1
|
Yu Y, Shen H, Qin Q, Wang J, Nie Y, Wen L, Tang Y, Qu M. The investigation of peripheral inflammatory and oxidative stress biomarkers in dementia with Lewy Bodies, compared with Alzheimer's Disease, and mild cognitive impairment. Neuroscience 2025; 568:209-218. [PMID: 39800047 DOI: 10.1016/j.neuroscience.2024.12.057] [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: 04/16/2024] [Revised: 12/05/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
Although inflammation and oxidative stress have been increasingly recognised as components of Alzheimer's disease (AD) and Parkinson's disease (PD) pathologies. Few studies have investigated peripheral inflammation, and none have examined oxidative stress in Dementia with Lewy bodies (DLB). The purpose of our study was to characterize and compare those biomarkers in DLB with those in AD and amnestic mild cognitive impairment (aMCI). Plasma samples were obtained from Chinese patients with DLB (n = 50), AD (n = 59), and aMCI (n = 30), and healthy controls (HCs) (n = 54). Peripheral inflammatory biomarkers, including interferon-gamma (IFN-γ), interleukins (IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12p70, IL-17A), tumor necrosis factor-alpha (TNF-α), and C-reactive protein (CRP). Oxidative stress markers, such as superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione peroxidase (GSH-Px), were also assessed. The findings revealed that DLB patients had higher IL-6 levels than AD and HCs and elevated IL-10 and IL-17A levels compared to HCs. In terms of oxidative stress, the levels of SOD were significantly lower and MDA were significantly higher in the DLB and AD compared with HCs. Significant positive correlations were found between Unified Parkinson's Disease Rating Scale (UPDRS) scores and CRP levels. Our study identifies a unique peripheral immune and oxidative stress profile in DLB, characterized by elevated IL-6, MDA, and reduced SOD levels, distinguishing it from AD. These findings, linked to α-synuclein (α-Syn) pathology, provide novel insights into DLB mechanisms and highlight potential biomarkers for disease monitoring, targeted therapies, and future clinical trials.
Collapse
Affiliation(s)
- Yueyi Yu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Huixin Shen
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Qi Qin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jing Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| | - Yuting Nie
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Lulu Wen
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Miao Qu
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Jimenez-Capdeville ME, Chi-Ahumada E, García-Ortega F, Castanedo-Cazares JP, Norman R, Rodríguez-Leyva I. Nuclear Alpha-Synuclein in Parkinson's Disease and the Malignant Transformation in Melanoma. Neurol Res Int 2025; 2025:1119424. [PMID: 39816956 PMCID: PMC11729518 DOI: 10.1155/nri/1119424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 12/11/2024] [Indexed: 01/18/2025] Open
Abstract
Background: Alpha-synuclein (ASyn), a marker of Parkinson's disease (PD) and other neurodegenerative processes, plays pivotal roles in neuronal nuclei and synapses. ASyn and its phosphorylated form at Serine 129 (p-ASyn) are involved in DNA protection and repair, processes altered in aging, neurodegeneration, and cancer. Objective: To analyze the localization of p-ASyn in skin biopsies of PD patients and melanoma. Methods: Biopsies from 26 PD patients, 20 melanoma patients, and 31 control subjects were probed and analyzed with a p-ASyn antibody by immunohistochemistry and immunofluorescence. Nuclear positivity was quantified by image analysis. Results: Peripheral nerve endings from healthy subjects show little p-ASyn immunopositivity but notable axonal presence in PD. Control subjects show immunopositivity to p-ASyn along all epidermic strata and scarce presence in their cytoplasm. In contrast, its nuclear presence in PD is weaker, with a higher cytoplasmic and intercellular presence. Nuclear p-ASyn in melanoma varied from similar to control skin in early stage melanoma to a higher rate of empty nuclei in the intermediate stage and total absence of nuclear p-ASyn in severe cases. Interpretation: These findings support the nuclear localization of p-ASyn in skin cells and show that its presence decreases PD and almost disappears in the malignant transformation of melanocytes, redistributing to the cytoplasm and intercellular spaces. This confirms the association between PD and melanoma, providing crucial insights into the role of p-ASyn in both diseases. Trial Registration: ClinicalTrials.gov identifier: NCT01380899.
Collapse
Affiliation(s)
- María E. Jimenez-Capdeville
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosi, Mexico
| | - Erika Chi-Ahumada
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosi, Mexico
| | - Francisco García-Ortega
- Coordinación Académica Región Altiplano, Universidad Autonóma de San Luis Potosi, Matehuala, San Luis Potosí, Mexico
| | | | - Robert Norman
- Center for Geriatric Dermatology, Integrative Dermatology and Euro-Dermatology, Tampa, Florida, USA
| | - Ildefonso Rodríguez-Leyva
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosi, Mexico
- Departamento de Neurología, Hospital “Ignacio Morones Prieto”, San Luis Potosí, Mexico
| |
Collapse
|
3
|
Paubel A, Marouillat S, Dangoumau A, Maurel C, Haouari S, Blasco H, Corcia P, Laumonnier F, Andres CR, Vourc’h P. Dynamic Expression of Genes Encoding Ubiquitin Conjugating Enzymes (E2s) During Neuronal Differentiation and Maturation: Implications for Neurodevelopmental Disorders and Neurodegenerative Diseases. Genes (Basel) 2024; 15:1381. [PMID: 39596581 PMCID: PMC11593721 DOI: 10.3390/genes15111381] [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: 09/30/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Background: The ubiquitination process plays a crucial role in neuronal differentiation and function. Numerous studies have focused on the expression and functions of E3 ligases during these different stages, far fewer on E2 conjugating enzymes. In mice, as in humans, these E2s belong to 17 conjugating enzyme families. Objectives: We analyzed by real-time PCR the expression dynamics of all known E2 genes during an in vitro differentiation of mouse hippocampal neuronal cultures, and after, we analyzed their stimulation with N-methyl-D-aspartate (NMDA). Results: We found that 36 of the 38 E2 genes were expressed in hippocampal neurons. Many were up-regulated during neuritogenesis and/or synaptogenesis stages, such as Ube2h, Ube2b, and Aktip. Rapid and delayed responses to NMDA stimulation were associated with the increased expression of several E2 genes, such as Ube2i, the SUMO-conjugating E2 enzyme. We also observed similar expression profiles within the same E2 gene family, consistent with the presence of similar transcription factor binding sites in their respective promoter sequences. Conclusions: Our study indicates that specific expression profiles of E2 genes are correlated with changes in neuronal differentiation and activity. A better understanding of the regulation and function of E2s is needed to better understand the role played by the ubiquitination process in physiological mechanisms and pathophysiological alterations involved in neurodevelopmental or neurodegenerative diseases.
Collapse
Affiliation(s)
- Agathe Paubel
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
| | - Sylviane Marouillat
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
| | - Audrey Dangoumau
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
| | - Cindy Maurel
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
| | - Shanez Haouari
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
| | - Hélène Blasco
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37032 Tours, France
| | - Philippe Corcia
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
- Service de Neurologie, Centre SLA, CHU de Tours, 37032 Tours, France
| | - Frédéric Laumonnier
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
| | - Christian R. Andres
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37032 Tours, France
| | - Patrick Vourc’h
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37044 Tours, France; (A.P.); (A.D.); (C.M.); (S.H.); (H.B.); (P.C.); (F.L.); (C.R.A.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37032 Tours, France
| |
Collapse
|
4
|
Lukashevich MV, Rudenok MM, Semenova EI, Partevian SA, Karabanov AV, Fedotova EY, Illarioshkin SN, Slominsky PA, Shadrina MI, Alieva AK. Analysis of Expression of the GRIPAP1, DLG4, KIF1B, NGFRAP1, and NRF1 Genes in Peripheral Blood of the Patients with Parkinson's Disease in the Early Clinical Stages. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1779-1788. [PMID: 39523115 DOI: 10.1134/s0006297924100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
Parkinson's disease (PD) is one of the most common progressive neurodegenerative diseases. An important feature of the disease is its long latent period, which necessitates search for prognostic biomarkers. One method of identifying biomarkers of PD is to study changes in gene expression in peripheral blood of the patients in early stages of the disease and have not been treated. In this study, we analyzed relative mRNA levels of the genes GRIPAP1, DLG4, KIF1B, NGFRAP1, and NRF1, which are associated with neurotransmitter transport, apoptosis, and mitochondrial dysfunction, in the peripheral blood of PD patients using reverse transcription and real-time PCR with TaqMan probes. The results of this study suggest that the GRIPAP1 and DLG4 genes could be considered as potential biomarkers for the early clinical stages of Parkinson's disease. The data obtained may indicate that NGFRAP1 is involved in pathogenesis of both PD and other neurodegenerative diseases. Furthermore, in the early clinical stages of the disease we studied, the KIF1B and NRF1 genes were found not to be involved in PD pathogenesis at the expression level.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Petr A Slominsky
- National Research Centre "Kurchatov Institute", Moscow, 123182, Russia
| | - Maria I Shadrina
- National Research Centre "Kurchatov Institute", Moscow, 123182, Russia
| | - Anelya Kh Alieva
- National Research Centre "Kurchatov Institute", Moscow, 123182, Russia
| |
Collapse
|
5
|
Huenchuguala S, Segura-Aguilar J. Natural Compounds That Activate the KEAP1/Nrf2 Signaling Pathway as Potential New Drugs in the Treatment of Idiopathic Parkinson's Disease. Antioxidants (Basel) 2024; 13:1125. [PMID: 39334784 PMCID: PMC11428591 DOI: 10.3390/antiox13091125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recently, a single-neuron degeneration model has been proposed to understand the development of idiopathic Parkinson's disease based on (i) the extremely slow development of the degenerative process before the onset of motor symptoms and during the progression of the disease and (ii) the fact that it is triggered by an endogenous neurotoxin that does not have an expansive character, limiting its neurotoxic effect to single neuromelanin-containing dopaminergic neurons. It has been proposed that aminochrome is the endogenous neurotoxin that triggers the neurodegenerative process in idiopathic Parkinson's disease by triggering mitochondrial dysfunction, oxidative stress, neuroinflammation, dysfunction of both lysosomal and proteasomal protein degradation, endoplasmic reticulum stress and formation of neurotoxic alpha-synuclein oligomers. Aminochrome is an endogenous neurotoxin that is rapidly reduced by flavoenzymes and/or forms adducts with proteins, which implies that it is impossible for it to have a propagative neurotoxic effect on neighboring neurons. Interestingly, the enzymes DT-diaphorase and glutathione transferase M2-2 prevent the neurotoxic effects of aminochrome. Natural compounds present in fruits, vegetables and other plant products have been shown to activate the KEAP1/Nrf2 signaling pathway by increasing the expression of antioxidant enzymes including DT-diaphorase and glutathione transferase. This review analyzes the possibility of searching for natural compounds that increase the expression of DT-diaphorase and glutathione transferase through activation of the KEAP1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Sandro Huenchuguala
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Santiago 8370003, Chile;
| | - Juan Segura-Aguilar
- Molecular & Clinical Pharmacology, Instituto de Ciencias Biomédicas (ICBM), Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
6
|
Leupold D, Buder S, Pfeifer L, Szyc L, Riederer P, Strobel S, Monoranu CM. New Aspects Regarding the Fluorescence Spectra of Melanin and Neuromelanin in Pigmented Human Tissue Concerning Hypoxia. Int J Mol Sci 2024; 25:8457. [PMID: 39126026 PMCID: PMC11313424 DOI: 10.3390/ijms25158457] [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: 06/06/2024] [Revised: 07/11/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Melanin is a crucial pigment in melanomagenesis. Its fluorescence in human tissue is exceedingly weak but can be detected through advanced laser spectroscopy techniques. The spectral profile of melanin fluorescence distinctively varies among melanocytes, nevomelanocytes, and melanoma cells, with melanoma cells exhibiting a notably "red" fluorescence spectrum. This characteristic enables the diagnosis of melanoma both in vivo and in histological samples. Neuromelanin, a brain pigment akin to melanin, shares similar fluorescence properties. Its fluorescence can also be quantified with high spectral resolution using the same laser spectroscopic methods. Documented fluorescence spectra of neuromelanin in histological samples from the substantia nigra substantiate these findings. Our research reveals that the spectral behavior of neuromelanin fluorescence mirrors that of melanin in melanomas. This indicates that the typical red fluorescence is likely influenced by the microenvironment around (neuro)melanin, rather than by direct pigment interactions. Our ongoing studies aim to further explore this distinctive "red" fluorescence. We have observed this red fluorescence spectrum in post-mortem measurements of melanin in benign nevus. The characteristic red spectrum is also evident here (unlike the benign nevus in vivo), suggesting that hypoxia may contribute to this phenomenon. Given the central role of hypoxia in both melanoma development and treatment, as well as in fundamental Parkinson's disease mechanisms, this study discusses strategies aimed at reinforcing the hypothesis that red fluorescence from (neuro)melanin serves as an indicator of hypoxia.
