1
|
Chang LC, Chiang SK, Chen SE, Hung MC. Exploring paraptosis as a therapeutic approach in cancer treatment. J Biomed Sci 2024; 31:101. [PMID: 39497143 PMCID: PMC11533606 DOI: 10.1186/s12929-024-01089-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
A variety of cell death pathways play critical roles in the onset and progression of multiple diseases. Paraptosis, a unique form of programmed cell death, has gained significant attention in recent years. Unlike apoptosis and necrosis, paraptosis is characterized by cytoplasmic vacuolization, swelling of the endoplasmic reticulum and mitochondria, and the absence of caspase activation. Numerous natural products, synthetic compounds, and newly launched nanomedicines have been demonstrated to prime cell death through the paraptotic program and may offer novel therapeutic strategies for cancer treatment. This review summarizes recent findings, delineates the intricate network of signaling pathways underlying paraptosis, and discusses the potential therapeutic implications of targeting paraptosis in cancer treatment. The aim of this review is to expand our understanding of this unique cell death process and explore the potential therapeutic implications of targeting paraptosis in cancer treatment.
Collapse
Affiliation(s)
- Ling-Chu Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 406040, Taiwan.
- Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan.
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40402, Taiwan.
| | - Shih-Kai Chiang
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Shuen-Ei Chen
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan
- Innovation and Development Center of Sustainable Agriculture (IDCSA), National Chung Hsing University, Taichung, 40227, Taiwan
- i-Center for Advanced Science and Technology (iCAST), National Chung Hsing University, Taichung, 40227, Taiwan
| | - Mien-Chie Hung
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 406040, Taiwan.
- Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan.
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40402, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan.
| |
Collapse
|
2
|
Alipour M, Hajipour-Verdom B, Javan M, Abdolmaleki P. Static and Electromagnetic Fields Differently Affect Proliferation and Cell Death Through Acid Enhancement of ROS Generation in Mesenchymal Stem Cells. Radiat Res 2022; 198:384-395. [PMID: 35867630 DOI: 10.1667/rade-21-00037.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Magnetic fields remotely influence cellular homeostasis as a physical agent through the changes in cell physicochemical reactions. Magnetic fields affect cell fate, which may provide an important and interesting challenge in stem cell behaviors. Here, we investigated the effects of the static magnetic field (SMF, 20 mT) and electromagnetic field (EMF, 20 mT-50 Hz) on reactive oxygen species (ROS) production and the acidic pH conditions as stimuli to change cell cycle progression and cell death in mesenchymal stem cells. Results show that SMF, EMF, and their simultaneous (SMF+EMF) administration increase ROS and expression of nuclear factor erythroid 2-related factor 2 (Nrf2), superoxide dismutase 2 (SOD2), and glutathione-S-transferase (GST) as an antioxidant defense system. Besides, intracellular pH (pHi) decreases in presence of either EMF or SMF+EMF, but not SMF. Decreased ROS content using ascorbic acid in these treatments leads to increased pH compared to the magnetic field treatments alone. Furthermore, each magnetic field has different effects on the cellular process of stem cells, including cell cycle, apoptosis and necrosis. Moreover, treatment by SMF enhances the cell viability after 24 h, while EMF or SMF+EMF decreases it. These observations indicate that fluctuations of ROS generation and acid enhancement during SMF and EMF treatments may reveal their beneficial and adverse effects on the molecular and cellular mechanisms involved in the growth, death, and differentiation of stem cells.
Collapse
Affiliation(s)
| | | | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14115-154, Iran
| | | |
Collapse
|
3
|
Wu H, Sun H, He Z, Chen X, Li Y, Zhao X, Kong W, Kong W. The effect and mechanism of 19S proteasome PSMD11/Rpn6 subunit in D-Galactose induced mimetic aging models. Exp Cell Res 2020; 394:112093. [PMID: 32450067 DOI: 10.1016/j.yexcr.2020.112093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/14/2020] [Accepted: 05/16/2020] [Indexed: 11/30/2022]
Abstract
Regulating proteasome activity is a potent therapeutic aspect of age-related hearing loss, which has been proven to protect neurons from age-related damaging. PSMD11, subunit of the 19S proteasome regulatory particle, is known to mainly up-regulate proteasome activity and prolong aging. However, the mechanism of PSMD11 in age-related hearing loss has not been deeply explored. In the present study, we explore the function and mechanism of PSMD11 protecting neurons in d-Galactose (D-Gal) mimetic aging models. Age-related pathologies were detected by Taq-PCR, ABR, Transmission electron microscopy, toluidine blue and β-galactosidase staining. The relative expressions of the proteins were explored by Western blotting, oxyblot, immunoprecipitation and immunofluorescence. Flow cytometry was used to manifest the oxidative state. We discovered that proteasome activity was impaired with aging, and that ROS and toxic protein accumulated in D-Gal induced aging models. PSMD11 changed with aging, and was associated with the metabolism of proteasome activity in the D-Gal treated models. Moreover, the knockdown or overexpression of PSMD11 was sufficient to change the oxidative state caused by D-Gal. Our results also demonstrated that PSMD11 could bond to AMPKα1/2 in the auditory cortex and PC12 cells, and AMPKα2 but not AMPKα1 was efficient to regulate the function of PSMD11. Deeper insights into the mechanisms of regulating PSMD11 for the anti-aging process are needed, and may offer novel therapeutic methods for central presbycusis.
