1
|
Cordeiro AMT, Singulani MP, Talib LL, Forlenza OV. Down syndrome and Alzheimer's disease: Oxidative stress in the neurodegenerative process. J Alzheimers Dis 2025; 105:258-267. [PMID: 40179214 DOI: 10.1177/13872877251326291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
BackgroundIndividuals with Down syndrome (DS) generally show neuropathological features of Alzheimer disease (AD). The trisomy of chromosome 21 causes an imbalance of antioxidant systems, which can be linked to AD pathophysiology.ObjectiveVerify the difference between the activity of antioxidant enzymes and products of the oxidation process in peripheral blood in non-trisomic (NT) and trisomic (DS) adults and elders and respective associations with cognitive impairment.MethodsA total of 120 subjects were included in this study. Sociodemographic and clinical information were collected as per protocol for participants in DS and NT groups. The cognitive state of the DS participants was established according to the Brazilian version of the Cambridge Examination for Mental Disorders of Older People with Down's syndrome and Others with Intellectual Disabilities (CAMDEX-DS). The CAMDEX interview was used for this purpose for participants in the NT group. Plasma samples were collected to evaluate protein carbonyl content, superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT), malondialdehyde (MDA), and 4-hydroxy-2-nonenal (HNE).ResultsWe found increased levels of SOD and CAT activity in the DS group. When the groups were stratified by cognitive decline, elevated levels of SOD and CAT activity were found both in DS groups with and without decline. The activity of GPx was similar between the groups, as well as MDA and HNE.ConclusionThe results suggest that alterations of the antioxidative processes can be implicated in the onset of neurodegeneration observed in individuals with DS.
Collapse
Affiliation(s)
- Augusto Magno Tranquezi Cordeiro
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
| | - Monique Patricio Singulani
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
- Faculdade de Medicina da Universidade de São Paulo (FMUSP), Centro de Neurociências Translacionais (CNT), São Paulo, Brazil
| | - Leda Leme Talib
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
| | - Orestes Vicente Forlenza
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
- Faculdade de Medicina da Universidade de São Paulo (FMUSP), Centro de Neurociências Translacionais (CNT), São Paulo, Brazil
| |
Collapse
|
2
|
Kayama E, Baoshuo N, Tatsuno R, Nishi K, Mohammed ESI, Abiko Y, Yanagawa T, Takahashi S, Warabi E. 4-Hydroxy-2-nonenal causes nuclear accumulation of p62 by inhibiting Xpo1 and promoting the proteolytic pathway in the nucleus. PLoS One 2025; 20:e0316558. [PMID: 39899539 PMCID: PMC11790078 DOI: 10.1371/journal.pone.0316558] [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: 08/31/2024] [Accepted: 12/12/2024] [Indexed: 02/05/2025] Open
Abstract
p62, an adapter protein involved in selective autophagy, is mainly found in the cytoplasm under normal conditions. Because p62 has nuclear localization signal (NLS) and a nuclear export signal, it has been suggested that p62 shuttles between the nucleus and cytoplasm. We studied the effect of 4-hydroxy-nonenal (4-HNE), an endogenous lipid peroxidation product, on intracellular p62 distribution in mouse embryonic fibroblasts. We found that treatment of 4-HNE causes p62 translocation from the cytoplasm to the nucleus. Further analysis revealed that 4-HNE directly binds to exportin-1 (Xpo1), essential protein for nuclear export of various proteins. Further analysis 4-HNE enhanced intranuclear EGFP-NLS-CL1 degradation in a p62-dependent manner. Our results suggest that 4-HNE changes p62 localization to the nucleus by inhibiting Xpo1 and might affect intranuclear protein quality control.
Collapse
Affiliation(s)
- Emi Kayama
- Department of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ning Baoshuo
- Department of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Rinna Tatsuno
- Department of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kanako Nishi
- Department of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Elsayed S. I. Mohammed
- Department of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Histology and Cytology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Yumi Abiko
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Toru Yanagawa
- Department of Clinical Medicine, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Department of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Eiji Warabi
- Department of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
3
|
Lanzillotta S, Esteve D, Lanzillotta C, Tramutola A, Lloret A, Forte E, Pesce V, Picca A, Di Domenico F, Perluigi M, Barone E. Altered mitochondrial unfolded protein response and protein quality control promote oxidative distress in down syndrome brain. Free Radic Biol Med 2025; 227:80-93. [PMID: 39586382 DOI: 10.1016/j.freeradbiomed.2024.11.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
Down Syndrome (DS) is a genetic disorder caused by the presence of an extra copy of chromosome 21, and leading to various developmental and cognitive defects. A critical feature of DS is the occurrence of oxidative distress particularly in the brain, which exacerbates neurodevelopmental processes. Mitochondria play a crucial role in cell energy metabolism and their impairment is one of the major causes of oxidative distress in several pathologies. Hence, this study investigates mitochondrial proteostasis by the mean of the mitochondrial Unfolded Protein Response (UPRmt) and the mitochondrial protein quality control (MQC) mechanisms in the context of DS, focusing on their implications in redox homeostasis in brain development. We analyzed key UPRmt markers and mitochondrial function in the frontal cortex isolated fromTs2Cje mice, a model for DS, across different developmental stages. Our results demonstrate significant alterations in UPRmt markers, particularly at postnatal day 0 (P0) and 1 month (1M). These changes indicate early UPRmt activation, primarily driven by the ATF5/GRP75 axis, although compromised by reduced levels of other components. Impaired UPRmt correlates with decreased mitochondrial activity, evidenced by reduced oxygen consumption rates and altered expression of OXPHOS complexes. Additionally, elevated oxidative stress markers such as 3-nitrotyrosine (3-NT), 4-hydroxynonenal (HNE), and protein carbonyls (PC) were observed, linking mitochondrial dysfunction to increased oxidative damage. Defects of MQC, including disrupted biogenesis, increased fission, and the activation of mitophagy were evident mostly at P0 and 1M consistent with UPRmt activation. Principal Component Analysis revealed distinct phenotypic differences between Ts2Cje and control mice, driven by these molecular alterations. Our findings underscore the critical role of UPRmt and MQC in DS brain development, highlighting potential therapeutic targets to mitigate mitochondrial dysfunction and oxidative distress, thereby alleviating some of the neurodevelopmental and cognitive impairments associated with DS.
Collapse
Affiliation(s)
- Simona Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Daniel Esteve
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain; Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Ana Lloret
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Elena Forte
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Vito Pesce
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
4
|
Micangeli G, Menghi M, Profeta G, Paparella R, Tarani F, Petrella C, Barbato C, Minni A, Greco A, Ferraguti G, Tarani L, Fiore M. Malignant and Benign Head and Neck Tumors of the Pediatric Age: A Narrative Review. Curr Pediatr Rev 2025; 21:118-132. [PMID: 38310547 DOI: 10.2174/0115733963258575231123043807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/22/2023] [Accepted: 11/02/2023] [Indexed: 02/06/2024]
Abstract
Malignant tumors of the head and neck are rare in children, but it is important to know these lesions and identify them early in order to have a good outcome for these patients. Benign lesions of the head and neck are much more frequent and have an excellent prognosis. For this reason, it is necessary to recognize the warning signs and symptoms and understand when to refer the patient to a reference center for the treatment of these pathologies. The clinical presentation of both benign and malignant lesions in children may be similar as usually, both categories have compressive effects. This confirms the fact that the clinical diagnosis is not sufficient and always requires instrumental investigations and biopsies. In this narrative review, we analyzed both malignant lesions such as lymphoma, rhabdomyosarcoma, thyroid tumors, salivary gland tumors, neuroblastoma, and nasopharyngeal carcinoma, and benign ones such as cystic dermoid teratoma, hemangioma, juvenile angiofibroma and fibrosis dysplasia. Indeed, we set out to discuss the most common lesions of this site by evaluating their characteristics to highlight the differentiation of malignant tumors from benign lesions and their correct clinical-therapeutic management. A literature search was carried out in the PubMed and Google Scholar databases to identify all narrative reviews addressing malignant and benign head and neck tumors of the pediatric age. In conclusion, the care of children affected by head and neck benign lesions and malignancy must be combined and multidisciplinary. It is essential to recognize the diseases early in order to differentiate and intervene as soon as possible for the correct clinical-therapeutic management.