Collapse
Affiliation(s)
- Dieter Leupold
- LTB Lasertechnik Berlin GmbH, 12489 Berlin, Germany; (D.L.); (L.P.)
| | - Susanne Buder
- Clinic for Dermatology and Venerology, Vivantes Klinikum Neukölln, 12351 Berlin, Germany;
| | - Lutz Pfeifer
- LTB Lasertechnik Berlin GmbH, 12489 Berlin, Germany; (D.L.); (L.P.)
| | | | - Peter Riederer
- Department and Research Unit of Psychiatry, University of Southern Denmark, 5230 Odense, Denmark;
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Sabrina Strobel
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Comprehensive Cancer Center (CCC) Mainfranken Wuerzburg, 97080 Wuerzburg, Germany;
| | - Camelia-Maria Monoranu
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Comprehensive Cancer Center (CCC) Mainfranken Wuerzburg, 97080 Wuerzburg, Germany;
| |
Collapse
|
7
|
Choza JI, Virani M, Kuhn NC, Adams M, Kochmanski J, Bernstein AI. Parkinson's disease-associated shifts between DNA methylation and DNA hydroxymethylation in human brain in PD-related genes, including PARK19 (DNAJC6) and PTPRN2 (IA-2β). RESEARCH SQUARE 2024:rs.3.rs-4572401. [PMID: 39070644 PMCID: PMC11275970 DOI: 10.21203/rs.3.rs-4572401/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background The majority of Parkinson's disease (PD) cases are due to a complex interaction between aging, genetics, and environmental factors; epigenetic mechanisms are thought to act as important mediators of these risk factors. While multiple studies to date have explored the role of DNA modifications in PD, few focus on 5-hydroxymethylcytosine (5hmC). Because 5hmC occurs at its highest levels in the brain and is thought to be particularly important in the central nervous system, particularly in the response to neurotoxicants, it is important to explore the potential role of 5hmC in PD. This study expands on our previously published epigenome-wide association study (EWAS) performed on DNA isolated from neuron-enriched nuclei from human postmortem parietal cortex from the Banner Sun Health Research Institute Brain Bank. The study aimed to identify paired changes in 5hmC and 5mC in PD in enriched neuronal nuclei isolated from PD post-mortem parietal cortex and age- and sex-matched controls. We performed oxidative bisulfite (oxBS) conversion and paired it with our previously published bisulfite (BS)-based EWAS on the same samples to identify cytosines with significant shifts between these two related epigenetic marks. Interaction differentially modified cytosines (iDMCs) were identified using our recently published mixed-effects model for co-analyzing βmC and βhmC data. Results We identified 1,030 iDMCs with paired changes in 5mC and 5hmC (FDR < 0.05) that map to 695 genes, including PARK19 (DNAJC6), a familial PD gene, and PTPRN2 (IA-2), which has been previously implicated in PD in both epigenetic and mechanistic studies. The majority of iDMC-containing genes have not previously been implicated in PD and were not identified in our previous BS-based EWAS. Conclusions These data potentially link epigenetic regulation of the PARK19 and PTPRN2 loci in the pathogenesis of idiopathic PD. In addition, iDMC-containing genes have known functions in synaptic formation and function, cell cycle and senescence, neuroinflammation, and epigenetic regulation. These data suggest that there are significant shifts between 5mC and 5hmC associated with PD in genes relevant to PD pathogenesis that are not captured by analyzing BS-based data alone or by analyzing each mark as a distinct dataset.
Collapse
|
8
|
Huenchuguala S, Segura-Aguilar J. Targets to Search for New Pharmacological Treatment in Idiopathic Parkinson's Disease According to the Single-Neuron Degeneration Model. Biomolecules 2024; 14:673. [PMID: 38927076 PMCID: PMC11201619 DOI: 10.3390/biom14060673] [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: 03/15/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
One of the biggest problems in the treatment of idiopathic Parkinson's disease is the lack of new drugs that slow its progression. L-Dopa remains the star drug in the treatment of this disease, although it induces severe side effects. The failure of clinical studies with new drugs depends on the use of preclinical models based on neurotoxins that do not represent what happens in the disease since they induce rapid and expansive neurodegeneration. We have recently proposed a single-neuron degeneration model for idiopathic Parkinson's disease that requires years to accumulate enough lost neurons for the onset of motor symptoms. This single-neuron degeneration model is based on the excessive formation of aminochrome during neuromelanin synthesis that surpass the neuroprotective action of the enzymes DT-diaphorase and glutathione transferase M2-2, which prevent the neurotoxic effects of aminochrome. Although the neurotoxic effects of aminochrome do not have an expansive effect, a stereotaxic injection of this endogenous neurotoxin cannot be used to generate a preclinical model in an animal. Therefore, the aim of this review is to evaluate the strategies for pharmacologically increasing the expression of DT diaphorase and GSTM2-2 and molecules that induce the expression of vesicular monoamine transporter 2, such as pramipexole.
Collapse
Affiliation(s)
- Sandro Huenchuguala
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Santiago 8370003, Chile;
| | - Juan Segura-Aguilar
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago 8330111, Chile
| |
Collapse
|
9
|
Fishman-Jacob T, Youdim MBH. A sporadic Parkinson's disease model via silencing of the ubiquitin-proteasome/E3 ligase component, SKP1A. J Neural Transm (Vienna) 2024; 131:675-707. [PMID: 37644186 PMCID: PMC11192832 DOI: 10.1007/s00702-023-02687-6] [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: 04/28/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Our and other's laboratory microarray-derived transcriptomic studies in human PD substantia nigra pars compacta (SNpc) samples have opened an avenue to concentrate on potential gene intersections or cross-talks along the dopaminergic (DAergic) neurodegenerative cascade in sporadic PD (SPD). One emerging gene candidate identified was SKP1A (p19, S-phase kinase-associated protein 1A), found significantly decreased in the SNpc as confirmed later at the protein level. SKP1 is part of the Skp1, Cullin 1, F-box protein (SCF) complex, the largest known class of sophisticated ubiquitin-proteasome/E3-ligases and was found to directly interact with FBXO7, a gene defective in PARK15-linked PD. This finding has led us to the hypothesis that a targeted site-specific reduction of Skp1 levels in DAergic neuronal cell culture and animal systems may result in a progressive loss of DAergic neurons and hopefully recreate motor disabilities in animals. The second premise considers the possibility that both intrinsic and extrinsic factors (e.g., manipulation of selected genes and mitochondria impairing toxins), alleged to play central roles in DAergic neurodegeneration in PD, may act in concert as modifiers of Skp1 deficiency-induced phenotype alterations ('dual-hit' hypothesis of neurodegeneration). To examine a possible role of Skp1 in DAergic phenotype, we have initially knocked down the expression of SKP1A gene in an embryonic mouse SN-derived cell line (SN4741) with short hairpin RNA (shRNA) lentiviruses (LVs). The deficiency of SKP1A closely recapitulated cardinal features of the DAergic pathology of human PD, such as decreased expression of DAergic phenotypic markers and cell cycle aberrations. Furthermore, the knocked down cells displayed a lethal phenotype when induced to differentiate exhibiting proteinaceous round inclusion structures, which were almost identical in composition to human Lewy bodies, a hallmark of PD. These findings support a role for Skp1 in neuronal phenotype, survival, and differentiation. The identification of Skp1 as a key player in DAergic neuron function suggested that a targeted site-specific reduction of Skp1 levels in mice SNpc may result in a progressive loss of DAergic neurons and terminal projections in the striatum. The injected LV SKP1shRNA to mouse SN resulted in decreased expression of Skp1 protein levels within DAergic neurons and loss of tyrosine hydroxylase immunoreactivity (TH-IR) in both SNpc and striatum that was accompanied by time-dependent motor disabilities. The reduction of the vertical movements, that is rearing, may be reminiscent of the early occurrence of hypokinesia and axial, postural instability in PD. According to the 'dual-hit' hypothesis of neurodegenerative diseases, it is predicted that gene-gene and/or gene-environmental factors would act in concert or sequentially to propagate the pathological process of PD. Our findings are compatible with this conjecture showing that the genetic vulnerability caused by knock down of SKP1A renders DAergic SN4741 cells especially sensitive to genetic reduction of Aldh1 and exposure to the external stressors MPP+ and DA, which have been implicated in PD pathology. Future consideration should be given in manipulation SKP1A expression as therapeutic window, via its induction genetically or pharmacological, to prevent degeneration of the nigra striatal dopamine neurons, since UPS is defective.
Collapse
Affiliation(s)
- Tali Fishman-Jacob
- Youdim Pharmaceutical Ltd, New Northern Industrial Park, 1 Ha- Tsmikha St, Stern Building, Fl-3, P. O. Box 72, 2069207, Yokneam, Israel
| | - Moussa B H Youdim
- Youdim Pharmaceutical Ltd, New Northern Industrial Park, 1 Ha- Tsmikha St, Stern Building, Fl-3, P. O. Box 72, 2069207, Yokneam, Israel.
| |
Collapse
|
10
|
Chapman MA, Sorg BA. A Systematic Review of Extracellular Matrix-Related Alterations in Parkinson's Disease. Brain Sci 2024; 14:522. [PMID: 38928523 PMCID: PMC11201521 DOI: 10.3390/brainsci14060522] [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: 04/17/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
The role of the extracellular matrix (ECM) in Parkinson's disease (PD) is not well understood, even though it is critical for neuronal structure and signaling. This systematic review identified the top deregulated ECM-related pathways in studies that used gene set enrichment analyses (GSEA) to document transcriptomic, proteomic, or genomic alterations in PD. PubMed and Google scholar were searched for transcriptomics, proteomics, or genomics studies that employed GSEA on data from PD tissues or cells and reported ECM-related pathways among the top-10 most enriched versus controls. Twenty-seven studies were included, two of which used multiple omics analyses. Transcriptomics and proteomics studies were conducted on a variety of tissue and cell types. Of the 17 transcriptomics studies (16 data sets), 13 identified one or more adhesion pathways in the top-10 deregulated gene sets or pathways, primarily related to cell adhesion and focal adhesion. Among the 8 proteomics studies, 5 identified altered overarching ECM gene sets or pathways among the top 10. Among the 4 genomics studies, 3 identified focal adhesion pathways among the top 10. The findings summarized here suggest that ECM organization/structure and cell adhesion (particularly focal adhesion) are altered in PD and should be the focus of future studies.
Collapse
Affiliation(s)
| | - Barbara A. Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, USA;
| |
Collapse
|
11
|
Vasilyeva OY, Tolmacheva EN, Dmitriev AE, Darkova YA, Sazhenova EA, Nikitina TV, Lebedev IN, Vasilyev SA. Aberrant methylation of placental development genes in chorionic villi of spontaneous abortions with trisomy 16. Vavilovskii Zhurnal Genet Selektsii 2024; 28:198-203. [PMID: 38680176 PMCID: PMC11043499 DOI: 10.18699/vjgb-24-24] [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: 11/17/2023] [Revised: 01/18/2024] [Accepted: 01/18/2023] [Indexed: 05/01/2024] Open
Abstract
In humans, aneuploidy is incompatible with the birth of healthy children and mainly leads to the death of embryos in the early stages of development in the first trimester of pregnancy. Trisomy 16 is the most common aneuploidy among spontaneous abortions of the first trimester of pregnancy. However, the mechanisms leading to the death of embryos with trisomy 16 remain insufficiently investigated. One of these potential mechanisms is abnormal placental development, including aberrant remodeling of spiral arteries. Spiral artery remodeling involves the migration of trophoblast cells into the maternal spiral arteries, replacing their endothelium and remodeling to ensure a stable embryonic nutrition and oxygen supply. This is a complex process which depends on many factors from both the embryo and the mother. We analyzed the methylation level of seven genes (ADORA2B, NPR3, PRDM1, PSG2, PHTLH, SV2C, and TICAM2) involved in placental development in the chorionic villi of spontaneous abortions with trisomy 16 (n = 14), compared with spontaneous abortions with a normal karyotype (n = 31) and the control group of induced abortions (n = 10). To obtain sequencing libraries, targeted amplification of individual gene regions using designed oligonucleotide primers for bisulfite-converted DNA was used. The analysis was carried out using targeted bisulfite massive parallel sequencing. In the group of spontaneous abortions with trisomy 16, the level of methylation of the PRDM1 and PSG2 genes was significantly increased compared to induced abortions (p = 0.0004 and p = 0.0015, respectively). In the group of spontaneous abortions, there was no increase in the level of methylation of the PRDM1 and PSG2 genes, but the level of methylation of the ADORA2B gene was significantly increased compared to the induced abortions (p = 0.032). The results obtained indicate the potential mechanisms of the pathogenetic effect of trisomy 16 on the placental development with the participation of the studied genes.
Collapse
Affiliation(s)
- O Yu Vasilyeva
- Research Institute of Medical Genetics of the Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - E N Tolmacheva
- Research Institute of Medical Genetics of the Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - A E Dmitriev
- National Research Tomsk State University, Tomsk, Russia
| | - Ya A Darkova
- National Research Tomsk State University, Tomsk, Russia
| | - E A Sazhenova
- Research Institute of Medical Genetics of the Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - T V Nikitina
- Research Institute of Medical Genetics of the Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - I N Lebedev
- Research Institute of Medical Genetics of the Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - S A Vasilyev
- Research Institute of Medical Genetics of the Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
12
|
Kon T, Lee S, Martinez-Valbuena I, Yoshida K, Tanikawa S, Lang AE, Kovacs GG. Molecular Behavior of α-Synuclein Is Associated with Membrane Transport, Lipid Metabolism, and Ubiquitin-Proteasome Pathways in Lewy Body Disease. Int J Mol Sci 2024; 25:2676. [PMID: 38473923 DOI: 10.3390/ijms25052676] [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: 01/19/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Lewy body diseases (LBDs) feature α-synuclein (α-syn)-containing Lewy bodies, with misfolded α-syn potentially propagating as seeds. Using a seeding amplification assay, we previously reported distinct α-syn seeding in LBD cases based on the area under seeding curves. This study revealed that LBD cases showing different α-syn seeding kinetics have distinct proteomics profiles, emphasizing disruptions in mitochondria and lipid metabolism in high-seeder cases. Though the mechanisms underlying LBD development are intricate, the factors influencing α-syn seeding activity remain elusive. To address this and complement our previous findings, we conducted targeted transcriptome analyses in the substantia nigra using the nanoString nCounter assay together with histopathological evaluations in high (n = 4) and low (n = 3) nigral α-syn seeders. Neuropathological findings (particularly the substantia nigra) were consistent between these groups and were characterized by neocortical LBD associated with Alzheimer's disease neuropathologic change. Among the 1811 genes assessed, we identified the top 20 upregulated and downregulated genes and pathways in α-syn high seeders compared with low seeders. Notably, alterations were observed in genes and pathways related to transmembrane transporters, lipid metabolism, and the ubiquitin-proteasome system in the high α-syn seeders. In conclusion, our findings suggest that the molecular behavior of α-syn is the driving force in the neurodegenerative process affecting the substantia nigra through these identified pathways. These insights highlight their potential as therapeutic targets for attenuating LBD progression.