Collapse
Affiliation(s)
- Han Wu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Haiying Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zuhong He
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xi Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yongqin Li
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xueyan Zhao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wen Kong
- Departments of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Weijia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
4
|
Zhu Y, Wu Y, Shi W, Wang J, Yan X, Wang Q, Liu Y, Yang L, Gao L, Li M. Inhibition of ubiquitin proteasome function prevents monocrotaline-induced pulmonary arterial remodeling. Life Sci 2017; 173:36-42. [PMID: 28212825 DOI: 10.1016/j.lfs.2017.02.007] [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: 10/05/2016] [Revised: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 11/28/2022]
Abstract
AIMS Previous study has indicated that inhibition of proteasome function ameliorates the development of pulmonary arterial hypertension (PAH), while its underlying mechanisms are still unclear. This study was performed to address these issues. MATERIAL AND METHODS Male Sprague-Dawley (SD) rats were divided into five groups: control group, PAH group, vehicle treated PAH group, MG-132 treated PAH group and bortezomib treated PAH group. PAH model was established by a single intraperitoneal injection of monocrotaline (MCT). MG-132 and bortezomib were administered to inhibit proteasome function. The right ventricular systolic pressure (RVSP), the right ventricle hypertrophy index (RVHI) and the percentage of medial wall thickness (%MT) were used to evaluate the development of PAH. Hematoxylin and eosin staining was performed to measure vascular remodeling. Immunoblotting was used to determine Akt phosphorylation, expression of PTEN and NEDD4, and the level of ubiquitinated-PTEN protein. KEY FINDINGS MCT increased RVSP, RVHI and %MT in rats, while these changes were suppressed by treatment of PAH rats with MG-132 or bortezomib. In PAH model, expression of PTEN was decreased and phosphorylation of Akt was increased, these were accompanied by an elevation of NEDD4 protein level. Treatment of PAH model with MG-132 or bortezomib increased PTEN expression and accumulation of ubiquitinated-PTEN protein and decreased Akt phosphorylation, while didn't change NEDD4 expression. SIGNIFICANCE Inhibition of proteasome function ameliorates pulmonary arterial remodeling by suppressing UPS-mediated PTEN degradation and subsequent inhibition of PI3K/Akt pathway, indicating that UPS might be a novel target for prevention of PAH.
Collapse
Affiliation(s)
- Yanting Zhu
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yinxia Wu
- Wuhan University of Science and Technology, Wuhan, Hubei 430081, People's Republic of China
| | - Wenhua Shi
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jian Wang
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Xin Yan
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Qingting Wang
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Ya Liu
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Lan Yang
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Li Gao
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Manxiang Li
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi 710061, People's Republic of China.
| |
Collapse
|
5
|
Morphological Changes within the Rat Lateral Ventricle after the Administration of Proteasome Inhibitors. PLoS One 2015; 10:e0140536. [PMID: 26479862 PMCID: PMC4610704 DOI: 10.1371/journal.pone.0140536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/28/2015] [Indexed: 01/07/2023] Open
Abstract
The broad variety of substances that inhibit the action of the ubiquitin-proteasome system (UPS)—known as proteasome inhibitors—have been used extensively in previous studies, and they are currently frequently proposed as a novel form of cancer treatment and as a protective factor in intracerebral hemorrhage treatment. The experimental data on the safest route of proteasome inhibitor administration, their associated side effects, and the possible ways of minimizing these effects have recently become a very important topic. The aim of our present study was to determine the effects of administering of MG-132, lactacystin and epoxomicin, compounds belonging to three different classes of proteasome inhibitors, on the ependymal walls of the lateral ventricle. Observations were made 2 and 8 weeks after the intraventricular administration of the studied substances dissolved in dimethyl sulfoxide (DMSO) into the lateral ventricle of adult Wistar rats. Qualitative and quantitative analysis of brain sections stained with histochemical and inmmunofluorescence techniques showed that the administration of proteasome inhibitors caused a partial occlusion of the injected ventricle in all of the studied animals. The occlusion was due to ependymal cells damage and subsequent ependymal discontinuity, which caused direct contact between the striatum and the lateral nuclei of the septum, mononuclear cell infiltration and the formation of a glial scar between these structures (with the activation of astroglia, microglia and oligodendroglia). Morphologically, the ubiquitin-positive aggregates corresponded to aggresomes, indicating impaired activity of the UPS and the accumulation and aggregation of ubiquitinated proteins that coincided with the occurrence of glial scars. The most significant changes were observed in the wall covering the striatum in animals that were administered epoxomicin, and milder changes were observed in animals administered lactacystin and MG-132. Interestingly, DMSO administration also caused damage to some of the ependymal cells, but the aggresome-like structures were not formed. Our results indicate that all of the studied classes of proteasome inhibitors are detrimental to ependymal cells to some extent, and may cause severe changes in the ventricular system. The safety implications of their usage in therapeutic strategies to attenuate intracerebral hemorrhagic injury and in brain cancer treatment will require further studies.