Collapse
Affiliation(s)
- Ginevra Micangeli
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Michela Menghi
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Giovanni Profeta
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Roberto Paparella
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesca Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185, Rome, Italy
| | - Christian Barbato
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185, Rome, Italy
| | - Antonio Minni
- Department of Sensory Organs, Sapienza University of Rome, 00185, Rome, Italy
- ASL Rieti-Sapienza University, Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, Viale Kennedy, 02100, Rieti, Italy
| | - Antonio Greco
- Department of Sensory Organs, Sapienza University of Rome, 00185, Roma, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00185, Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185, Rome, Italy
| |
Collapse
|
5
|
Di Domenico F, Lanzillotta C, Perluigi M. Redox imbalance and metabolic defects in the context of Alzheimer disease. FEBS Lett 2024; 598:2047-2066. [PMID: 38472147 DOI: 10.1002/1873-3468.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Redox reactions play a critical role for intracellular processes, including pathways involved in metabolism and signaling. Reactive oxygen species (ROS) act either as second messengers or generators of protein modifications, fundamental mechanisms for signal transduction. Disturbance of redox homeostasis is associated with many disorders. Among these, Alzheimer's disease is a neurodegenerative pathology that presents hallmarks of oxidative damage such as increased ROS production, decreased activity of antioxidant enzymes, oxidative modifications of macromolecules, and changes in mitochondrial homeostasis. Interestingly, alteration of redox homeostasis is closely associated with defects of energy metabolism, involving both carbohydrates and lipids, the major energy fuels for the cell. As the brain relies exclusively on glucose metabolism, defects of glucose utilization represent a harmful event for the brain. During aging, a progressive perturbation of energy metabolism occurs resulting in brain hypometabolism. This condition contributes to increase neuronal cell vulnerability ultimately resulting in cognitive impairment. The current review discusses the crosstalk between alteration of redox homeostasis and brain energy defects that seems to act in concert in promoting Alzheimer's neurodegeneration.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
6
|
Sukreet S, Rafii MS, Rissman RA. From understanding to action: Exploring molecular connections of Down syndrome to Alzheimer's disease for targeted therapeutic approach. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12580. [PMID: 38623383 PMCID: PMC11016820 DOI: 10.1002/dad2.12580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/17/2024]
Abstract
Down syndrome (DS) is caused by a third copy of chromosome 21. Alzheimer's disease (AD) is a neurodegenerative condition characterized by the deposition of amyloid-beta (Aβ) plaques and neurofibrillary tangles in the brain. Both disorders have elevated Aβ, tau, dysregulated immune response, and inflammation. In people with DS, Hsa21 genes like APP and DYRK1A are overexpressed, causing an accumulation of amyloid and neurofibrillary tangles, and potentially contributing to an increased risk of AD. As a result, people with DS are a key demographic for research into AD therapeutics and prevention. The molecular links between DS and AD shed insights into the underlying causes of both diseases and highlight potential therapeutic targets. Also, using biomarkers for early diagnosis and treatment monitoring is an active area of research, and genetic screening for high-risk individuals may enable earlier intervention. Finally, the fundamental mechanistic parallels between DS and AD emphasize the necessity for continued research into effective treatments and prevention measures for DS patients at risk for AD. Genetic screening with customized therapy approaches may help the DS population in current clinical studies and future biomarkers.
Collapse
Affiliation(s)
- Sonal Sukreet
- Department of NeurosciencesUniversity of California‐San DiegoLa JollaCaliforniaUSA
| | - Michael S. Rafii
- Department of Neurology, Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Robert A. Rissman
- Department of NeurosciencesUniversity of California‐San DiegoLa JollaCaliforniaUSA
- Department Physiology and Neuroscience, Alzheimer’s Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| |
Collapse
|
7
|
León-Paz-de-Rodríguez GE, Rodríguez-León E, Iñiguez-Palomares R. DNA Hyperstructure. ACS OMEGA 2024; 9:9013-9026. [PMID: 38434827 PMCID: PMC10905968 DOI: 10.1021/acsomega.3c07379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/29/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
This study presents a new procedure to condense DNA molecules and precipitate them onto a glass slide. The resulting DNA molecules undergo autonomous self-assembly, creating closed superstructures on the micrometer scale, which are called DNA hyperstructures. These structures can be observed using low-magnification (4×) light microscopy. Precisely controlling the alcohol/glacial acetic acid ratio and DNA concentration during precipitation enabled the regulation of structure compaction on the slide. The alcohol/glacial acetic acid ratio is inversely proportional to the DNA concentration to achieve optimal compaction on the slide. Confocal microscopy fluorescence analysis of DNA extracts stained with DAPI shows that nucleic acids self-assemble to form structures during precipitation on the slide. This methodology is relevant since it facilitates the precipitation and visualization of DNA, regardless of its origin or molecular weight. To confirm its versatility, results with DNA extracted from human peripheral blood, the Lambda virus, and plasmid pBR322 are presented. The study examined the morphological features of DNA hyperstructures in both healthy individuals and those diagnosed with different medical conditions or illnesses, revealing distinct patterns specific to each case. This innovative technology has potential for disease detection in peripheral blood samples, ranging from cancer and Alzheimer's disease to determining the gender of the gestational product at an early stage.
Collapse
|
8
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
9
|
Buczyńska A, Sidorkiewicz I, Krętowski AJ, Zbucka-Krętowska M. The Role of Oxidative Stress in Trisomy 21 Phenotype. Cell Mol Neurobiol 2023; 43:3943-3963. [PMID: 37819608 PMCID: PMC10661812 DOI: 10.1007/s10571-023-01417-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/17/2023] [Indexed: 10/13/2023]
Abstract
Extensive research has been conducted to gain a deeper understanding of the deregulated metabolic pathways in the development of trisomy 21 (T21) or Down syndrome. This research has shed light on the hypothesis that oxidative stress plays a significant role in the manifestation of the T21 phenotype. Although in vivo studies have shown promising results in mitigating the detrimental effects of oxidative stress, there is currently a lack of introduced antioxidant treatment options targeting cognitive impairments associated with T21. To address this gap, a comprehensive literature review was conducted to provide an updated overview of the involvement of oxidative stress in T21. The review aimed to summarize the insights into the pathogenesis of the Down syndrome phenotype and present the findings of recent innovative research that focuses on improving cognitive function in T21 through various antioxidant interventions. By examining the existing literature, this research seeks to provide a holistic understanding of the role oxidative stress plays in the development of T21 and to explore novel approaches that target multiple aspects of antioxidant intervention to improve cognitive function in individuals with Down syndrome. The guides -base systematic review process (Hutton et al. 2015).
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Białystok, ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland.
| | - Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Białystok, ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Białystok, ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, ul. Sklodowskiej-Curie 24a, 15-276, Białystok, Poland
| | - Monika Zbucka-Krętowska
- Department of Gynecological Endocrinology and Adolescent Gynecology, Medical University of Białystok, ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland.
| |
Collapse
|
10
|
Olasz B, Fiser B, Szőri M, Viskolcz B, Owen MC. Computational Elucidation of the Solvent-Dependent Addition of 4-Hydroxy-2-nonenal (HNE) to Cysteine and Cysteinate Residues. J Org Chem 2022; 87:12909-12920. [PMID: 36148484 DOI: 10.1021/acs.joc.2c01487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The lipid peroxidation end product, 4-hydroxy-2-nonenal (HNE), is a secondary mediator of oxidative stress due to its strong ability to form adducts to the side chains of lysine, histidine, and cysteine residues (Cys) at increasing reactivities. This reaction can take place in various cellular environments and may be dependent on solvent. Moreover, approximately 10% of cysteine residues within the cells exist as the negatively charged cysteinate, which may also have a distinct reactivity toward HNE. In this study, quantum chemical calculations are used to investigate the reactivity of HNE toward Cys and cysteinate in three distinct solvent environments to mimic the aqueous, polar, and hydrophobic regions within the cell. Water enhances the reactivity of HNE to cysteine compared to that of the polar and hydrophobic solvents, and the reactivity of HNE is further augmented when Cys is first ionized to cysteinate. This is also confirmed by the transition state rate constant calculations. This study reveals the role of solvent polarity in these reactions and how cysteinate can account for the seemingly high reactivity of HNE toward Cys compared to other amino acid residues and demonstrates how a strong nucleophile can enhance the reactivity of an antioxidant analogue of the Cys residue.