Collapse
Affiliation(s)
- Tomoya Kon
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Toronto, ON M5T 0S8, Canada
- Department of Neurology, Hirosaki University Graduate School of Medicine, 5 Zaifu, Hirosaki 036-8562, Japan
| | - Seojin Lee
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 200 Elizabeth St., Toronto, ON M5G 2C4, Canada
| | - Ivan Martinez-Valbuena
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Toronto, ON M5T 0S8, Canada
| | - Koji Yoshida
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Toronto, ON M5T 0S8, Canada
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Satoshi Tanikawa
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Toronto, ON M5T 0S8, Canada
| | - Anthony E Lang
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Toronto, ON M5T 0S8, Canada
- Edmond J Safra Program in Parkinson's Disease and Rossy Progressive Supranuclear Palsy Centre, Toronto Western Hospital, 399 Bathurst St., Toronto, ON M5T 2S8, Canada
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, 60 Leonard Ave., Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 200 Elizabeth St., Toronto, ON M5G 2C4, Canada
- Edmond J Safra Program in Parkinson's Disease and Rossy Progressive Supranuclear Palsy Centre, Toronto Western Hospital, 399 Bathurst St., Toronto, ON M5T 2S8, Canada
- Laboratory Medicine Program, University Health Network, 200 Elizabeth St., Toronto, ON M5G 2C4, Canada
- Krembil Brain Institute, University of Toronto, 60 Leonard Ave., Toronto, ON M5T 0S8, Canada
| |
Collapse
|
13
|
Janssen Daalen JM, Koopman WJH, Saris CGJ, Meinders MJ, Thijssen DHJ, Bloem BR. The Hypoxia Response Pathway: A Potential Intervention Target in Parkinson's Disease? Mov Disord 2024; 39:273-293. [PMID: 38140810 DOI: 10.1002/mds.29688] [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: 07/14/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder for which only symptomatic treatments are available. Both preclinical and clinical studies suggest that moderate hypoxia induces evolutionarily conserved adaptive mechanisms that enhance neuronal viability and survival. Therefore, targeting the hypoxia response pathway might provide neuroprotection by ameliorating the deleterious effects of mitochondrial dysfunction and oxidative stress, which underlie neurodegeneration in PD. Here, we review experimental studies regarding the link between PD pathophysiology and neurophysiological adaptations to hypoxia. We highlight the mechanistic differences between the rescuing effects of chronic hypoxia in neurodegeneration and short-term moderate hypoxia to improve neuronal resilience, termed "hypoxic conditioning". Moreover, we interpret these preclinical observations regarding the pharmacological targeting of the hypoxia response pathway. Finally, we discuss controversies with respect to the differential effects of hypoxia response pathway activation across the PD spectrum, as well as intervention dosing in hypoxic conditioning and potential harmful effects of such interventions. We recommend that initial clinical studies in PD should focus on the safety, physiological responses, and mechanisms of hypoxic conditioning, as well as on repurposing of existing pharmacological compounds. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jules M Janssen Daalen
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Werner J H Koopman
- Department of Pediatrics, Amalia Children's Hospital, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Christiaan G J Saris
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjan J Meinders
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| |
Collapse
|
14
|
Patterson JR, Kochmanski J, Stoll AC, Kubik M, Kemp CJ, Duffy MF, Thompson K, Howe JW, Cole-Strauss A, Kuhn NC, Miller KM, Nelson S, Onyekpe CU, Beck JS, Counts SE, Bernstein AI, Steece-Collier K, Luk KC, Sortwell CE. Transcriptomic profiling of early synucleinopathy in rats induced with preformed fibrils. NPJ Parkinsons Dis 2024; 10:7. [PMID: 38172128 PMCID: PMC10764951 DOI: 10.1038/s41531-023-00620-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Examination of early phases of synucleinopathy when inclusions are present, but long before neurodegeneration occurs, is critical to both understanding disease progression and the development of disease modifying therapies. The rat alpha-synuclein (α-syn) preformed fibril (PFF) model induces synchronized synucleinopathy that recapitulates the pathological features of Parkinson's disease (PD) and can be used to study synucleinopathy progression. In this model, phosphorylated α-syn (pSyn) inclusion-containing neurons and reactive microglia (major histocompatibility complex-II immunoreactive) peak in the substantia nigra pars compacta (SNpc) months before appreciable neurodegeneration. However, it remains unclear which specific genes are driving these phenotypic changes. To identify transcriptional changes associated with early synucleinopathy, we used laser capture microdissection of the SNpc paired with RNA sequencing (RNASeq). Precision collection of the SNpc allowed for the assessment of differential transcript expression in the nigral dopamine neurons and proximal glia. Transcripts upregulated in early synucleinopathy were mainly associated with an immune response, whereas transcripts downregulated were associated with neurotransmission and the dopamine pathway. A subset of 29 transcripts associated with neurotransmission/vesicular release and the dopamine pathway were verified in a separate cohort of males and females to confirm reproducibility. Within this subset, fluorescent in situ hybridization (FISH) was used to localize decreases in the Syt1 and Slc6a3 transcripts to pSyn inclusion-containing neurons. Identification of transcriptional changes in early synucleinopathy provides insight into the molecular mechanisms driving neurodegeneration.
Collapse
Affiliation(s)
- Joseph R Patterson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| | - Joseph Kochmanski
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Anna C Stoll
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Michael Kubik
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Christopher J Kemp
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Megan F Duffy
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Kajene Thompson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Jacob W Howe
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Allyson Cole-Strauss
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kathryn M Miller
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Seth Nelson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Christopher U Onyekpe
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John S Beck
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Alison I Bernstein
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
| | - Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
15
|
Salas-Leal AC, Salas-Pacheco SM, Hernández-Cosaín EI, Vélez-Vélez LM, Antuna-Salcido EI, Castellanos-Juárez FX, Méndez-Hernández EM, Llave-León OL, Quiñones-Canales G, Arias-Carrión O, Sandoval-Carrillo AA, Salas-Pacheco JM. Differential expression of PSMC4, SKP1, and HSPA8 in Parkinson's disease: insights from a Mexican mestizo population. Front Mol Neurosci 2023; 16:1298560. [PMID: 38115821 PMCID: PMC10728481 DOI: 10.3389/fnmol.2023.1298560] [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: 09/21/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative condition characterized by alpha-synuclein aggregation and dysfunctional protein degradation pathways. This study investigates the differential gene expression of pivotal components (UBE2K, PSMC4, SKP1, and HSPA8) within these pathways in a Mexican-Mestizo PD population compared to healthy controls. We enrolled 87 PD patients and 87 controls, assessing their gene expression levels via RT-qPCR. Our results reveal a significant downregulation of PSMC4, SKP1, and HSPA8 in the PD group (p = 0.033, p = 0.003, and p = 0.002, respectively). Logistic regression analyses establish a strong association between PD and reduced expression of PSMC4, SKP1, and HSPA8 (OR = 0.640, 95% CI = 0.415-0.987; OR = 0.000, 95% CI = 0.000-0.075; OR = 0.550, 95% CI = 0.368-0.823, respectively). Conversely, UBE2K exhibited no significant association or expression difference between the groups. Furthermore, we develop a gene expression model based on HSPA8, PSMC4, and SKP1, demonstrating robust discrimination between healthy controls and PD patients. Notably, the model's diagnostic efficacy is particularly pronounced in early-stage PD. In conclusion, our study provides compelling evidence linking decreased gene expression of PSMC4, SKP1, and HSPA8 to PD in the Mexican-Mestizo population. Additionally, our gene expression model exhibits promise as a diagnostic tool, particularly for early-stage PD diagnosis.
Collapse
Affiliation(s)
- Alma C. Salas-Leal
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - Sergio M. Salas-Pacheco
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - Erik I. Hernández-Cosaín
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - Lilia M. Vélez-Vélez
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | | | | | - Edna M. Méndez-Hernández
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - Osmel La Llave-León
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | | | - Oscar Arias-Carrión
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Ciudad de México, México
| | - Ada A. Sandoval-Carrillo
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - José M. Salas-Pacheco
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| |
Collapse
|
16
|
Riederer P, Nagatsu T, Youdim MBH, Wulf M, Dijkstra JM, Sian-Huelsmann J. Lewy bodies, iron, inflammation and neuromelanin: pathological aspects underlying Parkinson's disease. J Neural Transm (Vienna) 2023; 130:627-646. [PMID: 37062012 PMCID: PMC10121516 DOI: 10.1007/s00702-023-02630-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/29/2023] [Indexed: 04/17/2023]
Abstract
Since the description of some peculiar symptoms by James Parkinson in 1817, attempts have been made to define its cause or at least to enlighten the pathology of "Parkinson's disease (PD)." The vast majority of PD subtypes and most cases of sporadic PD share Lewy bodies (LBs) as a characteristic pathological hallmark. However, the processes underlying LBs generation and its causal triggers are still unknown. ɑ-Synuclein (ɑ-syn, encoded by the SNCA gene) is a major component of LBs, and SNCA missense mutations or duplications/triplications are causal for rare hereditary forms of PD. Thus, it is imperative to study ɑ-syn protein and its pathology, including oligomerization, fibril formation, aggregation, and spreading mechanisms. Furthermore, there are synergistic effects in the underlying pathogenic mechanisms of PD, and multiple factors-contributing with different ratios-appear to be causal pathological triggers and progression factors. For example, oxidative stress, reduced antioxidative capacity, mitochondrial dysfunction, and proteasomal disturbances have each been suggested to be causal for ɑ-syn fibril formation and aggregation and to contribute to neuroinflammation and neural cell death. Aging is also a major risk factor for PD. Iron, as well as neuromelanin (NM), show age-dependent increases, and iron is significantly increased in the Parkinsonian substantia nigra (SN). Iron-induced pathological mechanisms include changes of the molecular structure of ɑ-syn. However, more recent PD research demonstrates that (i) LBs are detected not only in dopaminergic neurons and glia but in various neurotransmitter systems, (ii) sympathetic nerve fibres degenerate first, and (iii) at least in "brain-first" cases dopaminergic deficiency is evident before pathology induced by iron and NM. These recent findings support that the ɑ-syn/LBs pathology as well as iron- and NM-induced pathology in "brain-first" cases are important facts of PD pathology and via their interaction potentiate the disease process in the SN. As such, multifactorial toxic processes posted on a personal genetic risk are assumed to be causal for the neurodegenerative processes underlying PD. Differences in ratios of multiple factors and their spatiotemporal development, and the fact that common triggers of PD are hard to identify, imply the existence of several phenotypical subtypes, which is supported by arguments from both the "bottom-up/dual-hit" and "brain-first" models. Therapeutic strategies are necessary to avoid single initiation triggers leading to PD.
Collapse
Affiliation(s)
- Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
- Department of Psychiatry, University of Southern Denmark Odense, J.B. Winslows Vey 18, 5000, Odense, Denmark.
| | - Toshiharu Nagatsu
- Center for Research Promotion and Support, School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | | | - Max Wulf
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801, Bochum, Germany
| | | | | |
Collapse
|
17
|
Rike WA, Stern S. Proteins and Transcriptional Dysregulation of the Brain Extracellular Matrix in Parkinson's Disease: A Systematic Review. Int J Mol Sci 2023; 24:ijms24087435. [PMID: 37108598 PMCID: PMC10138539 DOI: 10.3390/ijms24087435] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular matrix (ECM) of the brain is a dynamic structure made up of a vast network of bioactive macromolecules that modulate cellular events. Structural, organizational, and functional changes in these macromolecules due to genetic variation or environmental stressors are thought to affect cellular functions and may result in disease. However, most mechanistic studies to date usually focus on the cellular aspects of diseases and pay less attention to the relevance of the processes governing the dynamic nature of the extracellular matrix in disease pathogenesis. Thus, due to the ECM's diversified biological roles, increasing interest in its involvement in disease, and the lack of sufficient compiled evidence regarding its relationship with Parkinson's disease (PD) pathology, we aimed to compile the existing evidence to boost the current knowledge on the area and provide refined guidance for the future research. Here, in this review, we gathered postmortem brain tissue and induced pluripotent stem cell (iPSC)-related studies from PubMed and Google Scholar to identify, summarize and describe common macromolecular alterations in the expression of brain ECM components in Parkinson's disease (PD). A literature search was conducted up until 10 February 2023. The overall hits from the database and manual search for proteomic and transcriptome studies were 1243 and 1041 articles, respectively. Following a full-text review, 10 articles from proteomic and 24 from transcriptomic studies were found to be eligible for inclusion. According to proteomic studies, proteins such as collagens, fibronectin, annexins, and tenascins were recognized to be differentially expressed in Parkinson's disease. Transcriptomic studies displayed dysregulated pathways including ECM-receptor interaction, focal adhesion, and cell adhesion molecules in Parkinson's disease. A limited number of relevant studies were accessed from our search, indicating that much work remains to be carried out to better understand the roles of the ECM in neurodegeneration and Parkinson's disease. However, we believe that our review will elicit focused primary studies and thus support the ongoing efforts of the discovery and development of diagnostic biomarkers as well as therapeutic agents for Parkinson's disease.
Collapse
Affiliation(s)
- Wote Amelo Rike
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
18
|
Masato A, Plotegher N, Terrin F, Sandre M, Faustini G, Thor A, Adams S, Berti G, Cogo S, De Lazzari F, Fontana CM, Martinez PA, Strong R, Bandopadhyay R, Bisaglia M, Bellucci A, Greggio E, Dalla Valle L, Boassa D, Bubacco L. DOPAL initiates αSynuclein-dependent impaired proteostasis and degeneration of neuronal projections in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:42. [PMID: 36966140 PMCID: PMC10039907 DOI: 10.1038/s41531-023-00485-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 03/06/2023] [Indexed: 03/27/2023] Open
Abstract
Dopamine dyshomeostasis has been acknowledged among the determinants of nigrostriatal neuron degeneration in Parkinson's disease (PD). Several studies in experimental models and postmortem PD patients underlined increasing levels of the dopamine metabolite 3,4-dihydroxyphenylacetaldehyde (DOPAL), which is highly reactive towards proteins. DOPAL has been shown to covalently modify the presynaptic protein αSynuclein (αSyn), whose misfolding and aggregation represent a major trait of PD pathology, triggering αSyn oligomerization in dopaminergic neurons. Here, we demonstrated that DOPAL elicits αSyn accumulation and hampers αSyn clearance in primary neurons. DOPAL-induced αSyn buildup lessens neuronal resilience, compromises synaptic integrity, and overwhelms protein quality control pathways in neurites. The progressive decline of neuronal homeostasis further leads to dopaminergic neuron loss and motor impairment, as showed in in vivo models. Finally, we developed a specific antibody which detected increased DOPAL-modified αSyn in human striatal tissues from idiopathic PD patients, corroborating the translational relevance of αSyn-DOPAL interplay in PD neurodegeneration.