Collapse
|
6
|
Tan JY, Chen JL, Huang X, Yuan CL. Screening and verification of proteins that interact with HSPC238. Oncol Rep 2015; 34:3097-103. [PMID: 26398855 DOI: 10.3892/or.2015.4289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 07/14/2015] [Indexed: 11/06/2022] Open
Abstract
HSPC238 is a recently identified tumor suppressor and demonstrates ubiquitin ligase E3 enzyme activity. HSPC238 was found to be significantly downregulated in human hepatocellular carcinoma (HCC) in vivo and to inhibit the proliferation and invasion of hepatoma cells in vitro; however, the underlying molecular mechanism is largely unknown. In the present study, we screened for and identified proteins that physically interact with HSPC238. A bait vector for yeast two-hybrid was constructed with human HSPC238 gene cDNA. Yeast two-hybrid screening was performed using a human fetal liver cDNA library. Multiple reporter gene assays, DNA sequencing and BLAST comparison analysis were performed on positive clones. Protein interaction of screened candidates with HSPC238 was further validated by confocal microscopy, co-immunoprecipitation and pull-down assays. Yeast two-hybrid screening demonstrated 124 positive clones. Multiple reporter gene assays with LacZ, HIS and ADE2 selective media identified 12 genes. Further co-localization, co-immunoprecipitation and pull-down assays demonstrated that HMOX1, RPS27A, ubiquitinB and MT2A interacted with HSPC238. These four proteins are involved in tumor development and progression, and are associated with the ubiquitin-proteasome pathway. Our results suggest that HSPC238 may play a tumor suppressor role and interact with these proteins via the ubiquitin-proteasome pathway. The identification and validation of proteins interacting with HSP238 may lead to the discovery of novel mechanisms through which HSPC238 suppresses tumorigenesis in human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jia-Yu Tan
- Central Intensive Care Unit, Bo'ai Hospital of Zhongshan City Affiliated with Southern Medical University, Zhongshan 528403, P.R. China
| | - Jing-Lin Chen
- Prenatal Diagnosis Center, Bo'ai Hospital of Zhongshan City Affiliated with Southern Medical University, Zhongshan 528403, P.R. China
| | - Xiang Huang
- Prenatal Diagnosis Center, Bo'ai Hospital of Zhongshan City Affiliated with Southern Medical University, Zhongshan 528403, P.R. China
| | - Chun-Lei Yuan
- Department of Laboratory Medicine, Bo'ai Hospital of Zhongshan City Affiliated with Southern Medical University, Zhongshan 528403, P.R. China
| |
Collapse
|
7
|
Estepa A, Coll J. Innate Multigene Family Memories Are Implicated in the Viral-Survivor Zebrafish Phenotype. PLoS One 2015; 10:e0135483. [PMID: 26270536 PMCID: PMC4535885 DOI: 10.1371/journal.pone.0135483] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/22/2015] [Indexed: 11/18/2022] Open
Abstract
Since adaptive features such as memory were discovered in mammalian innate immunity, interest in the immunological status of primitive vertebrates after infections has grown. In this context, we used zebrafish (Danio rerio), a primitive vertebrate species suited to molecular and genetic studies to explore transcriptional memories of the immune system in long-term survivors of viral haemorrhagic septicemia virus infections. Immune-gene targeted microarrays designed in-house, multipath genes, gene set enrichment, and leading-edge analysis, reveal unexpected consistent correlations between the viral-survivor phenotype and several innate multigene families. Thus, here we describe in survivors of infections the upregulation of the multigene family of proteasome subunit macropains, zebrafish-specific novel gene sets, mitogen activated protein kinases, and epidermal growth factor. We also describe the downregulation of the multigene families of c-reactive proteins, myxovirus-induced proteins and novel immunoglobulin-type receptors. The strength of those immunological memories was reflected by the exceptional similarity of the transcriptional profiles of survivors before and after re-infection compared with primary infected fish. On the other hand, the high levels of neutralizing antibodies in the blood plasma of survivors contrasted with the depletion of transcripts specific for most cell types present in lymphoid organs. Therefore, long-term survivors maintained unexpected molecular/cellular memories of previous viral encounters by modulating the expression levels of innate multigene families as well as having specific adaptive antibodies. The implications of the so-called "trained immunity" for future research in this field are also discussed.
