prompted increased scrutiny over the immune-mediated processes underlying amyotrophic lateral sclerosis. Perhaps the biggest conundrum is that amyotrophic lateral sclerosis pathogenesis exhibits features of three otherwise distinct immune dysfunctions-excessive inflammation, autoimmunity and inefficient immune responses. Epidemiological and genome-wide association studies show only minimal overlap between amyotrophic lateral sclerosis and autoimmune diseases, so excessive inflammation is usually thought to be secondary to protein aggregation, mitochondrial damage or other stresses. In contrast, several recently characterized amyotrophic lateral sclerosis-linked mutations, including those in TBK1, OPTN, CYLD and C9orf72, could lead to inefficient immune responses and/or damage pile-up, suggesting that an innate immunodeficiency may also be a trigger and/or modifier of this disease. In such cases, non-selective immunosuppression would further restrict neuroprotective immune responses. Here we discuss multiple layers of immune-mediated neuroprotection and neurotoxicity in amyotrophic lateral sclerosis. Particular focus is placed on individual patient mutations that directly or indirectly affect the immune system, and the mechanisms by which these mutations influence disease progression. The topic of immunity in amyotrophic lateral sclerosis is timely and relevant, because it is one of the few common and potentially malleable denominators in this heterogenous disease. Importantly, amyotrophic lateral sclerosis progression has recently been intricately linked to patient T cell and monocyte profiles, as well as polymorphisms in cytokine and chemokine receptors. For this reason, precise patient stratification based on immunophenotyping will be crucial for efficient therapies.
Collapse
Affiliation(s)
| | - Andrea Markovinovic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Fabiola De Marchi
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Ervina Bilic
- Department of Neurology, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Letizia Mazzini
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Jasna Kriz
- CERVO Research Centre, Laval University, Quebec City, Quebec G1J 2G3, Canada
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
21
|
Liu L, Killoy KM, Vargas MR, Yamamoto Y, Pehar M. Effects of RAGE inhibition on the progression of the disease in hSOD1 G93A ALS mice. Pharmacol Res Perspect 2020; 8:e00636. [PMID: 32776498 PMCID: PMC7415959 DOI: 10.1002/prp2.636] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Astrocytes play a key role in the progression of amyotrophic lateral sclerosis (ALS) by actively inducing the degeneration of motor neurons. Motor neurons isolated from receptor for advanced glycation end products (RAGE)-knockout mice are resistant to the neurotoxic signal derived from ALS-astrocytes. Here, we confirmed that in a co-culture model, the neuronal death induced by astrocytes over-expressing the ALS-linked mutant hSOD1G93A is prevented by the addition of the RAGE inhibitors FPS-ZM1 or RAP. These inhibitors also prevented the motor neuron death induced by spinal cord extracts from symptomatic hSOD1G93A mice. To evaluate the relevance of this neurotoxic mechanism in ALS pathology, we assessed the therapeutic potential of FPS-ZM1 in hSOD1G93A mice. FPS-ZM1 treatment significantly improved hind-limb grip strength in hSOD1G93A mice during the progression of the disease, reduced the expression of atrophy markers in the gastrocnemius muscle, improved the survival of large motor neurons, and reduced gliosis in the ventral horn of the spinal cord. However, we did not observe a statistically significant effect of the drug in symptoms onset nor in the survival of hSOD1G93A mice. Maintenance of hind-limb grip strength was also observed in hSOD1G93A mice with RAGE haploinsufficiency [hSOD1G93A ;RAGE(+/-)], further supporting the beneficial effect of RAGE inhibition on muscle function. However, no benefits were observed after complete RAGE ablation. Moreover, genetic RAGE ablation significantly shortened the median survival of hSOD1G93A mice. These results indicate that the advance of new therapies targeting RAGE in ALS demands a better understanding of its physiological role in a cell type/tissue-specific context.
Collapse
Affiliation(s)
- Liping Liu
- Biomedical Sciences Training ProgramDepartment of Pharmacology and Experimental TherapeuticsMedical University of South CarolinaCharlestonSCUSA
| | - Kelby M. Killoy
- Biomedical Sciences Training ProgramDepartment of Pharmacology and Experimental TherapeuticsMedical University of South CarolinaCharlestonSCUSA
| | | | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular BiologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | - Mariana Pehar
- Division of Geriatrics and GerontologyDepartment of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- Geriatric Research Education Clinical CenterVeterans Affairs Medical CenterMadisonWIUSA
| |
Collapse
|
22
|
Lee JD, McDonald TS, Fung JNT, Woodruff TM. Absence of Receptor for Advanced Glycation End Product (RAGE) Reduces Inflammation and Extends Survival in the hSOD1 G93A Mouse Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2020; 57:4143-4155. [PMID: 32676989 DOI: 10.1007/s12035-020-02019-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing motor neuron degenerative disease that is without effective treatment. The receptor for advanced glycation end products (RAGE) is a major component of the innate immune system that has been implicated in ALS pathogenesis. However, the contribution of RAGE signalling to the neuroinflammation that underlies ALS neurodegeneration remains unknown. The present study therefore generated SOD1G93A mice lacking RAGE and compared them with SOD1G93A transgenic ALS mice in respect to disease progression (i.e. body weight, survival and muscle strength), neuroinflammation and denervation markers in the spinal cord and tibialis anterior muscle. We found that complete absence of RAGE signalling exerted a protective effect on SOD1G93A pathology, slowing disease progression and significantly extending survival by ~ 3 weeks and improving motor function (rotarod and grip strength). This was associated with reduced microgliosis, cytokines, innate immune factors (complement, TLRs, inflammasomes), and oxidative stress in the spinal cord, and a reduction of denervation markers in the tibialis anterior muscle. We also documented that RAGE mRNA expression was significantly increased in the spinal cord and muscles of preclinical SOD1 and TDP43 models of ALS, supporting a widespread involvement for RAGE in ALS pathology. In summary, our results indicate that RAGE signalling drives neuroinflammation and contributes to neurodegeneration in ALS and highlights RAGE as a potential immune therapeutic target for ALS.
Collapse
Affiliation(s)
- John D Lee
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Tanya S McDonald
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Jenny N T Fung
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia. .,Queensland Brain Institute, the University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
23
|
Damasiewicz-Bodzek A, Łabuz-Roszak B, Kumaszka B, Tyrpień-Golder K. Carboxymethyllysine and carboxyethyllysine in multiple sclerosis patients. Arch Med Sci 2020; 20:736-742. [PMID: 39050184 PMCID: PMC11264072 DOI: 10.5114/aoms.2020.95654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/19/2020] [Indexed: 07/27/2024] Open
Abstract
Introduction Advanced glycation end-products (AGE) are involved in the pathogenesis of many diseases, including neurodegenerative diseases such as multiple sclerosis (MS). The aim of the study was to evaluate the intensity of the protein glycation process in patients with multiple sclerosis and its possible involvement in disease activity. Material and methods The study group (n = 45) consisted of patients suffering from MS, and the control group (n = 31) consisted of healthy adults. Concentrations of selected glycation markers such as carboxymethyllysine (CML) and carboxyethyllysine (CEL) in sera of patients with MS and healthy volunteers were determined by enzyme-linked immunosorbent assay (ELISA). Results Serum CML and CEL concentrations in patients with MS were higher than in healthy volunteers but only for CML the difference was statistically significant. CML concentrations positively correlated with CEL concentrations only in the healthy persons. In MS patients the serum CML and CEL concentrations did not differ significantly depending on the duration of the disease and depending on the EDSS (Expanded Disability Status Scale) score. Conclusions Multiple sclerosis is accompanied by an intensification of protein glycation processes, especially within the pathways leading to the formation of carboxymethyllysine. The duration of the disease and the degree of motor impairment do not appear to affect the progression of the glycation processes. However, the disease process associated with multiple sclerosis may affect the relationship between CML and CEL concentrations.