Collapse
Affiliation(s)
- Anna Masato
- Department of Biology, University of Padova, Padova, 35131, Italy
| | - Nicoletta Plotegher
- Department of Biology, University of Padova, Padova, 35131, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | - Francesca Terrin
- Department of Biology, University of Padova, Padova, 35131, Italy
| | - Michele Sandre
- Department of Neuroscience, University of Padova, Padova, 35131, Italy
| | - Gaia Faustini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Andrea Thor
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0608, USA
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, 92093-0608, USA
| | - Stephen Adams
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093-0608, USA
| | - Giulia Berti
- Department of Biology, University of Padova, Padova, 35131, Italy
| | - Susanna Cogo
- Department of Biology, University of Padova, Padova, 35131, Italy
| | | | | | - Paul Anthony Martinez
- Department of Pharmacology and Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care Network, San Antonio, TX, 78229, USA
| | - Randy Strong
- Department of Pharmacology and Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care Network, San Antonio, TX, 78229, USA
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, WC1N 1PJ, UK
| | - Marco Bisaglia
- Department of Biology, University of Padova, Padova, 35131, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, 25123, Italy
| | - Elisa Greggio
- Department of Biology, University of Padova, Padova, 35131, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | | | - Daniela Boassa
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0608, USA.
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, 92093-0608, USA.
| | - Luigi Bubacco
- Department of Biology, University of Padova, Padova, 35131, Italy.
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy.
| |
Collapse
|
19
|
Kolacheva A, Pavlova E, Bannikova A, Bogdanov V, Troshev D, Ugrumov M. The Gene Expression of Proteins Involved in Intercellular Signaling and Neurodegeneration in the Substantia Nigra in a Mouse Subchronic Model of Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24033027. [PMID: 36769355 PMCID: PMC9917821 DOI: 10.3390/ijms24033027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Given the limited access to clinical material for studying the pathogenesis of Parkinson's disease (PD), these studies should be carried out on experimental models. We have recently developed a subchronic model of the progressive development of PD with a gradual transition from the preclinical (asymptomatic) stage to the clinical (symptomatic) one. The aim of this study was to evaluate changes in the expression of a wide range of genes in the substantia nigra (SN), the central link in the regulation of motor function, in mice in our subchronic model of PD. We have found changes in the expression of a number of genes encoding enzymes involved in the synthesis and degradation of dopamine as well as proteins involved in the vesicular cycle, axonal transport, protein degradation in the proteasome system, neuroinflammation, and cell death in the SN of our mouse model of the clinical stage of PD. Similar changes in gene expression were previously demonstrated in patients (postmortem), indicating good reproducibility of PD in our model. Further analysis of the gene expression in the SN of mice has shown that the expression of some genes also changes in the model of the preclinical stage, when dopaminergic neurons have not yet died. Thus, this study opens up broad prospects for further evaluation of the molecular mechanisms of PD pathogenesis and the development of a test system for drug screening.
Collapse
|
20
|
Zhu PA, Lu QQ, Li ZL, Hu RL, Xu S, Brodersen L, Liu YX, Liu H, Bao X. Efficacy of Tai Chi on lower limb function of Parkinson's disease patients: A systematic review and meta-analysis. Front Aging Neurosci 2023; 15:1096417. [PMID: 36819715 PMCID: PMC9929552 DOI: 10.3389/fnagi.2023.1096417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/09/2023] [Indexed: 02/04/2023] Open
Abstract
Background At present, the effect of Tai Chi (TC) on lower limb function in patients with Parkinson's disease (PD) is controversial. Therefore, we conducted a meta-analysis on the influence of TC on lower limb function in PD patients. Methods According to the PRISMA guidelines, seven databases were searched. Randomized controlled trials (RCTS) were selected and screened according to inclusion and exclusion criteria. We assessed the quality of the studies using the Cochrane Risk of Bias tool and then extracted the characteristics of the included studies. The random effect model was adopted, and heterogeneity was measured by I 2 statistic. Results A total of 441 articles were screened, and 10 high-quality RCTs were with a total of 532 patients with PD met Our inclusion criteria. Meta-analysis showed that compared To control groups TC improved several outcomes. TC significantly improved motor function (SMD = -0.70; 95% CI = -0.95, -0.45; p < 0.001; I 2 = 35%), although The results were not statistically significant for The subgroup analysis of TC duration (SMD = -0.70; 95% CI = -0.95, -0.45; p = 0.88; I 2 = 0%;). TC significantly improved balance function (SMD = 0.89; 95% CI = 0.51, 1.27; p < 0.001; I 2 = 54%), functional walking capacity (SMD = -1.24; 95% CI = -2.40, -0.09; p = 0.04; I 2 = 95%), and gait velocity (SMD = 0.48; 95% CI = -0.02, 0.94; p = 0.04; I 2 = 78%), But Did Not improve endurance (SMD = 0.31; 95% CI = -0.12, 0.75; p = 0.16; I 2 = 0%), step length (SMD = 0.01; 95% CI = -0.34, 0.37; p = 0.94; I 2 = 29%), and cadence (SMD = 0.06; 95% CI = -0.25, 0.36; p = 0.70; I 2 = 0%). Conclusion TC has beneficial effects on motor function, balance function, functional walking ability, and gait velocity, but does not improve walking endurance, stride length, and cadence.
Collapse
Affiliation(s)
- Ping-an Zhu
- Department of Rehabilitation Medicine, Yuebei People’s Hospital, Shaoguan, China
| | - Qi-qi Lu
- Department of Rehabilitation Medicine, Yuebei People’s Hospital, Shaoguan, China
| | - Zhi-liang Li
- Department of Rehabilitation Medicine, Yuebei People’s Hospital, Shaoguan, China
| | - Rong-liang Hu
- Department of Rehabilitation Medicine, Jiangmen Central Hospital, Guangdong, Jiangmen, China
| | - Shu Xu
- Department of Rehabilitation Medicine, Shaoguan Railway Hospital, Shaoguan, China
| | - Lisa Brodersen
- Physical Therapy Program, Allen College, Waterloo, IA, United States
| | - Yuan-xin Liu
- College of Sports and Health Science, Xi’an Physical Education University, Xi’an, China,*Correspondence: Yuan-xin Liu,
| | - Howe Liu
- Physical Therapy Program, Allen College, Waterloo, IA, United States,Howe Liu,
| | - Xiao Bao
- Department of Rehabilitation Medicine, Yuebei People’s Hospital, Shaoguan, China,Xiao Bao,
| |
Collapse
|
21
|
Parkinson's disease-associated, sex-specific changes in DNA methylation at PARK7 (DJ-1), SLC17A6 (VGLUT2), PTPRN2 (IA-2β), and NR4A2 (NURR1) in cortical neurons. NPJ Parkinsons Dis 2022; 8:120. [PMID: 36151217 PMCID: PMC9508164 DOI: 10.1038/s41531-022-00355-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 06/24/2022] [Indexed: 12/04/2022] Open
Abstract
Evidence for epigenetic regulation playing a role in Parkinson's disease (PD) is growing, particularly for DNA methylation. Approximately 90% of PD cases are due to a complex interaction between age, genes, and environmental factors, and epigenetic marks are thought to mediate the relationship between aging, genetics, the environment, and disease risk. To date, there are a small number of published genome-wide studies of DNA methylation in PD, but none accounted for cell type or sex in their analyses. Given the heterogeneity of bulk brain tissue samples and known sex differences in PD risk, progression, and severity, these are critical variables to account for. In this genome-wide analysis of DNA methylation in an enriched neuronal population from PD postmortem parietal cortex, we report sex-specific PD-associated methylation changes in PARK7 (DJ-1), SLC17A6 (VGLUT2), PTPRN2 (IA-2β), NR4A2 (NURR1), and other genes involved in developmental pathways, neurotransmitter packaging and release, and axon and neuron projection guidance.
Collapse
|
22
|
Gezen-Ak D, Yurttaş Z, Çamoǧlu T, Dursun E. Could Amyloid-β 1-42 or α-Synuclein Interact Directly with Mitochondrial DNA? A Hypothesis. ACS Chem Neurosci 2022; 13:2803-2812. [PMID: 36125124 PMCID: PMC9542719 DOI: 10.1021/acschemneuro.2c00512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The amyloid β (Aβ) and the α-synuclein (α-syn) are shown to be translocated into mitochondria. Even though their roles are widely investigated in pathological conditions, information on the presence and functions of Aβ and α-syn in mitochondria in endogenous levels is somewhat limited. We hypothesized that endogenous Aβ fragments or α-syn could interact with mitochondrial DNA (mtDNA) directly or influence RNAs or transcription factors in mitochondria and change the mtDNA transcription profile. In this review, we summarized clues of these possible interactions.
Collapse
Affiliation(s)
| | | | | | - Erdinç Dursun
- E.D.: email, ; phone, +90 212 414 30 00/68025, +90 533 339
98 82
| |
Collapse
|
23
|
Pantaleo E, Monaco A, Amoroso N, Lombardi A, Bellantuono L, Urso D, Lo Giudice C, Picardi E, Tafuri B, Nigro S, Pesole G, Tangaro S, Logroscino G, Bellotti R. A Machine Learning Approach to Parkinson’s Disease Blood Transcriptomics. Genes (Basel) 2022; 13:genes13050727. [PMID: 35627112 PMCID: PMC9141063 DOI: 10.3390/genes13050727] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 12/23/2022] Open
Abstract
The increased incidence and the significant health burden associated with Parkinson’s disease (PD) have stimulated substantial research efforts towards the identification of effective treatments and diagnostic procedures. Despite technological advancements, a cure is still not available and PD is often diagnosed a long time after onset when irreversible damage has already occurred. Blood transcriptomics represents a potentially disruptive technology for the early diagnosis of PD. We used transcriptome data from the PPMI study, a large cohort study with early PD subjects and age matched controls (HC), to perform the classification of PD vs. HC in around 550 samples. Using a nested feature selection procedure based on Random Forests and XGBoost we reached an AUC of 72% and found 493 candidate genes. We further discussed the importance of the selected genes through a functional analysis based on GOs and KEGG pathways.
Collapse
Affiliation(s)
- Ester Pantaleo
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento di Scienze Mediche di Base, Neuroscienze e Organi di Senso, Università degli Studi di Bari Aldo Moro, Piazza G. Cesare 11, 70124 Bari, Italy;
- Dipartimento Interateneo di Fisica M. Merlin, Università degli Studi di Bari Aldo Moro, Via G. Amendola 173, 70125 Bari, Italy
| | - Alfonso Monaco
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
| | - Nicola Amoroso
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy
| | - Angela Lombardi
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento Interateneo di Fisica M. Merlin, Università degli Studi di Bari Aldo Moro, Via G. Amendola 173, 70125 Bari, Italy
- Correspondence:
| | - Loredana Bellantuono
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento di Scienze Mediche di Base, Neuroscienze e Organi di Senso, Università degli Studi di Bari Aldo Moro, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Daniele Urso
- Centro per le Malattie Neurodegenerative e l’Invecchiamento Cerebrale, Dipartimento di Ricerca Clinica in Neurologia, Università degli Studi di Bari Aldo Moro, Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (D.U.); (B.T.); (S.N.)
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Claudio Lo Giudice
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy; (C.L.G.); (E.P.); (G.P.)
| | - Ernesto Picardi
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy; (C.L.G.); (E.P.); (G.P.)
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via G. Amendola 122/O, 70126 Bari, Italy
| | - Benedetta Tafuri
- Centro per le Malattie Neurodegenerative e l’Invecchiamento Cerebrale, Dipartimento di Ricerca Clinica in Neurologia, Università degli Studi di Bari Aldo Moro, Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (D.U.); (B.T.); (S.N.)
| | - Salvatore Nigro
- Centro per le Malattie Neurodegenerative e l’Invecchiamento Cerebrale, Dipartimento di Ricerca Clinica in Neurologia, Università degli Studi di Bari Aldo Moro, Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (D.U.); (B.T.); (S.N.)
- Istituto di Nanotecnologia (NANOTEC), Consiglio Nazionale delle Ricerche, Via Monteroni, 73100 Lecce, Italy
| | - Graziano Pesole
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy; (C.L.G.); (E.P.); (G.P.)
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via G. Amendola 122/O, 70126 Bari, Italy
| | - Sabina Tangaro
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti, Università degli Studi di Bari Aldo Moro, Via A. Orabona 4, 70125 Bari, Italy
| | - Giancarlo Logroscino
- Dipartimento di Scienze Mediche di Base, Neuroscienze e Organi di Senso, Università degli Studi di Bari Aldo Moro, Piazza G. Cesare 11, 70124 Bari, Italy;
- Centro per le Malattie Neurodegenerative e l’Invecchiamento Cerebrale, Dipartimento di Ricerca Clinica in Neurologia, Università degli Studi di Bari Aldo Moro, Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (D.U.); (B.T.); (S.N.)
| | - Roberto Bellotti
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Via A. Orabona 4, 70125 Bari, Italy; (E.P.); (A.M.); (N.A.); (L.B.); (S.T.); (R.B.)
- Dipartimento Interateneo di Fisica M. Merlin, Università degli Studi di Bari Aldo Moro, Via G. Amendola 173, 70125 Bari, Italy
| |
Collapse
|
24
|
Segura-Aguilar J, Mannervik B, Inzunza J, Varshney M, Nalvarte I, Muñoz P. Astrocytes protect dopaminergic neurons against aminochrome neurotoxicity. Neural Regen Res 2022; 17:1861-1866. [PMID: 35142659 PMCID: PMC8848618 DOI: 10.4103/1673-5374.335690] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Astrocytes protect neurons by modulating neuronal function and survival. Astrocytes support neurons in several ways. They provide energy through the astrocyte-neuron lactate shuttle, protect neurons from excitotoxicity, and internalize neuronal lipid droplets to degrade fatty acids for neuronal metabolic and synaptic support, as well as by their high capacity for glutamate uptake and the conversion of glutamate to glutamine. A recent reported astrocyte system for protection of dopamine neurons against the neurotoxic products of dopamine, such as aminochrome and other o-quinones, were generated under neuromelanin synthesis by oxidizing dopamine catechol structure. Astrocytes secrete glutathione transferase M2-2 through exosomes that transport this enzyme into dopaminergic neurons to protect these neurons against aminochrome neurotoxicity. The role of this new astrocyte protective mechanism in Parkinson´s disease is discussed.
