1
|
Jin M, Ye K, Hu D, Chen J, Wu S, Chi S. Identification of diagnose related therapeutic targets of Danggui buxue decoction in Parkinson's disease. Brain Res 2024; 1842:149097. [PMID: 38950810 DOI: 10.1016/j.brainres.2024.149097] [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: 03/13/2024] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is the fastest growing neurological disease. Currently, there is no disease-modifying therapy to slow the progression of the disease. Danggui buxue decoction (DBD) is widely used in the clinic because of its therapeutic effect. However, little is known about the molecular mechanism of DBD against PD. This study intends to explore the possible molecular mechanisms involved in DBD treatment of PD based on network pharmacology, and provide potential research directions for future research. METHODS Firstly, the active components and target genes of DBD were screened from the traditional Chinese medicine systems pharmacology (TCMSP), DrugBank and UniProt database. Secondly, target genes of PD were identified from the (GEO) dataset, followed by identification of common target genes of DBD and PD. Thirdly, analysis of protein-protein interaction (PPI), functional enrichment and diagnosis was performed on common target genes, followed by correlation analysis between core target genes, immune cell, miRNAs, and transcription factors (TFs). Finally, molecular docking between core target genes and active components, and real-time PCR were performed. RESULTS A total of 72 common target genes were identified between target genes of DBD and target genes of PD. Among which, 11 target genes with potential diagnostic value were further identified, including TP53, AKT1, IL1B, MMP9, NOS3, RELA, MAPK14, HMOX1, TGFB1, NOS2, and ERBB2. The combinations with the best docking binding were identified, including kaempferol-AKT1/HMOX1/NOS2/NOS3, quercetin-AKT1/ERBB2/IL1B/HMOX1/MMP9/TP53/NOS3/TGFB1. Moreover, IL1B and NOS2 respectively positively and negatively correlated with neutrophil and Type 1 T helper cell. Some miRNA-core target gene regulatory pairs were identified, such as hsa-miR-185-5p-TP53/TGFB1/RELA/MAPK14/IL1B/ERBB2/AKT1 and hsa-miR-214-3p-NOS3. These core target genes were significantly enriched in focal adhesion, TNF, HIF-1, and ErbB signaling pathway. CONCLUSION Diagnostic TP53, AKT1, IL1B, MMP9, NOS3, RELA, MAPK14, HMOX1, TGFB1, NOS2, and ERBB2 may be considered as potential therapeutic targets of DBD in the treatment of PD.
Collapse
Affiliation(s)
- Man Jin
- Department of Neurology, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310063, China
| | - Kaisheng Ye
- Department of Traditional Chinese Medicine, Hangzhou Kanghui Integrated Traditional and Western Medicine Clinic, Hangzhou, Zhejiang Province 310019, China.
| | - Defeng Hu
- Department of Psychiatry, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310063, China
| | - Jiefang Chen
- Department of Neurology, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310063, China
| | - Sha Wu
- Intensive Care Units, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310063, China
| | - Shumei Chi
- Department of Psychiatry, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310063, China
| |
Collapse
|
2
|
Martinez-Banaclocha MA. Targeting the Cysteine Redox Proteome in Parkinson's Disease: The Role of Glutathione Precursors and Beyond. Antioxidants (Basel) 2023; 12:1373. [PMID: 37507913 PMCID: PMC10376658 DOI: 10.3390/antiox12071373] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Encouraging recent data on the molecular pathways underlying aging have identified variants and expansions of genes associated with DNA replication and repair, telomere and stem cell maintenance, regulation of the redox microenvironment, and intercellular communication. In addition, cell rejuvenation requires silencing some transcription factors and the activation of pluripotency, indicating that hidden molecular networks must integrate and synchronize all these cellular mechanisms. Therefore, in addition to gene sequence expansions and variations associated with senescence, the optimization of transcriptional regulation and protein crosstalk is essential. The protein cysteinome is crucial in cellular regulation and plays unexpected roles in the aging of complex organisms, which show cumulative somatic mutations, telomere attrition, epigenetic modifications, and oxidative dysregulation, culminating in cellular senescence. The cysteine thiol groups are highly redox-active, allowing high functional versatility as structural disulfides, redox-active disulfides, active-site nucleophiles, proton donors, and metal ligands to participate in multiple regulatory sites in proteins. Also, antioxidant systems control diverse cellular functions, including the transcription machinery, which partially depends on the catalytically active cysteines that can reduce disulfide bonds in numerous target proteins, driving their biological integration. Since we have previously proposed a fundamental role of cysteine-mediated redox deregulation in neurodegeneration, we suggest that cellular rejuvenation of the cysteine redox proteome using GSH precursors, like N-acetyl-cysteine, is an underestimated multitarget therapeutic approach that would be particularly beneficial in Parkinson's disease.
Collapse
|
3
|
Checler F, Alves da Costa C. Parkin as a Molecular Bridge Linking Alzheimer’s and Parkinson’s Diseases? Biomolecules 2022; 12:biom12040559. [PMID: 35454148 PMCID: PMC9026546 DOI: 10.3390/biom12040559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s (AD) and Parkinson’s (PD) diseases are two distinct age-related pathologies that are characterized by various common dysfunctions. They are referred to as proteinopathies characterized by ubiquitinated protein accumulation and aggregation. This accumulation is mainly due to altered lysosomal and proteasomal clearing processes and is generally accompanied by ER stress disturbance, autophagic and mitophagic defects, mitochondrial structure and function alterations and enhanced neuronal cell death. Genetic approaches aimed at identifying molecular triggers responsible for familial forms of AD or PD have helped to understand the etiology of their sporadic counterparts. It appears that several proteins thought to contribute to one of these pathologies are also likely to contribute to the other. One such protein is parkin (PK). Here, we will briefly describe anatomical lesions and genetic advances linked to AD and PD as well as the main cellular processes commonly affected in these pathologies. Further, we will focus on current studies suggesting that PK could well participate in AD and thereby act as a molecular bridge between these two pathologies. In particular, we will focus on the transcription factor function of PK and its newly described transcriptional targets that are directly related to AD- and PD-linked cellular defects.
Collapse
|
4
|
Wu Z, Xia C, Zhang C, Tang D, Liu F, Ou Y, Gao J, Yi H, Yang D, Ma K. Adeno-associated virus-delivered alpha synuclein inhibits bladder cancer growth via the p53/p21 signaling pathway. Cancer Gene Ther 2022; 29:1193-1206. [PMID: 35064206 DOI: 10.1038/s41417-022-00425-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 11/09/2022]
|
5
|
Makav M, Eroğlu HA. Recuperative effect of estrogen on rotenone-induced experimental model of Parkinson's disease in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:21266-21275. [PMID: 33410082 DOI: 10.1007/s11356-020-11985-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 12/06/2020] [Indexed: 06/12/2023]
Abstract
Parkinson's disease (PD) is described as the loss of dopaminergic neurons located in the substantia nigra (SN) region of the brain and a progressive motor failure. Increased frequency of PD in women, especially after menopause, suggests the effect of estrogen. This view has been supported with empirical studies. Therefore, the effect of estrogen in an experimental model of Parkinson's disease induced by rotenone was investigated. A total of 32 female Wistar Albino rats were randomly assigned to four groups (control group, ovariectomy group, Parkinson's group, Parkinson's + estrogen group). The Parkinson's group received rotenone subcutanously at the dose of 2.5 mg/kg bw, on the 1st, 2nd, 3rd 4th, 6th, 9th, 12th, 15th, 18th, and 21st days animals in the Parkinson's + estrogen group received retonon as in the Parkinson's group and was additionally subcutaneously given estrogen (implant containing 0.5 mg 17 β-estradiol lasting for 21 days). The rats were subjected to rotarod, pole, and swimming tests at the end of the experiment for comparison of their motor activities, and then, histopathological and biochemical analyses were performed on the tissues that were extracted. The rotarod results revealed that Parkinson's group had the shortest time (32.33 ± 3.98 sn) than the groups of control (92.50 ± 12.60 s) ovariectomy (71.42 ± 10.58 s), and Parkinson's + estrogen (71.37 ± 9.26 s). The results of pole disclosed that return and landing time prolonged for Parkinson's group when compared with other groups (return time for control 2.98 ± 0.38 s, ovariectomy 3.02 ± 0.75 s, Parkinson 5.91 ± 0.33 s, Parkinson's + estrogen 3.48 ± 0.42 s and landing time for control 5.30 ± 0.59 s, ovariectomy 5.45 ± 0.73 s, Parkinson 9.80 ± 0.90 s, Parkinson's + estrogen 5.37 ± 1.02 s). Parkinson's group had longest (90.71 ± 12.56 s) swimming time to reach the target when compared with control (33.16 ± 8.68 s), ovariectomy (47.37 ± 12.19 s), and Parkinson's + estrogen (49.82 ± 5.78 s). Histopathological examination indicated a significant difference in tyrosine hydroxylase-stained cells (dopaminergic neurons and dopamine) between the Parkinson's + estrogen group and the Parkinson's group. The biochemical analyses of Caspas-3 activation in SN and striatum (STR) was significantly different between the Parkinson's + estrogen group and the Parkinson's group, but this difference was not observed in STR while evaluating Bcl-2. The results of this study suggested that estrogen may have a recuperative effect on PD.
