1
|
Chen J, Liang C, Wang F, Zhu Y, Zhu L, Chen J, Liu B, Yang X. Potential biofluid markers for cognitive impairment in Parkinson's disease. Neural Regen Res 2026; 21:281-295. [PMID: 39851136 PMCID: PMC12094573 DOI: 10.4103/nrr.nrr-d-24-00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/05/2024] [Accepted: 11/08/2024] [Indexed: 01/26/2025] Open
Abstract
Cognitive impairment is a particularly severe non-motor symptom of Parkinson's disease that significantly diminishes the quality of life of affected individuals. Identifying reliable biomarkers for cognitive impairment in Parkinson's disease is essential for early diagnosis, prognostic assessments, and the development of targeted therapies. This review aims to summarize recent advancements in biofluid biomarkers for cognitive impairment in Parkinson's disease, focusing on the detection of specific proteins, metabolites, and other biomarkers in blood, cerebrospinal fluid, and saliva. These biomarkers can shed light on the multifaceted etiology of cognitive impairment in Parkinson's disease, which includes protein misfolding, neurodegeneration, inflammation, and oxidative stress. The integration of biofluid biomarkers with neuroimaging and clinical data can facilitate the development of predictive models to enhance early diagnosis and monitor the progression of cognitive impairment in patients with Parkinson's disease. This comprehensive approach can improve the existing understanding of the mechanisms driving cognitive decline and support the development of targeted therapeutic strategies aimed at modifying the course of cognitive impairment in Parkinson's disease. Despite the promise of these biomarkers in characterizing the mechanisms underlying cognitive decline in Parkinson's disease, further research is necessary to validate their clinical utility and establish a standardized framework for early detection and monitoring of cognitive impairment in Parkinson's disease.
Collapse
Affiliation(s)
- Jieyu Chen
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Chunyu Liang
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Fang Wang
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Yongyun Zhu
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Liuhui Zhu
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jianzhun Chen
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Bin Liu
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xinglong Yang
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
2
|
Safi A, Giunti E, Melikechi O, Xia W, Melikechi N. Identification of blood plasma protein ratios for distinguishing Alzheimer's disease from healthy controls using machine learning. Heliyon 2025; 11:e42349. [PMID: 39981365 PMCID: PMC11840181 DOI: 10.1016/j.heliyon.2025.e42349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
Early detection of Alzheimer's disease is essential for effective treatment and the development of therapies that modify disease progression. Developing sensitive and specific noninvasive diagnostic tools is crucial for improving clinical outcomes and advancing our understanding of this condition. Liquid biopsy techniques, especially those involving plasma biomarkers, provide a promising noninvasive method for early diagnosis and disease monitoring. In this study, we analyzed the plasma proteomic profiles of 38 healthy individuals, with an average age of 66.5 years, and 22 patients with Alzheimer's disease, with an average age of 79.7 years. Proteins in the plasma were quantified using specialized panels designed for proteomic extension assays. Through computational analysis using a linear support vector machine algorithm, we identified 82 differentially expressed proteins between the two groups. From these, we calculated 6642 possible protein ratios and identified specific combinations of these ratios as significant features for distinguishing between individuals with Alzheimer's disease and healthy individuals. Notably, the protein ratios kynureninase to macrophage scavenger receptor type 1, Neurocan to protogenin, and interleukin-5 receptor alpha to glial cell line-derived neurotrophic factor receptor alpha 1 achieving accuracy up to 98 % in differentiating between the two groups. This study underscores the potential of leveraging protein relationships, expressed as ratios, in advancing Alzheimer's disease diagnostics. Furthermore, our findings highlight the promise of liquid biopsy techniques as a noninvasive and accurate approach for early detection and monitoring of Alzheimer's disease using blood plasma.