Collapse
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology ICBM Faculty of Medicine University of Chile, Santiago, Chile
| | - Bengt Mannervik
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, Stockholm, Sweden
| | - José Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Mukesh Varshney
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Patricia Muñoz
- Molecular and Clinical Pharmacology ICBM Faculty of Medicine University of Chile; Nucleo de Química y Bioquímica, Facultad de Estudios Interdisciplinarios, Universidad Mayor, Santiago, Chile
| |
Collapse
|
25
|
Li G, Huang P, Cui SS, Tan YY, He YC, Shen X, Jiang QY, Huang P, He GY, Li BY, Li YX, Xu J, Wang Z, Chen SD. Mechanisms of motor symptom improvement by long-term Tai Chi training in Parkinson’s disease patients. Transl Neurodegener 2022; 11:6. [PMID: 35125106 PMCID: PMC8819852 DOI: 10.1186/s40035-022-00280-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/05/2022] [Indexed: 11/10/2022] Open
Abstract
Background Tai Chi has been shown to improve motor symptoms in Parkinson’s disease (PD), but its long-term effects and the related mechanisms remain to be elucidated. In this study, we investigated the effects of long-term Tai Chi training on motor symptoms in PD and the underlying mechanisms. Methods Ninety-five early-stage PD patients were enrolled and randomly divided into Tai Chi (n = 32), brisk walking (n = 31) and no-exercise (n = 32) groups. At baseline, 6 months and 12 months during one-year intervention, all participants underwent motor symptom evaluation by Berg balance scale (BBS), Unified PD rating-scale (UPDRS), Timed Up and Go test (TUG) and 3D gait analysis, functional magnetic resonance imaging (fMRI), plasma cytokine and metabolomics analysis, and blood Huntingtin interaction protein 2 (HIP2) mRNA level analysis. Longitudinal self-changes were calculated using repeated measures ANOVA. GEE (generalized estimating equations) was used to assess factors associated with the longitudinal data of rating scales. Switch rates were used for fMRI analysis. False discovery rate correction was used for multiple correction. Results Participants in the Tai Chi group had better performance in BBS, UPDRS, TUG and step width. Besides, Tai Chi was advantageous over brisk walking in improving BBS and step width. The improved BBS was correlated with enhanced visual network function and downregulation of interleukin-1β. The improvements in UPDRS were associated with enhanced default mode network function, decreased L-malic acid and 3-phosphoglyceric acid, and increased adenosine and HIP2 mRNA levels. In addition, arginine biosynthesis, urea cycle, tricarboxylic acid cycle and beta oxidation of very-long-chain fatty acids were also improved by Tai Chi training. Conclusions Long-term Tai Chi training improves motor function, especially gait and balance, in PD. The underlying mechanisms may include enhanced brain network function, reduced inflammation, improved amino acid metabolism, energy metabolism and neurotransmitter metabolism, and decreased vulnerability to dopaminergic degeneration. Trial registration This study has been registered at Chinese Clinical Trial Registry (Registration number: ChiCTR2000036036; Registration date: August 22, 2020). Supplementary Information The online version contains supplementary material available at 10.1186/s40035-022-00280-7.
Collapse
|
26
|
Segura-Aguilar J, Muñoz P, Inzunza J, Varshney M, Nalvarte I, Mannervik B. Neuroprotection against Aminochrome Neurotoxicity: Glutathione Transferase M2-2 and DT-Diaphorase. Antioxidants (Basel) 2022; 11:296. [PMID: 35204179 PMCID: PMC8868244 DOI: 10.3390/antiox11020296] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Glutathione is an important antioxidant that plays a crucial role in the cellular protection against oxidative stress and detoxification of electrophilic mutagens, and carcinogens. Glutathione transferases are enzymes catalyzing glutathione-dependent reactions that lead to inactivation and conjugation of toxic compounds, processes followed by subsequent excretion of the detoxified products. Degeneration and loss of neuromelanin-containing dopaminergic neurons in the nigrostriatal neurons generally involves oxidative stress, neuroinflammation, alpha-synuclein aggregation to neurotoxic oligomers, mitochondrial dysfunction, protein degradation dysfunction, and endoplasmic reticulum stress. However, it is still unclear what triggers these neurodegenerative processes. It has been reported that aminochrome may elicit all of these mechanisms and, interestingly, aminochrome is formed inside neuromelanin-containing dopaminergic neurons during neuromelanin synthesis. Aminochrome is a neurotoxic ortho-quinone formed in neuromelanin synthesis. However, it seems paradoxical that the neurotoxin aminochrome is generated during neuromelanin synthesis, even though healthy seniors have these neurons intact when they die. The explanation of this paradox is the existence of protective tools against aminochrome neurotoxicity composed of the enzymes DT-diaphorase, expressed in these neurons, and glutathione transferase M2-2, expressed in astrocytes. Recently, it has been reported that dopaminergic neurons can be protected by glutathione transferase M2-2 from astrocytes, which secrete exosomes containing the protective enzyme.
Collapse
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology ICBM, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Patricia Muñoz
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8900000, Chile;
| | - Jose Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, SE-14157 Huddinge, Sweden; (J.I.); (M.V.); (I.N.)
| | - Mukesh Varshney
- Department of Biosciences and Nutrition, Karolinska Institutet, SE-14157 Huddinge, Sweden; (J.I.); (M.V.); (I.N.)
| | - Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institutet, SE-14157 Huddinge, Sweden; (J.I.); (M.V.); (I.N.)
| | - Bengt Mannervik
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, SE-10691 Stockholm, Sweden;
| |
Collapse
|
27
|
Ferraro F, Fevga C, Bonifati V, Mandemakers W, Mahfouz A, Reinders M. Correcting Differential Gene Expression Analysis for Cyto-Architectural Alterations in Substantia Nigra of Parkinson's Disease Patients Reveals Known and Potential Novel Disease-Associated Genes and Pathways. Cells 2022; 11:cells11020198. [PMID: 35053314 PMCID: PMC8774027 DOI: 10.3390/cells11020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 12/31/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
Several studies have analyzed gene expression profiles in the substantia nigra to better understand the pathological mechanisms causing Parkinson’s disease (PD). However, the concordance between the identified gene signatures in these individual studies was generally low. This might have been caused by a change in cell type composition as loss of dopaminergic neurons in the substantia nigra pars compacta is a hallmark of PD. Through an extensive meta-analysis of nine previously published microarray studies, we demonstrated that a big proportion of the detected differentially expressed genes was indeed caused by cyto-architectural alterations due to the heterogeneity in the neurodegenerative stage and/or technical artefacts. After correcting for cell composition, we identified a common signature that deregulated the previously unreported ammonium transport, as well as known biological processes such as bioenergetic pathways, response to proteotoxic stress, and immune response. By integrating with protein interaction data, we shortlisted a set of key genes, such as LRRK2, PINK1, PRKN, and FBXO7, known to be related to PD, others with compelling evidence for their role in neurodegeneration, such as GSK3β, WWOX, and VPC, and novel potential players in the PD pathogenesis. Together, these data show the importance of accounting for cyto-architecture in these analyses and highlight the contribution of multiple cell types and novel processes to PD pathology, providing potential new targets for drug development.
Collapse
Affiliation(s)
- Federico Ferraro
- Erasmus MC, Department of Clinical Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (F.F.); (C.F.); (V.B.); (W.M.)
| | - Christina Fevga
- Erasmus MC, Department of Clinical Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (F.F.); (C.F.); (V.B.); (W.M.)
| | - Vincenzo Bonifati
- Erasmus MC, Department of Clinical Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (F.F.); (C.F.); (V.B.); (W.M.)
| | - Wim Mandemakers
- Erasmus MC, Department of Clinical Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (F.F.); (C.F.); (V.B.); (W.M.)
| | - Ahmed Mahfouz
- Delft Bioinformatics Labaratory, Delft University of Technology, 2628 XE Delft, The Netherlands;
- Leiden Computational Biology Center, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marcel Reinders
- Delft Bioinformatics Labaratory, Delft University of Technology, 2628 XE Delft, The Netherlands;
- Leiden Computational Biology Center, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Section Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Correspondence:
| |
Collapse
|
28
|
Fernández-Santiago R, Esteve-Codina A, Fernández M, Valldeoriola F, Sanchez-Gómez A, Muñoz E, Compta Y, Tolosa E, Ezquerra M, Martí MJ. Transcriptome analysis in LRRK2 and idiopathic Parkinson's disease at different glucose levels. NPJ Parkinsons Dis 2021; 7:109. [PMID: 34853332 PMCID: PMC8636510 DOI: 10.1038/s41531-021-00255-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023] Open
Abstract
Type-2 diabetes (T2D) and glucose metabolic imbalances have been linked to neurodegenerative diseases, including Parkinson's disease (PD). To detect potential effects of different glucose levels on gene expression, by RNA-seq we analyzed the transcriptome of dermal fibroblasts from idiopathic PD (iPD) patients, LRRK2-associated PD (L2PD) patients, and healthy controls (total n = 21 cell lines), which were cultured at two different glucose concentrations (25 and 5 mM glucose). In PD patients we identified differentially expressed genes (DEGs) that were related to biological processes mainly involving the plasmatic cell membrane, the extracellular matrix, and also neuronal functions. Such pathway deregulation was largely similar in iPD or L2PD fibroblasts. Overall, the gene expression changes detected in this study were associated with PD independently of glucose concentration.
Collapse
Affiliation(s)
- Rubén Fernández-Santiago
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08036, Barcelona, Catalonia, Spain.
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028, Barcelona, Catalonia, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Manel Fernández
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
| | - Francesc Valldeoriola
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08036, Barcelona, Catalonia, Spain
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036, Barcelona, Catalonia, Spain
| | - Almudena Sanchez-Gómez
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08036, Barcelona, Catalonia, Spain
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036, Barcelona, Catalonia, Spain
| | - Esteban Muñoz
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08036, Barcelona, Catalonia, Spain
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036, Barcelona, Catalonia, Spain
| | - Yaroslau Compta
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08036, Barcelona, Catalonia, Spain
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036, Barcelona, Catalonia, Spain
| | - Eduardo Tolosa
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08036, Barcelona, Catalonia, Spain
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036, Barcelona, Catalonia, Spain
| | - Mario Ezquerra
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08036, Barcelona, Catalonia, Spain.
| | - María J Martí
- Lab of Parkinson disease and Other Neurodegenerative Movement Disorders: Clinical and Experimental Research, Department of Neurology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08036, Barcelona, Catalonia, Spain
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036, Barcelona, Catalonia, Spain
| |
Collapse
|
29
|
del Rey NLG, Balzano T, Martin-Rodriguez L, Salinas-Rebolledo C, Trigo-Damas I, Rojas-Fernandez A, Alvarez-Erviti L, Blesa J. Lack of Parkinsonian Pathology and Neurodegeneration in Mice After Long-Term Injections of a Proteasome Inhibitor in Olfactory Bulb and Amygdala. Front Aging Neurosci 2021; 13:698979. [PMID: 34744683 PMCID: PMC8570189 DOI: 10.3389/fnagi.2021.698979] [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: 04/22/2021] [Accepted: 09/27/2021] [Indexed: 11/24/2022] Open
Abstract
Proteinaceous inclusions, called Lewy bodies (LBs), are used as a pathological hallmark for Parkinson's disease (PD). Recent studies suggested a prion-like spreading mechanism for α-synucleinopathy where early neuropathological deposits occur, among others, in the olfactory bulb (OB) and amygdala. LBs contain insoluble α-synuclein and many other ubiquitinated proteins, suggesting a role of protein degradation system failure in PD pathogenesis. Therefore, we wanted to study the effects of a proteasomal inhibitor, lactacystin, on the aggregability and transmissibility of α-synuclein in the OB and amygdala. We performed injections of lactacystin in the OB and amygdala of wild-type mice. Motor behavior, markers of neuroinflammation, α-synuclein, and dopaminergic integrity were assessed by immunohistochemistry. Overall, there were no differences in the number of neurons and α-synuclein expression in these regions following injection of lactacystin into either the OB or amygdala. Microglial and astroglial labeling appeared to be correlated with surgery-induced inflammation or local effects of lactacystin. Consistent with the behavior and pathological findings, there was no loss of dopaminergic cell bodies in the substantia nigra and terminals in the striatum. Our data showed that long-term lactacystin injections in extra nigrostriatal regions may not mimic spreading aspects of PD and reinforce the special vulnerability of dopaminergic neurons of the substantia nigra pars compacta (SNc).
Collapse
Affiliation(s)
- Natalia Lopez-Gonzalez del Rey
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Tiziano Balzano
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Lucia Martin-Rodriguez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | | | - Ines Trigo-Damas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Lydia Alvarez-Erviti
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
30
|
Munson MJ, Mathai BJ, Ng MYW, Trachsel-Moncho L, de la Ballina LR, Schultz SW, Aman Y, Lystad AH, Singh S, Singh S, Wesche J, Fang EF, Simonsen A. GAK and PRKCD are positive regulators of PRKN-independent mitophagy. Nat Commun 2021; 12:6101. [PMID: 34671015 DOI: 10.1101/2020.11.05.369496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 09/29/2021] [Indexed: 05/28/2023] Open
Abstract
The mechanisms involved in programmed or damage-induced removal of mitochondria by mitophagy remains elusive. Here, we have screened for regulators of PRKN-independent mitophagy using an siRNA library targeting 197 proteins containing lipid interacting domains. We identify Cyclin G-associated kinase (GAK) and Protein Kinase C Delta (PRKCD) as regulators of PRKN-independent mitophagy, with both being dispensable for PRKN-dependent mitophagy and starvation-induced autophagy. We demonstrate that the kinase activity of both GAK and PRKCD are required for efficient mitophagy in vitro, that PRKCD is present on mitochondria, and that PRKCD facilitates recruitment of ULK1/ATG13 to early autophagic structures. Importantly, we demonstrate in vivo relevance for both kinases in the regulation of basal mitophagy. Knockdown of GAK homologue (gakh-1) in C. elegans or knockout of PRKCD homologues in zebrafish led to significant inhibition of basal mitophagy, highlighting the evolutionary relevance of these kinases in mitophagy regulation.