Collapse
Affiliation(s)
- Mustafa Makav
- Department of Physiology, Faculty of Veterinary Medicine, Kafkas University, Paşaçayırı Campus, TR-36100, Kars, Turkey.
| | - Hüseyin Avni Eroğlu
- Department of Physiology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
6
|
Lanni C, Masi M, Racchi M, Govoni S. Cancer and Alzheimer's disease inverse relationship: an age-associated diverging derailment of shared pathways. Mol Psychiatry 2021; 26:280-295. [PMID: 32382138 DOI: 10.1038/s41380-020-0760-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 04/06/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
Several epidemiological studies show an inverse association between cancer and Alzheimer's disease (AD). It is debated whether this association is the consequence of biological mechanisms shared by both these conditions or may be related to the pharmacological treatments carried out on the patients. The latter hypothesis, however, is not sustained by the available evidence. Hence, the focus of this review is to analyze common biological mechanisms for both cancer and AD and to build up a biological theory useful to explain the inverse correlation between AD and cancer. The review proposes a hypothesis, according to which several molecular players, prominently PIN1 and p53, have been investigated and considered involved in complex molecular interactions putatively associated with the inverse correlation. On the other hand, p53 involvement in both diseases seems to be a consequence of the aberrant activation of other proteins. Instead, PIN1 may be identified as a novel key regulator at the crossroad between cancer and AD. PIN1 is a peptidyl-prolyl cis-trans isomerase that catalyzes the cis-trans isomerization, thus regulating the conformation of different protein substrates after phosphorylation and modulating protein function. In particular, trans-conformations of Amyloid Precursor Protein (APP) and tau are functional and "healthy", while cis-conformations, triggered after phosphorylation, are pathogenic. As an example, PIN1 accelerates APP cis-to-trans isomerization thus favoring the non-amyloidogenic pathway, while, in the absence of PIN1, APP is processed through the amyloidogenic pathway, thus predisposing to neurodegeneration. Furthermore, a link between PIN1 and tau regulation has been found, since when PIN1 function is inhibited, tau is hyperphosphorylated. Data from brain specimens of subjects affected by mild cognitive impairment and AD have revealed a very low PIN1 expression. Moreover, polymorphisms in PIN1 promoter correlated with an increased PIN1 expression are associated with a delay of sporadic AD age of onset, while a polymorphism related to a reduced PIN1 expression is associated with a decreased risk of multiple cancers. In the case of dementias, in particular of Alzheimer's disease, new biological markers and targets based on the discussed players can be developed based on a theoretical approach relying on different grounds compared to the past. An unbiased expansion of the rationale and of the targets may help to achieve in the field of neurodegenerative dementias similar advances to those attained in the case of cancer treatment.
Collapse
Affiliation(s)
- Cristina Lanni
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12/14, 27100, Pavia, Italy
| | - Mirco Masi
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12/14, 27100, Pavia, Italy.,Scuola Universitaria Superiore IUSS Pavia, Piazza della Vittoria 15, 27100, Pavia, Italy
| | - Marco Racchi
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12/14, 27100, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12/14, 27100, Pavia, Italy.
| |
Collapse
|
7
|
The P53/microRNA network: A potential tumor suppressor with a role in anticancer therapy. Pharmacol Res 2020; 160:105179. [PMID: 32890739 DOI: 10.1016/j.phrs.2020.105179] [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: 06/09/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are endogenous and small non-coding RNAs that have been identified as mediators of tumor suppression as well as stress responses mediated by p53 suppressors. MiRNAs may act as tumor suppressors under certain conditions. MiRNAs regulated by p53 may control the expression of processes such as cell cycle progression, cell survival, and angiogenesis. P53 activity and expression are also controlled by miRNA; consequently alterations in the p53-miRNA network may be essential for tumor initiation and progression. Future studies on the p53-miRNA network presumably would find it helpful in diagnostic and therapeutic approaches or as tools for various cancers.
Collapse
|
8
|
Genome-wide association study identifies zonisamide responsive gene in Parkinson's disease patients. J Hum Genet 2020; 65:693-704. [PMID: 32355309 PMCID: PMC8075945 DOI: 10.1038/s10038-020-0760-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022]
Abstract
Long-term treatment of Parkinson's disease (PD) by levodopa leads to motor complication "wearing-off". Zonisamide is a nondopaminergic antiparkinsonian drug that can improve "wearing-off" although response to the treatment varies between individuals. To clarify the genetic basis of zonisamide responsiveness, we conducted a genome-wide association study (GWAS) on 200 PD patients from a placebo-controlled clinical trial, including 67 responders whose "off" time decreased ≥1.5 h after 12 weeks of zonisamide treatment and 133 poor responders. We genotyped and evaluated the association between 611,492 single nucleotide polymorphisms (SNPs) and "off" time reduction. We also performed whole-genome imputation, gene- and pathway-based analyses of GWAS data. For promising SNPs, we examined single-tissue expression quantitative trait loci (eQTL) data in the GTEx database. SNP rs16854023 (Mouse double minute 4, MDM4) showed genome-wide significant association with reduced "off" time (PAdjusted = 4.85 × 10-9). Carriers of responsive genotype showed >7-fold decrease in mean "off" time compared to noncarriers (1.42 h vs 0.19 h; P = 2.71 × 10-7). In silico eQTL data indicated that zonisamide sensitivity is associated with higher MDM4 expression. Among the 37 pathways significantly influencing "off" time, calcium and glutamate signaling have also been associated with anti-epileptic effect of zonisamide. MDM4 encodes a negative regulator of p53. The association between improved motor fluctuation and MDM4 upregulation implies that p53 inhibition may prevent dopaminergic neuron loss and consequent motor symptoms. This is the first genome-wide pharmacogenetics study on antiparkinsonian drug. The findings provide a basis for improved management of "wearing-off" in PD by genotype-guided zonisamide treatment.
Collapse
|
9
|
Benoit SM, Xu H, Schmid S, Alexandrova R, Kaur G, Thiruvahindrapuram B, Pereira SL, Jog M, Hebb MO. Expanding the search for genetic biomarkers of Parkinson's disease into the living brain. Neurobiol Dis 2020; 140:104872. [PMID: 32302674 DOI: 10.1016/j.nbd.2020.104872] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022] Open
Abstract
Altered gene expression related to Parkinson's Disease (PD) has not been described in the living brain, yet this information may support novel discovery pertinent to disease pathophysiology and treatment. This study compared the transcriptome in brain biopsies obtained from living PD and Control patients. To evaluate the novelty of this data, a comprehensive literature review also compared differentially expressed gene (DEGs) identified in the current study with those reported in PD cadaveric brain and peripheral tissues. RNA was extracted from rapidly cryopreserved frontal lobe specimens collected from PD and Control patients undergoing neurosurgical procedures. RNA sequencing (RNA-Seq) was performed and validated using quantitative polymerase chain reaction. DEG data was assessed using bioinformatics and subsequently included within a comparative analysis of PD RNA-Seq studies. 370 DEGs identified in living brain specimens reflected diverse gene groups and included key members of trophic signaling, apoptosis, inflammation and cell metabolism pathways. The comprehensive literature review yielded 7 RNA-Seq datasets generated from blood, skin and cadaveric brain but none from a living brain source. From the current dataset, 123 DEGs were identified only within the living brain and 267 DEGs were either newly found or had distinct directional change in living brain relative to other tissues. This is the first known study to analyze the transcriptome in brain tissue from living PD and Control patients. The data produced using these methods offer a unique, unexplored resource with potential to advance insight into the genetic associations of PD.
Collapse
Affiliation(s)
- Simon M Benoit
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, 339 Windermere Road, Suite C7-134, London N6A 5A5, Ontario, Canada
| | - Hu Xu
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, 339 Windermere Road, Suite C7-134, London N6A 5A5, Ontario, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, Western University, 1151 Richmond Street, Medical Sciences Building, Room 443, London N6A 3K7, Ontario, Canada
| | - Roumiana Alexandrova
- The Centre for Applied Genomics, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 656 Bay Street, Room 139800, Toronto M5G 0A4, Ontario, Canada
| | - Gaganjot Kaur
- The Centre for Applied Genomics, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 656 Bay Street, Room 139800, Toronto M5G 0A4, Ontario, Canada
| | - Bhooma Thiruvahindrapuram
- The Centre for Applied Genomics, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 656 Bay Street, Room 139800, Toronto M5G 0A4, Ontario, Canada
| | - Sergio L Pereira
- The Centre for Applied Genomics, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 656 Bay Street, Room 139800, Toronto M5G 0A4, Ontario, Canada
| | - Mandar Jog
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, 339 Windermere Road, Suite C7-134, London N6A 5A5, Ontario, Canada
| | - Matthew O Hebb
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, 339 Windermere Road, Suite C7-134, London N6A 5A5, Ontario, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, Western University, 1151 Richmond Street, Medical Sciences Building, Room 443, London N6A 3K7, Ontario, Canada.
| |
Collapse
|
10
|
Karim MR, Liao EE, Kim J, Meints J, Martinez HM, Pletnikova O, Troncoso JC, Lee MK. α-Synucleinopathy associated c-Abl activation causes p53-dependent autophagy impairment. Mol Neurodegener 2020; 15:27. [PMID: 32299471 PMCID: PMC7164361 DOI: 10.1186/s13024-020-00364-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 02/13/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Studies link c-Abl activation with the accumulation of pathogenic α-synuclein (αS) and neurodegeneration in Parkinson's disease (PD). Currently, c-Abl, a tyrosine kinase activated by cellular stress, is thought to promote αS pathology by either directly phosphorylating αS or by causing autophagy deficits. METHODS αS overexpressing transgenic (Tg) mice were used in this study. A53T Tg mice that express high levels of human mutant A53TαS under the control of prion protein promoter. Two different approaches were used in this study. Natural aging and seeding model of synucleinopathy. In seeding model, intracortical/intrastriatal (IC/IS) stereotaxic injection of toxic lysates was done using tissue lysates from end-stage symptomatic mice. In this study, nilotinib and pifithrin-α was used as a c-Abl and p53 inhibitor, respectively. Both Tg and non-transgenic (nTg) mice from each group were subjected to nilotinib (10 mg/kg) or vehicle (DMSO) treatment. Frozen brain tissues from PD and control human cases were analyzed. In vitro cells study was implied for c-Abl/p53 genetic manipulation to uncover signal transduction. RESULTS Herein, we show that the pathologic effects of c-Abl in PD also involve activation of p53, as c-Abl activation in a transgenic mouse model of α-synucleinopathy (TgA53T) and human PD cases are associated with the increased p53 activation. Significantly, active p53 in TgA53T neurons accumulates in the cytosol, which may lead to inhibition of autophagy. Thus, we hypothesized that c-Abl-dependent p53 activation contributes to autophagy impairment in α-synucleinopathy. In support of the hypothesis, we show that c-Abl activation is sufficient to inhibit autophagy in p53-dependent manner. Moreover, inhibition of either c-Abl, using nilotinib, or p53, using pifithrin-α, was sufficient to increase autophagic flux in neuronal cells by inducing phosphorylation of AMP-activated kinase (AMPK), ULK1 activation, and down-regulation of mTORC1 signaling. Finally, we show that pharmacological attenuation of c-Abl activity by nilotinib treatment in the TgA53T mouse model reduces activation of p53, stimulates autophagy, decreases accumulation αS pathology, and delays disease onset. CONCLUSION Collectively, our data show that c-Abl activation by α-synucleinopathy causes p53 dependent autophagy deficits and both c-Abl and p53 represent therapeutic target for PD.
