1
|
Gene-Morales J, Juesas A, Saez-Berlanga A, Martin EG, Garrigues-Pelufo L, Sandoval-Camargo BS, Martin-Rivera F, Chulvi-Medrano I, Jiménez-Martínez P, Alix-Fages C, Gargallo P, Fernandez-Garrido J, Caballero O, Jerez-Martínez A, Colado JC. Dietary Nucleotides Enhance Neurogenesis, Cognitive Capacity, Muscle Function, and Body Composition in Older Adults: A Randomized, Triple-Blind, Controlled Clinical Trial. Nutrients 2025; 17:1431. [PMID: 40362739 PMCID: PMC12073346 DOI: 10.3390/nu17091431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES this study evaluated the differential effects of two distinct dietary nucleotide supplements, combined with spontaneous physical activity, on neuromuscular, cognitive, and metabolic adaptations in older adults. METHODS Sixty-nine physically independent older adults (aged 60-75 years) were randomly assigned to three groups: (1) a yeast nucleotides formulation (YN) standardized in a high content of free nucleotides (>40%) rich in all macro and micro nutrients naturally occurring in yeast cell (amino acids, minerals and B-group vitamin); (2) a neuro-based formulation (NF) consisting of a blend of monophosphate nucleotides 5'; or (3) a placebo. Participants maintained their spontaneous physical activities without structured exercise during a 10-week intervention. Assessments included physical function, cognitive performance, body composition, quality of life, and serum biomarkers of oxidative stress, inflammation, and neurogenesis. RESULTS Both nucleotide-supplemented groups demonstrated significant improvements compared to placebo in physical performance (p ≤ 0.045), cognitive function (Trail Making Test B [TMT-B]: p ≤ 0.012), oxidative stress biomarkers (p ≤ 0.048), inflammatory cytokines (p ≤ 0.023), and quality-of-life parameters (p ≤ 0.047). Body composition remained stable in supplemented groups, whereas placebo increased fat mass (5.04%) and decreased muscle mass (-2.18%). CONCLUSIONS Dietary nucleotide supplementation enhances the benefits of spontaneous physical activity across all measured variables in older adults, highlighting nucleotides as promising nutritional support for healthy aging. YN exhibited a trend toward greater inflammatory modulation, whereas NF showed a tendency toward enhanced neurotrophic effects and functional improvements, with a statistically significant improvement in the Timed Up and Go Test (p = 0.014). These findings underscore the potential for tailored nucleotide-based interventions to optimize distinct physiological domains in aging populations.
Collapse
Affiliation(s)
- Javier Gene-Morales
- Department of Physical Education and Sports, University of Valencia, 46010 Valencia, Spain; (J.G.-M.); (A.S.-B.); (F.M.-R.); (I.C.-M.); (J.C.C.)
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| | - Alvaro Juesas
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
- Department of Education Sciences, CEU Cardenal Herrera University, 46115 Castellón, Spain
| | - Angel Saez-Berlanga
- Department of Physical Education and Sports, University of Valencia, 46010 Valencia, Spain; (J.G.-M.); (A.S.-B.); (F.M.-R.); (I.C.-M.); (J.C.C.)
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| | - Ezequiel G. Martin
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| | - Luis Garrigues-Pelufo
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| | - Brayan S. Sandoval-Camargo
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| | - Fernando Martin-Rivera
- Department of Physical Education and Sports, University of Valencia, 46010 Valencia, Spain; (J.G.-M.); (A.S.-B.); (F.M.-R.); (I.C.-M.); (J.C.C.)
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| | - Iván Chulvi-Medrano
- Department of Physical Education and Sports, University of Valencia, 46010 Valencia, Spain; (J.G.-M.); (A.S.-B.); (F.M.-R.); (I.C.-M.); (J.C.C.)