Collapse
Affiliation(s)
- Ali Safi
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Elisa Giunti
- Bedford VA Healthcare System, Bedford, MA, 01730, USA
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Omar Melikechi
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Weiming Xia
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
- Bedford VA Healthcare System, Bedford, MA, 01730, USA
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Noureddine Melikechi
- Kennedy College of Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| |
Collapse
|
3
|
Wang D, Wei S, Zhang L, Lang Z, Wang S, Cheng B, Lu Y, Wang X, Wang W, Li F, Zhang H. Impaired Basal Forebrain Cholinergic Neuron GDNF Signaling Contributes to Perioperative Sleep Deprivation-Induced Chronicity of Postsurgical Pain in Mice Through Regulating Cholinergic Neuronal Activity, Apoptosis, and Autophagy. CNS Neurosci Ther 2024; 30:e70147. [PMID: 39639706 PMCID: PMC11621383 DOI: 10.1111/cns.70147] [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: 06/10/2024] [Revised: 10/22/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024] Open
Abstract
AIMS This study investigated the roles of lateral basal forebrain glial cell line-derived neurotrophic factor (GDNF) signaling and cholinergic neuron activity, apoptosis, and autophagy dysfunction in sleep deprivation-induced increased risk of chronic postsurgical pain (CPSP) in mice. METHODS Sleep deprivation (6 h per day from -1 to 3 days postoperatively) was administered to mice receiving skin/muscle incision and retraction (SMIR) to determine whether perioperative sleep deprivation induces mechanical and thermal pain hypersensitivity, increases the risk of chronic pain, and causes changes of basal forebrain neurons activity (c-Fos immunostaining), apoptosis (cleaved Caspase-3 expression), autophagy (LC3 and p62 expression) and GDNF expression. Adeno-associated virus (AAV)-GDNF was microinjected into the basal forebrain to see whether increased GDNF expression could reverse sleep deprivation-induced changes in pain duration and cholinergic neuron apoptosis and autophagy. Cholinergic neurons were further depleted by mu p75-SAP to examine whether the pain-prolonging effects of sleep deprivation still exist. RESULTS Perioperative sleep deprivation enhanced pain sensation and prolonged pain duration in SMIR mice, which was accompanied by decreased cholinergic neuron activity and GDNF expression, increased apoptosis, and autophagy dysfunction in the substantia innominata (SI), magnocellular preoptic nucleus (MCPO), and horizontal diagonal band Broca (HDB) (hereafter lateral basal forebrain). Normalizing cholinergic neuron GDNF expression by AAV-GDNF in the lateral basal forebrain inhibited apoptosis and autophagy dysfunction and mitigated sleep deprivation-induced pain maintenance. Mice with selective lesion of lateral basal forebrain cholinergic neurons were resistant to the pain-enhancing and prolonging effects of sleep deprivation and the pain-alleviating effects of AAV-GDNF therapy. CONCLUSIONS Perioperative sleep deprivation promotes chronicity of postsurgical pain possibly through decreasing basal forebrain GDNF signaling and causing cholinergic neuronal apoptosis and autophagy dysfunction.
Collapse
Affiliation(s)
- Dong Wang
- The Postgraduate Training Base of Jinzhou Medical University and Department of AnesthesiologyThe PLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Shi‐Nan Wei
- Department of AnesthesiologyTangdu Hospital, Air Force Military Medical UniversityXianShanxiChina
| | - Lu Zhang
- Department of AnesthesiologyZibo Central HospitalZiboChina
| | - Zhi‐Chen Lang
- The Postgraduate Training Base of Jinzhou Medical University and Department of AnesthesiologyThe PLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Si‐Nian Wang
- Department of Nuclear Radiation Injury and MonitoringThe PLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Bo Cheng
- Department of PathologyThe PLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Yan Lu
- Department of NeurologyThe PLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Xiu Wang
- Department of PediatricsZhengzhou Central HospitalZhengzhouChina
| | - Wei Wang
- The Postgraduate Training Base of Jinzhou Medical University and Department of AnesthesiologyThe PLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Feng‐Sheng Li
- Department of Nuclear Radiation Injury and MonitoringThe PLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Hao Zhang
- The Postgraduate Training Base of Jinzhou Medical University and Department of AnesthesiologyThe PLA Rocket Force Characteristic Medical CenterBeijingChina
| |
Collapse
|
4
|
Sokolowski I, Kucharska-Lusina A, Miller E, Poplawski T, Majsterek I. Exploring the Gene Expression and Plasma Protein Levels of HSP90, HSP60, and GDNF in Multiple Sclerosis Patients and Healthy Controls. Curr Issues Mol Biol 2024; 46:11668-11680. [PMID: 39451573 PMCID: PMC11505768 DOI: 10.3390/cimb46100693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by immune-mediated inflammation and neurodegeneration in the central nervous system (CNS). In this study; we aimed to investigate the gene expression and plasma protein levels of three neuroprotective genes-heat shock proteins (HSP90 and HSP60) and glial cell line-derived neurotrophic factor (GDNF)-in MS patients compared to healthy controls. Forty patients with relapsing-remitting MS and 40 healthy volunteers participated in this study. Gene expression was measured using reverse transcription quantitative real-time PCR, and protein levels were assessed via ELISA. The results showed a significant increase in HSP90 (1.7-fold) and HSP60 (2-fold) gene expression in MS patients compared to controls, along with corresponding increases in protein levels (1.5-fold for both HSP90 and HSP60). In contrast, GDNF gene expression and protein levels were significantly reduced in MS patients, with a 7-fold decrease in gene expression and a 1.6-fold reduction in protein levels. Notably, a non-linear relationship between GDNF gene expression and protein concentration was observed in MS patients, suggesting complex regulatory mechanisms influencing GDNF in the disease. The upregulation of HSP90 and HSP60 in MS highlights their roles in immune regulation and stress responses, while the reduction in GDNF indicates impaired neuroprotection. These findings suggest that HSP90, HSP60, and GDNF could serve as biomarkers for disease progression and as potential therapeutic targets in MS, offering promising avenues for future research and treatment development.