Collapse
Affiliation(s)
- Md. Razaul Karim
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
| | - Elly E. Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
| | - Jaekwang Kim
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
- Present Address: Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, 41068 South Korea
| | - Joyce Meints
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
| | | | - Olga Pletnikova
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Juan C. Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN 55414 USA
| |
Collapse
|
11
|
Ye Q, Wen Y, Al-Kuwari N, Chen X. Association Between Parkinson's Disease and Melanoma: Putting the Pieces Together. Front Aging Neurosci 2020; 12:60. [PMID: 32210791 PMCID: PMC7076116 DOI: 10.3389/fnagi.2020.00060] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Patients with Parkinson’s disease (PD) generally have reduced risk of developing many types of cancers, except melanoma—a malignant tumor of melanin-producing cells in the skin. For decades, a large number of epidemiological studies have reported that the occurrence of melanoma is higher than expected among subjects with PD, and the occurrence of PD is reciprocally higher than expected among patients with melanoma. More recent epidemiological studies further indicated a bidirectional association, not only in the patients themselves but also in their relatives. This association between PD and melanoma offers a unique opportunity to understand PD. Here, we summarize epidemiological, clinical, and biological evidence in regard to shared risk factors and possible underlying mechanisms for these two seemingly distinct conditions.
Collapse
Affiliation(s)
- Qing Ye
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Ya Wen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Ietheory Institute, Burlington, MA, United States
| | - Nasser Al-Kuwari
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiqun Chen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Rahman RU, Liebhoff AM, Bansal V, Fiosins M, Rajput A, Sattar A, Magruder DS, Madan S, Sun T, Gautam A, Heins S, Liwinski T, Bethune J, Trenkwalder C, Fluck J, Mollenhauer B, Bonn S. SEAweb: the small RNA Expression Atlas web application. Nucleic Acids Res 2020; 48:D204-D219. [PMID: 31598718 PMCID: PMC6943056 DOI: 10.1093/nar/gkz869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/14/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
We present the Small RNA Expression Atlas (SEAweb), a web application that allows for the interactive querying, visualization and analysis of known and novel small RNAs across 10 organisms. It contains sRNA and pathogen expression information for over 4200 published samples with standardized search terms and ontologies. In addition, SEAweb allows for the interactive visualization and re-analysis of 879 differential expression and 514 classification comparisons. SEAweb's user model enables sRNA researchers to compare and re-analyze user-specific and published datasets, highlighting common and distinct sRNA expression patterns. We provide evidence for SEAweb's fidelity by (i) generating a set of 591 tissue specific miRNAs across 29 tissues, (ii) finding known and novel bacterial and viral infections across diseases and (iii) determining a Parkinson's disease-specific blood biomarker signature using novel data. We believe that SEAweb's simple semantic search interface, the flexible interactive reports and the user model with rich analysis capabilities will enable researchers to better understand the potential function and diagnostic value of sRNAs or pathogens across tissues, diseases and organisms.
Collapse
Affiliation(s)
- Raza-Ur Rahman
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anna-Maria Liebhoff
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Vikas Bansal
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- German Center for Neurodegenerative Diseases, 72076 Tübingen, Germany
| | - Maksims Fiosins
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- German Center for Neurodegenerative Diseases, 72076 Tübingen, Germany
- Genevention GmbH, 37079 Göttingen, Germany
| | - Ashish Rajput
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Abdul Sattar
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Daniel S Magruder
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Genevention GmbH, 37079 Göttingen, Germany
| | - Sumit Madan
- Fraunhofer Institute for Algorithms and Scientific Computing, Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- Rheinische Friedrich-Wilhelms-Universität Bonn, 53113 Bonn, Germany
| | - Ting Sun
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Abhivyakti Gautam
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sven Heins
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Timur Liwinski
- Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Jörn Bethune
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, 34128 Kassel, Germany
- Department of Neurosurgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Juliane Fluck
- Fraunhofer Institute for Algorithms and Scientific Computing, Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- Institute of Geodesy and Geoinformation, University of Bonn, 53115 Bonn, Germany
- German National Library of Medicine (ZB MED) - Information Centre for Life Sciences, 53115 Bonn, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, 34128 Kassel, Germany
- Institute of Neurology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- German Center for Neurodegenerative Diseases, 72076 Tübingen, Germany
| |
Collapse
|
13
|
Lange KW, Nakamura Y, Chen N, Guo J, Kanaya S, Lange KM, Li S. Diet and medical foods in Parkinson’s disease. FOOD SCIENCE AND HUMAN WELLNESS 2019. [DOI: 10.1016/j.fshw.2019.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
14
|
Baicalin alleviates 6-hydroxydopamine-induced neurotoxicity in PC12 cells by down-regulation of microRNA-192-5p. Brain Res 2018; 1708:84-92. [PMID: 30552896 DOI: 10.1016/j.brainres.2018.12.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD), which is caused by neurodegenerative disorder, has no effective treatment until now. Baicalin was reported to have neuroprotective effects. Hence, we investigated the effects of baicalin on PD in an in vitro cell model by using 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in rat pheochromocytoma PC12 cells. PC12 cells were stimulated by 6-OHDA and were treated with baicalin and/or transfected with miR-192-5p mimic or negative control (NC). Cell viability and apoptosis were examined by Cell Counting Kit-8 assay and Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) analysis, respectively. The expression of p62, ratio of light chain (LC)3-II/LC3-I, miR-192-5p was detected by qRT-PCR. All protein expression levels were analyzed by western blot. We found that 6-OHDA significantly inhibited cell viability, induced apoptosis and autophagy, while baicalin reversed the results led by 6-OHDA. Moreover, baicalin negatively regulated expression of miR-192-5p. Under baicalin treatment, transfection with miR-192-5p mimic decreased cell viability and induced apoptosis and autophagy in 6-OHDA-treated cells compared with NC. In addition, the phosphorylation of phosphatidylinositol 3'-kinase (PI3K) and protein kinase B (AKT) was statistically down-regulated by baicalin then thereafter reversed by miR-192-5p mimic. Baicalin reduced 6-OHDA-induced cell injury through down-regulation of miR-192-5p, as well as regulation of PI3K/AKT and MDM-2/p53 signal pathways.
Collapse
|
15
|
Brain Photobiomodulation Therapy: a Narrative Review. Mol Neurobiol 2018; 55:6601-6636. [PMID: 29327206 DOI: 10.1007/s12035-017-0852-4] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/19/2017] [Indexed: 12/20/2022]
Abstract
Brain photobiomodulation (PBM) therapy using red to near-infrared (NIR) light is an innovative treatment for a wide range of neurological and psychological conditions. Red/NIR light is able to stimulate complex IV of the mitochondrial respiratory chain (cytochrome c oxidase) and increase ATP synthesis. Moreover, light absorption by ion channels results in release of Ca2+ and leads to activation of transcription factors and gene expression. Brain PBM therapy enhances the metabolic capacity of neurons and stimulates anti-inflammatory, anti-apoptotic, and antioxidant responses, as well as neurogenesis and synaptogenesis. Its therapeutic role in disorders such as dementia and Parkinson's disease, as well as to treat stroke, brain trauma, and depression has gained increasing interest. In the transcranial PBM approach, delivering a sufficient dose to achieve optimal stimulation is challenging due to exponential attenuation of light penetration in tissue. Alternative approaches such as intracranial and intranasal light delivery methods have been suggested to overcome this limitation. This article reviews the state-of-the-art preclinical and clinical evidence regarding the efficacy of brain PBM therapy.
Collapse
|
16
|
Why should neuroscientists worry about iron? The emerging role of ferroptosis in the pathophysiology of neuroprogressive diseases. Behav Brain Res 2017; 341:154-175. [PMID: 29289598 DOI: 10.1016/j.bbr.2017.12.036] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/23/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022]
Abstract
Ferroptosis is a unique form of programmed death, characterised by cytosolic accumulation of iron, lipid hydroperoxides and their metabolites, and effected by the fatal peroxidation of polyunsaturated fatty acids in the plasma membrane. It is a major driver of cell death in neurodegenerative neurological diseases. Moreover, cascades underpinning ferroptosis could be active drivers of neuropathology in major psychiatric disorders. Oxidative and nitrosative stress can adversely affect mechanisms and proteins governing cellular iron homeostasis, such as the iron regulatory protein/iron response element system, and can ultimately be a source of abnormally high levels of iron and a source of lethal levels of lipid membrane peroxidation. Furthermore, neuroinflammation leads to the upregulation of divalent metal transporter1 on the surface of astrocytes, microglia and neurones, making them highly sensitive to iron overload in the presence of high levels of non-transferrin-bound iron, thereby affording such levels a dominant role in respect of the induction of iron-mediated neuropathology. Mechanisms governing systemic and cellular iron homeostasis, and the related roles of ferritin and mitochondria are detailed, as are mechanisms explaining the negative regulation of ferroptosis by glutathione, glutathione peroxidase 4, the cysteine/glutamate antiporter system, heat shock protein 27 and nuclear factor erythroid 2-related factor 2. The potential role of DJ-1 inactivation in the precipitation of ferroptosis and the assessment of lipid peroxidation are described. Finally, a rational approach to therapy is considered, with a discussion on the roles of coenzyme Q10, iron chelation therapy, in the form of deferiprone, deferoxamine (desferrioxamine) and deferasirox, and N-acetylcysteine.