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| | - Pablo Jiménez-Martínez
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
- ICEN Research Center, Department of Health Research, 38002 Santa Cruz de Tenerife, Spain
| | - Carlos Alix-Fages
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
- ICEN Research Center, Department of Health Research, 38002 Santa Cruz de Tenerife, Spain
| | - Pedro Gargallo
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| | - Julio Fernandez-Garrido
- Nursing Department, Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain; (J.F.-G.); (O.C.)
| | - Oscar Caballero
- Nursing Department, Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain; (J.F.-G.); (O.C.)
| | - Agustín Jerez-Martínez
- ICEN Research Center, Department of Health Research, 38002 Santa Cruz de Tenerife, Spain
- Faculty of Sports Sciences, Catholic University of Murcia (UCAM), 30107 Murcia, Spain
| | - Juan C. Colado
- Department of Physical Education and Sports, University of Valencia, 46010 Valencia, Spain; (J.G.-M.); (A.S.-B.); (F.M.-R.); (I.C.-M.); (J.C.C.)
- Research Group in Prevention and Health in Exercise and Sport (PHES), University of Valencia, 46010 Valencia, Spain; (A.J.); (E.G.M.); (L.G.-P.); (B.S.S.-C.); (P.J.-M.); (C.A.-F.); (P.G.)
| |
Collapse
|
2
|
Cheng F, Feng Y, Yang X, Flanagan M, Chen X, Bonakdarpour B, Jamshidi P, Castellani R, Mao Q, Chu X, Gao H, Liu Y, Dou L, Xu J, Hou Y, Martin W, Nelson P, Leverenz J, Hu M, Li Y, Pieper A, Cummings J. Genomic and epigenomic insights into purkinje and granule neurons in Alzheimer's disease and related dementia using single-nucleus multiome analysis. RESEARCH SQUARE 2025:rs.3.rs-6264481. [PMID: 40235507 PMCID: PMC11998783 DOI: 10.21203/rs.3.rs-6264481/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Although the human cerebellum is known to be neuropathologically impaired in Alzheimer's disease (AD) and AD-related dementias (ADRD), the cell type-specific transcriptional and epigenomic changes that contribute to this pathology are not well understood. Here, we report single-nucleus multiome (snRNA-seq and snATAC-seq) analysis of 103,861 nuclei isolated from both cerebellum and frontal cortex of AD/ADRD patients and normal controls. Using peak-to-gene linkage analysis, we identified 431,834 significant linkages between gene expression and cell subtype-specific chromatin accessibility regions enriched for candidate cis-regulatory elements (cCREs). These cCREs were associated with AD/ADRD-specific transcriptomic changes and disease-related gene regulatory networks, especially for RAR Related Orphan Receptor A (RORA) and E74 Like ETS Transcription Factor 1 (ELF1) in cerebellar Purkinje cells and granule cells, respectively. Trajectory analysis of granule cell populations further identified disease-relevant transcription factors, such as RORA, and their regulatory targets. Finally, we pinpointed two likely causal genes, Seizure Related 6 Homolog Like 2 (SEZ6L2) in Purkinje cells and KAT8 Regulatory NSL Complex Subunit 1 (KANSL1) in granule cells, through integrative analysis of cCREs derived from snATAC-seq, genome-wide AD/ADRD loci, and three-dimensional (3D) genome data. Via CRISPRi experiments, we found that perturbation of rs4788201 and rs62056801 significantly inhibited the expression of their target genes, SEZ6L2 and KANSL1, in human iPSC-derived neurons. This cell subtype-specific regulatory landscape in the human cerebellum identified here offers novel genomic and epigenomic insights into the neuropathology and pathobiology of AD/ADRD and other neurological disorders if broadly applied.