Collapse
Affiliation(s)
- Igor Sokolowski
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| | - Aleksandra Kucharska-Lusina
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Tomasz Poplawski
- Department of Microbiology and Pharmaceutical Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| |
Collapse
|
5
|
De Simone S, Alfieri L, Bosco MA, Cantatore S, Carpinteri M, Cipolloni L, Neri M. The forensic aspects of suicide and neurotrophin factors: a research study. Front Pharmacol 2024; 15:1392832. [PMID: 39170712 PMCID: PMC11335659 DOI: 10.3389/fphar.2024.1392832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction: Suicide represents a significant public health problem whose neurobiology is not yet fully understood. In many cases, suicidal behavior and psychiatric spectrum disorders are linked, in particular, to major depression. An emerging pathophysiological hypothesis underlines the role of neurotrophic factors, proteins involved in neurogenesis, in synaptic plasticity in response to stressors. Our research aims to evaluate the degree of expression of brain neurotrophic factor (BDNF) in brain areas involved in depressive disorder in suicidal subjects. Furthermore, we want to evaluate the expression of glial cell line-derived neurotrophic factor (GDNF) in suicidal subjects. Methods: We selected twenty confirmed cases of suicide among subjects with a clinical history of depressive pathology and possible psychopharmacological treatment, compared to ten controls of individuals who died of non-suicidal causes. For all selected cases and controls, immunohistochemical investigations were performed using a panel of antibodies against the BDNF and GDNF antigens on samples from the various brain areas. Results and discussion: The results show that BDNF was under-expressed in the cerebral parenchyma of subjects who died by suicide compared to controls, while there was an overexpression of GDNF in suicide victims, these data could be useful for a clinical application as potential markers for suicidal risk, to assess the severity of depression and development of specific pharmacological therapies for depression.
Collapse
Affiliation(s)
- Stefania De Simone
- Department of Clinical and Experimental Medicine, Section of Legal Medicine, University of Foggia, Foggia, Italy
| | - Letizia Alfieri
- Department of Medical Sciences, Section of Legal Medicine University of Ferrara, Ferrara, Italy
| | - Maria Antonella Bosco
- Department of Clinical and Experimental Medicine, Section of Legal Medicine, University of Foggia, Foggia, Italy
| | - Santina Cantatore
- Department of Clinical and Experimental Medicine, Section of Legal Medicine, University of Foggia, Foggia, Italy
| | - Michele Carpinteri
- Department of Biomedical, Metabolic and Neural Sciences, Institute of Legal Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Luigi Cipolloni
- Department of Clinical and Experimental Medicine, Section of Legal Medicine, University of Foggia, Foggia, Italy
| | - Margherita Neri
- Department of Medical Sciences, Section of Legal Medicine University of Ferrara, Ferrara, Italy
| |
Collapse
|
6
|
He M, Liu Z, Lian T, Guo P, Zhang W, Huang Y, Zhang Y, Liu G, Zhang W, Li J, Guan H, Zhang W, Luo D, Qi J, Yue H, Wang X, Zhang W. Role of nerve growth factor on cognitive impairment in patients with Alzheimer's disease carrying apolipoprotein E ε4. CNS Neurosci Ther 2024; 30:e14560. [PMID: 38112032 PMCID: PMC11163191 DOI: 10.1111/cns.14560] [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: 09/27/2023] [Revised: 11/23/2023] [Accepted: 11/25/2023] [Indexed: 12/20/2023] Open
Abstract
AIMS To investigate the roles of neurotrophic factors on cognition in patients with Alzheimer's disease (AD) carrying Apolipoprotein E (APOE) ε4. METHODS Totals of 173 patients with AD were divided into APOE ε4 carrier and non-carrier groups, and their demographics, cognition, and neurotrophic factors in cerebrospinal fluid (CSF) were compared. Multiple linear regression analyses were performed to assess correlations among APOE ε4, neurotrophic factors and cognition. Mediation analyses were conducted to assess the sequential associations among APOE ε4, nerve growth factor (NGF), and cognition. RESULTS Global cognition and multiple domains were impaired in the APOE ε4 carrier group (all p < 0.05). NGF level in the APOE ε4 carrier group was lower than that in the non-carrier group (p = 0.016). NGF level showed significant correlations with both global and multiple domains cognitions. Specifically, NGF mediated the association between APOE ε4 and Animal Fluency Test score (β, -0.45; 95% CI [-0.96, -0.07]; p < 0.001) and Trail Making Test-A (time) (β, 0.15; 95% CI [0.01, 0.33]; p < 0.001). CONCLUSION APOE ε4 is associated with cognitive impairment, and those carrying APOE ε4 have decreased NGF level in CSF. Declined NGF level is correlated with compromised cognition. NGF mediates APOE ε4-associated cognitive impairment.