Collapse
|
17
|
Magri F, Vanoli F, Corti S. miRNA in spinal muscular atrophy pathogenesis and therapy. J Cell Mol Med 2017; 22:755-767. [PMID: 29160009 PMCID: PMC5783860 DOI: 10.1111/jcmm.13450] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 10/03/2017] [Indexed: 02/06/2023] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disease characterized by the selective death of lower motor neurons in the brain stem and spinal cord. SMA is caused by mutations in the survival motor neuron 1 gene (SMN1), leading to the reduced expression of the full-length SMN protein. microRNAs (miRNAs) are small RNAs that regulate post-transcriptional gene expression. Recent findings have suggested an important role for miRNAs in the pathogenesis of motor neuron diseases, including SMA. Motor neuron-specific miRNA dysregulation in SMA might be implicated in their selective vulnerability. In this study, we discuss recent findings regarding the consequences of SMN defects on miRNAs and their target mRNAs in motor neurons. Taken together, these data suggest that cell-specific changes in miRNAs are not only involved in the SMA motor neuron phenotype but can also be used as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Francesca Magri
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Fiammetta Vanoli
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy.,Department of Neurological Sciences, Mental Health and Sensory Organs (NESMOS), "Sapienza" University of Rome, Rome, Italy
| | - Stefania Corti
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| |
Collapse
|
18
|
Phosphorylation of p53 by LRRK2 induces microglial tumor necrosis factor α-mediated neurotoxicity. Biochem Biophys Res Commun 2017; 482:1088-1094. [DOI: 10.1016/j.bbrc.2016.11.163] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 11/29/2016] [Indexed: 12/15/2022]
|
19
|
Alves da Costa C, Duplan E, Checler F. α-synuclein and p53 functional interplay in physiopathological contexts. Oncotarget 2016; 8:9001-9002. [PMID: 28053290 PMCID: PMC5354703 DOI: 10.18632/oncotarget.14385] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Cristine Alves da Costa
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Laboratory of Excellence (LABEX) Distalz", Sophia-Antipolis, Valbonne, France
| | - Eric Duplan
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Laboratory of Excellence (LABEX) Distalz", Sophia-Antipolis, Valbonne, France
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Laboratory of Excellence (LABEX) Distalz", Sophia-Antipolis, Valbonne, France
| |
Collapse
|
20
|
Qi X, Davis B, Chiang YH, Filichia E, Barnett A, Greig NH, Hoffer B, Luo Y. Dopaminergic neuron-specific deletion of p53 gene is neuroprotective in an experimental Parkinson's disease model. J Neurochem 2016; 138:746-57. [PMID: 27317935 DOI: 10.1111/jnc.13706] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/09/2016] [Accepted: 06/15/2016] [Indexed: 01/06/2023]
Abstract
p53, a stress response gene, is involved in diverse cell death pathways and its activation has been implicated in the pathogenesis of Parkinson's disease (PD). However, whether the neuronal p53 protein plays a direct role in regulating dopaminergic (DA) neuronal cell death is unknown. In this study, in contrast to the global inhibition of p53 function by pharmacological inhibitors and in traditional p53 knock-out (KO) mice, we examined the effect of DA specific p53 gene deletion in DAT-p53KO mice. These DAT-p53KO mice did not exhibit apparent changes in the general structure and neuronal density of DA neurons during late development and in aging. However, in DA-p53KO mice treated with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), we found that the induction of Bax and p53 up-regulated modulator of apoptosis (PUMA) mRNA and protein levels by MPTP were diminished in both striatum and substantia nigra of these mice. Notably, deletion of the p53 gene in DA neurons significantly reduced dopaminergic neuronal loss in substantia nigra, dopaminergic neuronal terminal loss at striatum and, additionally, decreased motor deficits in mice challenged with MPTP. In contrast, there was no difference in astrogliosis between WT and DAT-p53KO mice in response to MPTP treatment. These findings demonstrate a specific contribution of p53 activation in DA neuronal cell death by MPTP challenge. Our results further support the role of programmed cell death mediated by p53 in this animal model of PD and identify Bax, BAD and PUMA genes as downstream targets of p53 in modulating DA neuronal death in the in vivo MPTP-induced PD model. We deleted p53 gene in dopaminergic neurons in late developmental stages and found that DA specific p53 deletion is protective in acute MPTP animal model possibly through blocking MPTP-induced BAX and PUMA up-regulation. Astrocyte activation measured by GFAP positive cells and GFAP gene up-regulation in the striatum shows no difference between wt and DA-p53 ko mice.
Collapse
Affiliation(s)
- Xin Qi
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Brandon Davis
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yung-Hsiao Chiang
- Division of Neurosurgery, Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Emily Filichia
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
| | - Austin Barnett
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute of Aging, Baltimore, Maryland, USA
| | - Barry Hoffer
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yu Luo
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
21
|
Nair VD, Ge Y. Alterations of miRNAs reveal a dysregulated molecular regulatory network in Parkinson's disease striatum. Neurosci Lett 2016; 629:99-104. [PMID: 27369327 DOI: 10.1016/j.neulet.2016.06.061] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/12/2022]
Abstract
Molecular adaptations in the striatum mediated by dopamine (DA) denervation and/or levodopa (L-dopa) treatments have been implicated in the motor deficits found in Parkinson's disease (PD). Alterations in inflammatory response mechanisms and glutamatergic neurotransmission are reported to play important roles in mediating these changes. However, the mechanisms mediating the molecular adaptations in the striatum are not well understood. Small non-coding microRNAs (miRNAs) influence numerous biological processes including the development and maintenance of striatal neurons by regulating gene expression post-transcriptionally. To investigate miRNA function in human PD striatum, we examined the global expression of miRNAs in postmortem putamen (putamen along with caudate forms the striatum) tissues obtained from PD patients and neurologically normal controls using Nanostring miRNA assays. We found that 6 miRNAs were significantly (p≤0.05) upregulated and 7 miRNAs were downregulated in PD putamen when compared with control. The differential expression (DE) of the 4 highest scoring miRNAs was further confirmed by reverse transcription polymerase chain reaction. Ingenuity pathway analysis demonstrated that these miRNAs are enriched in the processes of inflammatory responses. We found that the expression of DE miRNAs in PD putamen negatively correlates with the expression of gene transcripts implicated in inflammatory response with p53 and NF-kB as central signaling molecules. Taken together, our results suggest that in PD striatum, the DE miRNAs are associated with the oxidative stress pathway. This mechanism may contribute to the molecular adaptations and related motor complications found in PD.
Collapse
Affiliation(s)
- Venugopalan D Nair
- Department of Neurology and Center for Translational Systems Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Yongchao Ge
- Department of Neurology and Center for Translational Systems Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
22
|
Pan Z, Niu Y, Liang Y, Zhang X, Dong M. β-Ecdysterone Protects SH-SY5Y Cells Against 6-Hydroxydopamine-Induced Apoptosis via Mitochondria-Dependent Mechanism: Involvement of p38(MAPK)-p53 Signaling Pathway. Neurotox Res 2016; 30:453-66. [PMID: 27229883 DOI: 10.1007/s12640-016-9631-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/24/2016] [Accepted: 05/17/2016] [Indexed: 11/28/2022]
Abstract
Parkinson's disease (PD) is a neurological disorder pathologically characterized by loss of dopaminergic neurons in the substantia nigra. No curative therapy is available for PD. We recently found that phytoestrogen β-ecdysterone (β-Ecd) is able to reduce MPP(+)-induced apoptosis in PC12 cells. This study investigated the potential of β-Ecd to protect against SH-SY5Y cell apoptosis induced by the PD-related neurotoxin 6-hydroxydopamine (6-OHDA) and the underlying mechanism for this cytoprotection. In the present study, pretreatment with β-Ecd significantly reduced 6-OHDA-induced apoptosis of SH-SY5Y cells by a mitochondria-dependent pathway, as indicated by downregulation of Bax and PUMA (p53 upregulated modulator of apoptosis) expression, suppressing ΔΨm loss, inhibiting cytochrome c release, and attenuating caspase-9 activation. Furthermore, we showed that the inhibition of p38 mitogen-activated protein kinase (p38(MAPK))-dependent p53 promoter activity contributed to the protection of SH-SY5Y cells from apoptosis, which was validated by the use of SB203580 or p38β dominant negative (DN) mutants. Additionally, knock-down apoptosis signal-regulating kinase 1 (ASK1) by specific shRNA and blockade reactive oxygen species (ROS) by pharmacological inhibitor competently prevented β-Ecd-mediated inhibition of p38(MAPK) and ASK1 phosphorylation, respectively. These data provide the first evidence that β-Ecd protects SH-SY5Y cells against 6-OHDA-induced apoptosis, possibly through mitochondria protection and p53 modulation via ROS-dependent ASK1-p38(MAPK) pathways. The neuroprotective effects of β-Ecd make it a promising candidate as a therapeutic agent for PD.