Collapse
Affiliation(s)
- Amparo Estepa
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Elche (UMH), Alicante, Spain
| | - Julio Coll
- Department of Biotechnology, Instituto Nacional Investigaciones Agrarias (INIA), Madrid, Spain
| |
Collapse
|
8
|
Hu Z, Holzschuh J, Driever W. Loss of DDB1 Leads to Transcriptional p53 Pathway Activation in Proliferating Cells, Cell Cycle Deregulation, and Apoptosis in Zebrafish Embryos. PLoS One 2015. [PMID: 26225764 PMCID: PMC4520591 DOI: 10.1371/journal.pone.0134299] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
DNA damage-binding protein 1 (DDB1) is a large subunit of the heterodimeric DDB complex that recognizes DNA lesions and initiates the nucleotide excision repair process. DDB1 is also a component of the CUL4 E3 ligase complex involved in a broad spectrum of cellular processes by targeted ubiquitination of key regulators. Functions of DDB1 in development have been addressed in several model organisms, however, are not fully understood so far. Here we report an ENU induced mutant ddb1 allele (ddb1m863) identified in zebrafish (Danio rerio), and analyze its effects on development. Zebrafish ddb1 is expressed broadly, both maternally and zygotically, with enhanced expression in proliferation zones. The (ddb1m863 mutant allele affects the splice acceptor site of exon 20, causing a splicing defect that results in truncation of the 1140 amino acid protein after residue 800, lacking part of the β-propeller domain BPC and the C-terminal helical domain CTD. ddb1m863 zygotic mutant embryos have a pleiotropic phenotype, including smaller and abnormally shaped brain, head skeleton, eyes, jaw, and branchial arches, as well as reduced dopaminergic neuron groups. However, early forming tissues develop normally in zygotic ddb1m863 mutant embryos, which may be due to maternal rescue. In ddb1m863 mutant embryos, pcna-expressing proliferating cell populations were reduced, concurrent with increased apoptosis. We also observed a concomitant strong up-regulation of transcripts of the tumor suppressor p53 (tp53) and the cell cycle inhibitor cdkn1a (p21a/bCIP1/WAF1) in proliferating tissues. In addition, transcription of cyclin genes ccna2 and ccnd1 was deregulated in ddb1m863 mutants. Reduction of p53 activity by anti-sense morpholinos alleviated the apoptotic phenotype in ddb1m863 mutants. These results imply that Ddb1 may be involved in maintaining proper cell cycle progression and viability of dividing cells during development through transcriptional mechanisms regulating genes involved in cell cycle control and cell survival.
Collapse
Affiliation(s)
- Zhilian Hu
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany; Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, 48109-5646, United States of America
| | - Jochen Holzschuh
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany
| | - Wolfgang Driever
- Developmental Biology, Institute Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| |
Collapse
|
9
|
Dal Vechio FH, Cerqueira F, Augusto O, Lopes R, Demasi M. Peptides that activate the 20S proteasome by gate opening increased oxidized protein removal and reduced protein aggregation. Free Radic Biol Med 2014; 67:304-13. [PMID: 24291399 DOI: 10.1016/j.freeradbiomed.2013.11.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 11/01/2013] [Accepted: 11/18/2013] [Indexed: 12/22/2022]
Abstract
The proteasome is a multicatalytic protease that is responsible for the degradation of the majority of intracellular proteins. Its role is correlated with several major regulatory pathways that are involved in cell cycle control, signaling, and antigen presentation, as well as in the removal of oxidatively damaged proteins. Although several proteasomal catalytic inhibitors have been described, very few activators have been reported to date. Some reports in the literature highlight the cellular protective effects of proteasome activation against oxidative stress and its effect on increased life span. In this work, we describe a peptide named proteasome-activating peptide 1 (PAP1), which increases the chymotrypsin-like proteasomal catalytic activity and, consequently, proteolytic rates both in vitro and in culture. PAP1 proteasomal activation is mediated by the opening of the proteasomal catalytic chamber. We also demonstrate that the observed proteasomal activation protected cells from oxidative stress; further, PAP1 prevented protein aggregation in a cellular model of amyotrophic lateral sclerosis. The role of 20SPT gate opening underlying protection against oxidative stress was also explored in yeast cells. The present data indicate the importance of proteasomal activators as potential drugs for the treatment of pathologies associated with the impaired removal of damaged proteins, which is observed in many neurodegenerative diseases.