Collapse
Affiliation(s)
| | - Beata Łabuz-Roszak
- Department of Basic Medical Sciences, School of Health Sciences in Bytom, Medical University of Silesia in Katowice, Poland
- Department of Neurology, Provincial Specialist Hospital, Opole, Poland
| | | | - Krystyna Tyrpień-Golder
- Department of Chemistry, School of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| |
Collapse
|
24
|
Paudel YN, Angelopoulou E, Piperi C, Othman I, Shaikh MF. Implication of HMGB1 signaling pathways in Amyotrophic lateral sclerosis (ALS): From molecular mechanisms to pre-clinical results. Pharmacol Res 2020; 156:104792. [PMID: 32278047 DOI: 10.1016/j.phrs.2020.104792] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 02/14/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and rapidly progressing neurodegenerative disorder with no effective disease-modifying treatment up to date. The underlying molecular mechanisms of ALS are not yet completely understood. However, the critical role of the innate immune system and neuroinflammation in ALS pathogenesis has gained increased attention. High mobility group box 1 (HMGB1) is a typical damage-associated molecular pattern (DAMP) molecule, acting as a pro-inflammatory cytokine mainly through activation of its principal receptors, the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) which are crucial components of the innate immune system. HMGB1 is an endogenous ligand for both RAGE and TLR4 that mediate its biological effects. Herein, on the ground of pre-clinical findings we unravel the underlying mechanisms behind the plausible contribution of HMGB1 and its receptors (RAGE and TLR4) in the ALS pathogenesis. Furthermore, we provide an account of the therapeutic outcomes associated with inhibition/blocking of HMGB1 receptor signalling in preventing motor neuron's death and delaying disease progression in ALS experimental models. There is strong evidence that HMGB1, RAGE and TLR4 signaling axes might present potential targets against ALS, opening a novel headway in ALS research that could plausibly bridge the current treatment gap.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
25
|
Allen SP, Hall B, Woof R, Francis L, Gatto N, Shaw AC, Myszczynska M, Hemingway J, Coldicott I, Willcock A, Job L, Hughes RM, Boschian C, Bayatti N, Heath PR, Bandmann O, Mortiboys H, Ferraiuolo L, Shaw PJ. C9orf72 expansion within astrocytes reduces metabolic flexibility in amyotrophic lateral sclerosis. Brain 2019; 142:3771-3790. [PMID: 31647549 PMCID: PMC6906594 DOI: 10.1093/brain/awz302] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/25/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022] Open
Abstract
It is important to understand how the disease process affects the metabolic pathways in amyotrophic lateral sclerosis and whether these pathways can be manipulated to ameliorate disease progression. To analyse the basis of the metabolic defect in amyotrophic lateral sclerosis we used a phenotypic metabolic profiling approach. Using fibroblasts and reprogrammed induced astrocytes from C9orf72 and sporadic amyotrophic lateral sclerosis cases we measured the production rate of reduced nicotinamide adenine dinucleotides (NADH) from 91 potential energy substrates simultaneously. Our screening approach identified that C9orf72 and sporadic amyotrophic lateral sclerosis induced astrocytes have distinct metabolic profiles compared to controls and displayed a loss of metabolic flexibility that was not observed in fibroblast models. This loss of metabolic flexibility, involving defects in adenosine, fructose and glycogen metabolism, as well as disruptions in the membrane transport of mitochondrial specific energy substrates, contributed to increased starvation induced toxicity in C9orf72 induced astrocytes. A reduction in glycogen metabolism was attributed to loss of glycogen phosphorylase and phosphoglucomutase at the protein level in both C9orf72 induced astrocytes and induced neurons. In addition, we found alterations in the levels of fructose metabolism enzymes and a reduction in the methylglyoxal removal enzyme GLO1 in both C9orf72 and sporadic models of disease. Our data show that metabolic flexibility is important in the CNS in times of bioenergetic stress.
Collapse
Affiliation(s)
- Scott P Allen
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Benjamin Hall
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Ryan Woof
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Francis
- The Living Systems Institute, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Noemi Gatto
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Allan C Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Monika Myszczynska
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Jordan Hemingway
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Ian Coldicott
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Amelia Willcock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Lucy Job
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Rachel M Hughes
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Camilla Boschian
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Nadhim Bayatti
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| |
Collapse
|
26
|
Hajizadeh-Sharafabad F, Sahebkar A, Zabetian-Targhi F, Maleki V. The impact of resveratrol on toxicity and related complications of advanced glycation end products: A systematic review. Biofactors 2019; 45:651-665. [PMID: 31185146 DOI: 10.1002/biof.1531] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022]
Abstract
Accumulation of advanced glycation end products (AGEs) promotes the generation of free radicals, which leads to chronic oxidative stress predisposing to chronic oxidative stress, inflammation, and related diseases. This systematic review aimed to determine the effect of resveratrol (RSV) on AGE-induced toxicity and its deleterious consequences. A comprehensive search was performed through literature were published until December 2018 using relevant keywords. The databases that were used for the search were PubMed, Scopus, Embase, ProQuest, and Google Scholar. A total of 29 eligible studies were found and included in the review for the analysis. Except one, all studies showed suppressing effects for RSV on the production of AGEs or receptor for advanced glycation end products (RAGE) and its detrimental consequences including oxidative stress, inflammatory response, cellular immune reactions, insulin response, and atherosclerosis. RSV exerts its effects through influencing RAGE, nuclear factor kappa B (NF-κB), peroxisome proliferator-activated receptor (PPAR) γ, and transforming growth factor (TGF)-β activities. This review suggests that RSV has got potential to decrease AGEs toxicity and inhibit the AGE-induced complications. More clinical trials are suggested to evaluate the beneficial effect of RSV on AGEs in chronic metabolic diseases.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Antioxidants/pharmacology
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Gene Expression Regulation
- Glycation End Products, Advanced/antagonists & inhibitors
- Glycation End Products, Advanced/genetics
- Glycation End Products, Advanced/metabolism
- Glycation End Products, Advanced/toxicity
- Humans
- Inflammation
- Mitogen-Activated Protein Kinases/genetics
- Mitogen-Activated Protein Kinases/metabolism
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Oxidative Stress
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Pyruvaldehyde/metabolism
- Resveratrol/pharmacology
- Signal Transduction
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
Collapse
Affiliation(s)
- Fatemeh Hajizadeh-Sharafabad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fateme Zabetian-Targhi
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Vahid Maleki
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Eleazu C, Omar N, Lim OZ, Yeoh BS, Nik Hussain NH, Mohamed M. Corrigendum: Obesity and Comorbidity: Could Simultaneous Targeting of esRAGE and sRAGE Be the Panacea? Front Physiol 2019; 10:1017. [PMID: 31417435 PMCID: PMC6692561 DOI: 10.3389/fphys.2019.01017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/23/2019] [Indexed: 02/05/2023] Open
Abstract
[This corrects the article DOI: 10.3389/fphys.2019.00787.].