Collapse
Affiliation(s)
- Michael J Munson
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway.
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway.
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Benan J Mathai
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Matthew Yoke Wui Ng
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Laura Trachsel-Moncho
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Laura R de la Ballina
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Sebastian W Schultz
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
- Department of Molecular Cell Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway
| | - Yahyah Aman
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478, Lørenskog, Norway
| | - Alf H Lystad
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Sakshi Singh
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Sachin Singh
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
- Department of Tumor Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway
| | - Jørgen Wesche
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
- Department of Tumor Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478, Lørenskog, Norway
| | - Anne Simonsen
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway.
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway.
- Department of Molecular Cell Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway.
| |
Collapse
|
31
|
Munson MJ, Mathai BJ, Ng MYW, Trachsel-Moncho L, de la Ballina LR, Schultz SW, Aman Y, Lystad AH, Singh S, Singh S, Wesche J, Fang EF, Simonsen A. GAK and PRKCD are positive regulators of PRKN-independent mitophagy. Nat Commun 2021; 12:6101. [PMID: 34671015 PMCID: PMC8528926 DOI: 10.1038/s41467-021-26331-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 09/29/2021] [Indexed: 12/26/2022] Open
Abstract
The mechanisms involved in programmed or damage-induced removal of mitochondria by mitophagy remains elusive. Here, we have screened for regulators of PRKN-independent mitophagy using an siRNA library targeting 197 proteins containing lipid interacting domains. We identify Cyclin G-associated kinase (GAK) and Protein Kinase C Delta (PRKCD) as regulators of PRKN-independent mitophagy, with both being dispensable for PRKN-dependent mitophagy and starvation-induced autophagy. We demonstrate that the kinase activity of both GAK and PRKCD are required for efficient mitophagy in vitro, that PRKCD is present on mitochondria, and that PRKCD facilitates recruitment of ULK1/ATG13 to early autophagic structures. Importantly, we demonstrate in vivo relevance for both kinases in the regulation of basal mitophagy. Knockdown of GAK homologue (gakh-1) in C. elegans or knockout of PRKCD homologues in zebrafish led to significant inhibition of basal mitophagy, highlighting the evolutionary relevance of these kinases in mitophagy regulation.
Collapse
Affiliation(s)
- Michael J Munson
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway.
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway.
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Benan J Mathai
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Matthew Yoke Wui Ng
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Laura Trachsel-Moncho
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Laura R de la Ballina
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Sebastian W Schultz
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
- Department of Molecular Cell Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway
| | - Yahyah Aman
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478, Lørenskog, Norway
| | - Alf H Lystad
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Sakshi Singh
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
| | - Sachin Singh
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
- Department of Tumor Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway
| | - Jørgen Wesche
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway
- Department of Tumor Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478, Lørenskog, Norway
| | - Anne Simonsen
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0372, Oslo, Norway.
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway.
- Department of Molecular Cell Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway.
| |
Collapse
|
32
|
Zaccaria A, Antinori P, Licker V, Kövari E, Lobrinus JA, Burkhard PR. Multiomic Analyses of Dopaminergic Neurons Isolated from Human Substantia Nigra in Parkinson's Disease: A Descriptive and Exploratory Study. Cell Mol Neurobiol 2021; 42:2805-2818. [PMID: 34528139 PMCID: PMC9561004 DOI: 10.1007/s10571-021-01146-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 09/02/2021] [Indexed: 11/21/2022]
Abstract
Dopaminergic neurons (DA) of the substantia nigra pars compacta (SNpc) selectively and progressively degenerate in Parkinson’s disease (PD). Until now, molecular analyses of DA in PD have been limited to genomic or transcriptomic approaches, whereas, to the best of our knowledge, no proteomic or combined multiomic study examining the protein profile of these neurons is currently available. In this exploratory study, we used laser capture microdissection to extract regions from DA in 10 human SNpc obtained at autopsy in PD patients and control subjects. Extracted RNA and proteins were identified by RNA sequencing and nanoliquid chromatography–mass spectrometry, respectively, and the differential expression between PD and control group was assessed. Qualitative analyses confirmed that the microdissection protocol preserves the integrity of our samples and offers access to specific molecular pathways. This multiomic analysis highlighted differential expression of 52 genes and 33 proteins, including molecules of interest already known to be dysregulated in PD, such as LRP2, PNMT, CXCR4, MAOA and CBLN1 genes, or the Aldehyde dehydrogenase 1 protein. On the other hand, despite the same samples were used for both analyses, correlation between RNA and protein expression was low, as exemplified by the CST3 gene encoding for the cystatin C protein. This is the first exploratory study analyzing both gene and protein expression of laser-dissected neuronal parts from SNpc in PD. Data are available via ProteomeXchange with identifier PXD024748 and via GEO with identifier GSE 169755.
Collapse
Affiliation(s)
- Affif Zaccaria
- Neuroproteomics Group, University Medical Center, Faculty of Medicine, Geneva University, Geneva, Switzerland.
| | - Paola Antinori
- Neuroproteomics Group, University Medical Center, Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Virginie Licker
- Neuroproteomics Group, University Medical Center, Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Enikö Kövari
- Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | | | - Pierre R Burkhard
- Neuroproteomics Group, University Medical Center, Faculty of Medicine, Geneva University, Geneva, Switzerland.,Department of Neurology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
33
|
Russo M, Sobh A, Zhang P, Loguinov A, Tagmount A, Vulpe CD, Liu B. Functional Pathway Identification With CRISPR/Cas9 Genome-wide Gene Disruption in Human Dopaminergic Neuronal Cells Following Chronic Treatment With Dieldrin. Toxicol Sci 2021; 176:366-381. [PMID: 32421776 DOI: 10.1093/toxsci/kfaa071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Organochlorine pesticides, once widely used, are extremely persistent and bio-accumulative in the environment. Epidemiological studies have implicated that environmental exposure to organochlorine pesticides including dieldrin is a risk factor for the development of Parkinson's disease. However, the pertinent mechanisms of action remain poorly understood. In this study, we carried out a genome-wide (Brunello library, 19 114 genes, 76 411 sgRNAs) CRISPR/Cas9 screen in human dopaminergic SH-SY5Y neuronal cells exposed to a chronic treatment (30 days) with dieldrin to identify cellular pathways that are functionally related to the chronic cellular toxicity. Our results indicate that dieldrin toxicity was enhanced by gene disruption of specific components of the ubiquitin proteasome system as well as, surprisingly, the protein degradation pathways previously implicated in inherited forms of Parkinson's disease, centered on Parkin. In addition, disruption of regulatory components of the mTOR pathway which integrates cellular responses to both intra- and extracellular signals and is a central regulator for cell metabolism, growth, proliferation, and survival, led to increased sensitivity to dieldrin-induced cellular toxicity. This study is one of the first to apply a genome-wide CRISPR/Cas9-based functional gene disruption screening approach in an adherent neuronal cell line to globally decipher cellular mechanisms that contribute to environmental toxicant-induced neurotoxicity and provides novel insight into the dopaminergic neurotoxicity associated with chronic exposure to dieldrin.
Collapse
Affiliation(s)
- Max Russo
- Department of Pharmacodynamics, College of Pharmacy
| | - Amin Sobh
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | - Ping Zhang
- Department of Pharmacodynamics, College of Pharmacy
| | - Alex Loguinov
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | - Abderrahmane Tagmount
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | - Chris D Vulpe
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | - Bin Liu
- Department of Pharmacodynamics, College of Pharmacy
| |
Collapse
|
34
|
Neurorescue Effects of Frondoside A and Ginsenoside Rg3 in C. elegans Model of Parkinson's Disease. Molecules 2021; 26:molecules26164843. [PMID: 34443430 PMCID: PMC8402114 DOI: 10.3390/molecules26164843] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/24/2021] [Accepted: 08/08/2021] [Indexed: 11/21/2022] Open
Abstract
Parkinson’s disease (PD) is a currently incurable neurodegenerative disorder characterized by the loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta and α-synuclein aggregation. Accumulated evidence indicates that the saponins, especially from ginseng, have neuroprotective effects against neurodegenerative disorders. Interestingly, saponin can also be found in marine organisms such as the sea cucumber, but little is known about its effect in neurodegenerative disease, including PD. In this study, we investigated the anti-Parkinson effects of frondoside A (FA) from Cucumaria frondosa and ginsenoside Rg3 (Rg3) from Panax notoginseng in C. elegans PD model. Both saponins were tested for toxicity and optimal concentration by food clearance assay and used to treat 6-OHDA-induced BZ555 and transgenic α-synuclein NL5901 strains in C. elegans. Treatment with FA and Rg3 significantly attenuated DAergic neurodegeneration induced by 6-OHDA in BZ555 strain, improved basal slowing rate, and prolonged lifespan in the 6-OHDA-induced wild-type strain with downregulation of the apoptosis mediators, egl-1 and ced-3, and upregulation of sod-3 and cat-2. Interestingly, only FA reduced α-synuclein aggregation, rescued lifespan in NL5901, and upregulated the protein degradation regulators, including ubh-4, hsf-1, hsp-16.1 and hsp-16.2. This study indicates that both FA and Rg3 possess beneficial effects in rescuing DAergic neurodegeneration in the 6-OHDA-induced C. elegans model through suppressing apoptosis mediators and stimulating antioxidant enzymes. In addition, FA could attenuate α-synuclein aggregation through the protein degradation process.
Collapse
|
35
|
Craig DW, Hutchins E, Violich I, Alsop E, Gibbs JR, Levy S, Robison M, Prasad N, Foroud T, Crawford KL, Toga AW, Whitsett TG, Kim S, Casey B, Reimer A, Hutten SJ, Frasier M, Kern F, Fehlman T, Keller A, Cookson MR, Van Keuren-Jensen K. RNA sequencing of whole blood reveals early alterations in immune cells and gene expression in Parkinson's disease. NATURE AGING 2021; 1:734-747. [PMID: 37117765 DOI: 10.1038/s43587-021-00088-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/21/2021] [Indexed: 04/30/2023]
Abstract
Changes in the blood-based RNA transcriptome have the potential to inform biomarkers of Parkinson's disease (PD) progression. Here we sequenced a discovery set of whole-blood RNA species in 4,871 longitudinally collected samples from 1,570 clinically phenotyped individuals from the Parkinson's Progression Marker Initiative (PPMI) cohort. Samples were sequenced to an average of 100 million read pairs to create a high-quality transcriptome. Participants with PD in the PPMI had significantly altered RNA expression (>2,000 differentially expressed genes), including an early and persistent increase in neutrophil gene expression, with a concomitant decrease in lymphocyte cell counts. This was validated in a cohort from the Parkinson's Disease Biomarkers Program (PDBP) consisting of 1,599 participants and by alterations in immune cell subtypes. This publicly available transcriptomic dataset, coupled with available detailed clinical data, provides new insights into PD biological processes impacting whole blood and new paths for developing diagnostic and prognostic PD biomarkers.
Collapse
Affiliation(s)
- David W Craig
- Institute of Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth Hutchins
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Ivo Violich
- Institute of Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - Eric Alsop
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - J Raphael Gibbs
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Shawn Levy
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Madison Robison
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Nripesh Prasad
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | - Karen L Crawford
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Timothy G Whitsett
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Seungchan Kim
- Center for Computational Systems Biology, Department of Electrical and Computer Engineering, Roy G. Perry College of Engineering, Prairie View A&M University, Prairie View, TX, USA
| | - Bradford Casey
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Alyssa Reimer
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Samantha J Hutten
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Mark Frasier
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Fabian Kern
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Tobias Fehlman
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
36
|
Lestón Pinilla L, Ugun-Klusek A, Rutella S, De Girolamo LA. Hypoxia Signaling in Parkinson's Disease: There Is Use in Asking "What HIF?". BIOLOGY 2021; 10:723. [PMID: 34439955 PMCID: PMC8389254 DOI: 10.3390/biology10080723] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022]
Abstract
Hypoxia is a condition characterized by insufficient tissue oxygenation, which results in impaired oxidative energy production. A reduction in cellular oxygen levels induces the stabilization of hypoxia inducible factor α (HIF-1α), master regulator of the molecular response to hypoxia, involved in maintaining cellular homeostasis and driving hypoxic adaptation through the control of gene expression. Due to its high energy requirement, the brain is particularly vulnerable to oxygen shortage. Thus, hypoxic injury can cause significant metabolic changes in neural cell populations, which are associated with neurodegeneration. Recent evidence suggests that regulating HIF-1α may ameliorate the cellular damage in neurodegenerative diseases. Indeed, the hypoxia/HIF-1α signaling pathway has been associated to several processes linked to Parkinson's disease (PD) including gene mutations, risk factors and molecular pathways such as mitochondrial dysfunction, oxidative stress and protein degradation impairment. This review will explore the impact of hypoxia and HIF-1α signaling on these specific molecular pathways that influence PD development and will evaluate different novel neuroprotective strategies involving HIF-1α stabilization.