Collapse
Affiliation(s)
- Francisco H Dal Vechio
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, SP 05503-001, Brazil
| | - Fernanda Cerqueira
- Laboratório Especial de Toxinologia Aplicada, Instituto Butantan, São Paulo, SP 05503-001, Brazil
| | - Ohara Augusto
- Laboratório Especial de Toxinologia Aplicada, Instituto Butantan, São Paulo, SP 05503-001, Brazil
| | - Robson Lopes
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, SP 05503-001, Brazil.
| |
Collapse
|
10
|
|
11
|
Loss of Usp9x disrupts cortical architecture, hippocampal development and TGFβ-mediated axonogenesis. PLoS One 2013; 8:e68287. [PMID: 23861879 PMCID: PMC3702552 DOI: 10.1371/journal.pone.0068287] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 05/28/2013] [Indexed: 01/17/2023] Open
Abstract
The deubiquitylating enzyme Usp9x is highly expressed in the developing mouse brain, and increased Usp9x expression enhances the self-renewal of neural progenitors in vitro. USP9X is a candidate gene for human neurodevelopmental disorders, including lissencephaly, epilepsy and X-linked intellectual disability. To determine if Usp9x is critical to mammalian brain development we conditionally deleted the gene from neural progenitors, and their subsequent progeny. Mating Usp9xloxP/loxP mice with mice expressing Cre recombinase from the Nestin promoter deleted Usp9x throughout the entire brain, and resulted in early postnatal lethality. Although the overall brain architecture was intact, loss of Usp9x disrupted the cellular organization of the ventricular and sub-ventricular zones, and cortical plate. Usp9x absence also led to dramatic reductions in axonal length, in vivo and in vitro, which could in part be explained by a failure in Tgf-β signaling. Deletion of Usp9x from the dorsal telencephalon only, by mating with Emx1-cre mice, was compatible with survival to adulthood but resulted in reduction or loss of the corpus callosum, a dramatic decrease in hippocampal size, and disorganization of the hippocampal CA3 region. This latter phenotypic aspect resembled that observed in Doublecortin knock-out mice, which is an Usp9x interacting protein. This study establishes that Usp9x is critical for several aspects of CNS development, and suggests that its regulation of Tgf-β signaling extends to neurons.
Collapse
|
12
|
Granese B, Scala I, Spatuzza C, Valentino A, Coletta M, Vacca RA, De Luca P, Andria G. Validation of microarray data in human lymphoblasts shows a role of the ubiquitin-proteasome system and NF-kB in the pathogenesis of Down syndrome. BMC Med Genomics 2013; 6:24. [PMID: 23830204 PMCID: PMC3717290 DOI: 10.1186/1755-8794-6-24] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 06/29/2013] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Down syndrome (DS) is a complex disorder caused by the trisomy of either the entire, or a critical region of chromosome 21 (21q22.1-22.3). Despite representing the most common cause of mental retardation, the molecular bases of the syndrome are still largely unknown. METHODS To better understand the pathogenesis of DS, we analyzed the genome-wide transcription profiles of lymphoblastoid cell lines (LCLs) from six DS and six euploid individuals and investigated differential gene expression and pathway deregulation associated with trisomy 21. Connectivity map and PASS-assisted exploration were used to identify compounds whose molecular signatures counteracted those of DS lymphoblasts and to predict their therapeutic potential. An experimental validation in DS LCLs and fetal fibroblasts was performed for the most deregulated GO categories, i.e. the ubiquitin mediated proteolysis and the NF-kB cascade. RESULTS We show, for the first time, that the level of protein ubiquitination is reduced in human DS cell lines and that proteasome activity is increased in both basal conditions and oxidative microenvironment. We also provide the first evidence that NF-kB transcription levels, a paradigm of gene expression control by ubiquitin-mediated degradation, is impaired in DS due to reduced IkB-alfa ubiquitination, increased NF-kB inhibitor (IkB-alfa) and reduced p65 nuclear fraction. Finally, the DSCR1/DYRK1A/NFAT genes were analysed. In human DS LCLs, we confirmed the presence of increased protein levels of DSCR1 and DYRK1A, and showed that the levels of the transcription factor NFATc2 were decreased in DS along with a reduction of its nuclear translocation upon induction of calcium fluxes. CONCLUSIONS The present work offers new perspectives to better understand the pathogenesis of DS and suggests a rationale for innovative approaches to treat some pathological conditions associated to DS.
Collapse
Affiliation(s)
- Barbara Granese
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| | - Iris Scala
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| | - Carmen Spatuzza
- Department of Biotechnological Sciences, Federico II University, Naples 80131, Italy
| | - Anna Valentino
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| | - Marcella Coletta
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes and Bioenergetics, National Council of Research, Bari 70126, Italy
| | - Pasquale De Luca
- Stazione Zoologica “A. Dohrn”, c/o BioGeM, Via Camporeale, Ariano Irpino 83031, Italy
| | - Generoso Andria
- Department of Pediatrics, Federico II University, Naples 80131, Italy
| |
Collapse
|
13
|
Bennett EJ, Mayor T. Proteomics propels protein degradation studies in San Diego. Mol Cell Proteomics 2012; 11:1523-8. [PMID: 22798279 DOI: 10.1074/mcp.o112.020008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Exquisite in vitro biochemical examinations of protein ubiquitylation and degradation have historically been the dominant methods for unraveling the mechanisms of protein destruction. The study of protein abundance alterations and protein modifications, a cornerstone of protein degradation pathways, naturally lends itself to global and systematic proteomic methods to decipher the emerging complexity of protein degradation pathways. Advances in proteomic technologies have fueled an explosion of systematic and quantitative studies aimed at understanding how the proteome is shaped and regulated by ubiquitin-dependent processes. These types of studies, as well as targeted analyses of cellular pathways, have revealed that alterations in protein degradation function can have a severe impact on human health and disease. The fusion of these two themes was the focus of the January 2012 conference on proteomics of protein degradation and ubiquitin pathways (PPDUP) held in San Diego. To gain insights into both the current state-of-the-art proteomic methods to investigate protein turnover, and how protein degradation function is altered within a range of human disorders a variety of speakers revealed the many connections between altered protein degradation function and human disease. Many of the sessions were framed by a consistent focus aimed at the discovery and development of novel therapeutics targeting protein degradation pathway components to treat various human maladies ranging from cancer to heart disease.