Collapse
Affiliation(s)
- Chinedum Eleazu
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Chemistry/Biochemistry/Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Norsuhana Omar
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Oon Zhi Lim
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Boon Seng Yeoh
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
28
|
Eleazu C, Omar N, Lim OZ, Yeoh BS, Nik Hussain NH, Mohamed M. Obesity and Comorbidity: Could Simultaneous Targeting of esRAGE and sRAGE Be the Panacea? Front Physiol 2019; 10:787. [PMID: 31293451 PMCID: PMC6603218 DOI: 10.3389/fphys.2019.00787] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/04/2019] [Indexed: 02/05/2023] Open
Abstract
Obesity, a chronic multifaceted disease, predisposes its patients to increased risk of metabolic disorders such as: diabetes mellitus, cardiovascular diseases, dyslipidemia, etc. Recent studies reported it to be amongst the leading causes of deaths in the world. Although several treatment options for obesity abound, many of them have not been able to successfully reverse the existing obesity and metabolic dysregulation. This has therefore warranted the need for either alternative therapies or diversification of the treatment approach for obesity and its comorbidity. When the receptor for advanced glycation end products (RAGE) interacts with its ligand, RAGE-ligand activates an inflammatory signaling cascade, that leads to the activation of nuclear factor kappa B (NF-κB) and transcription of inflammatory cytokines. This action has been associated with the development of obesity and its mediated metabolic dysregulation. In view of the increasing prevalence of obesity globally and the potential threat it places on life expectancy, this article reviewed the promising potentials of targeting endogenous secretory receptor for advanced glycation end products/soluble receptors for advanced glycation end products signaling as a treatment approach for obesity. We carried out a literature search in several electronic data bases such as: Pubmed, Pubmed Central, Google, Google Scholar, Scopus, and Medline from 1980 to 2019 to acquire the status of information concerning this. The article suggests the need for the development of an esRAGE/sRAGE targeted pharmacotherapy as a treatment approach for obesity and its comorbidity.
Collapse
Affiliation(s)
- Chinedum Eleazu
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Chemistry/Biochemistry/Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Norsuhana Omar
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Oon Zhi Lim
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Boon Seng Yeoh
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
29
|
MacLean M, Derk J, Ruiz HH, Juranek JK, Ramasamy R, Schmidt AM. The Receptor for Advanced Glycation End Products (RAGE) and DIAPH1: Implications for vascular and neuroinflammatory dysfunction in disorders of the central nervous system. Neurochem Int 2019; 126:154-164. [PMID: 30902646 PMCID: PMC10976457 DOI: 10.1016/j.neuint.2019.03.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 12/11/2022]
Abstract
The Receptor for Advanced Glycation End Products (RAGE) is expressed by multiple cell types in the brain and spinal cord that are linked to the pathogenesis of neurovascular and neurodegenerative disorders, including neurons, glia (microglia and astrocytes) and vascular cells (endothelial cells, smooth muscle cells and pericytes). Mounting structural and functional evidence implicates the interaction of the RAGE cytoplasmic domain with the formin, Diaphanous1 (DIAPH1), as the key cytoplasmic hub for RAGE ligand-mediated activation of cellular signaling. In aging and diabetes, the ligands of the receptor abound, both in the central nervous system (CNS) and in the periphery. Such accumulation of RAGE ligands triggers multiple downstream events, including upregulation of RAGE itself. Once set in motion, cell intrinsic and cell-cell communication mechanisms, at least in part via RAGE, trigger dysfunction in the CNS. A key outcome of endothelial dysfunction is reduction in cerebral blood flow and increased permeability of the blood brain barrier, conditions that facilitate entry of activated leukocytes into the CNS, thereby amplifying primary nodes of CNS cellular stress. This contribution details a review of the ligands of RAGE, the mechanisms and consequences of RAGE signal transduction, and cites multiple examples of published work in which RAGE contributes to the pathogenesis of neurovascular perturbation. Insights into potential therapeutic modalities targeting the RAGE signal transduction axis for disorders of CNS vascular dysfunction and neurodegeneration are also discussed.
Collapse
Affiliation(s)
- Michael MacLean
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Julia Derk
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Henry H Ruiz
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Judyta K Juranek
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
30
|
Festoff BW, Citron BA. Thrombin and the Coag-Inflammatory Nexus in Neurotrauma, ALS, and Other Neurodegenerative Disorders. Front Neurol 2019; 10:59. [PMID: 30804878 PMCID: PMC6371052 DOI: 10.3389/fneur.2019.00059] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
This review details our current understanding of thrombin signaling in neurodegeneration, with a focus on amyotrophic lateral sclerosis (ALS, Lou Gehrig's disease) as well as future directions to be pursued. The key factors are multifunctional and involved in regulatory pathways, namely innate immune and the coagulation cascade activation, that are essential for normal nervous system function and health. These two major host defense systems have a long history in evolution and include elements and regulators of the coagulation pathway that have significant impacts on both the peripheral and central nervous system in health and disease. The clotting cascade responds to a variety of insults to the CNS including injury and infection. The blood brain barrier is affected by these responses and its compromise also contributes to these detrimental effects. Important molecules in signaling that contribute to or protect against neurodegeneration include thrombin, thrombomodulin (TM), protease activated receptor 1 (PAR1), damage associated molecular patterns (DAMPs), such as high mobility group box protein 1 (HMGB1) and those released from mitochondria (mtDAMPs). Each of these molecules are entangled in choices dependent upon specific signaling pathways in play. For example, the particular cleavage of PAR1 by thrombin vs. activated protein C (APC) will have downstream effects through coupled factors to result in toxicity or neuroprotection. Furthermore, numerous interactions influence these choices such as the interplay between HMGB1, thrombin, and TM. Our hope is that improved understanding of the ways that components of the coagulation cascade affect innate immune inflammatory responses and influence the course of neurodegeneration, especially after injury, will lead to effective therapeutic approaches for ALS, traumatic brain injury, and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Barry W Festoff
- pHLOGISTIX LLC, Fairway, KS, United States.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Bruce A Citron
- Laboratory of Molecular Biology Research & Development, VA New Jersey Health Care System, East Orange, NJ, United States.,Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
31
|
Wang B, Lian YJ, Dong X, Peng W, Liu LL, Su WJ, Gong H, Zhang T, Jiang CL, Li JS, Wang YX. Glycyrrhizic acid ameliorates the kynurenine pathway in association with its antidepressant effect. Behav Brain Res 2018; 353:250-257. [PMID: 29366745 DOI: 10.1016/j.bbr.2018.01.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 12/20/2022]
Abstract
Our previous study implied the role of central high mobility group box 1 (HMGB1) in lipopolysaccharide (LPS)-induced depressive-like behaviors that could partially abrogate by glycyrrhizic acid (GZA). Here, we considered the potential mechanism underlying GZA ameliorating chronic stress-induced depression both in vivo and in vitro. Depression model was established with the 4-week chronic unpredictable mild stress (CUMS) mice. Sucrose preference test, tail suspension test and open field test were performed to reflect depressive-like behaviors. Enzyme activity of indoleamine-2,3-dioxygenase (IDO) was recorded with the ratio of kynurenine (KYN) / tryptophan (Trp). Transcription of gene was evaluated by RT-PCR. Along with depressive-like behaviors, IDO, the rate-limiting enzyme of the kynurenine pathway (KP), was upregulated at the level of mRNA expression, and enzyme activity was also elevated in stressed hippocampi and LPS/HMGB1-treated hippocampus slices. Treatment of mice with GZA, the inhibitor of HMGB1, prevented the activated enzymes in KP and the development of depressive-like behaviors. These experiments demonstrate that GZA may restrain HMGB1 thus improving chronic stress-induced depressive behavior through regulating KP.