Collapse
|
3
|
Feng Y, Flanagan ME, Bonakdarpour B, Jamshidi P, Castellani RJ, Mao Q, Chu X, Gao H, Liu Y, Xu J, Hou Y, Martin W, Nelson PT, Leverenz JB, Pieper AA, Cummings J, Cheng F. Single-nucleus multiome analysis of human cerebellum in Alzheimer's disease-related dementia. RESEARCH SQUARE 2024:rs.3.rs-4871032. [PMID: 39184089 PMCID: PMC11343296 DOI: 10.21203/rs.3.rs-4871032/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Although human cerebellum is known to be neuropathologically impaired in Alzheimer's disease (AD) and AD-related dementias (ADRD), the cell type-specific transcriptional and epigenomic changes that contribute to this pathology are not well understood. Here, we report single-nucleus multiome (snRNA-seq and snATAC-seq) analysis of 103,861 nuclei isolated from cerebellum from 9 human cases of AD/ADRD and 8 controls, and with frontal cortex of 6 AD donors for additional comparison. Using peak-to-gene linkage analysis, we identified 431,834 significant linkages between gene expression and cell subtype-specific chromatin accessibility regions enriched for candidate cis-regulatory elements (cCREs). These cCREs were associated with AD/ADRD-specific transcriptomic changes and disease-related gene regulatory networks, especially for RAR Related Orphan Receptor A (RORA) and E74 Like ETS Transcription Factor 1 (ELF1) in cerebellar Purkinje cells and granule cells, respectively. Trajectory analysis of granule cell populations further identified disease-relevant transcription factors, such as RORA, and their regulatory targets. Finally, we prioritized two likely causal genes, including Seizure Related 6 Homolog Like 2 (SEZ6L2) in Purkinje cells and KAT8 Regulatory NSL Complex Subunit 1 (KANSL1) in granule cells, through integrative analysis of cCREs derived from snATAC-seq, genome-wide AD/ADRD loci, and Hi-C looping data. This first cell subtype-specific regulatory landscape in the human cerebellum identified here offer novel genomic and epigenomic insights into the neuropathology and pathobiology of AD/ADRD and other neurological disorders if broadly applied.
Collapse
Affiliation(s)
- Yayan Feng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Margaret E Flanagan
- Biggs Institute, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Department of Pathology, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Borna Bonakdarpour
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pouya Jamshidi
- Department of Pathology and Northwestern Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rudolph J. Castellani
- Department of Pathology and Northwestern Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Qinwen Mao
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Xiaona Chu
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuan Hou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - William Martin
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Peter T Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - James B. Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Andrew A. Pieper
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland 44106, OH, USA
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, UNLV, Las Vegas, Nevada 89154, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
4
|
Vieira CP, Lelis CA, Ochioni AC, Rosário DKA, Rosario ILS, Vieira IRS, Carvalho APA, Janeiro JM, da Costa MP, Lima FRS, Mariante RM, Alves LA, Foguel D, Junior CAC. Estimating the therapeutic potential of NSAIDs and linoleic acid-isomers supplementation against neuroinflammation. Biomed Pharmacother 2024; 177:116884. [PMID: 38889635 DOI: 10.1016/j.biopha.2024.116884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) regulate inflammation, which is associated with their role in preventing neurodegenerative diseases in epidemiological studies. It has sparked interest in their unconventional application for reducing neuroinflammation, opening up new avenues in biomedical research. However, given the pharmacological drawbacks of NSAIDs, the development of formulations with naturally antioxidant/anti-inflammatory dietary fatty acids has been demonstrated to be advantageous for the clinical translation of anti-inflammatory-based therapies. It includes improved blood-brain barrier (BBB) permeability and reduced toxicity. It permits us to speculate about the value of linoleic acid (LA)-isomers in preventing and treating neuroinflammatory diseases compared to NSAIDs. Our research delved into the impact of various factors, such as administration route, dosage, timing of intervention, and BBB permeability, on the efficacy of NSAIDs and LA-isomers in preclinical and clinical settings. We conducted a systematic comparison between NSAIDs and LA-isomers regarding their therapeutic effectiveness, BBB compatibility, and side effects. Additionally, we explored their underlying mechanisms in addressing neuroinflammation. Through our analysis, we've identified challenges and drawn conclusions that could propel advancements in treating neurodegenerative diseases and inform the development of future alternative therapeutic strategies.