Collapse
Affiliation(s)
- Mingyue He
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Zhan Liu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Tenghong Lian
- Center for Cognitive Neurology, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Peng Guo
- Center for Cognitive Neurology, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Wenjing Zhang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Yue Huang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- Department of Pharmacology, School of Medical Sciences, Faculty of Medicine & HealthUNSW SydneySydneyNew South WalesAustralia
| | - Yanan Zhang
- Department of Blood TransfusionBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Gaifen Liu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Weijiao Zhang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Jinghui Li
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Huiying Guan
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Weijia Zhang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Dongmei Luo
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Jing Qi
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Hao Yue
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Xiaomin Wang
- Department of PhysiologyCapital Medical UniversityBeijingChina
| | - Wei Zhang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- Center for Cognitive Neurology, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- Center of Parkinson's DiseaseBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory on Parkinson DiseaseBeijingChina
| |
Collapse
|
7
|
Delaby C, Lehmann S. [On the road to biological blood diagnosis of Alzheimer's disease?]. Med Sci (Paris) 2024; 40:351-360. [PMID: 38651960 DOI: 10.1051/medsci/2024037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
The growing number of people suffering from Alzheimer's disease (AD) represents a major public health problem. The diagnosis of AD is multidisciplinary and involves the use of amyloid and tau biomarkers measured in cerebrospinal fluid. Recent advances in analytical techniques now allow us to measure these biomarkers in blood. Blood biomarkers offer particularly promising potential for early, minimally invasive detection of AD, as well as for differential diagnosis of dementia and patient follow-up. The aim of this review is to provide an overview of current and candidate blood biomarkers for AD, their informative value, and their potential to be integrated into clinical practice in the near future.
Collapse
Affiliation(s)
- Constance Delaby
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm U1298, Montpellier, France - Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelone, Espagne
| | - Sylvain Lehmann
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm U1298, Montpellier, France
| |
Collapse
|
8
|
Alster P, Otto-Ślusarczyk D, Szlufik S, Duszyńska-Wąs K, Drzewińska A, Wiercińska-Drapało A, Struga M, Kutyłowski M, Friedman A, Madetko-Alster N. The significance of glial cell line-derived neurotrophic factor analysis in Progressive Supranuclear Palsy. Sci Rep 2024; 14:2805. [PMID: 38307947 PMCID: PMC10837430 DOI: 10.1038/s41598-024-53355-y] [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: 11/26/2023] [Accepted: 01/31/2024] [Indexed: 02/04/2024] Open
Abstract
Progressive Supranuclear Palsy (PSP) is an atypical parkinsonism. Major subtypes of the disease: PSP-Richardson's Syndrome (PSP-RS) and PSP Parkinsonism Predominant (PSP-P) vary in clinical features, the pathomechanism remains unexplored. The aim of this work is to analyze the relevance of glial cell line-derived neurotrophic factor (GDNF) evaluation in the serum and cerebrospinal fluid (CSF) in PSP subtypes and to verify its significance as a possible factor in the in vivo examination. Authors assessed the concentration of GDNF in the serum and CSF of 12 patients with PSP-RS, 12 with PSP-P and 12 controls. Additionally authors evaluated patients using Unified Parkinson's Disease Rating Scale-III part (UPDRS-III), Frontal Assessment Battery (FAB) and Magnetic Resonance Imaging (MRI). The evaluation revealed significantly increased concentrations of GDNF in the CSF among PSP-RS patients and substantially increased concentrations of GDNF in the serum in PSP-P. Though the GDNF concentrations differentiated PSP subtypes, no correlations between with clinical factors were observed however certain correlations with atrophic changes in MRI were detected. GDNF is a factor which may impact the pathogenesis of PSP. Possible implementation of GDNF as a therapeutic factor could be a perspective in the search for therapy in this currently incurable disease.
Collapse
Affiliation(s)
- Piotr Alster
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242, Warsaw, Poland.
| | | | - Stanisław Szlufik
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242, Warsaw, Poland
| | - Karolina Duszyńska-Wąs
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242, Warsaw, Poland
| | - Agnieszka Drzewińska
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242, Warsaw, Poland
| | - Alicja Wiercińska-Drapało
- Department of Hepatology and Infectious and Tropical Diseases, Medical University of Warsaw, Provincial Infectious Diseases Hospital in Warsaw, Warsaw, Poland
| | - Marta Struga
- Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Michał Kutyłowski
- Department of Diagnostic Imaging, Brodno Mazovian Hospital, Warsaw, Poland
| | - Andrzej Friedman
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242, Warsaw, Poland
| | - Natalia Madetko-Alster
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242, Warsaw, Poland
| |
Collapse
|
9
|
Syed RA, Hayat M, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Kaleem I, Bashir S. Aging-Related Protein Alterations in the Brain. J Alzheimers Dis 2024; 99:S5-S22. [PMID: 38339930 DOI: 10.3233/jad-230801] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Aging is an intrinsic aspect of an organism's life cycle and is characterized by progressive physiological decline and increased susceptibility to mortality. Many age-associated disorders, including neurological disorders, are most commonly linked with the aging process, such as Alzheimer's disease (AD). This review aims to provide a comprehensive overview of the effects of aging and AD on the molecular pathways and levels of different proteins in the brain, including metalloproteins, neurotrophic factors, amyloid proteins, and tau proteins. AD is caused by the aggregation of amyloid proteins in the brain. Factors such as metal ions, protein ligands, and the oligomerization state of amyloid precursor protein significantly influence the proteolytic processing of amyloid-β protein precursor (AβPP). Tau, a disordered cytosolic protein, serves as the principal microtubule-associated protein in mature neurons. AD patients exhibit decreased levels of nerve growth factor within their nervous systems and cerebrospinal fluid. Furthermore, a significant increase in brain-derived neurotrophic factor resulting from the neuroprotective effect of glial cell line-derived neurotrophic factor suggests that the synergistic action of these proteins plays a role in inhibiting neuronal degeneration and atrophy. The mechanism through which Aβ and AβPP govern Cu2+ transport and their influence on Cu2+ and other metal ion pools requires elucidation in future studies. A comprehensive understanding of the influence of aging and AD on molecular pathways and varying protein levels may hold the potential for the development of novel diagnostic and therapeutic methods for the treatment of AD.