Collapse
Affiliation(s)
- Bo Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China
| | - Yong-Jie Lian
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China; Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xin Dong
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wei Peng
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China; Department of Psychiatry, The 92nd Hospital of PLA, Nanping 353000, China
| | - Lin-Lin Liu
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China; Department of Nursing, The 474th Hospital of PLA, Urumqi 830012, China
| | - Wen-Jun Su
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China
| | - Hong Gong
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China
| | - Ting Zhang
- Department of Navy Medicine, Second Military Medical University, Shanghai 200433, China
| | - Chun-Lei Jiang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China
| | - Jia-Si Li
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | - Yun-Xia Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
32
|
Zhu Q, Smith EA. Diaphanous-1 affects the nanoscale clustering and lateral diffusion of receptor for advanced glycation endproducts (RAGE). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:43-49. [PMID: 30401627 DOI: 10.1016/j.bbamem.2018.10.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022]
Abstract
The interactions between the cytoplasmic protein diaphanous-1 (Diaph1) and the receptor for advanced glycation endproducts (RAGE) drive the negative consequences of RAGE signaling in several disease processes. Reported in this work is how Diaph1 affects the nanoscale clustering and diffusion of RAGE measured using super-resolution stochastic optical reconstruction microscopy (STORM) and single particle tracking (SPT). Altering the Diaph1 binding site has a different impact on RAGE diffusion compared to when Diaph1 expression is reduced in HEK293 cells. In cells with reduced Diaph1 expression (RAGE-Diaph1-/-), the average RAGE diffusion coefficient is increased by 35%. RAGE diffusion is known to be influenced by the dynamics of the actin cytoskeleton. Actin labeling shows that a reduced Diaph1 expression leads to cells with reduced filopodia density and length. In contrast, when two RAGE amino acids that interact with Diaph1 are mutated (RAGERQ/AA), the average RAGE diffusion coefficient is decreased by 16%. Since RAGE diffusion is slowed when the interaction between Diaph1 and RAGE is disrupted, the interaction of the two proteins results in faster RAGE diffusion. In both RAGERQ/AA and RAGE-Diaph1-/- cells the number and size of RAGE clusters are decreased compared to cells expressing RAGE and native concentrations of Diaph1. This work shows that Diaph1 has a role in affecting RAGE clusters and diffusion.
Collapse
Affiliation(s)
- Qiaochu Zhu
- Department of Chemistry, Iowa State University, Ames, IA 50011, United States
| | - Emily A Smith
- Department of Chemistry, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
33
|
Brambilla L, Martorana F, Guidotti G, Rossi D. Dysregulation of Astrocytic HMGB1 Signaling in Amyotrophic Lateral Sclerosis. Front Neurosci 2018; 12:622. [PMID: 30210286 PMCID: PMC6123379 DOI: 10.3389/fnins.2018.00622] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
Astrocytes have emerged as critical elements for the maintenance and function of the central nervous system. The expression on their cell membrane of RAGE and TLR4 receptors makes astrocytes susceptible to High-mobility group box 1 (HMGB1), a nuclear protein typically released in the extracellular milieu by living cells experiencing physiological stress conditions or by damaged cells. Here, we show that the interaction of HMGB1 with normal spinal cord astrocytes induces the astrocytic production of neurotrophic factors, particularly brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF). Multiple investigations suggest a role for HMGB1 in amyotrophic lateral sclerosis (ALS). Yet, no mechanistic information on the implication of HMGB1 signaling in this disorder is currently available. We demonstrate that non-transgenic and transgenic SOD1WT spinal motor neurons exhibit only a basal nucleus-to-cytoplasm shuttling of the HMGB1 protein. Conversely, in SOD1G93A ALS mouse spinal cords, HMGB1 significantly translocates from the nucleus to the cytoplasm of motor neurons, thereby suggesting that it may be eventually released in the extracellular environment during the progression of the disease. We postulate that extracellular HMGB1 can paracrinally interact with the neighboring astrocytes in an attempt to counteract the neurodegenerative process. Yet, at variance with normal cells, SOD1G93A-expressing astrocytes show impaired capacity to raise BDNF and GDNF levels upon HMGB1 stimulation. Our data suggest that HMGB1 have a potential to promote neuroprotective actions by healthy astrocytes. However, this neurotrophic response is disrupted in ALS astrocytes. This indicates that diseased astroglial cells may exacerbate motor neuron degeneration in ALS because of the loss of their neurosupportive functions.
Collapse
Affiliation(s)
- Liliana Brambilla
- Laboratory for Research on Neurodegenerative Disorders, IRCCS Istituti Clinici Scientifici Maugeri (ICS Maugeri), Pavia, Italy
| | - Francesca Martorana
- Laboratory for Research on Neurodegenerative Disorders, IRCCS Istituti Clinici Scientifici Maugeri (ICS Maugeri), Pavia, Italy
| | - Giulia Guidotti
- Laboratory for Research on Neurodegenerative Disorders, IRCCS Istituti Clinici Scientifici Maugeri (ICS Maugeri), Pavia, Italy
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, IRCCS Istituti Clinici Scientifici Maugeri (ICS Maugeri), Pavia, Italy
| |
Collapse
|
34
|
Wang B, Lian YJ, Su WJ, Peng W, Dong X, Liu LL, Gong H, Zhang T, Jiang CL, Wang YX. HMGB1 mediates depressive behavior induced by chronic stress through activating the kynurenine pathway. Brain Behav Immun 2018; 72:51-60. [PMID: 29195782 DOI: 10.1016/j.bbi.2017.11.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022] Open
Abstract
Our previous study has reported that the proactive secretion and role of central high mobility group box 1 (HMGB1) in lipopolysaccharide-induced depressive behavior. Here, the potential mechanism of HMGB1 mediating chronic-stress-induced depression through the kynurenine pathway (KP) was further explored both in vivo and in vitro. Depression model was established with the 4-week chronic unpredictable mild stress (CUMS). Sucrose preference and Barnes maze test were performed to reflect depressive behaviors. The ratio of kynurenine (KYN)/tryptophan (Trp) represented the enzyme activity of indoleamine-2,3-dioxygenase (IDO). Gene transcription and protein expression were assayed by real-time RT-PCR and western-blot or ELISA kit respectively. Along with depressive behaviors, HMGB1 concentrations in the hippocampus and serum substantially increased post 4-week CUMS exposure. Concurrent with the upregulated HMGB1 protein, the regulator of translocation of HMGB1, sirtuin 1 (SIRT1) concentration in the hippocampus remarkably increased. In addition to HMGB1 and SIRT1, IDO, the rate limiting enzyme of KP, was upregulated at the level of mRNA expression and enzyme activity in stressed hippocampi and LPS/HMGB1-treated hippocampal slices. The gene transcription of kynurenine monooxygenase (KMO) and kynureninase (KYNU) in the downstream of KP also increased both in vivo and in vitro. Mice treated with ethyl pyruvate (EP), the inhibitor of HMGB1 releasing, were observed with lower tendency of developing depressive behaviors and reduced activation of enzymes in KP. All of these experiments demonstrate that the role of HMGB1 on the induction of depressive behavior is mediated by KP activation.