Collapse
Affiliation(s)
- Zhi Pan
- Center for New Medicine Research, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yingcai Niu
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, Jianhua District, Qiqihar, 161006, China
| | - Yini Liang
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, Jianhua District, Qiqihar, 161006, China
| | - Xiaojie Zhang
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, Jianhua District, Qiqihar, 161006, China
| | - Miaoxian Dong
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, Jianhua District, Qiqihar, 161006, China.
| |
Collapse
|
23
|
Chu Y, Morfini GA, Kordower JH. Alterations in Activity-Dependent Neuroprotective Protein in Sporadic and Experimental Parkinson’s Disease. JOURNAL OF PARKINSONS DISEASE 2016; 6:77-97. [DOI: 10.3233/jpd-160812] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Yaping Chu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Gerardo A. Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Jeffrey H. Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
24
|
Feng C, Luo T, Zhang S, Liu K, Zhang Y, Luo Y, Ge P. Lycopene protects human SH‑SY5Y neuroblastoma cells against hydrogen peroxide‑induced death via inhibition of oxidative stress and mitochondria‑associated apoptotic pathways. Mol Med Rep 2016; 13:4205-14. [PMID: 27035331 PMCID: PMC4838073 DOI: 10.3892/mmr.2016.5056] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 03/14/2016] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress, which is characterized by excessive production of reactive oxygen species (ROS), is a common pathway that results in neuronal injury or death due to various types of pathological stress. Although lycopene has been identified as a potent antioxidant, its effect on hydrogen peroxide (H2O2)-induced neuronal damage remains unclear. In the present study, pretreatment with lycopene was observed to protect SH-SY5Y neuroblastoma cells against H2O2-induced death via inhibition of apoptosis resulting from activation of caspase-3 and translocation of apoptosis inducing factor (AIF) to the nucleus. Furthermore, the over-produced ROS, as well as the reduced activities of anti-oxidative enzymes, superoxide dismutase and catalase, were demonstrated to be alleviated by lycopene. Additionally, lycopene counteracted H2O2-induced mitochondrial dysfunction, which was evidenced by suppression of mitochondrial permeability transition pore opening, attenuation of the decline of the mitochondrial membrane potential, and inhibition of the increase of Bax and decrease of Bcl-2 levels within the mitochondria. The release of cytochrome c and AIF from the mitochondria was also reduced. These results indicate that lycopene is a potent neuroprotectant against apoptosis, oxidative stress and mitochondrial dysfunction, and could be administered to prevent neuronal injury or death.
Collapse
Affiliation(s)
- Chunsheng Feng
- Department of Anesthesiology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Tianfei Luo
- Department of Neurology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shuyan Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Kai Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yanhong Zhang
- Department of Emergent Medicine, People's Hospital of Jilin Province, Changchun, Jilin 130021, P.R. China
| | - Yinan Luo
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
25
|
Duplan E, Giordano C, Checler F, Alves da Costa C. Direct α-synuclein promoter transactivation by the tumor suppressor p53. Mol Neurodegener 2016; 11:13. [PMID: 26833254 PMCID: PMC4736712 DOI: 10.1186/s13024-016-0079-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/27/2016] [Indexed: 11/21/2022] Open
Abstract
Background Parkinson’s disease (PD) is a motor disease associated with the degeneration of dopaminergic neurons of the substantia nigra pars compacta. p53 is a major neuronal pro-apoptotic factor that is at the center of gravity of multiple physiological and pathological cascades, some of which implying several key PD-linked proteins. Since p53 is up-regulated in PD-affected brain, we have examined its ability to regulate the transcription of α-synuclein, a key protein that accumulates in PD-related Lewy bodies. Results We show that pharmacological and genetic up-regulation of p53 expression lead to a strong increase of α-synuclein protein, promoter activity and mRNA levels. Several lines of evidence indicate that this transcriptional control is due to the DNA-binding properties of p53. Firstly, p53 DNA-binding dead mutations abolish p53 regulation of α-synuclein. Secondly, the deletion of p53 responsive element from α-synuclein promoter abrogates p53-mediated α-synuclein regulation. Thirdly, gel shift and chromatin immunoprecipitation studies indicate that p53 interacts physically with α-synuclein promoter both in vitro and in a physiological context. Furthermore, we show that the depletion of endogenous p53 in cells as well as in knockout mice down-regulates α-synuclein transcription. Conclusions Overall, we have identified α-synuclein as a new transcriptional target of p53 and delineated a cellular mechanism feeding the accumulation of toxic aggregated α-synuclein in PD. This original α-syn regulatory mechanism may be central to PD-related cell death and may lead to novel opportunities to design alternative neuroprotective strategies in PD. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0079-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eric Duplan
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Cécile Giordano
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France.
| | - Cristine Alves da Costa
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France.
| |
Collapse
|
26
|
Bhinge A, Namboori SC, Bithell A, Soldati C, Buckley NJ, Stanton LW. MiR-375 is Essential for Human Spinal Motor Neuron Development and May Be Involved in Motor Neuron Degeneration. Stem Cells 2016; 34:124-134. [DOI: 10.1002/stem.2233] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
The transcription factor REST is a key suppressor of neuronal genes in non-neuronal tissues. REST has been shown to suppress proneuronal microRNAs in neural progenitors indicating that REST-mediated neurogenic suppression may act in part via microRNAs. We used neural differentiation of Rest-null mouse ESC to identify dozens of microRNAs regulated by REST during neural development. One of the identified microRNAs, miR-375, was upregulated during human spinal motor neuron development. We found that miR-375 facilitates spinal motor neurogenesis by targeting the cyclin kinase CCND2 and the transcription factor PAX6. Additionally, miR-375 inhibits the tumor suppressor p53 and protects neurons from apoptosis in response to DNA damage. Interestingly, motor neurons derived from a spinal muscular atrophy patient displayed depressed miR-375 expression and elevated p53 protein levels. Importantly, SMA motor neurons were significantly more susceptible to DNA damage induced apoptosis suggesting that miR-375 may play a protective role in motor neurons.
Collapse
Affiliation(s)
- Akshay Bhinge
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Seema C. Namboori
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Angela Bithell
- Department of Neuroscience, Centre for the Cellular Basis of Behaviour, Institute of Psychiatry, King's College London, London, UK
| | - Chiara Soldati
- Department of Neuroscience, Centre for the Cellular Basis of Behaviour, Institute of Psychiatry, King's College London, London, UK
| | - Noel J. Buckley
- Department of Psychiatry, University of Oxford, Warneford Hospital, Headington, Oxford, United Kingdom
| | - Lawrence W. Stanton
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
27
|
Ho DH, Kim H, Kim J, Sim H, Ahn H, Kim J, Seo H, Chung KC, Park BJ, Son I, Seol W. Leucine-Rich Repeat Kinase 2 (LRRK2) phosphorylates p53 and induces p21(WAF1/CIP1) expression. Mol Brain 2015; 8:54. [PMID: 26384650 PMCID: PMC4575451 DOI: 10.1186/s13041-015-0145-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/04/2015] [Indexed: 11/25/2022] Open
Abstract
Background Leucine-rich repeat kinase 2 (LRRK2) is a gene in which a mutation causes Parkinson’s disease (PD), and p53 is a prototype tumor suppressor. In addition, activation of p53 in patient with PD has been reported by several studies. Because phosphorylation of p53 is critical for regulating its activity and LRRK2 is a kinase, we tested whether p53 is phosphorylated by LRRK2. Results LRRK2 phosphorylates threonine (Thr) at TXR sites in an in vitro kinase assay, and the T304 and T377 were identified as putative phosphorylated residues. An increase of phospho-Thr in the p53 TXR motif was confirmed in the cells overexpressing G2019S, and human induced pluripotent stem (iPS) cells of a G2019S carrier. Interactions between LRRK2 and p53 were confirmed by co-immunoprecipitation of lysates of differentiated SH-SY5Y cells. LRRK2 mediated p53 phosphorylation translocalizes p53 predominantly to nucleus and increases p21WAF1/CIP1 expression in SH-SY5Y cells based on reverse transcription-polymerase chain reaction and Western blot assay results. The luciferase assay using the p21WAF1/CIP1 promoter-reporter also confirmed that LRRK2 kinase activity increases p21 expression. Exogenous expression of G2019S and the phosphomimetic p53 T304/377D mutants increased expression of p21WAF1/CIP1 and cleaved PARP, and cytotoxicity in the same cells. We also observed increase of p21 expression in rat primary neuron cells after transient expression of p53 T304/377D mutants and the mid-brain lysates of the G2019S transgenic mice. Conclusion p53 is a LRRK2 kinase substrate. Phosphorylation of p53 by LRRK2 induces p21WAF1/CIP1 expression and apoptosis in differentiated SH-SY5Y cells and rat primary neurons. Electronic supplementary material The online version of this article (doi:10.1186/s13041-015-0145-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dong Hwan Ho
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.,Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Hyejung Kim
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea
| | - Jisun Kim
- Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Hyuna Sim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hyunjun Ahn
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.,Korea University of Science & Technology (UST), 113 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Ansanshi, Gyeonggido, Republic of Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seodaemun-gu, Seoul, Republic of Korea
| | - Bum-Joon Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Pusan, Republic of Korea
| | - Ilhong Son
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea. .,Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, 321 Sanbon-ro, Gunposhi, Gyeonggido, Republic of Korea.