Collapse
Affiliation(s)
- Balázs Olasz
- Institute of Chemistry, University of Miskolc, Miskolc-Egyetemváros, H-3515 Miskolc, Hungary
| | - Béla Fiser
- Institute of Chemistry, University of Miskolc, Miskolc-Egyetemváros, H-3515 Miskolc, Hungary.,Higher Education and Industrial Cooperation Centre, University of Miskolc, Miskolc-Egyetemváros, H-3515 Miskolc, Hungary.,Ferenc Rákóczi II Transcarpathian Hungarian College of Higher Education, UA-90200 Beregszász, Transcarpathia, Ukraine
| | - Milán Szőri
- Institute of Chemistry, University of Miskolc, Miskolc-Egyetemváros, H-3515 Miskolc, Hungary
| | - Béla Viskolcz
- Institute of Chemistry, University of Miskolc, Miskolc-Egyetemváros, H-3515 Miskolc, Hungary.,Higher Education and Industrial Cooperation Centre, University of Miskolc, Miskolc-Egyetemváros, H-3515 Miskolc, Hungary
| | - Michael C Owen
- Institute of Chemistry, University of Miskolc, Miskolc-Egyetemváros, H-3515 Miskolc, Hungary.,Higher Education and Industrial Cooperation Centre, University of Miskolc, Miskolc-Egyetemváros, H-3515 Miskolc, Hungary
| |
Collapse
|
11
|
Pagnotta S, Tramutola A, Barone E, Di Domenico F, Pittalà V, Salerno L, Folgiero V, Caforio M, Locatelli F, Petrini S, Butterfield DA, Perluigi M. CAPE and its synthetic derivative VP961 restore BACH1/NRF2 axis in Down Syndrome. Free Radic Biol Med 2022; 183:1-13. [PMID: 35283228 DOI: 10.1016/j.freeradbiomed.2022.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/17/2022] [Accepted: 03/08/2022] [Indexed: 12/19/2022]
Abstract
The cells possess several mechanisms to counteract the over-production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), including enzymes such as superoxide dismutase, catalase and glutathione peroxidase. Moreover, an important sensor involved in the anti-oxidant response is KEAP1-NRF2-ARE signaling complex. Under oxidative stress (OS), the transcription factor NRF2 can dissociate from the KEAP1-complex in the cytosol and translocate into the nucleus to promote the transcriptional activation of anti-oxidant genes, such as heme oxygenase 1 and NADPH quinone oxidoreductase. Within this context, the activation of NRF2 response is further regulated by BACH1, a transcription repressor, that compete with the KEAP1-NRF2-ARE complex. In this work, we focused on the role of BACH1/NRF2 ratio in the regulation of the anti-oxidant response, proposing their antithetical relation as a valuable target for a therapeutic strategy to test drugs able to exert neuroprotective effects, notably in aging and neurodegenerative diseases. Among these, Down syndrome (DS) is a complex genetic disorder characterized by BACH1 gene triplication that likely results in the impairment of NRF2 causing increased OS. Our results revealed that BACH1 overexpression alters the BACH1/NRF2 ratio in the nucleus and disturbs the induction of antioxidant response genes ultimately resulting in the accumulation of oxidative damage both in Ts2Cje mice (a mouse model of DS) and human DS lymphoblastoid cell lines (LCLs). Based on this evidence, we tested Caffeic Acid Phenethyl Ester (CAPE) and the synthetic analogue VP961, which have been proven to modulate NRF2 activity. We showed that CAPE and VP961 administration to DS LCLs was able to promote NRF2 nuclear translocation, which resulted in the amelioration of antioxidant response. Overall, our study supports the hypothesis that BACH1 triplication in DS subjects is implicated in the alteration of redox homeostasis and therapeutic strategies to overcome this effect are under investigation in our laboratory.
Collapse
Affiliation(s)
- Sara Pagnotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Valentina Folgiero
- Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Matteo Caforio
- Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy; Department of Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Laboratory affiliiated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy.
| |
Collapse
|
12
|
Fehsel K, Christl J. Comorbidity of osteoporosis and Alzheimer's disease: Is `AKT `-ing on cellular glucose uptake the missing link? Ageing Res Rev 2022; 76:101592. [PMID: 35192961 DOI: 10.1016/j.arr.2022.101592] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2023]
Abstract
Osteoporosis and Alzheimer's disease (AD) are both degenerative diseases. Osteoporosis often proceeds cognitive deficits, and multiple studies have revealed common triggers that lead to energy deficits in brain and bone. Risk factors for osteoporosis and AD, such as obesity, type 2 diabetes, aging, chemotherapy, vitamin deficiency, alcohol abuse, and apolipoprotein Eε4 and/or Il-6 gene variants, reduce cellular glucose uptake, and protective factors, such as estrogen, insulin, exercise, mammalian target of rapamycin inhibitors, hydrogen sulfide, and most phytochemicals, increase uptake. Glucose uptake is a fine-tuned process that depends on an abundance of glucose transporters (Gluts) on the cell surface. Gluts are stored in vesicles under the plasma membrane, and protective factors cause these vesicles to fuse with the membrane, resulting in presentation of Gluts on the cell surface. This translocation depends mainly on AKT kinase signaling and can be affected by a range of factors. Reduced AKT kinase signaling results in intracellular glucose deprivation, which causes endoplasmic reticulum stress and iron depletion, leading to activation of HIF-1α, the transcription factor necessary for higher Glut expression. The link between diseases and aging is a topic of growing interest. Here, we show that diseases that affect the same biochemical pathways tend to co-occur, which may explain why osteoporosis and/or diabetes are often associated with AD.
Collapse
|
13
|
Buczyńska A, Sidorkiewicz I, Krętowski AJ, Zbucka-Krętowska M, Adamska A. Metformin Intervention—A Panacea for Cancer Treatment? Cancers (Basel) 2022; 14:cancers14051336. [PMID: 35267644 PMCID: PMC8909770 DOI: 10.3390/cancers14051336] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 02/07/2023] Open
Abstract
The molecular mechanism of action and the individual influence of various metabolic pathways related to metformin intervention are under current investigation. The available data suggest that metformin provides many advantages, exhibiting anti-inflammatory, anti-cancer, hepatoprotective, cardioprotective, otoprotective, radioprotective, and radio-sensitizing properties depending on cellular context. This literature review was undertaken to provide novel evidence concerning metformin intervention, with a particular emphasis on cancer treatment and prevention. Undoubtedly, the pleiotropic actions associated with metformin include inhibiting inflammatory processes, increasing antioxidant capacity, and improving glycemic and lipid metabolism. Consequently, these characteristics make metformin an attractive medicament to translate to human trials, the promising results of which were also summarized in this review.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
- Correspondence: (A.B.); (A.A.); Tel.: +48-85-746-8513 (A.B.); +48-85-746-8660 (A.A.)
| | - Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Monika Zbucka-Krętowska
- Department of Gynecological Endocrinology and Adolescent Gynecology, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Agnieszka Adamska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
- Correspondence: (A.B.); (A.A.); Tel.: +48-85-746-8513 (A.B.); +48-85-746-8660 (A.A.)
| |
Collapse
|
14
|
Ferroptosis and NRF2: an emerging battlefield in the neurodegeneration of Alzheimer's disease. Essays Biochem 2021; 65:925-940. [PMID: 34623415 DOI: 10.1042/ebc20210017] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022]
Abstract
Ferroptosis is an iron- and lipid peroxidation-dependent cell death modality and emerging evidence indicates that ferroptosis has great explanatory potential for neuronal loss and associated CNS dysfunction in a range of neurodegenerative diseases (e.g., Alzheimer's, Parkinson's and Huntington's diseases, Motor neuron disease, Friedreich ataxia (FRDA)). Ferroptotic death results from lethal levels of phospholipid hydroperoxides that are generated by iron-dependent peroxidation of polyunsaturated fatty acids (PUFAs), such as arachidonic and adrenic acids, which are conjugated to specific phospholipids (e.g., phosphatidylethanolamines (PEs)). The major cellular protector against ferroptosis is glutathione peroxidase 4 (GPX4), a membrane-associated selenoenzyme that reduces deleterious phospholipid hydroperoxides to their corresponding benign phospholipid alcohols in a glutathione-dependent manner. Other complementary protective systems have also been identified that act to bolster cellular defences against ferroptosis. Many pharmacological modulators of the ferroptosis pathway have been identified, targeting proteins involved in iron homoeostasis and autophagy; the production and detoxification of lipid peroxides, and cyst(e)ine/glutathione metabolism. While a growing number of cell signalling pathways converge to regulate the ferroptosis cascade, an emerging understanding of ferroptosis regulation suggests that the ferroptotic 'tone' of cells can be set by the transcription factor, nuclear factor erythroid 2-related factor 2 (NRF2), which transcriptionally controls many key components of the ferroptosis pathway. In this review, we provide a critical overview of the relationship between ferroptosis and NRF2 signalling. With a focus on the role of ferroptosis in Alzheimer's disease (AD), we discuss how therapeutic modulation of the NRF2 pathway is a viable strategy to explore in the treatment of ferroptosis-driven neurodegeneration.