Collapse
Affiliation(s)
- Bo Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Yong-Jie Lian
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Wen-Jun Su
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Wei Peng
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China; Department of Psychiatry, The 92nd Hospital of PLA, Nanping 353000, PR China
| | - Xin Dong
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Lin-Lin Liu
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China; Department of Nursing, The 474th Hospital of PLA, Urumqi 830012, PR China
| | - Hong Gong
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Ting Zhang
- Department of Navy Medicine, Second Military Medical University, Shanghai 200433, PR China
| | - Chun-Lei Jiang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Yun-Xia Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China.
| |
Collapse
|
35
|
Kim MJ, Vargas MR, Harlan BA, Killoy KM, Ball LE, Comte-Walters S, Gooz M, Yamamoto Y, Beckman JS, Barbeito L, Pehar M. Nitration and Glycation Turn Mature NGF into a Toxic Factor for Motor Neurons: A Role for p75 NTR and RAGE Signaling in ALS. Antioxid Redox Signal 2018; 28:1587-1602. [PMID: 28537420 PMCID: PMC5962334 DOI: 10.1089/ars.2016.6966] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Glycating stress can occur together with oxidative stress during neurodegeneration and contribute to the pathogenic mechanism. Nerve growth factor (NGF) accumulates in several neurodegenerative diseases. Besides promoting survival, NGF can paradoxically induce cell death by signaling through the p75 neurotrophin receptor (p75NTR). The ability of NGF to induce cell death is increased by nitration of its tyrosine residues under conditions associated with increased peroxynitrite formation. AIMS Here we investigated whether glycation also changes the ability of NGF to induce cell death and assessed the ability of post-translational modified NGF to signal through the receptor for advanced glycation end products (RAGEs). We also explored the potential role of RAGE-p75NTR interaction in the motor neuron death occurring in amyotrophic lateral sclerosis (ALS) models. RESULTS Glycation promoted NGF oligomerization and ultimately allowed the modified neurotrophin to signal through RAGE and p75NTR to induce motor neuron death at low physiological concentrations. A similar mechanism was observed for nitrated NGF. We provide evidence for the interaction of RAGE with p75NTR at the cell surface. Moreover, we observed that post-translational modified NGF was present in the spinal cord of an ALS mouse model. In addition, NGF signaling through RAGE and p75NTR was involved in astrocyte-mediated motor neuron toxicity, a pathogenic feature of ALS. INNOVATION Oxidative modifications occurring under stress conditions can enhance the ability of mature NGF to induce neuronal death at physiologically relevant concentrations, and RAGE is a new p75NTR coreceptor contributing to this pathway. CONCLUSION Our results indicate that NGF-RAGE/p75NTR signaling may be a therapeutic target in ALS. Antioxid. Redox Signal. 28, 1587-1602.
Collapse
Affiliation(s)
- Mi Jin Kim
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Marcelo R Vargas
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Benjamin A Harlan
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Kelby M Killoy
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Lauren E Ball
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Susana Comte-Walters
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| | - Monika Gooz
- 2 Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , Charleston, South Carolina
| | - Yasuhiko Yamamoto
- 3 Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences , Kanazawa, Japan
| | - Joseph S Beckman
- 4 Department of Biochemistry and Biophysics, Linus Pauling Institute, Environmental Health Sciences Center, Oregon State University , Corvallis, Oregon
| | - Luis Barbeito
- 5 Institut Pasteur de Montevideo , Montevideo, Uruguay
| | - Mariana Pehar
- 1 Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina , Charleston, South Carolina
| |
Collapse
|
36
|
Liu L, Zhao Z, Lu L, Liu J, Wu X, Sun J, Wei Y, Dong J. The role of HMGB1 in neuroinflammation and tissue repair: A potential therapeutic target for depression? TRADITIONAL MEDICINE AND MODERN MEDICINE 2018. [DOI: 10.1142/s2575900018300035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
High mobility group protein box 1 (HMGB1), a sophisticated danger signal with pleiotropic functions, has been proved to function as a pro-inflammatory cytokine. In the central neural system (CNS), HMGB1 can stimulate microglia, the immune cell in the CNS, to release inflammatory factors and to cause chronic neurodegeneration. The evidence showed that HMGB1 can act as a pro-inflammatory cytokine mainly through its receptors like advanced glycation end product (RAGE), Toll-like 4 (TLR4), and so on. Moreover, HMGB1 contributed to the priming effects of stress-pretreatment and played a key role in neurodegeneration diseases via mediating neuroinflammation. However, the evidence also showed that HMGB1 played a role in tissue repair, with the ability to promote cell migration and proliferation, to induce the differentiation of mesenchymal stem cells (MSCs), and to regenerate spinal cord. These pleiotropic functions of HMGB1 make it possible to play a role from cell death to new life. Depression is a chronic, severe, and often life-threatening disease accompanied with impaired neurogenesis. The evidence showed that neuroinflammation played a key role in the process of depression. Depressive patients often showed a high expression of inflammatory cytokines in the blood and an activation of microglia in the brain. Meanwhile, they also showed a neuron deficit in the brain. Though they lack direct evidence linking HMGB1 with depression, the ability of HMGB1 that can function from neuroinflammation to tissue repair makes HMGB1 a promising therapeutic target of depression.
Collapse
Affiliation(s)
- Lumei Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, P. R. China
| | - Zhengxiao Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, P. R. China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, P. R. China
| | - Jiaqi Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, P. R. China
| | - Xiao Wu
- The Respiratory Department of the TCM Hospital of Jiangsu, Nanjing 210000, P. R. China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, P. R. China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, P. R. China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, P. R. China
| |
Collapse
|
37
|
Derk J, MacLean M, Juranek J, Schmidt AM. The Receptor for Advanced Glycation Endproducts (RAGE) and Mediation of Inflammatory Neurodegeneration. JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2018; 8:421. [PMID: 30560011 PMCID: PMC6293973 DOI: 10.4172/2161-0460.1000421] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Julia Derk
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, 550 First Avenue, Smilow 906, New York, NY, 10016, USA
| | - Michael MacLean
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, 550 First Avenue, Smilow 906, New York, NY, 10016, USA
| | - Judyta Juranek
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, 550 First Avenue, Smilow 906, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, 550 First Avenue, Smilow 906, New York, NY, 10016, USA
| |
Collapse
|
38
|
VanPatten S, Al-Abed Y. High Mobility Group Box-1 (HMGb1): Current Wisdom and Advancement as a Potential Drug Target. J Med Chem 2018; 61:5093-5107. [PMID: 29268019 DOI: 10.1021/acs.jmedchem.7b01136] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
High mobility group box-1 (HMGb1) protein, a nuclear non-histone protein that is released or secreted from the cell in response to damage or stress, is a sentinel for the immune system that plays a critical role in cell survival/death pathways. This review highlights key features of the endogenous danger-associated molecular pattern (DAMP) protein, HMGb1 in the innate inflammatory response along with various cofactors and receptors that regulate its downstream effects. The evidence demonstrating increased levels of HMGb1 in human inflammatory diseases and conditions is presented, along with a summary of current small molecule or peptide-like antagonists proven to specifically target HMGb1. Additionally, we delineate the measures needed toward validating this protein as a clinically relevant biomarker or bioindicator and as a relevant drug target.