Collapse
Affiliation(s)
- Carla Paulo Vieira
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil; Cellular Communication Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ 21040-900, Brazil
| | - Carini A Lelis
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Alan Clavelland Ochioni
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Denes Kaic A Rosário
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Iuri L S Rosario
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil; Laboratory of Inspection and Technology of Milk and Derivatives (LaITLácteos), School of Veterinary Medicine and Zootechnies, Universidade Federal da Bahia (UFBA), Ondina, Salvador, BA 40170-110, Brazil
| | - Italo Rennan S Vieira
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - Anna Paula A Carvalho
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil
| | - José Marcos Janeiro
- Glial Cell Biology Laboratory, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-590, Brazil
| | - Marion P da Costa
- Laboratory of Inspection and Technology of Milk and Derivatives (LaITLácteos), School of Veterinary Medicine and Zootechnies, Universidade Federal da Bahia (UFBA), Ondina, Salvador, BA 40170-110, Brazil; Graduate Program in Food Science (PGAli), Faculty of Pharmacy, Universidade Federal da Bahia (UFBA), Ondina, Salvador, BA 40170-110, Brazil
| | - Flavia R S Lima
- Glial Cell Biology Laboratory, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-590, Brazil
| | - Rafael M Mariante
- Laboratory of Structural Biology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ 21040-900, Brazil
| | - Luiz Anastácio Alves
- Cellular Communication Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ 21040-900, Brazil
| | - Debora Foguel
- Laboratory of Protein Aggregation and Amyloidosis, Institute of Medical Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-590, Brazil
| | - Carlos Adam Conte Junior
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Biochemistry, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-909, Brazil; Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-598, Brazil.
| |
Collapse
|
5
|
Zamanian MY, Golmohammadi M, Amin RS, Bustani GS, Romero-Parra RM, Zabibah RS, Oz T, Jalil AT, Soltani A, Kujawska M. Therapeutic Targeting of Krüppel-Like Factor 4 and Its Pharmacological Potential in Parkinson's Disease: a Comprehensive Review. Mol Neurobiol 2024; 61:3596-3606. [PMID: 37996730 PMCID: PMC11087351 DOI: 10.1007/s12035-023-03800-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023]
Abstract
Krüppel-like factor 4 (KLF4), a zinc finger transcription factor, is found in different human tissues and shows diverse regulatory activities in a cell-dependent manner. In the brain, KLF4 controls various neurophysiological and neuropathological processes, and its contribution to various neurological diseases has been widely reported. Parkinson's disease (PD) is an age-related neurodegenerative disease that might have a connection with KLF4. In this review, we discussed the potential implication of KLF4 in fundamental molecular mechanisms of PD, including aberrant proteostasis, neuroinflammation, apoptosis, oxidative stress, and iron overload. The evidence collected herein sheds new light on KLF4-mediated pathways, which manipulation appears to be a promising therapeutic target for PD management. However, there is a gap in the knowledge on this topic, and extended research is required to understand the translational value of the KLF4-oriented therapeutical approach in PD.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | - Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1988873554, Iran
| | | | | | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Tuba Oz
- Department of Toxicology, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznan, Poland
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Afsaneh Soltani
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1988873554, Iran.
| | - Małgorzata Kujawska
- Department of Toxicology, Poznan University of Medical Sciences, Rokietnicka 3, 60-806, Poznan, Poland.