Collapse
|
15
|
Rujeedawa T, Carrillo Félez E, Clare ICH, Fortea J, Strydom A, Rebillat AS, Coppus A, Levin J, Zaman SH. The Clinical and Neuropathological Features of Sporadic (Late-Onset) and Genetic Forms of Alzheimer's Disease. J Clin Med 2021; 10:4582. [PMID: 34640600 PMCID: PMC8509365 DOI: 10.3390/jcm10194582] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022] Open
Abstract
The purpose of this review is to compare and highlight the clinical and pathological aspects of genetic versus acquired Alzheimer's disease: Down syndrome-associated Alzheimer's disease in (DSAD) and Autosomal Dominant Alzheimer's disease (ADAD) are compared with the late-onset form of the disease (LOAD). DSAD and ADAD present in a younger population and are more likely to manifest with non-amnestic (such as dysexecutive function features) in the prodromal phase or neurological features (such as seizures and paralysis) especially in ADAD. The very large variety of mutations associated with ADAD explains the wider range of phenotypes. In the LOAD, age-associated comorbidities explain many of the phenotypic differences.
Collapse
Affiliation(s)
- Tanzil Rujeedawa
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
| | - Eva Carrillo Félez
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
| | - Isabel C. H. Clare
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
- Cambridgeshire and Peterborough Foundation NHS Trust, Fulbourn CB21 5EF, UK
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, 08029 Barcelona, Spain
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK;
- South London and the Maudsley NHS Foundation Trust, The LonDowns Consortium, London SE5 8AZ, UK
| | | | - Antonia Coppus
- Department for Primary and Community Care, Department of Primary and Community Care (149 ELG), Radboud University Nijmegen Medical Center, P.O. Box 9101, 6525 GA Nijmegen, The Netherlands;
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, 80539 Munich, Germany;
- German Center for Neurodegenerative Diseases, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Shahid H. Zaman
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
- Cambridgeshire and Peterborough Foundation NHS Trust, Fulbourn CB21 5EF, UK
| |
Collapse
|
16
|
Novel Approaches to an Integrated Route for Trisomy 21 Evaluation. Biomolecules 2021; 11:biom11091328. [PMID: 34572541 PMCID: PMC8465311 DOI: 10.3390/biom11091328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/16/2021] [Accepted: 09/06/2021] [Indexed: 12/31/2022] Open
Abstract
Trisomy 21 (T21) is one of the most commonly occurring genetic disorders, caused by the partial or complete triplication of chromosome 21. Despite the significant progress in the diagnostic tools applied for prenatal screening, commonly used methods are still imprecise and involve invasive diagnostic procedures that are related to a maternal risk of miscarriage. In this case, novel prenatal biomarkers are still being evaluated using highly specialized techniques, which could increase the diagnostic usefulness of biochemical prenatal screening for T21. From the other hand, the T21′s pathogenesis, caused by the improper division of genetic material, disrupting many metabolic pathways, could be further evaluated with the use of omics methods, which could result in bringing relevant insights for the evaluation of potential medical targets. Accordingly, a literature search was undertaken to collect novel information about prenatal screening for Down syndrome with the use of advanced technology, with a particular emphasis on the evaluation of novel screening biomarkers and the discovery of potential medical targets. These meta-analyses are focused on novel approaches designed with the use of omics techniques, representing the most rapidly developing and promising field in research today. Considering the limitations and progress of these methods, the use of omics techniques in evaluating T21 pathogenesis could bring beneficial results in prenatal screening, simultaneously uncovering novel potential medical targets.
Collapse
|
17
|
Maciejewska K, Czarnecka K, Szymański P. A review of the mechanisms underlying selected comorbidities in Alzheimer's disease. Pharmacol Rep 2021; 73:1565-1581. [PMID: 34121170 PMCID: PMC8599320 DOI: 10.1007/s43440-021-00293-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the central nervous system (CNS) leading to mental deterioration and devastation, and eventually a fatal outcome. AD affects mostly the elderly. AD is frequently accompanied by hypercholesterolemia, hypertension, atherosclerosis, and diabetes mellitus, and these are significant risk factors of AD. Other conditions triggered by the progression of AD include psychosis, sleep disorders, epilepsy, and depression. One important comorbidity is Down’s syndrome, which directly contributes to the severity and rapid progression of AD. The development of new therapeutic strategies for AD includes the repurposing of drugs currently used for the treatment of comorbidities. A better understanding of the influence of comorbidities on the pathogenesis of AD, and the medications used in its treatment, might allow better control of disease progression, and more effective pharmacotherapy.
Collapse
Affiliation(s)
- Karolina Maciejewska
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Kamila Czarnecka
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St, 01-163, Warsaw, Poland
| | - Paweł Szymański
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland.
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St, 01-163, Warsaw, Poland.
| |
Collapse
|
18
|
Buczyńska A, Sidorkiewicz I, Ławicki S, Krętowski AJ, Zbucka-Krętowska M. Prenatal Screening of Trisomy 21: Could Oxidative Stress Markers Play a Role? J Clin Med 2021; 10:jcm10112382. [PMID: 34071365 PMCID: PMC8198847 DOI: 10.3390/jcm10112382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 11/23/2022] Open
Abstract
Despite significant progress in trisomy 21 (T21) diagnostic tools, amniocentesis is still used for the confirmation of an abnormal fetal karyotype. Invasive tests carry the potential risk of miscarriage; thus, screening biomarkers are commonly used before undergoing invasive procedures. In our study, we investigated the possible application of oxidative stress markers in the prenatal screening of trisomy 21. The DNA/RNA oxidative stress damage products (OSDPs), advanced glycation end (AGE) products, ischemia-modified albumin (IMA), alfa-1-antitrypsin (A1AT), asprosin, and vitamin D concentrations were measured in both maternal plasma and amniotic fluid in trisomy 21 (T21) and euploid pregnancies. The obtained results indicated increased levels of DNA/RNA OSDPs and asprosin with simultaneous decreased levels of vitamin D and A1AT in the study group. The diagnostic utility of the plasma measurement based on the area under the received operative characteristic (ROC) curve (AUC) calculation of asprosin (AUC = 0.965), IMA (AUC = 0.880), AGE (AUC = 0.846) and DNA/RNA OSDPs (AUC = 0.506) in T21 screening was demonstrated. The obtained results indicate a potential role for the application of oxidative stress markers in the prenatal screening of T21 with the highest screening utility of plasma asprosin.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (A.B.); (I.S.); (A.J.K.)
| | - Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (A.B.); (I.S.); (A.J.K.)
| | - Sławomir Ławicki
- Department of Population Medicine and Civilization Diseases Prevention, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (A.B.); (I.S.); (A.J.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Monika Zbucka-Krętowska
- Department of Gynecological Endocrinology and Adolescent Gynecology, Medical University of Bialystok, 15-276 Bialystok, Poland
- Correspondence: ; Tel.: +48 85-746-83-36
| |
Collapse
|
19
|
Ferrari M, Stagi S. Oxidative Stress in Down and Williams-Beuren Syndromes: An Overview. Molecules 2021; 26:molecules26113139. [PMID: 34073948 PMCID: PMC8197362 DOI: 10.3390/molecules26113139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress is the result of an imbalance in the redox state in a cell or a tissue. When the production of free radicals, which are physiologically essential for signaling, exceeds the antioxidant capability, pathological outcomes including oxidative damage to macromolecules, aberrant signaling, and inflammation can occur. Down syndrome (DS) and Williams-Beuren syndrome (WBS) are well-known and common genetic conditions with multi-systemic involvement. Their etiology is linked to oxidative stress with important causative genes, such as SOD-1 and NCF-1, respectively, of the diseases being primarily involved in the regulation of the redox state. Early aging, dementia, autoimmunity, and chronic inflammation are some of the main characteristics of these conditions that can be associated with oxidative stress. In recent decades, there has been a growing interest in the possible role of oxidative stress and inflammation in the pathology of these conditions. However, at present, few studies have investigated these correlations. We provide an overview of the current literature concerning the role of oxidative stress and oxidative damage in genetic syndromes with a focus on Down syndrome and WBS. We hope to provide new insights to improve the management of complications related to these diseases.