Collapse
Affiliation(s)
- Sonya VanPatten
- Center for Molecular Innovation , The Feinstein Institute for Medical Research , 350 Community Drive , Manhasset , New York 11030 , United States
| | - Yousef Al-Abed
- Center for Molecular Innovation , The Feinstein Institute for Medical Research , 350 Community Drive , Manhasset , New York 11030 , United States
| |
Collapse
|
39
|
Bulc M, Palus K, Całka J, Zielonka Ł. Changes in Immunoreactivity of Sensory Substances within the Enteric Nervous System of the Porcine Stomach during Experimentally Induced Diabetes. J Diabetes Res 2018; 2018:4735659. [PMID: 30140706 PMCID: PMC6081574 DOI: 10.1155/2018/4735659] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/30/2018] [Accepted: 05/15/2018] [Indexed: 01/28/2023] Open
Abstract
One of the most frequently reported disorders associated with diabetes is gastrointestinal (GI) disturbance. Although pathogenesis of these complications is multifactorial, the complicity of the enteric nervous system (ENS) in this respect has significant importance. Therefore, this paper analysed changes in substance P- (SP-), calcitonin gene-related peptide- (CGRP-), and leu5-enkephalin- (L-ENK-) like immunoreactivity (LI) in enteric stomach neurons caused by chemically induced diabetes in a porcine model. Using double immunofluorescent labelling, it was found that acute hyperglycaemia led to significant changes in the chemical coding of stomach enteric neurons. Generally, the response to artificially inducted diabetes depended on the "kind" of enteric plexus as well as the stomach region studied. A clear increase in the percentage of neurons immunoreactive to SP and CGRP was visible in the myenteric plexus (MP) in the antrum, corpus, and pylorus as well as in the submucosal plexus (SmP) in the corpus. For L-ENK, an increase in the number of L-ENK-LI neurons was observed in the MP of the antrum and SmP in the corpus, while in the MP of the corpus and pylorus, a decrease in the percentage of L-ENK-LI neurons was noted.
Collapse
Affiliation(s)
- Michał Bulc
- Department of Clinical Physiology Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str. 13, 10-719 Olsztyn, Poland
| | - Katarzyna Palus
- Department of Clinical Physiology Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str. 13, 10-719 Olsztyn, Poland
| | - Jarosław Całka
- Department of Clinical Physiology Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str. 13, 10-719 Olsztyn, Poland
| | - Łukasz Zielonka
- Department of Veterinary Prevention and Feed Hygiene, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-718 Olsztyn, Poland
| |
Collapse
|
40
|
Maguire G. Amyotrophic lateral sclerosis as a protein level, non-genomic disease: Therapy with S2RM exosome released molecules. World J Stem Cells 2017; 9:187-202. [PMID: 29312526 PMCID: PMC5745587 DOI: 10.4252/wjsc.v9.i11.187] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/10/2017] [Accepted: 09/04/2017] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease that leads to death. No effective treatments are currently available. Based on data from epidemiological, etiological, laboratory, and clinical studies, I offer a new way of thinking about ALS and its treatment. This paper describes a host of extrinsic factors, including the exposome, that disrupt the extracellular matrix and protein function such that a spreading, prion-like disease leads to neurodegeneration in the motor tracts. A treatment regimen is described using the stem cell released molecules from a number of types of adult stem cells to provide tissue dependent molecules that restore homeostasis, including proteostasis, in the ALS patient. Because stem cells themselves as a therapeutic are cumbersome and expensive, and when implanted in a host cause aging of the host tissue and often fail to engraft or remain viable, only the S2RM molecules are used. Rebuilding of the extracellular matrix and repair of the dysfunctional proteins in the ALS patient ensues.
Collapse
Affiliation(s)
- Greg Maguire
- BioRegenerative Sciences, Inc., La Jolla, CA 92037, United States
| |
Collapse
|
41
|
Gasparotto J, Ribeiro CT, Bortolin RC, Somensi N, Rabelo TK, Kunzler A, Souza NC, Pasquali MADB, Moreira JCF, Gelain DP. Targeted inhibition of RAGE in substantia nigra of rats blocks 6-OHDA-induced dopaminergic denervation. Sci Rep 2017; 7:8795. [PMID: 28821831 PMCID: PMC5562811 DOI: 10.1038/s41598-017-09257-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 07/19/2017] [Indexed: 11/18/2022] Open
Abstract
The receptor for advanced glycation endproducts (RAGE) is a pattern-recognition receptor associated with inflammation in most cell types. RAGE up-regulates the expression of proinflammatory mediators and its own expression via activation of NF-kB. Recent works have proposed a role for RAGE in Parkinson's disease (PD). In this study, we used the multimodal blocker of RAGE FPS-ZM1, which has become available recently, to selectively inhibit RAGE in the substantia nigra (SN) of rats intracranially injected with 6-hydroxydopamine (6-OHDA). FPS-ZM1 (40 μg per rat), injected concomitantly with 6-OHDA (10 μg per rat) into the SN, inhibited the increase in RAGE, activation of ERK1/2, Src and nuclear translocation of NF-kB p65 subunit in the SN. RAGE inhibition blocked glial fibrillary acidic protein and Iba-1 upregulation as well as associated astrocyte and microglia activation. Circulating cytokines in serum and CSF were also decreased by FPS-ZM1 injection. The loss of tyrosine hydroxylase and NeuN-positive neurons was significantly inhibited by RAGE blocking. Finally, FPS-ZM1 attenuated locomotory and exploratory deficits induced by 6-OHDA. Our results demonstrate that RAGE is an essential component in the neuroinflammation and dopaminergic denervation induced by 6-OHDA in the SN. Selective inhibition of RAGE may offer perspectives for therapeutic approaches.
Collapse
Affiliation(s)
- Juciano Gasparotto
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Camila Tiefensee Ribeiro
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rafael Calixto Bortolin
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 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, Porto Alegre, RS, Brazil
| | - Thallita Kelly Rabelo
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alice Kunzler
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Natália Cabral Souza
- Instituto de Medicina Tropical, Departamento de Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Matheus Augusto de Bittencourt Pasquali
- Unidade Acadêmica de Engenharia de Alimentos, Centro de Tecnologia e Recursos Naturais, Universidade Federal de Campina Grande - UFCG, Campina Grande, Paraíba, Brazil
| | - José Claudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 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, Porto Alegre, RS, Brazil
| |
Collapse
|
42
|
Aguilera G, Santamaria A. Potential Therapeutic Targets of the Endocannabinoid System in Common Neurodegenerative Disorders and Organic Acidemias. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2017. [DOI: 10.1177/2326409817723667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Gabriela Aguilera
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
- Departamento de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| |
Collapse
|
43
|
The Astrocytic S100B Protein with Its Receptor RAGE Is Aberrantly Expressed in SOD1 G93A Models, and Its Inhibition Decreases the Expression of Proinflammatory Genes. Mediators Inflamm 2017; 2017:1626204. [PMID: 28713206 PMCID: PMC5496121 DOI: 10.1155/2017/1626204] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/30/2017] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is one of the major players in amyotrophic lateral sclerosis (ALS) pathogenesis, and astrocytes are significantly involved in this process. The astrocytic protein S100B can be released in pathological states activating the receptor for advanced glycation end products (RAGE). Different indications point to an aberrant expression of S100B and RAGE in ALS. In this work, we observed that S100B and RAGE are progressively and selectively upregulated in astrocytes of diseased rats with a tissue-specific timing pattern, correlated to the level of neurodegeneration. The expression of the full-length and soluble RAGE isoforms could also be linked to the degree of tissue damage. The mere presence of mutant SOD1 is able to increase the intracellular levels and release S100B from astrocytes, suggesting the possibility that an increased astrocytic S100B expression might be an early occurring event in the disease. Finally, our findings indicate that the protein may exert a proinflammatory role in ALS, since its inhibition in astrocytes derived from SOD1G93A mice limits the expression of reactivity-linked/proinflammatory genes. Thus, our results propose the S100B-RAGE axis as an effective contributor to the pathogenesis of the disease, suggesting its blockade as a rational target for a therapeutic intervention in ALS.