| |
Collapse
|
28
|
MAUGERI GRAZIA, D'AMICO AGATAGRAZIA, MAGRO GAETANO, SALVATORELLI LUCIA, BARBAGALLO GIUSEPPEM, SACCONE SALVATORE, DRAGO FILIPPO, CAVALLARO SEBASTIANO, D'AGATA VELIA. Expression profile of parkin isoforms in human gliomas. Int J Oncol 2015; 47:1282-92. [DOI: 10.3892/ijo.2015.3105] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/12/2015] [Indexed: 11/05/2022] Open
|
29
|
Khan MZ, He L. The role of polyunsaturated fatty acids and GPR40 receptor in brain. Neuropharmacology 2015; 113:639-651. [PMID: 26005184 DOI: 10.1016/j.neuropharm.2015.05.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/02/2015] [Accepted: 05/08/2015] [Indexed: 01/15/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) are found in abundance in the nervous system. They perform significant functions for example boosting synaptogenesis, neurogenesis, inducing antinociception, stimulating gene expression and neuronal activity, preventing apoptosis and neuroinflammation. G-protein-coupled receptor 40 (GPR40), also called free fatty acid receptor 1 (FFA1), is ubiquitously expressed in various regions of the human brain including the olfactory bulb, midbrain, medulla oblongata, hippocampus, hypothalamus, cerebral cortex, cerebellum and in the spinal cord. GPR40, when binding with polyunsaturated fatty acids (PUFAs) has shown promising therapeutic potential. This review presents current knowledge regarding the pharmacological properties of GPR40 and addresses its functions in brain, with a focus on neurodevelopment & neurogenesis. Furthermore, the demonstration of GPR40 involvement in several neuropathological conditions such as apoptosis, inflammatory pain, Alzheimer's disease and Parkinson's disease. Although the results are encouraging, further research is needed to clarify their role in the treatment of inflammatory pain, Alzheimer's disease and Parkinson's disease. This article is part of the Special Issue entitled 'Lipid Sensing G Protein-Coupled Receptors in the CNS'.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
30
|
Srivastav S, Singh SK, Yadav AK, Srikrishna S. Folic Acid Supplementation Ameliorates Oxidative Stress, Metabolic Functions and Developmental Anomalies in a Novel Fly Model of Parkinson's Disease. Neurochem Res 2015; 40:1350-9. [PMID: 25963948 DOI: 10.1007/s11064-015-1598-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 04/27/2015] [Accepted: 05/02/2015] [Indexed: 10/23/2022]
Abstract
Mutations in parkin cause early-onset Parkinson's disease. Studies involving Drosophila model have emphasised mitochondrial dysfunction as a critical event in disease pathogenesis. In this context, we employed a novel recessive allele of parkin, park (c00062) , for the current study. The piggyBac insertion at 3rd intron of parkin in park (c00062) was confirmed by PCR. Homozygous park (c00062) has diminished levels of truncated parkin transcript with no detectable protein as confirmed by qRT-PCR and western blot analysis, respectively. The homozygous park (c00062) displayed severe developmental anomalies involving reduced body size, ~45 % pupal lethality, high mortality with locomotory defect, elevated oxidative stress, low metabolic active cell status with low mitochondrial respiration as reflected from reduced ATP levels. Further, folic acid therapeutic potential was analysed in park (c00062) . Here we show that dietary folic acid provided protection against disparities involving pupal lethality, high mortality, locomotory defect, elevated oxidative stress and low metabolic active cell status associated with park (c00062) . Further mitochondrial respiration was enhanced as reflected from improved ATP levels in folate supplemented park (c00062) . To corroborate mitochondrial functioning further our analysis regarding transcript status of p53 and spargel by qRT-PCR, revealed down regulation of p53 and up regulation of spargel in folate supplemented park (c00062) , which was originally vice a versa. Our data thus support the potential of FA in alleviating the disparities associated with parkin loss of function in fly model. Further, FA role in alleviating mitochondrial dysfunction is encouraging to further explore FA mechanistic role to be utilized as potential therapeutics for parkin mediated neurodegenerative diseases.
Collapse
Affiliation(s)
- Saurabh Srivastav
- Cell and Neurobiology Laboratory, Department of Biochemistry, Faculty of Science, Banaras Hindu University, Varanasi, India
| | | | | | | |
Collapse
|
31
|
Srivastav S, Singh SK, Yadav AK, Srikrishna S. Folic acid supplementation rescues anomalies associated with knockdown of parkin in dopaminergic and serotonergic neurons in Drosophila model of Parkinson's disease. Biochem Biophys Res Commun 2015; 460:780-5. [DOI: 10.1016/j.bbrc.2015.03.106] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/20/2015] [Indexed: 10/23/2022]
|
32
|
Lawrence K, Jackson T, Jamieson D, Stevens A, Owens G, Sayan B, Locke I, Townsend P. Urocortin – From Parkinson's disease to the skeleton. Int J Biochem Cell Biol 2015; 60:130-8. [DOI: 10.1016/j.biocel.2014.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/12/2014] [Accepted: 12/13/2014] [Indexed: 01/04/2023]
|
33
|
Dopamine midbrain neurons in health and Parkinson’s disease: Emerging roles of voltage-gated calcium channels and ATP-sensitive potassium channels. Neuroscience 2015; 284:798-814. [DOI: 10.1016/j.neuroscience.2014.10.037] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/20/2014] [Accepted: 10/22/2014] [Indexed: 12/14/2022]
|
34
|
Labandeira-Garcia JL, Rodríguez-Perez AI, Villar-Cheda B, Borrajo A, Dominguez-Meijide A, Guerra MJ. Rho Kinase and Dopaminergic Degeneration. Neuroscientist 2014; 21:616-29. [DOI: 10.1177/1073858414554954] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The small GTP-binding protein Rho plays an important role in several cellular functions. RhoA, which is a member of the Rho family, initiates cellular processes that act on its direct downstream effector Rho-associated kinase (ROCK). ROCK inhibition protects against dopaminergic cell death induced by dopaminergic neurotoxins. It has been suggested that ROCK inhibition activates neuroprotective survival cascades in dopaminergic neurons. Axon-stabilizing effects in damaged neurons may represent another mechanism of neuroprotection of dopaminergic neurons by ROCK inhibition. However, it has been shown that microglial cells play a crucial role in neuroprotection by ROCK inhibition and that activation of microglial ROCK mediates major components of the microglial inflammatory response. Additional mechanisms such as interaction with autophagy may also contribute to the neuroprotective effects of ROCK inhibition. Interestingly, ROCK interacts with several brain factors that play a major role in dopaminergic neuron vulnerability such as NADPH-oxidase, angiotensin, and estrogen. ROCK inhibition may provide a new neuroprotective strategy for Parkinson’s disease. This is of particular interest because ROCK inhibitors are currently used against vascular diseases in clinical practice. However, it is necessary to develop more potent and selective ROCK inhibitors to reduce side effects and enhance the efficacy.
Collapse
Affiliation(s)
- Jose L. Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Ana I. Rodríguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Begoña Villar-Cheda
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Ana Borrajo
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Antonio Dominguez-Meijide
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Maria J. Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| |
Collapse
|
35
|
Zhou D, Zhan C, Zhong Q, Li S. Upregulation of sestrin-2 expression via P53 protects against 1-methyl-4-phenylpyridinium (MPP+) neurotoxicity. J Mol Neurosci 2014; 51:967-75. [PMID: 23959424 DOI: 10.1007/s12031-013-0081-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 07/18/2013] [Indexed: 01/20/2023]
Abstract
Sestrin-2 (SESN2) is a conserved antioxidant protein that is activated upon oxidative stress and protects cells against reactive oxygen species (ROS). However, the role of SESN2 in neurodegenerative diseases, especially in Parkinson's disease (PD), has not yet been elucidated. In this study, we found that expression of SESN2 is elevated in the midbrain of patients with PD. Moreover, in vitro experiments display that the drug 1-methyl-4-phenylpyridinium (MPP+) induces the expression of SESN2 in SH-SY5Y cells in a time- and dose-dependent manner. Our results show that p53 is activated by MPP+. Importantly, inhibition of p53 using small RNA interferences abolishes the increased SESN2 levels induced by MPP+, suggesting that the inductive effect of MPP+ on SESN2 is mediated by p53. Furthermore, knockdown of SESN2 using small RNA interferences promotes MPP+-related neurotoxicity by attenuating oxidative stress, mitochondrial dysfunction, and apoptosis. All these data imply that the induction of SESN2 produces a protective effect in PD.
Collapse
|
36
|
Niedzwiedz A, Jaworski Z, Tykalowski B, Smialek M. Neutrophil and macrophage apoptosis in bronchoalveolar lavage fluid from healthy horses and horses with recurrent airway obstruction (RAO). BMC Vet Res 2014; 10:29. [PMID: 24460911 PMCID: PMC3903020 DOI: 10.1186/1746-6148-10-29] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 01/22/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Dysregulation of apoptosis has been implicated in a range of diseases including tumors, neurodegenerative and autoimmune diseases, as well as allergic asthma and chronic obstructive pulmonary disease (COPD) in humans. Although it has a different pathophysiology, delayed apoptosis of various inflammatory cells may play a pivotal role in the development of recurrent airway obstruction (RAO) in horses. Reduction of inflammatory cell apoptosis or a dysregulation of this process could lead to chronic inflammation and tissue injury. Therefore, the aim of this study was to investigate the rate of apoptosis and necrosis of neutrophils and macrophages in bronchoalveolar lavage fluid obtained from seven horses suffering from RAO (study group) and seven control horses. RESULTS We demonstrated that neutrophil/macrophage apoptosis is altered in RAO-affected horses compared with the control group in the BAL fluid. We found a significant difference between the median percentage of early and late apoptosis of neutrophils between the study and control group of horses. Moreover, we found a positive correlation between the rate of apoptosis and the median percentage of macrophages in RAO-affected horses. CONCLUSION The findings suggest that apoptosis dysregulation may play a significant role in the pathogenesis of RAO. However, further studies are needed to clarify the role of altered apoptosis in the course of equine recurrent airway obstruction.
Collapse
Affiliation(s)
- Artur Niedzwiedz
- Department of Internal Diseases with Clinic for Horses, Dogs and Cats, The Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Pl, Grunwaldzki 47, Wrocław 50-366, Poland.
| | | | | | | |
Collapse
|
37
|
Naoi M, Maruyama W, Inaba-Hasegawa K. Revelation in the neuroprotective functions of rasagiline and selegiline: the induction of distinct genes by different mechanisms. Expert Rev Neurother 2014; 13:671-84. [PMID: 23739004 DOI: 10.1586/ern.13.60] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In Parkinson's disease, cell death of dopamine neurons in the substantia nigra progresses and neuroprotective therapy is required to halt neuronal loss. In cellular and animal models, selegiline [(-)deprenyl] and rasagiline, inhibitors of type B monoamine oxidase (MAO)-B, protect neuronal cells from programmed cell death. In this paper, the authors review their recent results on the molecular mechanisms by which MAO inhibitors prevent the cell death through the induction of antiapoptotic, prosurvival genes. MAO-A mediates the induction of antiapoptotic bcl-2 and mao-a itself by rasagiline, whereas a different mechanism is associated with selegiline. Rasagiline and selegiline preferentially increase GDNF and BDNF in nonhuman primates and Parkinsonian patients, respectively. Enhanced neurotrophic factors might be applicable to monitor the neurorescuing activity of neuroprotection.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, Nisshin, Aichi, Japan.
| | | | | |
Collapse
|
38
|
p53 in neurodegenerative diseases and brain cancers. Pharmacol Ther 2013; 142:99-113. [PMID: 24287312 DOI: 10.1016/j.pharmthera.2013.11.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 11/07/2013] [Indexed: 12/21/2022]
Abstract
More than thirty years elapsed since a protein, not yet called p53 at the time, was detected to bind SV40 during viral infection. Thousands of papers later, p53 evolved as the main tumor suppressor involved in growth arrest and apoptosis. A lot has been done but the protein has not yet revealed all its secrets. Particularly important is the observation that in totally distinct pathologies where apoptosis is either exacerbated or impaired, p53 appears to play a central role. This is exemplified for Alzheimer's and Parkinson's diseases that represent the two main causes of age-related neurodegenerative affections, where cell death enhancement appears as one of the main etiological paradigms. Conversely, in cancers, about half of the cases are linked to mutations in p53 leading to the impairment of p53-dependent apoptosis. The involvement of p53 in these pathologies has driven a huge amount of studies aimed at designing chemical tools or biological approaches to rescue p53 defects or over-activity. Here, we describe the data linking p53 to neurodegenerative diseases and brain cancers, and we document the various strategies to interfere with p53 dysfunctions in these disorders.