Collapse
Affiliation(s)
- Laura Lestón Pinilla
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Aslihan Ugun-Klusek
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Luigi A. De Girolamo
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| |
Collapse
|
37
|
Yan Q, Forno E, Cardenas A, Qi C, Han YY, Acosta-Pérez E, Kim S, Zhang R, Boutaoui N, Canino G, Vonk JM, Xu CJ, Chen W, Marsland A, Oken E, Gold DR, Koppelman GH, Celedón JC. Exposure to violence, chronic stress, nasal DNA methylation, and atopic asthma in children. Pediatr Pulmonol 2021; 56:1896-1905. [PMID: 33751861 PMCID: PMC8217314 DOI: 10.1002/ppul.25372] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/01/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Exposure to violence (ETV) or chronic stress may influence asthma through unclear mechanisms. METHODS Epigenome-wide association study (EWAS) of ETV or chronic stress measures and DNA methylation in nasal epithelium from 487 Puerto Ricans aged 9-20 years who participated in the Epigenetic Variation and Childhood Asthma in Puerto Ricans study [EVA-PR]). We assessed four measures of ETV and chronic stress in children (ETV scale, gun violence, and perceived stress) and their mothers (perceived stress). Each EWAS was conducted using linear regression, with CpGs as dependent variables and the stress/violence measure as a predictor, adjusting for age, sex, the top five principal components, and SVA latent factors. We then selected the top 100 CpGs (by p value) associated with each stress/violence measure in EVA-PR and conducted a meta-analysis of the selected CpGs and atopic asthma using data from EVA-PR and two additional cohorts (Project Viva and PIAMA). RESULTS Three CpGs (in SNN, PTPRN2, and LINC01164) were associated with maternal perceived stress or gun violence (p = 1.28-3.36 × 10-7 ), but not with atopic asthma, in EVA-PR. In a meta-analysis of three cohorts, which included the top CpGs associated with stress/violence measures in EVA-PR, 12 CpGs (in STARD3NL, SLC35F4, TSR3, CDC42SE2, KLHL25, PLCB1, BUD13, OR2B3, GALR1, TMEM196, TEAD4, and ANAPC13) were associated with atopic asthma at FDR-p < .05. CONCLUSIONS Pending confirmation in longitudinal studies, our findings suggest that nasal epithelial methylation markers associated with measures of ETV and chronic stress may be linked to atopic asthma in children and adolescents.
Collapse
Affiliation(s)
- Qi Yan
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andres Cardenas
- Division of Environmental Health Sciences, University of California, Berkeley, California, USA.,Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Cancan Qi
- Department of Pediatric Pulmonology and Pediatric Allergy, University Medical Center Groningen, Beatrix Children's Hospital, University of Groningen, Groningen, The Netherlands.,University Medical Center Groningen, GRIAC Research Institute, University of Groningen, Groningen, The Netherlands
| | - Yueh-Ying Han
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Edna Acosta-Pérez
- Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Soyeon Kim
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rong Zhang
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Statistics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nadia Boutaoui
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Glorisa Canino
- Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Judith M Vonk
- University Medical Center Groningen, GRIAC Research Institute, University of Groningen, Groningen, The Netherlands.,Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cheng-Jian Xu
- Department of Pediatric Pulmonology and Pediatric Allergy, University Medical Center Groningen, Beatrix Children's Hospital, University of Groningen, Groningen, The Netherlands.,University Medical Center Groningen, GRIAC Research Institute, University of Groningen, Groningen, The Netherlands
| | - Wei Chen
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anna Marsland
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Diane R Gold
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergy, University Medical Center Groningen, Beatrix Children's Hospital, University of Groningen, Groningen, The Netherlands.,University Medical Center Groningen, GRIAC Research Institute, University of Groningen, Groningen, The Netherlands
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
38
|
1,4-dihydropyridine derivatives increase mRNA expression of Psma3, Psmb5, and Psmc6 in rats. ACTA ACUST UNITED AC 2021; 72:148-156. [PMID: 34187104 PMCID: PMC8265202 DOI: 10.2478/aiht-2021-72-3422] [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: 04/01/2020] [Accepted: 06/01/2021] [Indexed: 11/21/2022]
Abstract
The ubiquitin-proteasome system modifies different cellular and protein functions. Its dysregulation may lead to disrupted proteostasis associated with multiple pathologies and aging. Pharmacological regulation of proteasome functions is already an important part of the treatment of several diseases. 1,4-dihydropyridine (1,4-DHP) derivatives possess different pharmacological activities, including antiaging and neuroprotective. The aim of this study was to investigate the effects of several 1,4-DHP derivatives on mRNA expression levels of proteasomal genes Psma3, Psmb5, and Psmc6 in several organs of rats. Rats were treated with metcarbatone, etcarbatone, glutapyrone, styrylcarbatone, AV-153-Na, or AV-153-Ca per os for three days. mRNA expression levels were determined with real-time polymerase chain reaction (PCR). For AV-153-Na and AV-153-Ca, we also determined the expression of the Psma6 gene. In the kidney, metcarbatone, etcarbatone, styrylcarbatone, and AV-153-Na increased the expression of all analysed genes. Glutapyrone increased the expression of Psmb5 and Psmc6 but did not affect the expression of Psma3. In the blood, glutapyrone increased Psmb5 expression. In the liver, AV-153-Na increased the expression of Psma6 and Psmc6 but lowered the expression of Psmb5, while AV-153-Ca only increased Psma6 expression. The ability of 1,4-DHP derivatives to increase the expression of proteasome subunit genes might hold a therapeutic potential in conditions associated with impaired proteasomal functions, but further research is needed.
Collapse
|
39
|
Gezen-Ak D, Alaylıoğlu M, Genç G, Şengül B, Keskin E, Sordu P, Güleç ZEK, Apaydın H, Bayram-Gürel Ç, Ulutin T, Yılmazer S, Ertan S, Dursun E. Altered Transcriptional Profile of Mitochondrial DNA-Encoded OXPHOS Subunits, Mitochondria Quality Control Genes, and Intracellular ATP Levels in Blood Samples of Patients with Parkinson's Disease. J Alzheimers Dis 2021; 74:287-307. [PMID: 32007957 DOI: 10.3233/jad-191164] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mitochondrial dysfunctions are significant contributors to neurodegeneration. One result or a cause of mitochondrial dysfunction might be the disruption of mtDNA transcription. Limited data indicated an altered expression of mtDNA encoded transcripts in Alzheimer's disease (AD) or Parkinson's disease (PD). The number of mitochondria is high in cells with a high energy demand, such as muscle or nerve cells. AD or PD involves increased risk of cardiomyopathy, suggesting that mitochondrial dysfunction might be systemic. If it is systemic, we should observe it in different cell types. Given that, we wanted to investigate any disruption in the regulation of mtDNA encoded gene expression in addition to PINK1, PARKIN, and ATP levels in peripheral blood samples of PD cases who are affected by a neurodegenerative disorder that is very well known by its mitochondrial aspects. Our results showed for the first time that: 1) age of onset > 50 PD sporadic (PDS) cases: mtDNA transcription and quality control genes were affected; 2) age of onset <50 PDS cases: only mtDNA transcription was affected; and 3) PD cases with familial background: only quality control genes were affected. mtDNA copy number was not a confounder. Intracellular ATP levels of PD case subgroups were significantly higher than those of healthy subjects. We suggest that a systemic dysregulation of transcription of mtDNA or mitochondrial quality control genes might result in the development of a sporadic form of the disease. Additionally, ATP elevation might be an independent compensatory and response mechanism. Hyperactive cells in AD and PD require further investigation.
Collapse
Affiliation(s)
- Duygu Gezen-Ak
- Department of Medical Biology, Brain and Neurodegenerative Disorders Research Laboratories, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Merve Alaylıoğlu
- Department of Medical Biology, Brain and Neurodegenerative Disorders Research Laboratories, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gençer Genç
- Department of Neurology, Şişli Etfal Training and Research Hospital, Istanbul, Turkey
| | - Büşra Şengül
- Department of Medical Biology, Brain and Neurodegenerative Disorders Research Laboratories, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ebru Keskin
- Department of Medical Biology, Brain and Neurodegenerative Disorders Research Laboratories, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Pelin Sordu
- Department of Medical Biology, Brain and Neurodegenerative Disorders Research Laboratories, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeynep Ece Kaya Güleç
- Department of Neurology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Hülya Apaydın
- Department of Neurology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Çiğdem Bayram-Gürel
- Department of Medical Biology, Brain and Neurodegenerative Disorders Research Laboratories, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Turgut Ulutin
- Department of Medical Biology, Brain and Neurodegenerative Disorders Research Laboratories, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Selma Yılmazer
- Department of Medical Biology, Faculty of Medicine, Altınbaş University, Istanbul, Turkey
| | - Sibel Ertan
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul, Turkey
| | - Erdinç Dursun
- Department of Medical Biology, Brain and Neurodegenerative Disorders Research Laboratories, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.,Department of Neuroscience, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
40
|
Jiang J, Hu X, Liu L, He Z, Wu Q, Li Q, Hu X, Jiang N, Liu C. Age-related changes in expression of lysine48 and lysine63 ubiquitin linkages in dopamine neurons of midbrain in mice. Neuroreport 2021; 32:569-576. [PMID: 33657079 DOI: 10.1097/wnr.0000000000001626] [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: 02/05/2023]
Abstract
Ubiquitination of target proteins is mediated via different ubiquitin lysine (K) linkages and determines the protein fates. In particular, K48 ubiquitin linkage targets proteins for degradation, whereas K63 ubiquitin linkage plays a nondegradative role. Parkinson's disease is an age-onset neurodegenerative disorder, which shows selective loss of dopamine neurons in substantia nigra pars compacta (SNC) and ubiquitinated protein aggregates. However, age-related expression of K48 and K63 ubiquitin linkages in SNC dopamine neurons remains elusive. We thus sought to explore the expression of K48 and K63 ubiquitin linkages in dopamine neurons in SNCs of mice at different ages with morphological and biochemical assays. Here our results indicated that in 5-week-old mice, dopamine neurons presented higher levels of K48 and K63 ubiquitin linkages than nondopamine neural cells. Aging promoted the formation of protein aggregates that are positive for both K48 and K63 ubiquitin linkages, together with tyrosine hydroxylase, a dopamine neuron marker. Moreover, 21-month-old mice showed fewer neural cells and tyrosine hydroxylase positive neurons in the SNCs than younger mice. Through biochemical analysis, the 21-month-old mice were shown to express more K48 ubiquitin linkages and less tyrosine hydroxylase and NeuN than the 5-week-old mice. These results suggest the first time that expression of K48 and K63 ubiquitin lysine linkages in midbrain dopamine neurons is age-related and may be involved in the loss of dopamine neurons.
Collapse
Affiliation(s)
- Jing Jiang
- School of Basic Medical Sciences
- Department of Histology and Embryology
- Institute of Stem Cell and Tissue Engineering
| | - Xiujuan Hu
- School of Basic Medical Sciences
- Department of Histology and Embryology
- Institute of Stem Cell and Tissue Engineering
| | | | - Zhicheng He
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Qingjie Wu
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Qunchao Li
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Xianwei Hu
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Nan Jiang
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Chao Liu
- School of Basic Medical Sciences
- Department of Histology and Embryology
- Institute of Stem Cell and Tissue Engineering
| |
Collapse
|
41
|
Miller KM, Mercado NM, Sortwell CE. Synucleinopathy-associated pathogenesis in Parkinson's disease and the potential for brain-derived neurotrophic factor. NPJ PARKINSONS DISEASE 2021; 7:35. [PMID: 33846345 PMCID: PMC8041900 DOI: 10.1038/s41531-021-00179-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The lack of disease-modifying treatments for Parkinson’s disease (PD) is in part due to an incomplete understanding of the disease’s etiology. Alpha-synuclein (α-syn) has become a point of focus in PD due to its connection to both familial and idiopathic cases—specifically its localization to Lewy bodies (LBs), a pathological hallmark of PD. Within this review, we will present a comprehensive overview of the data linking synuclein-associated Lewy pathology with intracellular dysfunction. We first present the alterations in neuronal proteins and transcriptome associated with LBs in postmortem human PD tissue. We next compare these findings to those associated with LB-like inclusions initiated by in vitro exposure to α-syn preformed fibrils (PFFs) and highlight the profound and relatively unique reduction of brain-derived neurotrophic factor (BDNF) in this model. Finally, we discuss the multitude of ways in which BDNF offers the potential to exert disease-modifying effects on the basal ganglia. What remains unknown is the potential for BDNF to mitigate inclusion-associated dysfunction within the context of synucleinopathy. Collectively, this review reiterates the merit of using the PFF model as a tool to understand the physiological changes associated with LBs, while highlighting the neuroprotective potential of harnessing endogenous BDNF.
Collapse
Affiliation(s)
- Kathryn M Miller
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA. .,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
42
|
Overview of the Neuroprotective Effects of the MAO-Inhibiting Antidepressant Phenelzine. Cell Mol Neurobiol 2021; 42:225-242. [PMID: 33839994 PMCID: PMC8732914 DOI: 10.1007/s10571-021-01078-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
Abstract
Phenelzine (PLZ) is a monoamine oxidase (MAO)-inhibiting antidepressant with anxiolytic properties. This multifaceted drug has a number of pharmacological and neurochemical effects in addition to inhibition of MAO, and findings on these effects have contributed to a body of evidence indicating that PLZ also has neuroprotective/neurorescue properties. These attributes are reviewed in this paper and include catabolism to the active metabolite β-phenylethylidenehydrazine (PEH) and effects of PLZ and PEH on the GABA-glutamate balance in brain, sequestration of reactive aldehydes, and inhibition of primary amine oxidase. Also discussed are the encouraging findings of the effects of PLZ in animal models of stroke, spinal cord injury, traumatic brain injury, and multiple sclerosis, as well other actions such as reduction of nitrative stress, reduction of the effects of a toxin on dopaminergic neurons, potential anticonvulsant actions, and effects on brain-derived neurotrophic factor, neural cell adhesion molecules, an anti-apoptotic factor, and brain levels of ornithine and N-acetylamino acids.