Collapse
|
20
|
Illouz T, Nicola R, Ben-Shushan L, Madar R, Biragyn A, Okun E. Maternal antibodies facilitate Amyloid-β clearance by activating Fc-receptor-Syk-mediated phagocytosis. Commun Biol 2021; 4:329. [PMID: 33712740 PMCID: PMC7955073 DOI: 10.1038/s42003-021-01851-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Maternal antibodies (MAbs) protect against infections in immunologically-immature neonates. Maternally transferred immunity may also be harnessed to target diseases associated with endogenous protein misfolding and aggregation, such as Alzheimer's disease (AD) and AD-pathology in Down syndrome (DS). While familial early-onset AD (fEOAD) is associated with autosomal dominant mutations in the APP, PSEN1,2 genes, promoting cerebral Amyloid-β (Aβ) deposition, DS features a life-long overexpression of the APP and DYRK1A genes, leading to a cognitive decline mediated by Aβ overproduction and tau hyperphosphorylation. Although no prenatal screening for fEOAD-related mutations is in clinical practice, DS can be diagnosed in utero. We hypothesized that anti-Aβ MAbs might promote the removal of early Aβ accumulation in the central nervous system of human APP-expressing mice. To this end, a DNA-vaccine expressing Aβ1-11 was delivered to wild-type female mice, followed by mating with 5xFAD males, which exhibit early Aβ plaque formation. MAbs reduce the offspring's cortical Aβ levels 4 months after antibodies were undetectable, along with alleviating short-term memory deficits. MAbs elicit a long-term shift in microglial phenotype in a mechanism involving activation of the FcγR1/Syk/Cofilin pathway. These data suggest that maternal immunization can alleviate cognitive decline mediated by early Aβ deposition, as occurs in EOAD and DS.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar-Ilan University, Ramat Gan, Israel
| | - Raneen Nicola
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar-Ilan University, Ramat Gan, Israel
| | - Linoy Ben-Shushan
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar-Ilan University, Ramat Gan, Israel
- The Mina and Everard Goodman faculty of Life sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Ravit Madar
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar-Ilan University, Ramat Gan, Israel
- The Mina and Everard Goodman faculty of Life sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel.
- The Paul Feder Laboratory on Alzheimer's disease research, Bar-Ilan University, Ramat Gan, Israel.
- The Mina and Everard Goodman faculty of Life sciences, Bar-Ilan University, Ramat Gan, Israel.
| |
Collapse
|
21
|
Lanzillotta C, Di Domenico F. Stress Responses in Down Syndrome Neurodegeneration: State of the Art and Therapeutic Molecules. Biomolecules 2021; 11:biom11020266. [PMID: 33670211 PMCID: PMC7916967 DOI: 10.3390/biom11020266] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Down syndrome (DS) is the most common genomic disorder characterized by the increased incidence of developing early Alzheimer’s disease (AD). In DS, the triplication of genes on chromosome 21 is intimately associated with the increase of AD pathological hallmarks and with the development of brain redox imbalance and aberrant proteostasis. Increasing evidence has recently shown that oxidative stress (OS), associated with mitochondrial dysfunction and with the failure of antioxidant responses (e.g., SOD1 and Nrf2), is an early signature of DS, promoting protein oxidation and the formation of toxic protein aggregates. In turn, systems involved in the surveillance of protein synthesis/folding/degradation mechanisms, such as the integrated stress response (ISR), the unfolded stress response (UPR), and autophagy, are impaired in DS, thus exacerbating brain damage. A number of pre-clinical and clinical studies have been applied to the context of DS with the aim of rescuing redox balance and proteostasis by boosting the antioxidant response and/or inducing the mechanisms of protein re-folding and clearance, and at final of reducing cognitive decline. So far, such therapeutic approaches demonstrated their efficacy in reverting several aspects of DS phenotype in murine models, however, additional studies aimed to translate these approaches in clinical practice are still needed.
Collapse
|
22
|
Abstract
Proteins succumb to numerous post-translational modifications (PTMs). These relate to enzymatic or non-enzymatic reactions taking place in either the intracellular or extracellular compartment. While intracellular oxidative changes are mainly due to redox stress, extracellular PTMs may be induced in an inflammatory micro milieu that is rich in reactive species. The increasing recognition of oxidative modifications as a causing agent or side-effect of pathophysiological states and diseases puts oxidative PTMS (oxPTMs) into the spotlight of inflammation research. Pathological hyper-modification of proteins can lead to accumulation, aggregation, cell stress, altered antigenic peptides, and damage-associated molecular pattern (DAMP)-like recognition by host immunity. Such processes are linked to cardiovascular disease and autoinflammation. At the same time, a detailed understanding of the mechanisms governing inflammatory responses to oxPTMs may capitalize on new therapeutic routes for enhancing adaptive immune responses as needed, for instance, in oncology. We here summarize some of the latest developments of oxPTMs in disease diagnosis and therapy. Potential target proteins and upcoming technologies, such as gas plasmas, are outlined for future research that may aid in identifying the molecular basis of immunogenic vs. tolerogenic oxPTMs.
Collapse
|
23
|
The BACH1/Nrf2 Axis in Brain in Down Syndrome and Transition to Alzheimer Disease-Like Neuropathology and Dementia. Antioxidants (Basel) 2020; 9:antiox9090779. [PMID: 32839417 PMCID: PMC7554729 DOI: 10.3390/antiox9090779] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is the most common genetic cause of intellectual disability that is associated with an increased risk to develop early-onset Alzheimer-like dementia (AD). The brain neuropathological features include alteration of redox homeostasis, mitochondrial deficits, inflammation, accumulation of both amyloid beta-peptide oligomers and senile plaques, as well as aggregated hyperphosphorylated tau protein-containing neurofibrillary tangles, among others. It is worth mentioning that some of the triplicated genes encoded are likely to cause increased oxidative stress (OS) conditions that are also associated with reduced cellular responses. Published studies from our laboratories propose that increased oxidative damage occurs early in life in DS population and contributes to age-dependent neurodegeneration. This is the result of damaged, oxidized proteins that belong to degradative systems, antioxidant defense system, neuronal trafficking. and energy metabolism. This review focuses on a key element that regulates redox homeostasis, the transcription factor Nrf2, which is negatively regulated by BACH1, encoded on chromosome 21. The role of the Nrf2/BACH1 axis in DS is under investigation, and the effects of triplicated BACH1 on the transcriptional regulation of Nrf2 are still unknown. In this review, we discuss the physiological relevance of BACH1/Nrf2 signaling in the brain and how the dysfunction of this system affects the redox homeostasis in DS neurons and how this axis may contribute to the transition of DS into DS with AD neuropathology and dementia. Further, some of the evidence collected in AD regarding the potential contribution of BACH1 to neurodegeneration in DS are also discussed.
Collapse
|
24
|
O'Bryant SE, Zhang F, Silverman W, Lee JH, Krinsky‐McHale SJ, Pang D, Hall J, Schupf N. Proteomic profiles of incident mild cognitive impairment and Alzheimer's disease among adults with Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12033. [PMID: 32490140 PMCID: PMC7241058 DOI: 10.1002/dad2.12033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 11/09/2022]
Abstract
INTRODUCTION We sought to determine if proteomic profiles could predict risk for incident mild cognitive impairment (MCI) and Alzheimer's disease (AD) among adults with Down syndrome (DS). METHODS In a cohort of 398 adults with DS, a total of n = 186 participants were determined to be non-demented and without MCI or AD at baseline and throughout follow-up; n = 103 had incident MCI and n = 81 had incident AD. Proteomics were conducted on banked plasma samples from a previously generated algorithm. RESULTS The proteomic profile was highly accurate in predicting incident MCI (area under the curve [AUC] = 0.92) and incident AD (AUC = 0.88). For MCI risk, the support vector machine (SVM)-based high/low cut-point yielded an adjusted hazard ratio (HR) = 6.46 (P < .001). For AD risk, the SVM-based high/low cut-point score yielded an adjusted HR = 8.4 (P < .001). DISCUSSION The current results provide support for our blood-based proteomic profile for predicting risk for MCI and AD among adults with DS.