Collapse
|
44
|
Measurement and Clinical Significance of Biomarkers of Oxidative Stress in Humans. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6501046. [PMID: 28698768 PMCID: PMC5494111 DOI: 10.1155/2017/6501046] [Citation(s) in RCA: 487] [Impact Index Per Article: 60.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/26/2017] [Accepted: 05/21/2017] [Indexed: 12/11/2022]
Abstract
Oxidative stress is the result of the imbalance between reactive oxygen species (ROS) formation and enzymatic and nonenzymatic antioxidants. Biomarkers of oxidative stress are relevant in the evaluation of the disease status and of the health-enhancing effects of antioxidants. We aim to discuss the major methodological bias of methods used for the evaluation of oxidative stress in humans. There is a lack of consensus concerning the validation, standardization, and reproducibility of methods for the measurement of the following: (1) ROS in leukocytes and platelets by flow cytometry, (2) markers based on ROS-induced modifications of lipids, DNA, and proteins, (3) enzymatic players of redox status, and (4) total antioxidant capacity of human body fluids. It has been suggested that the bias of each method could be overcome by using indexes of oxidative stress that include more than one marker. However, the choice of the markers considered in the global index should be dictated by the aim of the study and its design, as well as by the clinical relevance in the selected subjects. In conclusion, the clinical significance of biomarkers of oxidative stress in humans must come from a critical analysis of the markers that should give an overall index of redox status in particular conditions.
Collapse
|
45
|
Pinto S, Cunha C, Barbosa M, Vaz AR, Brites D. Exosomes from NSC-34 Cells Transfected with hSOD1-G93A Are Enriched in miR-124 and Drive Alterations in Microglia Phenotype. Front Neurosci 2017; 11:273. [PMID: 28567000 PMCID: PMC5434170 DOI: 10.3389/fnins.2017.00273] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/28/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disorder affecting motor neurons (MNs). Evidences indicate that ALS is a non-cell autonomous disease in which glial cells participate in both disease onset and progression. Exosomal transfer of mutant copper-zinc superoxide dismutase 1 (mSOD1) from cell-to-cell was suggested to contribute to disease dissemination. Data from our group and others showed that exosomes from activated cells contain inflammatory-related microRNAs (inflamma-miRNAs) that recapitulate the donor cell. While glia-derived exosomes and their effects in neurons have been addressed by several studies, only a few investigated the influence of motor neuron (MN)-derived exosomes in other cell function, the aim of the present study. We assessed a set of inflamma-miRs in NSC-34 MN-like cells transfected with mutant SOD1(G93A) and extended the study into their derived exosomes (mSOD1 exosomes). Then, the effects produced by mSOD1 exosomes in the activation and polarization of the recipient N9 microglial cells were investigated. Exosomes in coculture with N9 microglia and NSC-34 cells [either transfected with either wild-type (wt) human SOD1 or mutant SOD1(G93A)] showed to be transferred into N9 cells. Increased miR-124 expression was found in mSOD1 NSC-34 cells and in their derived exosomes. Incubation of mSOD1 exosomes with N9 cells determined a sustained 50% reduction in the cell phagocytic ability. It also caused a persistent NF-kB activation and an acute generation of NO, MMP-2, and MMP-9 activation, as well as upregulation of IL-1β, TNF-α, MHC-II, and iNOS gene expression, suggestive of induced M1 polarization. Marked elevation of IL-10, Arginase 1, TREM2, RAGE, and TLR4 mRNA levels, together with increased miR-124, miR-146a, and miR-155, at 24 h incubation, suggest the switch to mixed M1 and M2 subpopulations in the exosome-treated N9 microglial cells. Exosomes from mSOD1 NSC-34 MNs also enhanced the number of senescent-like positive N9 cells. Data suggest that miR-124 is translocated from the mSOD1 MNs to exosomes, which determine early and late phenotypic alterations in the recipient N9-microglial cells. In conclusion, modulation of the inflammatory-associated miR-124, in mSOD1 NSC-34 MNs, with potential benefits in the cargo of their exosomes may reveal a promising therapeutic strategy in halting microglia activation and associated effects in MN degeneration.
Collapse
Affiliation(s)
- Sara Pinto
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Carolina Cunha
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Marta Barbosa
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Ana R Vaz
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Dora Brites
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| |
Collapse
|
46
|
Shekhtman A, Ramasamy R, Schmidt AM. Glycation & the RAGE axis: targeting signal transduction through DIAPH1. Expert Rev Proteomics 2016; 14:147-156. [PMID: 27967251 DOI: 10.1080/14789450.2017.1271719] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION The consequences of chronic disease are vast and unremitting; hence, understanding the pathogenic mechanisms mediating such disorders holds promise to identify therapeutics and diminish the consequences. The ligands of the receptor for advanced glycation end products (RAGE) accumulate in chronic diseases, particularly those characterized by inflammation and metabolic dysfunction. Although first discovered and reported as a receptor for advanced glycation end products (AGEs), the expansion of the repertoire of RAGE ligands implicates the receptor in diverse milieus, such as autoimmunity, chronic inflammation, obesity, diabetes, and neurodegeneration. Areas covered: This review summarizes current knowledge regarding the ligand families of RAGE and data from human subjects and animal models on the role of the RAGE axis in chronic diseases. The recent discovery that the cytoplasmic domain of RAGE binds to the formin homology 1 (FH1) domain, DIAPH1, and that this interaction is essential for RAGE ligand-stimulated signal transduction, is discussed. Finally, we review therapeutic opportunities targeting the RAGE axis as a means to mitigate chronic diseases. Expert commentary: With the aging of the population and the epidemic of cardiometabolic disease, therapeutic strategies to target molecular pathways that contribute to the sequelae of these chronic diseases are urgently needed. In this review, we propose that the ligand/RAGE axis and its signaling nexus is a key factor in the pathogenesis of chronic disease and that therapeutic interruption of this pathway may improve quality and duration of life.
Collapse
Affiliation(s)
- Alexander Shekhtman
- a Department of Chemistry , University at Albany, State University of New York , Albany , NY , 12222 , USA
| | - Ravichandran Ramasamy
- b Diabetes Research Program, Division of Endocrinology, Department of Medicine , NYU Langone Medical Center , New York , NY , 10016 , USA
| | - Ann Marie Schmidt
- b Diabetes Research Program, Division of Endocrinology, Department of Medicine , NYU Langone Medical Center , New York , NY , 10016 , USA
| |
Collapse
|
47
|
Papatheodorou A, Stein A, Bank M, Sison CP, Gibbs K, Davies P, Bloom O. High-Mobility Group Box 1 (HMGB1) Is Elevated Systemically in Persons with Acute or Chronic Traumatic Spinal Cord Injury. J Neurotrauma 2016; 34:746-754. [PMID: 27673428 DOI: 10.1089/neu.2016.4596] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Inflammation in traumatic spinal cord injury (SCI) has been proposed to promote damage acutely and oppose functional recovery chronically. However, we do not yet understand the signals that initiate or prolong inflammation in persons with SCI. High-Mobility Group Box 1 (HMGB1) is a potent systemic inflammatory cytokine-or damage-associated molecular pattern molecule (DAMP)-studied in a variety of clinical settings. It is elevated in pre-clinical models of traumatic spinal cord injury (SCI), where it promotes secondary injury, and strategies that block HMGB1 improve functional recovery. To investigate the potential translational relevance of these observations, we measured HMGB1 in plasma from adults with acute (≤ 1 week post-SCI, n = 16) or chronic (≥ 1 year post-SCI, n = 47) SCI. Plasma from uninjured persons (n = 51) served as controls for comparison. In persons with acute SCI, average HMGB1 levels were significantly elevated within 0-3 days post-injury (6.00 ± 1.8 ng/mL, mean ± standard error of the mean [SEM]) or 4-7 (6.26 ± 1.3 ng/mL, mean ± SEM), compared with controls (1.26 ± 0.24 ng/mL, mean ± SEM; p ≤ 0.001 and p ≤ 0.01, respectively). In persons with chronic SCI who were injured for 15 ± 1.5 years (mean ± SEM), HMGB1 also was significantly elevated, compared with uninjured persons (3.7 ± 0.69 vs. 1.26 ± 0.24 ng/mL, mean ± SEM; p ≤ 0.0001). Together, these data suggest that HMGB1 may be a common, early, and persistent danger signal promoting inflammation in individuals with SCI.