| |
Collapse
|
6
|
Murugan R, Haridevamuthu B, Kumar RS, Almutairi BO, Arokiyaraj S, Arockiaraj J. Deacetyl epoxyazadiradione ameliorates BPA-induced neurotoxicity by mitigating ROS and inflammatory markers in N9 cells and zebrafish larvae. Comp Biochem Physiol C Toxicol Pharmacol 2023; 271:109692. [PMID: 37394128 DOI: 10.1016/j.cbpc.2023.109692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/24/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
Bisphenol A (BPA) leaches from plastic products have become a major inevitable concern among the research society. Human exposure to BPA leads to deleterious effects on multiple organs by the induced hyper inflammatory and oxidative stress responses. Due to the compromised antioxidant mechanism, the brain environment was highly susceptible and required special concern to ameliorate the effects of BPA. Hence, this study investigates the potential of neem-derived semi natural deacetyl epoxyazadiradione (DEA) against the oxidative stress and inflammatory response induced by BPA exposure in N9 cells and zebrafish larvae. The results from the in vitro analyses showed a decrease in cell viability in the MTT assay and a decline in mitochondrial damage in BPA-exposed N9 cells. Further in vivo, results revealed that pre-treatment of DEA to zebrafish larvae has significantly reduced the level of superoxide anion and increased the production of antioxidant enzymes such as SOD, CAT, GST, GPx and GR. We also found a significant decrease in the production of nitric oxide (p < 0.0001) and iNOS gene expression at 150 μM concentration. Further, DEA pre-treatment improved the behaviour of zebrafish larvae by ameliorating the production of the AChE enzyme. In conclusion, DEA protected zebrafish larvae from BPA toxicity by ameliorating oxidative stress and inflammatory responses.
Collapse
Affiliation(s)
- Raghul Murugan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603203 Chengalpattu District, Tamil Nadu, India. https://twitter.com/Raghul0210
| | - B Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603203 Chengalpattu District, Tamil Nadu, India. https://twitter.com/haridevamuthub
| | - Rajendran Saravana Kumar
- Chemistry Division, School of Advanced Sciences, VIT University, Chennai Campus, Chennai 600 127, Tamil Nadu, India
| | - Bader O Almutairi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, 11451 Riyadh, Saudi Arabia
| | - Selvaraj Arokiyaraj
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603203 Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
7
|
Zamanian MY, Parra RMR, Soltani A, Kujawska M, Mustafa YF, Raheem G, Al-Awsi L, Lafta HA, Taheri N, Heidari M, Golmohammadi M, Bazmandegan G. Targeting Nrf2 signaling pathway and oxidative stress by resveratrol for Parkinson's disease: an overview and update on new developments. Mol Biol Rep 2023; 50:5455-5464. [PMID: 37155008 DOI: 10.1007/s11033-023-08409-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/24/2023] [Indexed: 05/10/2023]
Abstract
Parkinson's disease (PD) as a prevalent neurodegenerative condition impairs motor function and is caused by the progressive deterioration of nigrostriatal dopaminergic (DAergic) neurons. The current therapy solutions for PD are ineffective because they could not inhibit the disease's progression and they even have adverse effects. Natural polyphenols, a group of phytochemicals, have been found to offer various health benefits, including neuroprotection against PD. Among these, resveratrol (RES) has neuroprotective properties owing to its capacity to protect mitochondria and act as an antioxidant. An increase in the formation of reactive oxygen species (ROS) leads to oxidative stress (OS), which is responsible for cellular damage resulting in lipid peroxidation, oxidative protein alteration, and DNA damage. In PD models, it's been discovered that RES pretreatment can diminish oxidative stress by boosting endogenous antioxidant status and directly scavenging ROS. Several studies have examined the involvement of RES in the modulation of the transcriptional factor Nrf2 in PD models because this protein recognizes oxidants and controls the antioxidant defense. In this review, we have examined the molecular mechanisms underlying the RES activity and reviewed its effects in both in vitro and in vivo models of PD. The gathered evidence herein showed that RES treatment provides neuroprotection against PD by reducing OS and upregulation of Nrf2. Moreover, in the present study, scientific proof of the neuroprotective properties of RES against PD and the mechanism supporting clinical development consideration has been described.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | | | - Afsaneh Soltani
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Małgorzata Kujawska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, Poznan, 60-631, Poland
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Ghaidaa Raheem
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | - Lateef Al-Awsi
- Department of Radiological Techniques, Al-Mustaqbal University College, Babylon, Iraq
| | - Holya A Lafta
- Department of Pharmacy, Al-Nisour University College, Baghdad, Iraq
| | - Niloofar Taheri
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mahsa Heidari
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholamreza Bazmandegan
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