Collapse
Affiliation(s)
- Sid E. O'Bryant
- Department of Pharmacology & Neuroscience I Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Fan Zhang
- Vermont Genetics NetworkUniversity of VermontBurlingtonVermontUSA
| | | | - Joseph H. Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- G.H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Department of EpidemiologyMailman School of Public Health Columbia UniversityNew YorkNew YorkUSA
| | - Sharon J. Krinsky‐McHale
- Department of PsychologyStaten IslandNYS Institute for Basic Research in Developmental DisabilitiesNew YorkNew YorkUSA
| | - Deborah Pang
- Department of PsychologyStaten IslandNYS Institute for Basic Research in Developmental DisabilitiesNew YorkNew YorkUSA
| | - James Hall
- Department of Pharmacology & Neuroscience I Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- G.H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Department of EpidemiologyMailman School of Public Health Columbia UniversityNew YorkNew YorkUSA
- Departments of Neurology and PsychiatryColumbia University Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
25
|
Amuno S, Shekh K, Kodzhahinchev V, Niyogi S. Neuropathological changes in wild muskrats (Ondatra zibethicus) and red squirrels (Tamiasciurus hudsonicus) breeding in arsenic endemic areas of Yellowknife, Northwest Territories (Canada): Arsenic and cadmium accumulation in the brain and biomarkers of oxidative stress. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 704:135426. [PMID: 31822412 DOI: 10.1016/j.scitotenv.2019.135426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/03/2019] [Accepted: 11/06/2019] [Indexed: 06/10/2023]
Abstract
The brain is one of the critical organs particularly susceptible to the damaging effects of chronic arsenic poisoning and there is a growing body of evidence that suggest that oxidative stress plays a key role in the pathogenesis of neurodegenerative disorders. The aim of this present work was to comparatively assess biomarkers of oxidative stress and status of antioxidant enzyme activities in the brains of muskrats and squirrels breeding in arsenic endemic areas, specifically near the vicinity of the abandoned Giant mine site (~2 km radius), and an intermediate location approximately 20 km from the mine area and in reference locations spanning 52-105 km from the city of Yellowknife, Northwest Territories (Canada). Analysis included measurement of total arsenic and cadmium concentration in the nails, brain, and stomach content of muskrats and squirrels, in addition to biochemical evaluation of lipid peroxidation levels and antioxidant enzymes defense: catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx) in the brain tissues. The results revealed that arsenic concentration in the nails of muskrats collected closest to the vicinity of the mine area was in the range of 11 to 35.1 times higher than those from the reference site. The maximum concentration of arsenic in the nails of muskrats from the intermediate location was 47.6 times higher than the maximum concentration observed in the reference muskrats. Cadmium was generally undetected in the nails of muskrats and squirrels from the three sampling locations. Arsenic in the gut contents of muskrats from the arsenic affected area was 4.5 to 49.1 times higher than those from the reference site. Cadmium levels in the guts of muskrats from the mine area almost doubled those from the reference site. Arsenic accumulated in the nails of squirrels from the areas closest to the mine but was undetected in the squirrel nails from the reference location. The maximum arsenic levels in the stomach content of squirrels from the mine area was ~40 times higher than those from the reference site. Arsenic did not accumulate in the brains of muskrats, but cadmium was detected in a few brains of muskrats. Brains of squirrels from the mine area and intermediate locations accumulated both arsenic and cadmium. The brains of squirrels and muskrats from the arsenic affected area showed no evidence of increased lipid peroxidation compared to the animals from the reference site. However, SOD, CAT and GPx activities in the brains of animals from the arsenic endemic areas tended to be higher compared to the control sites. This is the first study documenting evidence of oxidative stress and altered antioxidant enzyme activities in brains of wild rodent population in arsenic endemic areas of Canada.
Collapse
Affiliation(s)
- S Amuno
- School of Environment and Sustainability, University of Saskatchewan, Saskatoon, Canada.
| | - K Shekh
- Department of Biology, University of Saskatchewan, Saskatoon, Canada; Toxicology Centre, University of Saskatchewan, Saskatoon, Canada
| | - V Kodzhahinchev
- Department of Biology, University of Saskatchewan, Saskatoon, Canada
| | - S Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, Canada; Toxicology Centre, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
26
|
Tramutola A, Lanzillotta C, Di Domenico F, Head E, Butterfield DA, Perluigi M, Barone E. Brain insulin resistance triggers early onset Alzheimer disease in Down syndrome. Neurobiol Dis 2020; 137:104772. [PMID: 31987911 DOI: 10.1016/j.nbd.2020.104772] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/03/2020] [Accepted: 01/23/2020] [Indexed: 01/08/2023] Open
Abstract
Dysregulation of insulin signaling pathway with reduced downstream neuronal survival and plasticity mechanisms is a fundamental abnormality observed in Alzheimer's disease (AD) brain. This phenomenon, known as brain insulin resistance, is associated with poor cognitive performance and is driven by the uncoupling of insulin receptor (IR) from its direct substrate (IRS1). Considering that Down syndrome (DS) and AD neuropathology share many common features, we investigated metabolic aspects of neurodegeneration, i.e., brain insulin resistance, in DS and whether it would contribute to early onset AD in DS population. Changes of levels and activation of main brain proteins belonging to the insulin signaling pathway (i.e., IR, IRS1, PTEN, GSK3β, PKCζ, AS160, GLUT4) were evaluated. Furthermore, we analyzed whether changes of these proteins were associated with alterations of: (i) proteins regulating brain energy metabolism; (ii) APP cleavage; and (ii) regulation of synaptic plasticity mechanisms in post-mortem brain samples collected from people with DS before and after the development of AD pathology (DSAD) compared with their age-matched controls. We found that DS cases were characterized by key markers of brain insulin resistance (reduced IR and increased IRS1 inhibition) early in life. Furthermore, downstream from IRS1, an overall uncoupling among the proteins of insulin signaling was observed. Dysregulated brain insulin signaling was associated with reduced hexokinase II (HKII) levels and proteins associated with mitochondrial complexes levels as well as with reduced levels of syntaxin in DS cases. Tellingly, these alterations precede the development of AD neuropathology and clinical presentations in DS. We propose that markers of brain insulin resistance rise earlier with age in DS compared with the general population and may contribute to the cognitive impairment associated with the early development of AD in DS.
Collapse
Affiliation(s)
- Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - D Allan Butterfield
- Department of Chemistry, Markey Cancer Center, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy.
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy.
| |
Collapse
|
27
|
Abstract
Alzheimer disease (AD) is a major cause of age-related dementia. We do not fully understand AD aetiology and pathogenesis, but oxidative damage is a key component. The brain mostly uses glucose for energy, but in AD and amnestic mild cognitive impairment glucose metabolism is dramatically decreased, probably owing, at least in part, to oxidative damage to enzymes involved in glycolysis, the tricarboxylic acid cycle and ATP biosynthesis. Consequently, ATP-requiring processes for cognitive function are impaired, and synaptic dysfunction and neuronal death result, with ensuing thinning of key brain areas. We summarize current research on the interplay and sequence of these processes and suggest potential pharmacological interventions to retard AD progression.
Collapse
|
28
|
Yang B, Fritsche KL, Beversdorf DQ, Gu Z, Lee JC, Folk WR, Greenlief CM, Sun GY. Yin-Yang Mechanisms Regulating Lipid Peroxidation of Docosahexaenoic Acid and Arachidonic Acid in the Central Nervous System. Front Neurol 2019; 10:642. [PMID: 31275232 PMCID: PMC6591372 DOI: 10.3389/fneur.2019.00642] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/31/2019] [Indexed: 12/11/2022] Open
Abstract
Phospholipids in the central nervous system (CNS) are rich in polyunsaturated fatty acids (PUFAs), particularly arachidonic acid (ARA) and docosahexaenoic acid (DHA). Besides providing physical properties to cell membranes, these PUFAs are metabolically active and undergo turnover through the “deacylation-reacylation (Land's) cycle”. Recent studies suggest a Yin-Yang mechanism for metabolism of ARA and DHA, largely due to different phospholipases A2 (PLA2s) mediating their release. ARA and DHA are substrates of cyclooxygenases and lipoxygenases resulting in an array of lipid mediators, which are pro-inflammatory and pro-resolving. The PUFAs are susceptible to peroxidation by oxygen free radicals, resulting in the production of 4-hydroxynonenal (4-HNE) from ARA and 4-hydroxyhexenal (4-HHE) from DHA. These alkenal electrophiles are reactive and capable of forming adducts with proteins, phospholipids and nucleic acids. The perceived cytotoxic and hormetic effects of these hydroxyl-alkenals have impacted cell signaling pathways, glucose metabolism and mitochondrial functions in chronic and inflammatory diseases. Due to the high levels of DHA and ARA in brain phospholipids, this review is aimed at providing information on the Yin-Yang mechanisms for regulating these PUFAs and their lipid peroxidation products in the CNS, and implications of their roles in neurological disorders.