Collapse
|
39
|
Sunico CR, Nakamura T, Rockenstein E, Mante M, Adame A, Chan SF, Newmeyer TF, Masliah E, Nakanishi N, Lipton SA. S-Nitrosylation of parkin as a novel regulator of p53-mediated neuronal cell death in sporadic Parkinson's disease. Mol Neurodegener 2013; 8:29. [PMID: 23985028 PMCID: PMC3765907 DOI: 10.1186/1750-1326-8-29] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 08/16/2013] [Indexed: 12/21/2022] Open
Abstract
Background Mutations in the gene encoding parkin, a neuroprotective protein with dual functions as an E3 ubiquitin ligase and transcriptional repressor of p53, are linked to familial forms of Parkinson’s disease (PD). We hypothesized that oxidative posttranslational modification of parkin by environmental toxins may contribute to sporadic PD. Results We first demonstrated that S-nitrosylation of parkin decreased its activity as a repressor of p53 gene expression, leading to upregulation of p53. Chromatin immunoprecipitation as well as gel-shift assays showed that parkin bound to the p53 promoter, and this binding was inhibited by S-nitrosylation of parkin. Additionally, nitrosative stress induced apoptosis in cells expressing parkin, and this death was, at least in part, dependent upon p53. In primary mesencephalic cultures, pesticide-induced apoptosis was prevented by inhibition of nitric oxide synthase (NOS). In a mouse model of pesticide-induced PD, both S-nitrosylated (SNO-)parkin and p53 protein levels were increased, while administration of a NOS inhibitor mitigated neuronal death in these mice. Moreover, the levels of SNO-parkin and p53 were simultaneously elevated in postmortem human PD brain compared to controls. Conclusions Taken together, our data indicate that S-nitrosylation of parkin, leading to p53-mediated neuronal cell death, contributes to the pathophysiology of sporadic PD.
Collapse
Affiliation(s)
- Carmen R Sunico
- Sanford-Burnham Medical Research Institute, Del E, Webb Center for Neuroscience, Aging, and Stem Cell Research, 10901, North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Parkinson's disease (PD) is one of the most common degenerative disorders of the central nervous system that produces motor and non-motor symptoms. The majority of cases are idiopathic and characterized by the presence of Lewy bodies containing fibrillar α-synuclein. Small ubiquitin-related modifier (SUMO) immunoreactivity was observed among others in cases with PD. Key disease-associated proteins are SUMO-modified, linking this posttranslational modification to neurodegeneration. SUMOylation and SUMO-mediated mechanisms have been intensively studied in recent years, revealing nuclear and extranuclear functions for SUMO in a variety of cellular processes, including the regulation of transcriptional activity, modulation of signal transduction pathways, and response to cellular stress. This points to a role for SUMO more than just an antagonist to ubiquitin and proteasomal degradation. The identification of risk and age-at-onset gene loci was a breakthrough in PD and promoted the understanding of molecular mechanisms in the pathology. PD has been increasingly linked with mitochondrial dysfunction and impaired mitochondrial quality control. Interestingly, SUMO is involved in many of these processes and up-regulated in response to cellular stress, further emphasizing the importance of SUMOylation in physiology and disease.
Collapse
Affiliation(s)
- Katrin Eckermann
- Department of Neurology, University Medical Center Goettingen, Waldweg 33, 37073, Goettingen, Germany,
| |
Collapse
|
41
|
Lanni C, Necchi D, Pinto A, Buoso E, Buizza L, Memo M, Uberti D, Govoni S, Racchi M. Zyxin is a novel target for β-amyloid peptide: characterization of its role in Alzheimer's pathogenesis. J Neurochem 2013; 125:790-9. [PMID: 23330981 DOI: 10.1111/jnc.12154] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 01/11/2013] [Accepted: 01/11/2013] [Indexed: 11/26/2022]
Abstract
Zyxin is an adaptor protein recently identified as a novel regulator of the homeodomain-interacting protein kinase 2 (HIPK2)-p53 signaling in response to DNA damage. We recently reported an altered conformational state of p53 in tissues from patients with Alzheimer 's disease (AD), because of a deregulation of HIPK2 activity, leading to an impaired and dysfunctional response to stressors. Here, we examined the molecular mechanisms underlying the deregulation of HIPK2 activity in two cellular models, HEK-293 cells and SH-SY5Y neuroblastoma cells differentiated with retinoic acid over-expressing the amyloid precursor protein, focusing on the evidence that zyxin expression is important to maintain HIPK2 protein stability. We demonstrated that both beta-amyloid (Aβ) 1-40 and 1-42 induce zyxin deregulation, thus affecting the transcriptional repressor activity of HIPK2 onto its target promoter, metallothionein 2A, which is in turn responsible for the induction of an altered conformational state of p53. We demonstrate for the first time that zyxin is a novel target of Aβ activities in AD. These results may help the studies on the pathogenesis of AD, through the fine dissection of events related to beta-amyloid activities.
Collapse
Affiliation(s)
- Cristina Lanni
- Department of Drug Sciences, Centre of Excellence in Applied Biology, University of Pavia, 27100 Pavia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Almeida A. Genetic determinants of neuronal vulnerability to apoptosis. Cell Mol Life Sci 2013; 70:71-88. [PMID: 22695677 PMCID: PMC11113535 DOI: 10.1007/s00018-012-1029-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 04/22/2012] [Accepted: 05/07/2012] [Indexed: 12/18/2022]
Abstract
Apoptosis is a common mode of cell death that contributes to neuronal loss associated with neurodegeneration. Single-nucleotide polymorphisms (SNPs) in chromosomal DNA are contributing factors dictating natural susceptibility of humans to disease. Here, the most common SNPs affecting neuronal vulnerability to apoptosis are reviewed in the context of neurological disorders. Polymorphic variants in genes encoding apoptotic proteins, either from the extrinsic (FAS, TNF-α, CASP8) or the intrinsic (BAX, BCL2, CASP3, CASP9) pathways could be highly valuable in the diagnosis of neurodegenerative diseases and stroke. Interestingly, the Arg72Pro SNP in TP53, the gene encoding tumor suppressor p53, was recently revealed a biomarker of poor prognosis in stroke due to its ability to modulate neuronal apoptotic death. Search for new SNPs responsible for genetic variability to apoptosis will ensure the implementation of novel diagnostic and prognostic tools, as well as therapeutic strategies against neurological diseases.
Collapse
Affiliation(s)
- Angeles Almeida
- Instituto de Investigación Biomédica de Salamanca, Hospital Universitario de Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
43
|
D'Amelio M, Sheng M, Cecconi F. Caspase-3 in the central nervous system: beyond apoptosis. Trends Neurosci 2012; 35:700-9. [PMID: 22796265 DOI: 10.1016/j.tins.2012.06.004] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 06/14/2012] [Accepted: 06/15/2012] [Indexed: 01/06/2023]
Abstract
Caspase-3 has been identified as a key mediator of neuronal programmed cell death. This protease plays a central role in the developing nervous system and its activation is observed early in neural tube formation and persists during postnatal differentiation of the neural network. Caspase-3 activation, a crucial event of neuronal cell death program, is also a feature of many chronic neurodegenerative diseases. This traditional apoptotic function of caspase-3 is challenged by recent studies that reveal new cell death-independent roles for mitochondrial-activated caspase-3 in neurite pruning and synaptic plasticity. These findings underscore the need for further research into the mechanism of action and functions of caspase-3 that may prove useful in the development of novel pharmacological treatments for a diverse range of neurological disorders.
Collapse
Affiliation(s)
- Marcello D'Amelio
- Istituto di Ricovero e Cura a Carattere Scientifico, S. Lucia Foundation, via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | | | | |
Collapse
|
44
|
Janda E, Isidoro C, Carresi C, Mollace V. Defective autophagy in Parkinson's disease: role of oxidative stress. Mol Neurobiol 2012; 46:639-61. [PMID: 22899187 DOI: 10.1007/s12035-012-8318-1] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 07/30/2012] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a paradigmatic example of neurodegenerative disorder with a critical role of oxidative stress in its etiopathogenesis. Genetic susceptibility factors of PD, such as mutations in Parkin, PTEN-induced kinase 1, and DJ-1 as well as the exposure to pesticides and heavy metals, both contribute to altered redox balance and degeneration of dopaminergic neurons in the substantia nigra. Dysregulation of autophagy, a lysosomal-driven process of self degradation of cellular organelles and protein aggregates, is also implicated in PD and PD-related mutations, and environmental toxins deregulate autophagy. However, experimental evidence suggests a complex and ambiguous role of autophagy in PD since either impaired or abnormally upregulated autophagic flux has been shown to cause neuronal loss. Finally, it is generally believed that oxidative stress is a strong proautophagic stimulus. However, some evidence coming from neurobiology as well as from other fields indicate an inhibitory role of reactive oxygen species and reactive nitrogen species on the autophagic machinery. This review examines the scientific evidence supporting different concepts on how autophagy is dysregulated in PD and attempts to reconcile apparently contradictory views on the role of oxidative stress in autophagy regulation. The complex relationship between autophagy and oxidative stress is also considered in the context of the ongoing search for a novel PD therapy.