Collapse
Affiliation(s)
- Rafay Ali Syed
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Mahnoor Hayat
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Imdad Kaleem
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
10
|
Luo BL, Zhang ZZ, Chen J, Liu X, Zhang YM, Yang QG, Chen GH. Effects of gestational inflammation on age-related cognitive decline and hippocampal Gdnf-GFRα1 levels in F1 and F2 generations of CD-1 Mice. BMC Neurosci 2023; 24:26. [PMID: 37055728 PMCID: PMC10103445 DOI: 10.1186/s12868-023-00793-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/23/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND It has been reported that age-associated cognitive decline (AACD) accelerated by maternal lipopolysaccharide (LPS) insult during late pregnancy can be transmitted to the second generation in a sex-specificity manner. In turn, recent studies indicated that glial cell line-derived neurotrophic factor (GDNF) and its cognate receptor (GFRα1) are critical for normal cognitive function. Based on this evidence, we aimed to explore whether Gdnf-GFRα1 expression contributes to cognitive decline in the F1 and F2 generations of mouse dams exposed to lipopolysaccharide (LPS) during late gestation, and to evaluate also the potential interference effect of pro-inflammatory cytokines. METHODS During gestational days 15-17, pregnant CD-1 mice (8-10 weeks old) received a daily intraperitoneal injection of LPS (50 μg/kg) or saline (control). In utero LPS-exposed F1 generation mice were selectively mated to produce F2 generation mice. In F1 and F2 mice aged 3 and 15 months, the Morris water maze (MWM) was used to evaluated the spatial learning and memory ability, the western blotting and RT-PCR were used for analyses of hippocampal Gdnf and GFRα1 expression, and ELISA was used to analyse IL-1β, IL-6 and TNF-α levels in serum. RESULTS Middle-aged F1 offspring from LPS-treated mothers exhibited longer swimming latency and distance during the learning phase, lower percentage swimming time and distance in targe quadrant during memory phase, and lower hippocampal levels of Gdnf and GFRα1 gene products compared to age-matched controls. Similarly, the middle-aged F2 offspring from the Parents-LPS group had longer swimming latency and distance in the learning phase, and lower percentage swimming time and distance in memory phase than the F2-CON group. Moreover, the 3-month-old Parents-LPS and 15-month-old Parents- and Father-LPS groups had lower GDNF and GFRα1 protein and mRNAs levels compared to the age-matched F2-CON group. Furthermore, hippocampal levels of Gdnf and GFRα1 were correlated with impaired cognitive performance in the Morris water maze after controlling for circulating pro-inflammatory cytokine levels. CONCLUSIONS Our findings indicate that accelerated AACD by maternal LPS exposure can be transmitted across at least two generations through declined Gdnf and GFRα1 expression, mainly via paternal linage.
Collapse
Affiliation(s)
- Bao-Ling Luo
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Zhe-Zhe Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Jing Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Xue Liu
- Department of Geriatrics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, People's Republic of China
| | - Yue-Ming Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Qi-Gang Yang
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China.
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China.
| |
Collapse
|
11
|
Delaby C, Hirtz C, Lehmann S. Overview of the blood biomarkers in Alzheimer's disease: Promises and challenges. Rev Neurol (Paris) 2023; 179:161-172. [PMID: 36371265 DOI: 10.1016/j.neurol.2022.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
The increasing number of people with advanced Alzheimer's disease (AD) represents a significant psychological and financial cost to the world population. Accurate detection of the earliest phase of preclinical AD is of major importance for the success of preventive and therapeutic strategies (Cullen et al., 2021). Advances in analytical techniques have been essential for the development of sensitive, specific and reliable diagnostic tests for AD biomarkers in biological fluids (cerebrospinal fluid and blood). Blood biomarkers hold promising potential for early and minimally invasive detection of AD, but also for differential diagnosis of dementia and for monitoring the course of the disease. The aim of this review is to provide an overview of current blood biomarkers of AD, from tau proteins and amyloid peptides to biomarkers of neuronal degeneration and inflammation, reactive and metabolic factors. We thus discuss the informative value of currently candidate blood biomarkers and their potential to be integrated into clinical practice for the management of AD in the near future.
Collapse
Affiliation(s)
- C Delaby
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France; Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
| | - C Hirtz
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France
| | - S Lehmann
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France.
| |
Collapse
|
12
|
Andronie-Cioara FL, Ardelean AI, Nistor-Cseppento CD, Jurcau A, Jurcau MC, Pascalau N, Marcu F. Molecular Mechanisms of Neuroinflammation in Aging and Alzheimer's Disease Progression. Int J Mol Sci 2023; 24:ijms24031869. [PMID: 36768235 PMCID: PMC9915182 DOI: 10.3390/ijms24031869] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/01/2023] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Aging is the most prominent risk factor for late-onset Alzheimer's disease. Aging associates with a chronic inflammatory state both in the periphery and in the central nervous system, the evidence thereof and the mechanisms leading to chronic neuroinflammation being discussed. Nonetheless, neuroinflammation is significantly enhanced by the accumulation of amyloid beta and accelerates the progression of Alzheimer's disease through various pathways discussed in the present review. Decades of clinical trials targeting the 2 abnormal proteins in Alzheimer's disease, amyloid beta and tau, led to many failures. As such, targeting neuroinflammation via different strategies could prove a valuable therapeutic strategy, although much research is still needed to identify the appropriate time window. Active research focusing on identifying early biomarkers could help translating these novel strategies from bench to bedside.