Collapse
Affiliation(s)
- Bo Yang
- Department of Chemistry, University of Missouri, Columbia, MO, United States
| | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - David Q Beversdorf
- Departments of Radiology, Neurology and Psychological Sciences, and the Thompson Center, Columbia, MO, United States
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, United States
| | - James C Lee
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - William R Folk
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| | - C Michael Greenlief
- Department of Chemistry, University of Missouri, Columbia, MO, United States
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| |
Collapse
|
29
|
Nunes J, Charneira C, Morello J, Rodrigues J, Pereira SA, Antunes AMM. Mass Spectrometry-Based Methodologies for Targeted and Untargeted Identification of Protein Covalent Adducts (Adductomics): Current Status and Challenges. High Throughput 2019; 8:ht8020009. [PMID: 31018479 PMCID: PMC6631461 DOI: 10.3390/ht8020009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/18/2019] [Accepted: 04/20/2019] [Indexed: 12/12/2022] Open
Abstract
Protein covalent adducts formed upon exposure to reactive (mainly electrophilic) chemicals may lead to the development of a wide range of deleterious health outcomes. Therefore, the identification of protein covalent adducts constitutes a huge opportunity for a better understanding of events underlying diseases and for the development of biomarkers which may constitute effective tools for disease diagnosis/prognosis, for the application of personalized medicine approaches and for accurately assessing human exposure to chemical toxicants. The currently available mass spectrometry (MS)-based methodologies, are clearly the most suitable for the analysis of protein covalent modifications, providing accuracy, sensitivity, unbiased identification of the modified residue and conjugates along with quantitative information. However, despite the huge technological advances in MS instrumentation and bioinformatics tools, the identification of low abundant protein covalent adducts is still challenging. This review is aimed at summarizing the MS-based methodologies currently used for the identification of protein covalent adducts and the strategies developed to overcome the analytical challenges, involving not only sample pre-treatment procedures but also distinct MS and data analysis approaches.
Collapse
Affiliation(s)
- João Nunes
- Centro de Química Estrutural, Instituto Superior Técnico, ULisboa, 1049-001 Lisboa, Portugal.
| | - Catarina Charneira
- Centro de Química Estrutural, Instituto Superior Técnico, ULisboa, 1049-001 Lisboa, Portugal.
| | - Judit Morello
- Centro de Química Estrutural, Instituto Superior Técnico, ULisboa, 1049-001 Lisboa, Portugal.
| | - João Rodrigues
- Clarify Analytical, Rua dos Mercadores 128A, 7000-872 Évora, Portugal.
| | - Sofia A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-006 Lisboa, Portugal.
| | - Alexandra M M Antunes
- Centro de Química Estrutural, Instituto Superior Técnico, ULisboa, 1049-001 Lisboa, Portugal.
| |
Collapse
|
30
|
Di Domenico F, Tramutola A, Barone E, Lanzillotta C, Defever O, Arena A, Zuliani I, Foppoli C, Iavarone F, Vincenzoni F, Castagnola M, Butterfield DA, Perluigi M. Restoration of aberrant mTOR signaling by intranasal rapamycin reduces oxidative damage: Focus on HNE-modified proteins in a mouse model of down syndrome. Redox Biol 2019; 23:101162. [PMID: 30876754 PMCID: PMC6859577 DOI: 10.1016/j.redox.2019.101162] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/26/2019] [Accepted: 03/05/2019] [Indexed: 01/05/2023] Open
Abstract
Increasing evidences support the notion that the impairment of intracellular degradative machinery is responsible for the accumulation of oxidized/misfolded proteins that ultimately results in the deposition of protein aggregates. These events are key pathological aspects of "protein misfolding diseases", including Alzheimer disease (AD). Interestingly, Down syndrome (DS) neuropathology shares many features with AD, such as the deposition of both amyloid plaques and neurofibrillary tangles. Studies from our group and others demonstrated, in DS brain, the dysfunction of both proteasome and autophagy degradative systems, coupled with increased oxidative damage. Further, we observed the aberrant increase of mTOR signaling and of its down-stream pathways in both DS brain and in Ts65Dn mice. Based on these findings, we support the ability of intranasal rapamycin treatment (InRapa) to restore mTOR pathway but also to restrain oxidative stress resulting in the decreased accumulation of lipoxidized proteins. By proteomics approach, we were able to identify specific proteins that showed decreased levels of HNE-modification after InRapa treatment compared with vehicle group. Among MS-identified proteins, we found that reduced oxidation of arginase-1 (ARG-1) and protein phosphatase 2A (PP2A) might play a key role in reducing brain damage associated with synaptic transmission failure and tau hyperphosphorylation. InRapa treatment, by reducing ARG-1 protein-bound HNE levels, rescues its enzyme activity and conceivably contribute to the recovery of arginase-regulated functions. Further, it was shown that PP2A inhibition induces tau hyperphosphorylation and spatial memory deficits. Our data suggest that InRapa was able to rescue PP2A activity as suggested by reduced p-tau levels. In summary, considering that mTOR pathway is a central hub of multiple intracellular signaling, we propose that InRapa treatment is able to lower the lipoxidation-mediated damage to proteins, thus representing a valuable therapeutic strategy to reduce the early development of AD pathology in DS population.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy; Universidad Autònoma de Chile, Instituto de Ciencias Biomédicas, Facultad de alud, Providencia, Santiago, Chile
| | - Chiara Lanzillotta
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Olivia Defever
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Andrea Arena
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Ilaria Zuliani
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Cesira Foppoli
- CNR Institute of Molecular Biology and Pathology, Sapienza University of Rome, Rome, Italy
| | - Federica Iavarone
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Federica Vincenzoni
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Massimo Castagnola
- Laboratorio di Proteomica e Metabonomica, IRCCS, Fondazione Santa Lucia - Rome and Istituto per la Chimica del Riconoscimento Molecolare, CNR, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
31
|
Li X, Tang Y, Li J, Hu X, Yin C, Yang Z, Wang Q, Wu Z, Lu X, Wang W, Huang W, Fan Q. A small-molecule probe for ratiometric photoacoustic imaging of hydrogen sulfide in living mice. Chem Commun (Camb) 2019; 55:5934-5937. [DOI: 10.1039/c9cc02224d] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A small molecule ratiometric photoacoustic probe was developed for real-time monitoring of hydrogen sulfide in living mice.
Collapse
|
32
|
Tramutola A, Lanzillotta C, Barone E, Arena A, Zuliani I, Mosca L, Blarzino C, Butterfield DA, Perluigi M, Di Domenico F. Intranasal rapamycin ameliorates Alzheimer-like cognitive decline in a mouse model of Down syndrome. Transl Neurodegener 2018; 7:28. [PMID: 30410750 PMCID: PMC6218962 DOI: 10.1186/s40035-018-0133-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023] Open
Abstract
Background Down syndrome (DS) individuals, by the age of 40s, are at increased risk to develop Alzheimer-like dementia, with deposition in brain of senile plaques and neurofibrillary tangles. Our laboratory recently demonstrated the disturbance of PI3K/AKT/mTOR axis in DS brain, prior and after the development of Alzheimer Disease (AD). The aberrant modulation of the mTOR signalling in DS and AD age-related cognitive decline affects crucial neuronal pathways, including insulin signaling and autophagy, involved in pathology onset and progression. Within this context, the therapeutic use of mTOR-inhibitors may prevent/attenuate the neurodegenerative phenomena. By our work we aimed to rescue mTOR signalling in DS mice by a novel rapamycin intranasal administration protocol (InRapa) that maximizes brain delivery and reduce systemic side effects. Methods Ts65Dn mice were administered with InRapa for 12 weeks, starting at 6 months of age demonstrating, at the end of the treatment by radial arms maze and novel object recognition testing, rescued cognition. Results The analysis of mTOR signalling, after InRapa, demonstrated in Ts65Dn mice hippocampus the inhibition of mTOR (reduced to physiological levels), which led, through the rescue of autophagy and insulin signalling, to reduced APP levels, APP processing and APP metabolites production, as well as, to reduced tau hyperphosphorylation. In addition, a reduction of oxidative stress markers was also observed. Discussion These findings demonstrate that chronic InRapa administration is able to exert a neuroprotective effect on Ts65Dn hippocampus by reducing AD pathological hallmarks and by restoring protein homeostasis, thus ultimately resulting in improved cognition. Results are discussed in term of a potential novel targeted therapeutic approach to reduce cognitive decline and AD-like neuropathology in DS individuals.