Collapse
Affiliation(s)
- Eric J Bennett
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA.
| | | |
Collapse
|
14
|
Tanabe F, Yone K, Kawabata N, Sakakima H, Matsuda F, Ishidou Y, Maeda S, Abematsu M, Komiya S, Setoguchi T. Accumulation of p62 in degenerated spinal cord under chronic mechanical compression: functional analysis of p62 and autophagy in hypoxic neuronal cells. Autophagy 2012; 7:1462-71. [PMID: 22082874 DOI: 10.4161/auto.7.12.17892] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Intracellular accumulation of altered proteins, including p62 and ubiquitinated proteins, is the basis of most neurodegenerative disorders. The relationship among the accumulation of altered proteins, autophagy, and spinal cord dysfunction by cervical spondylotic myelopathy has not been clarified. We examined the expression of p62 and autophagy markers in the chronically compressed spinal cord of tiptoe-walking Yoshimura mice. In addition, we examined the expression and roles of p62 and autophagy in hypoxic neuronal cells. Western blot analysis showed the accumulation of p62, ubiquitinated proteins, and microtubule-associated protein 1 light chain 3 (LC3), an autophagic marker, in the compressed spinal cord. Immunohistochemical examinations showed that p62 accumulated in neurons, axons, astrocytes, and oligodendrocytes. Electron microscopy showed the expression of autophagy markers, including autolysosomes and autophagic vesicles, in the compressed spinal cord. These findings suggest the presence of p62 and autophagy in the degenerated compressed spinal cord. Hypoxic stress increased the expression of p62, ubiquitinated proteins, and LC3-II in neuronal cells. In addition, LC3 turnover assay and GFP-LC3 cleavage assay showed that hypoxic stress increased autophagy flux in neuronal cells. These findings suggest that hypoxic stress induces accumulation of p62 and autophagy in neuronal cells. The forced expression of p62 decreased the number of neuronal cells under hypoxic stress. These findings suggest that p62 accumulation under hypoxic stress promotes neuronal cell death. Treatment with 3-methyladenine, an autophagy inhibitor decreased the number of neuronal cells, whereas lithium chloride, an autophagy inducer increased the number of cells under hypoxic stress. These findings suggest that autophagy promotes neuronal cell survival under hypoxic stress. Our findings suggest that pharmacological inducers of autophagy may be useful for treating cervical spondylotic myelopathy patients.
Collapse
Affiliation(s)
- Fumito Tanabe
- Department of Orthopaedic Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Kagoshima Prefecture, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Silva GM, Netto LES, Simões V, Santos LFA, Gozzo FC, Demasi MAA, Oliveira CLP, Bicev RN, Klitzke CF, Sogayar MC, Demasi M. Redox control of 20S proteasome gating. Antioxid Redox Signal 2012; 16:1183-94. [PMID: 22229461 PMCID: PMC3324812 DOI: 10.1089/ars.2011.4210] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED The proteasome is the primary contributor in intracellular proteolysis. Oxidized or unstructured proteins can be degraded via a ubiquitin- and ATP-independent process by the free 20S proteasome (20SPT). The mechanism by which these proteins enter the catalytic chamber is not understood thus far, although the 20SPT gating conformation is considered to be an important barrier to allowing proteins free entrance. We have previously shown that S-glutathiolation of the 20SPT is a post-translational modification affecting the proteasomal activities. AIMS The goal of this work was to investigate the mechanism that regulates 20SPT activity, which includes the identification of the Cys residues prone to S-glutathiolation. RESULTS Modulation of 20SPT activity by proteasome gating is at least partially due to the S-glutathiolation of specific Cys residues. The gate was open when the 20SPT was S-glutathiolated, whereas following treatment with high concentrations of dithiothreitol, the gate was closed. S-glutathiolated 20SPT was more effective at degrading both oxidized and partially unfolded proteins than its reduced form. Only 2 out of 28 Cys were observed to be S-glutathiolated in the proteasomal α5 subunit of yeast cells grown to the stationary phase in glucose-containing medium. INNOVATION We demonstrate a redox post-translational regulatory mechanism controlling 20SPT activity. CONCLUSION S-glutathiolation is a post-translational modification that triggers gate opening and thereby activates the proteolytic activities of free 20SPT. This process appears to be an important regulatory mechanism to intensify the removal of oxidized or unstructured proteins in stressful situations by a process independent of ubiquitination and ATP consumption. Antioxid. Redox Signal. 16, 1183-1194.