Collapse
Affiliation(s)
- Felicia Liana Andronie-Cioara
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Adriana Ioana Ardelean
- Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Carmen Delia Nistor-Cseppento
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Correspondence: (C.D.N.-C.); (N.P.)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | | | - Nicoleta Pascalau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Correspondence: (C.D.N.-C.); (N.P.)
| | - Florin Marcu
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
13
|
Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Bryukhovetskiy I, Manzhulo I, Wiklund L, Sharma HS. Nanowired Delivery of Cerebrolysin Together with Antibodies to Amyloid Beta Peptide, Phosphorylated Tau, and Tumor Necrosis Factor Alpha Induces Superior Neuroprotection in Alzheimer's Disease Brain Pathology Exacerbated by Sleep Deprivation. ADVANCES IN NEUROBIOLOGY 2023; 32:3-53. [PMID: 37480458 DOI: 10.1007/978-3-031-32997-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Sleep deprivation induces amyloid beta peptide and phosphorylated tau deposits in the brain and cerebrospinal fluid together with altered serotonin metabolism. Thus, it is likely that sleep deprivation is one of the predisposing factors in precipitating Alzheimer's disease (AD) brain pathology. Our previous studies indicate significant brain pathology following sleep deprivation or AD. Keeping these views in consideration in this review, nanodelivery of monoclonal antibodies to amyloid beta peptide (AβP), phosphorylated tau (p-tau), and tumor necrosis factor alpha (TNF-α) in sleep deprivation-induced AD is discussed based on our own investigations. Our results suggest that nanowired delivery of monoclonal antibodies to AβP with p-tau and TNF-α induces superior neuroprotection in AD caused by sleep deprivation, not reported earlier.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Department Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
14
|
Amyloidogenesis and Neurotrophic Dysfunction in Alzheimer’s Disease: Do They have a Common Regulating Pathway? Cells 2022; 11:cells11203201. [PMID: 36291068 PMCID: PMC9600014 DOI: 10.3390/cells11203201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
The amyloid cascade hypothesis has predominately been used to describe the pathogenesis of Alzheimer’s disease (AD) for decades, as Aβ oligomers are thought to be the prime cause of AD. Meanwhile, the neurotrophic factor hypothesis has also been proposed for decades. Accumulating evidence states that the amyloidogenic process and neurotrophic dysfunction are mutually influenced and may coincidently cause the onset and progress of AD. Meanwhile, there are intracellular regulators participating both in the amyloidogenic process and neurotrophic pathways, which might be the common original causes of amyloidogenesis and neurotrophic dysfunction. In this review, the current understanding regarding the role of neurotrophic dysfunction and the amyloidogenic process in AD pathology is briefly summarized. The mutual influence of these two pathogenesis pathways and their potential common causal pathway are further discussed. Therapeutic strategies targeting the common pathways to simultaneously prevent amyloidogenesis and neurotrophic dysfunction might be anticipated for the disease-modifying treatment of AD.
Collapse
|
15
|
Tang CX, Chen J, Shao KQ, Liu YH, Zhou XY, Ma CC, Liu MT, Shi MY, Kambey PA, Wang W, Ayanlaja AA, Liu YF, Xu W, Chen G, Wu J, Li X, Gao DS. Blunt dopamine transmission due to decreased GDNF in the PFC evokes cognitive impairment in Parkinson's disease. Neural Regen Res 2022; 18:1107-1117. [PMID: 36255000 PMCID: PMC9827775 DOI: 10.4103/1673-5374.355816] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Studies have found that the absence of glial cell line-derived neurotrophic factor may be the primary risk factor for Parkinson's disease. However, there have not been any studies conducted on the potential relationship between glial cell line-derived neurotrophic factor and cognitive performance in Parkinson's disease. We first performed a retrospective case-control study at the Affiliated Hospital of Xuzhou Medical University between September 2018 and January 2020 and found that a decreased serum level of glial cell line-derived neurotrophic factor was a risk factor for cognitive disorders in patients with Parkinson's disease. We then established a mouse model of Parkinson's disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and analyzed the potential relationships among glial cell line-derived neurotrophic factor in the prefrontal cortex, dopamine transmission, and cognitive function. Our results showed that decreased glial cell line-derived neurotrophic factor in the prefrontal cortex weakened dopamine release and transmission by upregulating the presynaptic membrane expression of the dopamine transporter, which led to the loss and primitivization of dendritic spines of pyramidal neurons and cognitive impairment. In addition, magnetic resonance imaging data showed that the long-term lack of glial cell line-derived neurotrophic factor reduced the connectivity between the prefrontal cortex and other brain regions, and exogenous glial cell line-derived neurotrophic factor significantly improved this connectivity. These findings suggested that decreased glial cell line-derived neurotrophic factor in the prefrontal cortex leads to neuroplastic degeneration at the level of synaptic connections and circuits, which results in cognitive impairment in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Chuan-Xi Tang
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jing Chen
- Experinental Teaching Center of Morphology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Kai-Quan Shao
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ye-Hao Liu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xiao-Yu Zhou
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Department of Neurology, Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu Province, China
| | - Cheng-Cheng Ma
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Meng-Ting Liu
- Department of Rehabilitation, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ming-Yu Shi
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Piniel Alphayo Kambey
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Wei Wang
- Department of Medicine, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Abiola Abdulrahman Ayanlaja
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yi-Fang Liu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Wei Xu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jiao Wu
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xue Li
- Department of Nursing Care, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Dian-Shuai Gao
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Correspondence to: Dian-Shuai Gao, .