Collapse
Affiliation(s)
- Antonella Tramutola
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Chiara Lanzillotta
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Eugenio Barone
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,2Universidad Autònoma de Chile, Instituto de Ciencias Biomédicas, Facultad de alud, Avenida Pedro de Valdivia 425, Providencia, Santiago, Chile
| | - Andrea Arena
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Ilaria Zuliani
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Luciana Mosca
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Carla Blarzino
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - D Allan Butterfield
- 3Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055 USA
| | - Marzia Perluigi
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Fabio Di Domenico
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
33
|
Butterfield DA. Perspectives on Oxidative Stress in Alzheimer’s Disease and Predictions of Future Research Emphases. J Alzheimers Dis 2018; 64:S469-S479. [DOI: 10.3233/jad-179912] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
34
|
Gebre AK, Altaye BM, Atey TM, Tuem KB, Berhe DF. Targeting Renin-Angiotensin System Against Alzheimer's Disease. Front Pharmacol 2018; 9:440. [PMID: 29760662 PMCID: PMC5937164 DOI: 10.3389/fphar.2018.00440] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
Renin Angiotensin System (RAS) is a hormonal system that regulates blood pressure and fluid balance through a coordinated action of renal, cardiovascular, and central nervous systems. In addition to its hemodynamic regulatory role, RAS involves in many brain activities, including memory acquisition and consolidation. This review has summarized the involvement of RAS in the pathology of Alzheimer’s disease (AD), and the outcomes of treatment with RAS inhibitors. We have discussed the effect of brain RAS in the amyloid plaque (Aβ) deposition, oxidative stress, neuroinflammation, and vascular pathology which are directly and indirectly associated with AD. Angiotensin II (AngII) via AT1 receptor is reported to increase brain Aβ level via different mechanisms including increasing amyloid precursor protein (APP) mRNA, β-secretase activity, and presenilin expression. Similarly, it was associated with tau phosphorylation, and reactive oxygen species generation. However, these effects are counterbalanced by Ang II mediated AT2 signaling. The protective effect observed with angiotensin receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACEIs) could be as the result of inhibition of Ang II signaling. ARBs also offer additional benefit by shifting the effect of Ang II toward AT2 receptor. To conclude, targeting RAS in the brain may benefit patients with AD though it still requires further in depth understanding.
Collapse
Affiliation(s)
- Abadi Kahsu Gebre
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Birhanetensay Masresha Altaye
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Tesfay Mehari Atey
- Clinical Pharmacy Unit, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Kald Beshir Tuem
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Derbew Fikadu Berhe
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
35
|
Butterfield DA, Boyd-Kimball D. Oxidative Stress, Amyloid-β Peptide, and Altered Key Molecular Pathways in the Pathogenesis and Progression of Alzheimer's Disease. J Alzheimers Dis 2018; 62:1345-1367. [PMID: 29562527 PMCID: PMC5870019 DOI: 10.3233/jad-170543] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2017] [Indexed: 12/12/2022]
Abstract
Oxidative stress is implicated in the pathogenesis and progression of Alzheimer's disease (AD) and its earlier stage, amnestic mild cognitive impairment (aMCI). One source of oxidative stress in AD and aMCI brains is that associated with amyloid-β peptide, Aβ1-42 oligomers. Our laboratory first showed in AD elevated oxidative stress occurred in brain regions rich in Aβ1-42, but not in Aβ1-42-poor regions, and was among the first to demonstrate Aβ peptides led to lipid peroxidation (indexed by HNE) in AD and aMCI brains. Oxidatively modified proteins have decreased function and contribute to damaged key biochemical and metabolic pathways in which these proteins normally play a role. Identification of oxidatively modified brain proteins by the methods of redox proteomics was pioneered in the Butterfield laboratory. Four recurring altered pathways secondary to oxidative damage in brain from persons with AD, aMCI, or Down syndrome with AD are interrelated and contribute to neuronal death. This "Quadrilateral of Neuronal Death" includes altered: glucose metabolism, mTOR activation, proteostasis network, and protein phosphorylation. Some of these pathways are altered even in brains of persons with preclinical AD. We opine that targeting these pathways pharmacologically and with lifestyle changes potentially may provide strategies to slow or perhaps one day, prevent, progression or development of this devastating dementing disorder. This invited review outlines both in vitro and in vivo studies from the Butterfield laboratory related to Aβ1-42 and AD and discusses the importance and implications of some of the major achievements of the Butterfield laboratory in AD research.
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Debra Boyd-Kimball
- Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH, USA
| |
Collapse
|
36
|
Barone E, Arena A, Head E, Butterfield DA, Perluigi M. Disturbance of redox homeostasis in Down Syndrome: Role of iron dysmetabolism. Free Radic Biol Med 2018; 114:84-93. [PMID: 28705658 PMCID: PMC5748256 DOI: 10.1016/j.freeradbiomed.2017.07.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 02/08/2023]
Abstract
Down Syndrome (DS) is the most common genetic form of intellectual disability that leads in the majority of cases to development of early-onset Alzheimer-like dementia (AD). The neuropathology of DS has several common features with AD including alteration of redox homeostasis, mitochondrial deficits, and inflammation among others. Interestingly, some of the genes encoded by chromosome 21 are responsible of increased oxidative stress (OS) conditions that are further exacerbated by decreased antioxidant defense. Previous studies from our groups showed that accumulation of oxidative damage is an early event in DS neurodegeneration and that oxidative modifications of selected proteins affects the integrity of the protein degradative systems, antioxidant response, neuronal integrity and energy metabolism. In particular, the current review elaborates recent findings demonstrating the accumulation of oxidative damage in DS and we focus attention on specific deregulation of iron metabolism, which affects both the central nervous system and the periphery. Iron dysmetabolism is a well-recognized factor that contributes to neurodegeneration; thus we opine that better understanding how and to what extent the concerted loss of iron dyshomeostasis and increased OS occur in DS could provide novel insights for the development of therapeutic strategies for the treatment of Alzheimer-like dementia.
Collapse
Affiliation(s)
- Eugenio Barone
- Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Rome, Italy; Universidad Autónoma de Chile, Instituto de Ciencias Biomédicas, Facultad de alud, Avenida Pedro de Valdivia 425, Providencia, Santiago, Chile
| | - Andrea Arena
- Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Rome, Italy
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA; Department of Neurology, University of Kentucky, Lexington, KY 40506 USA
| | - D Allan Butterfield
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA; Department of Chemistry, University of Kentucky, Lexington, KY 40506 USA
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Rome, Italy.
| |
Collapse
|
37
|
Valacchi G, Pecorelli A, Cervellati C, Hayek J. 4-hydroxynonenal protein adducts: Key mediator in Rett syndrome oxinflammation. Free Radic Biol Med 2017; 111:270-280. [PMID: 28063942 DOI: 10.1016/j.freeradbiomed.2016.12.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 12/24/2016] [Accepted: 12/30/2016] [Indexed: 12/13/2022]
Abstract
In the last 15 years a strong correlation between oxidative stress (OxS) and Rett syndrome (RTT), a rare neurodevelopmental disorder known to be caused in 95% of the cases, by a mutation in the methyl-CpG-binding protein 2 (MECP2) gene, has been well documented. Here, we revised, summarized and discussed the current knowledge on the role of lipid peroxidation byproducts, with special emphasis on 4-hydroxynonenal (4HNE), in RTT pathophysiology. The posttranslational modifications of proteins via 4HNE, known as 4HNE protein adducts (4NHE-PAs), causing detrimental effects on protein functions, appear to contribute to the clinical severity of the syndrome, since their levels increase significantly during the subsequent 4 clinical stages, reaching the maximum degree at stage 4, represented by a late motor deterioration. In addition, 4HNE-PA are only partially removed due to the compromised functionality of the proteasome activity, contributing therefore to the cellular damage in RTT. All this will lead to a characteristic subclinical inflammation, defined "OxInflammation", derived by a positive feedback loop between OxS byproducts and inflammatory mediators that in a long run further aggravates the clinical features of RTT patients. Therefore, in a pathology completely orphan of any therapy, aiming 4HNE as a therapeutic target could represent a coadjuvant treatment with some beneficial impact in these patients..
Collapse
Affiliation(s)
- Giuseppe Valacchi
- Plants for Human Health Institute, Department of Animal Sciences, NC State University, NC Research Campus, 600 Laureate Way, Kannapolis, NC 28081, USA; Department of Life Sciences and Biotechnology, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy.
| | - Alessandra Pecorelli
- Plants for Human Health Institute, Department of Animal Sciences, NC State University, NC Research Campus, 600 Laureate Way, Kannapolis, NC 28081, USA; Department of Life Sciences and Biotechnology, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Carlo Cervellati
- Department of Biomedical and Specialist Surgical Sciences, Section of Medical Biochemistry, Molecular Biology and Genetics, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| | - Joussef Hayek
- Child Neuropsychiatry Unit, University Hospital, AOUS, Viale Mario Bracci, 53100 Siena, Italy
| |
Collapse
|
38
|
Poli G, Zarkovic N. Editorial Introduction to the Special Issue on 4-Hydroxynonenal and Related Lipid Oxidation Products. Free Radic Biol Med 2017; 111:2-5. [PMID: 28576671 DOI: 10.1016/j.freeradbiomed.2017.05.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Giuseppe Poli
- Department of Clinical and Biological Sciences, School of Medicine, University of Torino, Italy
| | - Neven Zarkovic
- Laboratory for Oxidative Stress (LabOS), Rudjer Boskovic Institute, Bijenicka 54, HR-1000 Zagreb, Croatia.
| |
Collapse
|