8
|
The Interplay between Gut Microbiota and Parkinson's Disease: Implications on Diagnosis and Treatment. Int J Mol Sci 2022; 23:ijms232012289. [PMID: 36293176 PMCID: PMC9603886 DOI: 10.3390/ijms232012289] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
Abstract
The bidirectional interaction between the gut microbiota (GM) and the Central Nervous System, the so-called gut microbiota brain axis (GMBA), deeply affects brain function and has an important impact on the development of neurodegenerative diseases. In Parkinson’s disease (PD), gastrointestinal symptoms often precede the onset of motor and non-motor manifestations, and alterations in the GM composition accompany disease pathogenesis. Several studies have been conducted to unravel the role of dysbiosis and intestinal permeability in PD onset and progression, but the therapeutic and diagnostic applications of GM modifying approaches remain to be fully elucidated. After a brief introduction on the involvement of GMBA in the disease, we present evidence for GM alterations and leaky gut in PD patients. According to these data, we then review the potential of GM-based signatures to serve as disease biomarkers and we highlight the emerging role of probiotics, prebiotics, antibiotics, dietary interventions, and fecal microbiota transplantation as supportive therapeutic approaches in PD. Finally, we analyze the mutual influence between commonly prescribed PD medications and gut-microbiota, and we offer insights on the involvement also of nasal and oral microbiota in PD pathology, thus providing a comprehensive and up-to-date overview on the role of microbial features in disease diagnosis and treatment.
Collapse
|
9
|
Rahman MU, Bilal M, Shah JA, Kaushik A, Teissedre PL, Kujawska M. CRISPR-Cas9-Based Technology and Its Relevance to Gene Editing in Parkinson's Disease. Pharmaceutics 2022; 14:1252. [PMID: 35745824 PMCID: PMC9229276 DOI: 10.3390/pharmaceutics14061252] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) and other chronic and debilitating neurodegenerative diseases (NDs) impose a substantial medical, emotional, and financial burden on individuals and society. The origin of PD is unknown due to a complex combination of hereditary and environmental risk factors. However, over the last several decades, a significant amount of available data from clinical and experimental studies has implicated neuroinflammation, oxidative stress, dysregulated protein degradation, and mitochondrial dysfunction as the primary causes of PD neurodegeneration. The new gene-editing techniques hold great promise for research and therapy of NDs, such as PD, for which there are currently no effective disease-modifying treatments. As a result, gene therapy may offer new treatment options, transforming our ability to treat this disease. We present a detailed overview of novel gene-editing delivery vehicles, which is essential for their successful implementation in both cutting-edge research and prospective therapeutics. Moreover, we review the most recent advancements in CRISPR-based applications and gene therapies for a better understanding of treating PD. We explore the benefits and drawbacks of using them for a range of gene-editing applications in the brain, emphasizing some fascinating possibilities.
Collapse
Affiliation(s)
- Mujeeb ur Rahman
- Department of Toxicology, Faculty of Pharmacy, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznan, Poland;
| | - Muhammad Bilal
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China;
| | - Junaid Ali Shah
- College of Life Sciences, Jilin University, Changchun 130012, China;
- Fergana Medical Institute of Public Health Uzbekistan, Fergana 150110, Uzbekistan
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Health System Engineering, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805, USA;
- School of Engineering, University of Petroleum and Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| | - Pierre-Louis Teissedre
- Institut des Sciences de la Vigne et du Vin, Université de Bordeaux, EA 4577, Œnologie, 210 Chemin de Leysotte, F-33140 Villenave d’Ornon, France;
- Institut des Sciences de la Vigne et du Vin, INRA, USC 1366 INRA, IPB, 210 Chemin de Leysotte, F-33140 Villenave d’Ornon, France
| | - Małgorzata Kujawska
- Department of Toxicology, Faculty of Pharmacy, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznan, Poland;
| |
Collapse
|
10
|
Pomilio AB, Vitale AA, Lazarowski AJ. Uncommon Noninvasive Biomarkers for the Evaluation and Monitoring of the Etiopathogenesis of Alzheimer's Disease. Curr Pharm Des 2022; 28:1152-1169. [DOI: 10.2174/1381612828666220413101929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer´s disease (AD) is the most widespread dementia in the world, followed by vascular dementia. Since AD is a heterogeneous disease that shows several varied phenotypes, it is not easy to make an accurate diagnosis, so it arises when the symptoms are clear and the disease is already very advanced. Therefore, it is important to find out biomarkers for AD early diagnosis that facilitate treatment or slow down the disease. Classic biomarkers are obtained from cerebrospinal fluid and plasma, along with brain imaging by positron emission tomography. Attempts have been made to discover uncommon biomarkers from other body fluids, which are addressed in this update.