Collapse
|
43
|
Fan HH, Guo Q, Zheng J, Lian YZ, Huang SS, Sun Y, Zou M, Zhu JH, Zhang X. ALDH1A1 Genetic Variations May Modulate Risk of Parkinson's Disease in Han Chinese Population. Front Neurosci 2021; 15:620929. [PMID: 33815038 PMCID: PMC8017280 DOI: 10.3389/fnins.2021.620929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/10/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Studies in animal models have suggested that aldehyde dehydrogenase 1 (encoded by ALDH1A1) protects against Parkinson’s disease (PD) by reducing toxic metabolites of dopamine. Herein we aimed to investigate whether ALDH1A1 was genetically associated with PD susceptibility in humans. Methods: A Han Chinese population of 1,039 subjects was recruited to analyze six tag-single nucleotide polymorphisms (SNPs), followed by haplotype analyses and variants interaction analyses. Real-time PCR was used to analyze mRNA levels of ALDH1A1 in peripheral blood of 42 subjects. Results: The tag-SNP rs7043217 of ALDH1A1 was significantly associated with PD susceptibility with the T serving as a risk allele (genotype frequency, P = 0.030; allele frequency, P = 0.013, OR = 1.258, 95% CI = 1.050–1.508). Multiple haplotypes were linked to abnormalities of PD risk, topped by a 4-SNP GGTA module in the order of rs4646547, rs1888202, rs7043217, and rs647880 (P = 9.610 × 10–8, OR = 6.420, 95% CI = 2.944–13.998). Interaction analyses showed that a simultaneous presence of the CC genotype of rs7043217 and the TT genotype of ALDH2 variant rs4767944 conferred an elevated protection against PD (P = 4.68 × 10–4, OR = 0.378, 95% CI = 0.219–0.652). The mRNA expression of ALDH1A1 showed a trend of reduction (P = 0.084) in PD patients compared to the controls. Conclusion: Our results provide novel genetic insights into the role of ALDH1 in PD pathogenesis.
Collapse
Affiliation(s)
- Hui-Hui Fan
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, China.,Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qing Guo
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jing Zheng
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yi-Zhi Lian
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shi-Shi Huang
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yue Sun
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ming Zou
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jian-Hong Zhu
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, China.,Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiong Zhang
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, China.,Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
44
|
Karaaslan Z, Kahraman ÖT, Şanlı E, Ergen HA, Ulusoy C, Bilgiç B, Yılmaz V, Tüzün E, Hanağası HA, Küçükali Cİ. Inflammation and regulatory T cell genes are differentially expressed in peripheral blood mononuclear cells of Parkinson's disease patients. Sci Rep 2021; 11:2316. [PMID: 33504893 PMCID: PMC7841172 DOI: 10.1038/s41598-021-81961-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Our aim was to identify the differentially expressed genes (DEGs) in peripheral blood mononuclear cells (PBMC) of Parkinson’s disease (PD) patients and healthy controls by microarray technology and analysis of related molecular pathways by functional annotation. Thirty PD patients and 30 controls were enrolled. Agilent Human 8X60 K Oligo Microarray was used for gene level expression identification. Gene ontology and pathway enrichment analyses were used for functional annotation of DEGs. Protein–protein interaction analyses were performed with STRING. Expression levels of randomly selected DEGs were quantified by real time quantitative polymerase chain reaction (RT-PCR) for validation. Flow cytometry was done to determine frequency of regulatory T cells (Tregs) in PBMC. A total of 361 DEGs (143 upregulated and 218 downregulated) were identified after GeneSpring analysis. DEGs were involved in 28 biological processes, 12 cellular components and 26 molecular functions. Pathway analyses demonstrated that upregulated genes mainly enriched in p53 (CASP3, TSC2, ATR, MDM4, CCNG1) and PI3K/Akt (IL2RA, IL4R, TSC2, VEGFA, PKN2, PIK3CA, ITGA4, BCL2L11) signaling pathways. TP53 and PIK3CA were identified as most significant hub proteins. Expression profiles obtained by RT-PCR were consistent with microarray findings. PD patients showed increased proportions of CD49d+ Tregs, which correlated with disability scores. Survival pathway genes were upregulated putatively to compensate neuronal degeneration. Bioinformatics analysis showed an association between survival and inflammation genes. Increased CD49d+ Treg ratios might signify the effort of the immune system to suppress ongoing neuroinflammation.
Collapse
Affiliation(s)
- Zerrin Karaaslan
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Özlem Timirci Kahraman
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Elif Şanlı
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Hayriye Arzu Ergen
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Canan Ulusoy
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Başar Bilgiç
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Vuslat Yılmaz
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Haşmet Ayhan Hanağası
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Cem İsmail Küçükali
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
45
|
Phung DM, Lee J, Hong S, Kim YE, Yoon J, Kim YJ. Meta-Analysis of Differentially Expressed Genes in the Substantia Nigra in Parkinson's Disease Supports Phenotype-Specific Transcriptome Changes. Front Neurosci 2020; 14:596105. [PMID: 33390883 PMCID: PMC7775392 DOI: 10.3389/fnins.2020.596105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/16/2020] [Indexed: 01/26/2023] Open
Abstract
Background Studies regarding differentially expressed genes (DEGs) in Parkinson’s disease (PD) have focused on common upstream regulators or dysregulated pathways or ontologies; however, the relationships between DEGs and disease-related or cell type-enriched genes have not been systematically studied. Meta-analysis of DEGs (meta-DEGs) are expected to overcome the limitations, such as replication failure and small sample size of previous studies. Purpose Meta-DEGs were performed to investigate dysregulated genes enriched with neurodegenerative disorder causative or risk genes in a phenotype-specific manner. Methods Six microarray datasets from PD patients and controls, for which substantia nigra sample transcriptome data were available, were downloaded from the NINDS data repository. Meta-DEGs were performed using two methods, combining p-values and combing effect size, and common DEGs were used for secondary analyses. Gene sets of cell type-enriched or disease-related genes for PD, Alzheimer’s disease (AD), and hereditary progressive ataxia were constructed by curation of public databases and/or published literatures. Results Our meta-analyses revealed 449 downregulated and 137 upregulated genes. Overrepresentation analyses with cell type-enriched genes were significant in neuron-enriched genes but not in astrocyte- or microglia-enriched genes. Meta-DEGs were significantly enriched in causative genes for hereditary disorders accompanying parkinsonism but not in genes associated with AD or hereditary progressive ataxia. Enrichment of PD-related genes was highly significant in downregulated DEGs but insignificant in upregulated genes. Conclusion Downregulated meta-DEGs were associated with PD-related genes, but not with other neurodegenerative disorder genes. These results highlight disease phenotype-specific changes in dysregulated genes in PD.
Collapse
Affiliation(s)
- Duong My Phung
- Department of Biomedical Gerontology, Ilsong Institute of Life and Science, Hallym University, Anyang, South Korea
| | - Jinwoo Lee
- Department of Computer Engineering, Hallym University, Chuncheon, South Korea
| | - SangKyoon Hong
- Hallym Institute of Translational Genomics and Bioinformatics, Anyang, South Korea
| | - Young Eun Kim
- Laboratory of Parkinson's Disease and Neurogenetics, Department of Neurology, Hallym University, Anyang, South Korea
| | - Jeehee Yoon
- Department of Computer Engineering, Hallym University, Chuncheon, South Korea
| | - Yun Joong Kim
- Department of Neurology, Yonsei University College of Medicine, Yongin, South Korea.,Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| |
Collapse
|
46
|
Monaco A, Pantaleo E, Amoroso N, Bellantuono L, Lombardi A, Tateo A, Tangaro S, Bellotti R. Identifying potential gene biomarkers for Parkinson's disease through an information entropy based approach. Phys Biol 2020; 18:016003. [PMID: 33049726 DOI: 10.1088/1478-3975/abc09a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD) is a chronic, progressive neurodegenerative disease and represents the most common disease of this type, after Alzheimer's dementia. It is characterized by motor and nonmotor features and by a long prodromal stage that lasts many years. Genetic research has shown that PD is a complex and multisystem disorder. To capture the molecular complexity of this disease we used a complex network approach. We maximized the information entropy of the gene co-expression matrix betweenness to obtain a gene adjacency matrix; then we used a fast greedy algorithm to detect communities. Finally we applied principal component analysis on the detected gene communities, with the ultimate purpose of discriminating between PD patients and healthy controls by means of a random forests classifier. We used a publicly available substantia nigra microarray dataset, GSE20163, from NCBI GEO database, containing gene expression profiles for 10 PD patients and 18 normal controls. With this methodology we identified two gene communities that discriminated between the two groups with mean accuracy of 0.88 ± 0.03 and 0.84 ± 0.03, respectively, and validated our results on an independent microarray experiment. The two gene communities presented a considerable reduction in size, over 100 times, compared to the initial network and were stable within a range of tested parameters. Further research focusing on the restricted number of genes belonging to the selected communities may reveal essential mechanisms responsible for PD at a network level and could contribute to the discovery of new biomarkers for PD.
Collapse
Affiliation(s)
- A Monaco
- Istituto Nazionale di Fisica Nucleare (INFN), Sezione di Bari, Bari, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Yan Q, Forno E, Cardenas A, Qi C, Han YY, Acosta-Pérez E, Kim S, Zhang R, Boutaoui N, Canino G, Vonk JM, Xu CJ, Chen W, Oken E, Gold DR, Koppelman GH, Celedón JC. Exposure to violence, chronic stress, nasal DNA methylation, and atopic asthma in children. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 33173928 DOI: 10.1101/2020.11.03.20225250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background Exposure to violence (ETV) or stress may cause asthma through unclear mechanisms. Methods Epigenome-wide association study (EWAS) of DNA methylation in nasal epithelium and four ETV or chronic stress measures in 487 Puerto Ricans aged 9-20 years who participated in the Epigenetic Variation and Childhood Asthma in Puerto Ricans study [EVA-PR]). We assessed measures of ETV or chronic stress in children (ETV scale, gun violence, and perceived stress) and their mothers (perceived stress). Each EWAS was conducted using linear regression, with CpGs as dependent variables and the stress/violence measure as a predictor, adjusting for age, sex, the top five principal components, and SVA latent factors. We then selected the top 100 CpGs (by P-value) associated with each stress/violence measure in EVA-PR and conducted a meta-analysis of the selected CpGs and atopic asthma using data from EVA-PR and two additional cohorts (Project Viva and PIAMA). Results In the EWAS of stress/violence in EVA-PR, gun violence was associated with methylation of cg18961589 in LINC01164 (β=0.03, P =1.28×10 -7 ), and maternal stress was associated with methylation of cg03402351 in SNN (β=0.04, P =1.69×10 -7 ) and cg19064846 in PTPRN2 (β=0.03, P =3.36×10 -7 ). In a meta-analysis of three cohorts, which included the top CpGs associated with stress/violence in EVA-PR, CpGs in STARD3NL, SLC35F4, TSR3, CDC42SE2, KLHL25, PLCB1, BUD13, OR2B3, GALR1, TMEM196, TEAD4 and ANAPC13 were associated with atopic asthma at FDR- P < 0.05. Conclusions ETV and chronic stress may increase the risk of atopic asthma through DNA methylation in airway epithelium, though this needs confirmation in future longitudinal studies.
Collapse
|
48
|
Proteasome Subunits Involved in Neurodegenerative Diseases. Arch Med Res 2020; 52:1-14. [PMID: 32962866 DOI: 10.1016/j.arcmed.2020.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/25/2020] [Accepted: 09/04/2020] [Indexed: 12/29/2022]
Abstract
The ubiquitin-proteasome system is the major pathway for the maintenance of protein homeostasis. Its inhibition causes accumulation of ubiquitinated proteins; this accumulation has been associated with several of the most common neurodegenerative diseases. Several genetic factors have been identified for most neurodegenerative diseases, however, most cases are considered idiopathic, thus making the study of the mechanisms of protein accumulation a relevant field of research. It is often mentioned that the biggest risk factor for neurodegenerative diseases is aging, and several groups have reported an age-related alteration of the expression of some of the 26S proteasome subunits and a reduction of its activity. Proteasome subunits interact with proteins that are known to accumulate in neurodegenerative diseases such as α-synuclein in Parkinson's, tau in Alzheimer's, and huntingtin in Huntington's diseases. These interactions have been explored for several years, but only until recently, we are beginning to understand them. In this review, we discuss the known interactions, the underlying patterns, and the phenotypes associated with the 26S proteasome subunits in the etiology and progression of neurodegenerative diseases where there is evidence of proteasome involvement. Special emphasis is made in reviewing proteasome subunits that interact with proteins known to have an age-related altered expression or to be involved in neurodegenerative diseases to explore key effectors that may trigger or augment their progression. Interestingly, while the causes of age-related reduction of some of the proteasome subunits are not known, there are specific relationships between the observed neurodegenerative disease and the affected proteasome subunits.
Collapse
|
49
|
Kumari B, Mandal M, Dholaniya PS. Analysis of multiple transcriptome data to determine age-associated genes for the progression of Parkinson's disease. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
50
|
Dabool L, Hakim-Mishnaevski K, Juravlev L, Flint-Brodsly N, Mandel S, Kurant E. Drosophila Skp1 Homologue SkpA Plays a Neuroprotective Role in Adult Brain. iScience 2020; 23:101375. [PMID: 32739834 PMCID: PMC7399183 DOI: 10.1016/j.isci.2020.101375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/14/2020] [Accepted: 07/14/2020] [Indexed: 11/17/2022] Open
Abstract
Skp1, a component of the ubiquitin E3 ligases, was found to be decreased in the brains of sporadic Parkinson's disease (PD) patients, and its overexpression prevented death of murine neurons in culture. Here we expose the neuroprotective role of the Drosophila skp1 homolog, skpA, in the adult brain. Neuronal knockdown of skpA leads to accumulation of ubiquitinated protein aggregates and loss of dopaminergic neurons accompanied by motor dysfunction and reduced lifespan. Conversely, neuronal overexpression of skpA reduces aggregate load, improves age-related motor decline, and prolongs lifespan. Moreover, SkpA rescues neurodegeneration in a Drosophila model of PD. We also show that a Drosophila homolog of FBXO7, the F Box protein, Nutcracker (Ntc), works in the same pathway with SkpA. However, skpA overexpression rescues ntc knockdown phenotype, suggesting that SkpA interacts with additional F box proteins in the adult brain neurons. Collectively, our study discloses Skp1/SkpA as a potential therapeutic target in neurodegenerative diseases.
SkpA-mediated protein degradation is required for normal function of the adult brain SkpA overexpression rescues neurodegeneration in α-synuclein-induced fly PD model SkpA and Ntc work in the same pathway of protein degradation in adult brain neurons
Collapse
Affiliation(s)
- Lital Dabool
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel; The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Ketty Hakim-Mishnaevski
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel
| | - Liza Juravlev
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel
| | - Naama Flint-Brodsly
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel
| | - Silvia Mandel
- The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Estee Kurant
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel; The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|