Collapse
Affiliation(s)
- Gustavo M Silva
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brasil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ramachandran U, Manavalan A, Sundaramurthi H, Sze SK, Feng ZW, Hu JM, Heese K. Tianma modulates proteins with various neuro-regenerative modalities in differentiated human neuronal SH-SY5Y cells. Neurochem Int 2012; 60:827-36. [PMID: 22710396 DOI: 10.1016/j.neuint.2012.03.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/29/2012] [Accepted: 03/19/2012] [Indexed: 12/15/2022]
Abstract
Tianma (Rhizoma gastrodiae) is the dried rhizome of the plant Gastrodia elata Blume (Orchidaceae family). As a medicinal herb in traditional Chinese medicine (TCM) its functions are to control convulsions, pain, headache, dizziness, vertigo, seizure, epilepsy and others. In addition, tianma is frequently used for the treatment of neurodegenerative disorders though the mechanism of action is widely unknown. Accordingly, this study was designed to examine the effects of tianma on the proteome metabolism in differentiated human neuronal SH-SY5Y cells to explore its specific effects on neuronal signaling pathways. Using an iTRAQ (isobaric tags for relative and absolute quantitation)-based proteomics research approach, we identified 2390 modulated proteins, out of which 406 were found to be altered by tianma in differentiated human neuronal SH-SY5Y cells. Based on the observed data, we hypothesize that tianma promotes neuro-regenerative signaling cascades by controlling chaperone/proteasomal degradation pathways (e.g. CALR, FKBP3/4, HSP70/90) and mobilizing neuro-protective genes (such as AIP5) as well as modulating other proteins (RTN1/4, NCAM, PACSIN2, and PDLIM1/5) with various regenerative modalities and capacities related to neuro-synaptic plasticity.
Collapse
|
17
|
Dysregulation of gene expression in a lysosomal storage disease varies between brain regions implicating unexpected mechanisms of neuropathology. PLoS One 2012; 7:e32419. [PMID: 22403656 PMCID: PMC3293807 DOI: 10.1371/journal.pone.0032419] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/30/2012] [Indexed: 12/19/2022] Open
Abstract
The characteristic neurological feature of many neurogenetic diseases is intellectual disability. Although specific neuropathological features have been described, the mechanisms by which specific gene defects lead to cognitive impairment remain obscure. To gain insight into abnormal functions occurring secondary to a single gene defect, whole transcriptome analysis was used to identify molecular and cellular pathways that are dysregulated in the brain in a mouse model of a lysosomal storage disorder (LSD) (mucopolysaccharidosis [MPS] VII). We assayed multiple anatomical regions separately, in a large cohort of normal and diseased mice, which greatly increased the number of significant changes that could be detected compared to past studies in LSD models. We found that patterns of aberrant gene expression and involvement of multiple molecular and cellular systems varied significantly between brain regions. A number of changes revealed unexpected system and process alterations, such as up-regulation of the immune system with few inflammatory changes (a significant difference from the closely related MPS IIIb model), down-regulation of major oligodendrocyte genes even though white matter changes are not a feature histopathologically, and a plethora of developmental gene changes. The involvement of multiple neural systems indicates that the mechanisms of neuropathology in this type of disease are much broader than previously appreciated. In addition, the variation in gene dysregulation between brain regions indicates that different neuropathologic mechanisms may predominate within different regions of a diseased brain caused by a single gene mutation.
Collapse
|
18
|
Protein microarrays for the identification of praja1 e3 ubiquitin ligase substrates. Cell Biochem Biophys 2011; 60:127-35. [PMID: 21461837 DOI: 10.1007/s12013-011-9180-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Although they are the primary determinants of substrate specificity, few E3-substrate pairs have been positively identified, and few E3's profiled in a proteomic fashion. Praja1 is an E3 implicated in bone development and highly expressed in brain. Although it has been well studied relative to the majority of E3's, little is known concerning the repertoire of proteins it ubiquitylates. We sought to identify high confidence substrates for Praja1 from an unbiased proteomic profile of thousands of human proteins using protein microarrays. We first profiled Praja1 activity against a panel of E2's to identify its optimal partner in vitro. We then ubiquitylated multiple, identical protein arrays and detected putative substrates with reagents that vary in ubiquitin recognition according to the extent of chain formation. Gene ontology clustering identified putative substrates consistent with information previously known about Praja1 function, and provides clues into novel aspects of this enzyme's function.