| |
Collapse
|
16
|
Bonanni R, Cariati I, Tarantino U, D’Arcangelo G, Tancredi V. Physical Exercise and Health: A Focus on Its Protective Role in Neurodegenerative Diseases. J Funct Morphol Kinesiol 2022; 7:jfmk7020038. [PMID: 35645300 PMCID: PMC9149968 DOI: 10.3390/jfmk7020038] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023] Open
Abstract
Scientific evidence has demonstrated the power of physical exercise in the prevention and treatment of numerous chronic and/or age-related diseases, such as musculoskeletal, metabolic, and cardiovascular disorders. In addition, regular exercise is known to play a key role in the context of neurodegenerative diseases, as it helps to reduce the risk of their onset and counteracts their progression. However, the underlying molecular mechanisms have not yet been fully elucidated. In this regard, neurotrophins, such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), glia cell line-derived neurotrophic factor (GDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4), have been suggested as key mediators of brain health benefits, as they are involved in neurogenesis, neuronal survival, and synaptic plasticity. The production of these neurotrophic factors, known to be increased by physical exercise, is downregulated in neurodegenerative disorders, suggesting their fundamental importance in maintaining brain health. However, the mechanism by which physical exercise promotes the production of neurotrophins remains to be understood, posing limits on their use for the development of potential therapeutic strategies for the treatment of neurodegenerative diseases. In this literature review, we analyzed the most recent evidence regarding the relationship between physical exercise, neurotrophins, and brain health, providing an overview of their involvement in the onset and progression of neurodegeneration.
Collapse
Affiliation(s)
- Roberto Bonanni
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (R.B.); (U.T.)
| | - Ida Cariati
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (R.B.); (U.T.)
- Correspondence:
| | - Umberto Tarantino
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (R.B.); (U.T.)
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, 00133 Rome, Italy
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (G.D.); (V.T.)
| | - Giovanna D’Arcangelo
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (G.D.); (V.T.)
- Department of Systems Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy
| | - Virginia Tancredi
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (G.D.); (V.T.)
- Department of Systems Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy
| |
Collapse
|
17
|
The Effects of the Task Balance Training Program on the Glial Cell Line-Derived Neurotrophic Factor Levels, Cognitive Function, and Postural Balance in Old People. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9887985. [PMID: 35360515 PMCID: PMC8964155 DOI: 10.1155/2022/9887985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 02/10/2022] [Accepted: 02/19/2022] [Indexed: 11/23/2022]
Abstract
Exercise in the form of physical activity can provide neuroprotective benefits. The purpose of this study is to determine the effect of the task balance training program (TBT program) on the glial cell-derived neurotrophic factor levels, cognitive function, and postural balance in old people. The population of this study was the old people members of the Batara Hati Mulia Gowa Foundation who were willing to participate in the study (n = 66). The sample of this study was obtained through a random sampling technique to determine the treatment (n = 32) and control (n = 34) groups. Before and after implementing the TBT program, glial cell-derived neurotrophic factor (GDNF) level measurement and cognitive function and postural balance assessment were performed. Cognitive function was measured by using Montreal cognitive assessment (MoCA). Postural balance was measured in two ways by using the timed up and go (TUG) test and Tinetti performance-oriented mobility assessment (POMA). The treatment group showed significantly greater changes than the control group in GDNF levels (2.24 (±0.63) vs. 1.24 (±0.43), P = 0.001), cognitive function (24.66 (±3.42) vs. 19.18 (±2.67), P = 0.001), and postural balance (TUG [14.00 (±4.04) vs. 18.68 (±3.98)]; POMA [26.53 (±1.74) vs. 23.47 (±3.06)], P = 0.001) after training. The treatment group also showed a significant relationship between GDNF levels and cognitive function (r = 0.840, P = 0.001) and postural balance (TUG [r = 0.814, P = 0.001]; POMA [r = 0.630, P = 0.001]). The TBT program affects the levels of GDNF in old people. The TBT program involves cognitive function improvement and affects postural balance changes in old people.