Objective:
This update aims to describe recent biomarkers from minimally invasive body fluids for the patients, such as saliva, urine, eye fluid or tears.
Methods:
Biomarkers were determined in patients versus controls by single tandem mass spectrometry, and immunoassays. Metabolites were identified by nuclear magnetic resonance, and microRNAs with genome-wide high-throughput real-time polymerase chain reaction-based platforms.
Results:
Biomarkers from urine, saliva, and eye fluid were described, including peptides/proteins, metabolites, and some microRNAs. The association with AD neuroinflammation and neurodegeneration was analyzed, highlighting the contribution of matrix metalloproteinases, the immune system and microglia, as well as the vascular system.
Conclusion:
Unusual biomarkers have been developed, which distinguish each stage and progression of the disease, and are suitable for the early AD diagnosis. An outstanding relationship of biomarkers with neuroinflammation and neurodegeneration was assessed, clearing up concerns of the etiopathogenesis of AD.
Collapse
Affiliation(s)
- Alicia B. Pomilio
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Arturo A. Vitale
- Departamento de Bioquímica Clínica, Área Hematología, Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Av. Córdoba 2351, C1120AAF Buenos Aires, Argentina
| | - Alberto J. Lazarowski
- Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Córdoba 2351, C1120AAF Buenos Aires, Argentina
| |
Collapse
|
11
|
Viana MDB, de Aquino PEA, Estadella D, Ribeiro DA, Viana GSDB. Cannabis sativa and Cannabidiol: A Therapeutic Strategy for the Treatment of Neurodegenerative Diseases? Med Cannabis Cannabinoids 2022; 5:207-219. [PMID: 36467781 PMCID: PMC9710321 DOI: 10.1159/000527335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/04/2022] [Indexed: 08/27/2023] Open
Abstract
This work is a literature review, presenting the current state of the use of cannabinoids on neurodegenerative diseases. The emphasis is on Parkinson's (PD) and Alzheimer's (AD) diseases, the two most prevalent neurological diseases. The review goes from Cannabis sativa and its hundreds of bioactive compounds to Δ9-tetrahydrocannabinol (THC) and mainly cannabidiol (CBD) and their interactions with the endocannabinoid receptors (CB1 and CB2). CBD molecular targets were also focused on to explain its neuroprotective action mechanism on neurodegenerative diseases. Although THC is the main psychoactive component of C. sativa, and it may induce transient psychosis-like symptoms, growing evidence suggests that CBD may have protective effects against the psychotomimetic effects of THC and therapeutic properties. Furthermore, a great number of recent works on the neuroprotective and anti-inflammatory CBD effects and its molecular targets are also reviewed. We analyzed CBD actions in preclinical and in clinical trials, conducted with PD and AD patients. Although the data on preclinical assays are more convincing, the same is not true with the clinical data. Despite the consensus among researchers on the potential of CBD as a neuroprotective agent, larger and well-designed randomized clinical trials will be necessary to gather conclusive results concerning the use of CBD as a therapeutic strategy for the treatment of diseases such as PD and AD.
Collapse
Affiliation(s)
- Milena de Barros Viana
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Campus Santos, Santos, Brazil
| | | | - Débora Estadella
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Campus Santos, Santos, Brazil
| | - Daniel Araki Ribeiro
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Campus Santos, Santos, Brazil
| | | |
Collapse
|