Collapse
Affiliation(s)
- Angelos Papatheodorou
- 1 Department of Autoimmune and Musculoskeletal Disorders, North Shore University Hospital , Northwell Health, Manhasset, New York
| | - Adam Stein
- 2 Department of Physical Medicine and Rehabilitation, North Shore University Hospital , Northwell Health, Manhasset, New York
| | - Matthew Bank
- 3 Department of Surgery, North Shore University Hospital , Northwell Health, Manhasset, New York
| | - Cristina P Sison
- 4 Department of Molecular Medicine, Hofstra Northwell School of Medicine, North Shore University Hospital , Northwell Health, Manhasset, New York
| | - Katie Gibbs
- 2 Department of Physical Medicine and Rehabilitation, North Shore University Hospital , Northwell Health, Manhasset, New York
| | - Peter Davies
- 5 Litwin-Zucker Research Center for the Study of Alzheimer's Disease, the Feinstein Institute for Medical Research, North Shore University Hospital , Northwell Health, Manhasset, New York
| | - Ona Bloom
- 1 Department of Autoimmune and Musculoskeletal Disorders, North Shore University Hospital , Northwell Health, Manhasset, New York.,2 Department of Physical Medicine and Rehabilitation, North Shore University Hospital , Northwell Health, Manhasset, New York.,4 Department of Molecular Medicine, Hofstra Northwell School of Medicine, North Shore University Hospital , Northwell Health, Manhasset, New York
| |
Collapse
|
48
|
RAGE Expression and ROS Generation in Neurons: Differentiation versus Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9348651. [PMID: 27313835 PMCID: PMC4897723 DOI: 10.1155/2016/9348651] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/03/2016] [Indexed: 12/27/2022]
Abstract
RAGE is a multiligand receptor able to bind advanced glycation end-products (AGEs), amphoterin, calgranulins, and amyloid-beta peptides, identified in many tissues and cells, including neurons. RAGE stimulation induces the generation of reactive oxygen species (ROS) mainly through the activity of NADPH oxidases. In neuronal cells, RAGE-induced ROS generation is able to favor cell survival and differentiation or to induce death through the imbalance of redox state. The dual nature of RAGE signaling in neurons depends not only on the intensity of RAGE activation but also on the ability of RAGE-bearing cells to adapt to ROS generation. In this review we highlight these aspects of RAGE signaling regulation in neuronal cells.
Collapse
|
49
|
Wan W, Cao L, Khanabdali R, Kalionis B, Tai X, Xia S. The Emerging Role of HMGB1 in Neuropathic Pain: A Potential Therapeutic Target for Neuroinflammation. J Immunol Res 2016; 2016:6430423. [PMID: 27294160 PMCID: PMC4887637 DOI: 10.1155/2016/6430423] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023] Open
Abstract
Neuropathic pain (NPP) is intolerable, persistent, and specific type of long-term pain. It is considered to be a direct consequence of pathological changes affecting the somatosensory system and can be debilitating for affected patients. Despite recent progress and growing interest in understanding the pathogenesis of the disease, NPP still presents a major diagnostic and therapeutic challenge. High mobility group box 1 (HMGB1) mediates inflammatory and immune reactions in nervous system and emerging evidence reveals that HMGB1 plays an essential role in neuroinflammation through receptors such as Toll-like receptors (TLR), receptor for advanced glycation end products (RAGE), C-X-X motif chemokines receptor 4 (CXCR4), and N-methyl-D-aspartate (NMDA) receptor. In this review, we present evidence from studies that address the role of HMGB1 in NPP. First, we review studies aimed at determining the role of HMGB1 in NPP and discuss the possible mechanisms underlying HMGB1-mediated NPP progression where receptors for HMGB1 are involved. Then we review studies that address HMGB1 as a potential therapeutic target for NPP.
Collapse
Affiliation(s)
- Wenbin Wan
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lan Cao
- State Key Laboratory of Medical Neurobiology, Department of Neurobiology and Institutes of Brain Science, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Ramin Khanabdali
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre and Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, VIC 3052, Australia
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre and Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, VIC 3052, Australia
| | - Xiantao Tai
- School of Acupuncture, Massage and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| | - Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
50
|
Richards RI, Robertson SA, O'Keefe LV, Fornarino D, Scott A, Lardelli M, Baune BT. The Enemy within: Innate Surveillance-Mediated Cell Death, the Common Mechanism of Neurodegenerative Disease. Front Neurosci 2016; 10:193. [PMID: 27242399 PMCID: PMC4862319 DOI: 10.3389/fnins.2016.00193] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/18/2016] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative diseases comprise an array of progressive neurological disorders all characterized by the selective death of neurons in the central nervous system. Although, rare (familial) and common (sporadic) forms can occur for the same disease, it is unclear whether this reflects several distinct pathogenic pathways or the convergence of different causes into a common form of nerve cell death. Remarkably, neurodegenerative diseases are increasingly found to be accompanied by activation of the innate immune surveillance system normally associated with pathogen recognition and response. Innate surveillance is the cell's quality control system for the purpose of detecting such danger signals and responding in an appropriate manner. Innate surveillance is an "intelligent system," in that the manner of response is relevant to the magnitude and duration of the threat. If possible, the threat is dealt with within the cell in which it is detected, by degrading the danger signal(s) and restoring homeostasis. If this is not successful then an inflammatory response is instigated that is aimed at restricting the spread of the threat by elevating degradative pathways, sensitizing neighboring cells, and recruiting specialized cell types to the site. If the danger signal persists, then the ultimate response can include not only the programmed cell death of the original cell, but the contents of this dead cell can also bring about the death of adjacent sensitized cells. These responses are clearly aimed at destroying the ability of the detected pathogen to propagate and spread. Innate surveillance comprises intracellular, extracellular, non-cell autonomous and systemic processes. Recent studies have revealed how multiple steps in these processes involve proteins that, through their mutation, have been linked to many familial forms of neurodegenerative disease. This suggests that individuals harboring these mutations may have an amplified response to innate-mediated damage in neural tissues, and renders innate surveillance mediated cell death a plausible common pathogenic pathway responsible for neurodegenerative diseases, in both familial and sporadic forms. Here we have assembled evidence in favor of the hypothesis that neurodegenerative disease is the cumulative result of chronic activation of the innate surveillance pathway, triggered by endogenous or environmental danger or damage associated molecular patterns in a progressively expanding cascade of inflammation, tissue damage and cell death.
Collapse
Affiliation(s)
- Robert I Richards
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide Adelaide, SA, Australia
| | - Sarah A Robertson
- School of Paediatrics and Reproductive Health, Robinson Research Institute, The University of Adelaide Adelaide, SA, Australia
| | - Louise V O'Keefe
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide Adelaide, SA, Australia
| | - Dani Fornarino
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide Adelaide, SA, Australia
| | - Andrew Scott
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide Adelaide, SA, Australia
| | - Michael Lardelli
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide Adelaide, SA, Australia
| | - Bernhard T Baune
- School of Medicine, Discipline of Psychiatry, The University of Adelaide Adelaide, SA, Australia
| |
Collapse
|