Collapse
|
18
|
Therapeutic potential of neurotrophic factors in Alzheimer's Disease. Mol Biol Rep 2021; 49:2345-2357. [PMID: 34826049 DOI: 10.1007/s11033-021-06968-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia among the elderly population. AD is accompanied with the dysregulation of specific neurotrophic factors (NTFs) and their receptors, which plays a critical role in neuronal degeneration. NTFs are small proteins with therapeutic potential for human neurodegenerative diseases. These growth factors are categorized into four families: neurotrophins, neurokines, the glial cell line-derived NTF family of ligands, and the newly discovered cerebral dopamine NTF/mesencephalic astrocyte-derived NTF family. NTFs are capable of preventing cell death in degenerative conditions and can increase the neuronal growth and function in these disorders. Nevertheless, the adverse side effects of NTFs delivery and poor diffusion of these factors in the brain restrict the efficacy of NTFs therapy in clinical situations. MATERIALS AND METHODS In this review, we focus on the current advances in the use of NTFs to treat AD and summarize previous experimental and clinical studies for supporting the protective and therapeutic effects of these factors. CONCLUSION Based on reports, NTFs are considered as new and promising candidates for treating AD and AD-associated cognitive impairment.
Collapse
|
19
|
The Potential Role of Cytokines and Growth Factors in the Pathogenesis of Alzheimer's Disease. Cells 2021; 10:cells10102790. [PMID: 34685770 PMCID: PMC8534363 DOI: 10.3390/cells10102790] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/06/2021] [Accepted: 10/10/2021] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most prominent neurodegenerative diseases, which impairs cognitive function in afflicted individuals. AD results in gradual decay of neuronal function as a consequence of diverse degenerating events. Several neuroimmune players (such as cytokines and growth factors that are key players in maintaining CNS homeostasis) turn aberrant during crosstalk between the innate and adaptive immunities. This aberrance underlies neuroinflammation and drives neuronal cells toward apoptotic decline. Neuroinflammation involves microglial activation and has been shown to exacerbate AD. This review attempted to elucidate the role of cytokines, growth factors, and associated mechanisms implicated in the course of AD, especially with neuroinflammation. We also evaluated the propensities and specific mechanism(s) of cytokines and growth factors impacting neuron upon apoptotic decline and further shed light on the availability and accessibility of cytokines across the blood-brain barrier and choroid plexus in AD pathophysiology. The pathogenic and the protective roles of macrophage migration and inhibitory factors, neurotrophic factors, hematopoietic-related growth factors, TAU phosphorylation, advanced glycation end products, complement system, and glial cells in AD and neuropsychiatric pathology were also discussed. Taken together, the emerging roles of these factors in AD pathology emphasize the importance of building novel strategies for an effective therapeutic/neuropsychiatric management of AD in clinics.
Collapse
|
20
|
Shityakov S, Hayashi K, Störk S, Scheper V, Lenarz T, Förster CY. The Conspicuous Link between Ear, Brain and Heart-Could Neurotrophin-Treatment of Age-Related Hearing Loss Help Prevent Alzheimer's Disease and Associated Amyloid Cardiomyopathy? Biomolecules 2021; 11:biom11060900. [PMID: 34204299 PMCID: PMC8235707 DOI: 10.3390/biom11060900] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/05/2021] [Accepted: 06/14/2021] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia in the elderly, is a neurodegenerative disorder associated with neurovascular dysfunction and cognitive decline. While the deposition of amyloid β peptide (Aβ) and the formation of neurofibrillary tangles (NFTs) are the pathological hallmarks of AD-affected brains, the majority of cases exhibits a combination of comorbidities that ultimately lead to multi-organ failure. Of particular interest, it can be demonstrated that Aβ pathology is present in the hearts of patients with AD, while the formation of NFT in the auditory system can be detected much earlier than the onset of symptoms. Progressive hearing impairment may beget social isolation and accelerate cognitive decline and increase the risk of developing dementia. The current review discusses the concept of a brain-ear-heart axis by which Aβ and NFT inhibition could be achieved through targeted supplementation of neurotrophic factors to the cochlea and the brain. Such amyloid inhibition might also indirectly affect amyloid accumulation in the heart, thus reducing the risk of developing AD-associated amyloid cardiomyopathy and cardiovascular disease.
Collapse
Affiliation(s)
- Sergey Shityakov
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, D-97080 Würzburg, Germany;
- Infochemistry Scientific Center, Laboratory of Chemoinformatics, ITMO University, 191002 Saint-Petersburg, Russia
| | - Kentaro Hayashi
- Advanced Stroke Center, Shimane University Hospital, 89-1 Enya, Shimane, Izumo 693-8501, Japan;
| | - Stefan Störk
- Comprehensive Heart Failure Q9 Center, University of Würzburg, D-97080 Würzburg, Germany;
| | - Verena Scheper
- Department of Otolaryngology, Hannover Medical School and Cluster of Excellence “Hearing4All”, 30625 Hannover, Germany;
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School and Cluster of Excellence “Hearing4All”, 30625 Hannover, Germany;
- Correspondence: (T.L.); (C.Y.F.)
| | - Carola Y. Förster
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, D-97080 Würzburg, Germany;
- Correspondence: (T.L.); (C.Y.F.)
| |
Collapse
|