Collapse
Affiliation(s)
- Elzbieta Janda
- Department of Health Sciences, University Magna Graecia, Edificio Bioscienze, viale Europa, Campus Salvatore Venuta, Germaneto, 88100 Catanzaro, Italy.
| | | | | | | |
Collapse
|
45
|
Redox Regulation of Protein Function via Cysteine S-Nitrosylation and Its Relevance to Neurodegenerative Diseases. Int J Cell Biol 2012; 2012:463756. [PMID: 22956959 PMCID: PMC3431077 DOI: 10.1155/2012/463756] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/17/2012] [Indexed: 12/21/2022] Open
Abstract
Debilitating neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), can be attributed to neuronal cell damage in specific brain regions. An important hallmark of these diseases is increased oxidative and nitrosative stress that occurs via overproduction of highly reactive free radicals known as reactive oxygen species (ROS) and reactive nitrogen species (RNS). These molecules are normally removed by cellular antioxidant systems. Under physiological conditions, ROS/RNS are present at low levels, mediating several neurotrophic and neuroprotective signaling pathways. In contrast, under pathological conditions, there is a pronounced increase in ROS/RNS generation, impairing normal neurological function. Nitric oxide (NO) is one such molecule that functions as a signaling agent under physiological conditions but causes nitrosative stress under pathological conditions due to its enhanced production. As first reported by our group and colleagues, the toxic effects of NO can be in part attributed to thiol S-nitrosylation, a posttranslational modification of cysteine residues on specific proteins. Here, we review several reports appearing over the past decade showing that S-nitrosylation of an increasing number of proteins compromises important cellular functions, including mitochondrial dynamics, endoplasmic reticulum (ER) protein folding, and signal transduction, thereby promoting synaptic damage, cell death, and neurodegeneration.
Collapse
|
46
|
Lanni C, Racchi M, Memo M, Govoni S, Uberti D. p53 at the crossroads between cancer and neurodegeneration. Free Radic Biol Med 2012; 52:1727-33. [PMID: 22387179 DOI: 10.1016/j.freeradbiomed.2012.02.034] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 02/17/2012] [Accepted: 02/22/2012] [Indexed: 12/20/2022]
Abstract
Aging, dementia, and cancer share a critical set of altered cellular functions in response to DNA damage, genotoxic stress, and other insults. Recent data suggest that the molecular machinery involved in maintaining neural function in neurodegenerative disease may be shared with oncogenic pathways. Cancer and neurodegenerative diseases may be influenced by common signaling pathways regulating the balance of cell survival versus death, a decision often governed by checkpoint proteins. This paper focuses on one such protein, p53, which represents one of the most extensively studied proteins because of its role in cancer prevention and which, furthermore, has been recently shown to be involved in aging and Alzheimer disease (AD). The contribution of a conformational change in p53 to aging and neurodegenerative processes has yet to be elucidated. In this review we discuss the multiple functions of p53 and how these correlate between cancer and neurodegeneration, focusing on various factors that may have a role in regulating p53 activity. The observation that aging and AD interfere with proteins controlling duplication and cell cycle may lead to the speculation that, in senescent neurons, aberrations in proteins generally dealing with cell cycle control and apoptosis could affect neuronal plasticity and functioning rather than cell duplication.
Collapse
Affiliation(s)
- Cristina Lanni
- Department of Drug Sciences, Centre of Excellence in Applied Biology, University of Pavia, 27100 Pavia, Italy.
| | | | | | | | | |
Collapse
|
47
|
The bad, the good, and the ugly about oxidative stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:163913. [PMID: 22619696 PMCID: PMC3350994 DOI: 10.1155/2012/163913] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 01/16/2012] [Accepted: 02/07/2012] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD), Parkinson's disease (PD), and cancer (e.g., leukemia) are the most devastating disorders affecting millions of people worldwide. Except for some kind of cancers, no effective and/or definitive therapeutic treatment aimed to reduce or to retard the clinic and pathologic symptoms induced by AD and PD is presently available. Therefore, it is urgently needed to understand the molecular basis of these disorders. Since oxidative stress (OS) is an important etiologic factor of the pathologic process of AD, PD, and cancer, understanding how intracellular signaling pathways respond to OS will have a significant implication in the therapy of these diseases. Here, we propose a model of minimal completeness of cell death signaling induced by OS as a mechanistic explanation of neuronal and cancer cell demise. This mechanism might provide the basis for therapeutic design strategies. Finally, we will attempt to associate PD, cancer, and OS. This paper critically analyzes the evidence that support the “oxidative stress model” in neurodegeneration and cancer.
Collapse
|
48
|
Zhu G, Wang X, Wu S, Li Q. Involvement of activation of PI3K/Akt pathway in the protective effects of puerarin against MPP+-induced human neuroblastoma SH-SY5Y cell death. Neurochem Int 2012; 60:400-8. [PMID: 22265823 DOI: 10.1016/j.neuint.2012.01.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 12/28/2011] [Accepted: 01/04/2012] [Indexed: 01/01/2023]
Abstract
In an attempt to clarify the protective effect of puerarin on toxin-insulted dopaminergic neuronal death, this present study was carried out by using a typical Parkinson's disease (PD) model - 1-methyl-4-phenylpyridinium iodide (MPP(+))-induced dopaminergic SH-SY5Y cellular model. Data are presented, which showed that puerarin up-regulated Akt phosphorylation in both of MPP(+)-treated and non-MPP(+)-treated cells. The presence of PI3K inhibitor LY294002 completely blocked puerarin-induced activation of Akt phosphorylation. Moreover, puerarin decreased MPP(+)-induced cell death, which was blocked by phosphoinositide 3-kinase (PI3K) inhibitor LY294002. We further demonstrated that puerarin protected against MPP(+)-induced p53 nuclear accumulation, Puma (p53-upregulated mediator of apoptosis) and Bax expression and caspase-3-dependent programmed cell death (PCD). This protection was blocked by applying a PI3K/Akt inhibitor. Additionally, it was Pifithrin-α, but not Pifithrin-μ, which blocked MPP(+)-induced Puma and Bax expression, caspase-3 activation and cell death. Collectively, these data suggest that the activation of PI3K/Akt pathway is involved in the protective effect of puerarin against MPP(+)-induced neuroblastoma SH-SY5Y cell death through inhibiting nuclear p53 accumulation and subsequently caspase-3-dependent PCD. Puerarin might be a potential therapeutic agent for PD.
Collapse
Affiliation(s)
- Guoqi Zhu
- Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Traditional Chinese Medicine, Hefei, China
| | | | | | | |
Collapse
|
49
|
Ser46 phosphorylation and prolyl-isomerase Pin1-mediated isomerization of p53 are key events in p53-dependent apoptosis induced by mutant huntingtin. Proc Natl Acad Sci U S A 2011; 108:17979-84. [PMID: 22011578 DOI: 10.1073/pnas.1106198108] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the gene coding for huntingtin protein. Several mechanisms have been proposed by which mutant huntingtin (mHtt) may trigger striatal neurodegeneration, including mitochondrial dysfunction, oxidative stress, and apoptosis. Furthermore, mHtt induces DNA damage and activates a stress response. In this context, p53 plays a crucial role in mediating mHtt toxic effects. Here we have dissected the pathway of p53 activation by mHtt in human neuronal cells and in HD mice, with the aim of highlighting critical nodes that may be pharmacologically manipulated for therapeutic intervention. We demonstrate that expression of mHtt causes increased phosphorylation of p53 on Ser46, leading to its interaction with phosphorylation-dependent prolyl isomerase Pin1 and consequent dissociation from the apoptosis inhibitor iASPP, thereby inducing the expression of apoptotic target genes. Inhibition of Ser46 phosphorylation by targeting homeodomain-interacting protein kinase 2 (HIPK2), PKCδ, or ataxia telangiectasia mutated kinase, as well as inhibition of the prolyl isomerase Pin1, prevents mHtt-dependent apoptosis of neuronal cells. These results provide a rationale for the use of small-molecule inhibitors of stress-responsive protein kinases and Pin1 as a potential therapeutic strategy for HD treatment.
Collapse
|
50
|
Bousquet M, Calon F, Cicchetti F. Impact of ω-3 fatty acids in Parkinson's disease. Ageing Res Rev 2011; 10:453-63. [PMID: 21414422 DOI: 10.1016/j.arr.2011.03.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 03/03/2011] [Accepted: 03/07/2011] [Indexed: 01/29/2023]
Abstract
Current epidemiological, preclinical and clinical data suggest that omega-3 polyunsaturated fatty acids (n-3 PUFAs) may constitute therapeutic strategy for several disorders of the central nervous system, including Parkinson's disease (PD). PD is a neurodegenerative disorder primarily characterized by motor symptoms but which also includes several other pathological features such as autonomic system failures, mood disorders, and cognitive deficits. Current pharmacological options for the disease are limited to symptom management and their long-term use leads to important side effects. In this review, we discuss the evidence for the effects of n-3 PUFAs in PD both from an epidemiological perspective as well as in light of data gathered on various pathological features of the disease. Effects of n-3 PUFAs on the dopaminergic system, α-synucleinopathy, their possible mechanisms of action as well as their therapeutic potential for PD patients are also reviewed. n-3 PUFAs are inexpensive, readily transferable to the clinical setting and their use could represent a neuroprotective strategy or a disease-modifying option to delay the appearance of symptoms. It could also be beneficial as a symptomatologic treatment or serve as an add-on therapy to current pharmacological approaches. Review of the current literature as well as the undertaking of future clinical trials will shed light on these possibilities.
Collapse
|