Collapse
|
19
|
Inhibition of ubiquitin proteasome function suppresses proliferation of pulmonary artery smooth muscle cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2011; 384:517-23. [PMID: 21850573 DOI: 10.1007/s00210-011-0678-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 05/10/2011] [Indexed: 10/24/2022]
Abstract
Inhibition of proteasome function has been shown to suppress several types of cells proliferation; this study investigates whether this also occurs in pulmonary artery smooth muscle cells (PASMCs) and its potential mechanisms. Serotonin induced 4.27-fold increase in DNA synthesis in PASMCs, and this effect was dose-dependently blocked by prior incubation of cells with MG132, a specific proteasome inhibitor. Inhibition of proteasome function did not modulate serotonin-triggered pro-proliferation signaling pathways, such as extracellular signal-regulated mitogen-activated protein kinase (ERK1/2 MAPK) and Ras homolog gene family member A (RhoA). Further study indicated that treatment of PASMCs with serotonin reduced p21(WAF1) protein level but not its transcription; this was reversed by inhibiting ERK1/2 MAPK or RhoA cascade equally. In addition, MG132 increased the protein level of p21(WAF1) in a dose-dependent manner in the presence of serotonin, 10 μM MG132 led to a 4.2-fold increase in p21(WAF1) protein level, and this effect was not mediated by increasing p21(WAF1) mRNA level. More importantly, cell lacking p21(WAF1) by siRNA transfection abolished the inhibitive effect of MG132 on cells proliferation. Our study suggests that accumulation of p21(WAF1) protein level caused by proteasome inhibition particularly mediated its inhibitive effect on PASMCs proliferation, and inhibition of proteasome function might have potential value in the treatment of pulmonary hypertension.
Collapse
|
20
|
Dysfunction of the ubiquitin-proteasome system in the cerebellum of aging Ts65Dn mice. Exp Neurol 2011; 232:114-8. [PMID: 21867700 DOI: 10.1016/j.expneurol.2011.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 07/22/2011] [Accepted: 08/08/2011] [Indexed: 01/09/2023]
Abstract
In the cerebellum of adult-aging Ts65Dn mice, a murine model of Down syndrome, Purkinje cells undergo degeneration. Searching for the cause of Purkinje cell degeneration, we have studied the ubiquitin-proteasome system (UPS) in the cerebellum of aging Ts65Dn mice. Inhibition of UPS is sufficient to induce neuron degeneration and death. Proteasome chymotrypsin-like proteolytic activity was reduced by 35% in the cerebellum of Ts65Dn mice in comparison with euploid animals. Accordingly, Western blot analysis of ubiquitin showed an increase in ubiquitinated proteins. Immunocytochemistry for ubiquitin revealed strongly positive intranuclear inclusions in Purkinje cells and large neurons of cerebellar nuclei. The Western blot analysis of ubiquitin in nuclear protein extracts confirmed the increase of ubiquitinated proteins in the cell nuclei. After FUS immunocytochemistry, large intranuclear inclusions were visible in Purkinje cells and large neurons of cerebellar nuclei in Ts65Dn mice. Together, data indicate a possible role for proteasome inhibition in the cerebellar neurodegeneration in Ts65Dn mice.
Collapse
|
21
|
Ito Y, Shibata N, Saito K, Kobayashi M, Osawa M. New insights into the pathogenesis of spinal muscular atrophy. Brain Dev 2011; 33:321-31. [PMID: 20605078 DOI: 10.1016/j.braindev.2010.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2009] [Revised: 06/10/2010] [Accepted: 06/12/2010] [Indexed: 12/01/2022]
Abstract
To clarify the pathomechanism of spinal muscular atrophy (SMA) with mutations in the gene for survival motor neuron (SMN) protein, postmortem neuropathological analyses were performed on spinal cords obtained at autopsy from 2 fetuses with SMA, 5 infants and a low teenager with SMA type 1, and a higher teenager with SMA type 2; the diagnosis of all of them was confirmed clinically and genetically. Histopathologically, it was noted that lower motor neurons (LMNs) in the SMA cases showed immature profiles characterized by fine Nissl bodies restricted to the periphery of small round somata with a few cell processes in the fetal period, and showed small-sized profiles in the postnatal period. LMNs began to reduce in size and number in the fetal period, ballooned neurons (BNs) appeared postnatally, and the remaining LMNs including BNs diminished with age. BNs were filled with phosphorylated neurofilament protein, and morphologically similar to but smaller than typical chromatolytic neurons as axonal reaction. The population of survived LMNs was relatively preserved in an SMA type 2 case, who lived to 17-year-old, as compared to SMA type 1 cases. Immunohistochemical analysis demonstrated expression of Bcl-2, Bax, activated caspase-3 and SMN in the LMNs prominent in the fetal cases. There was no significant difference in staining for these substances between the control and SMA cases. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay revealed no significant signal in the control and SMA cases. Given that downregulation of SMN leads to a failure in neurite outgrowth and neuromuscular contact of LMNs, the present results suggest the involvement of a fetal developmental maturation error as well as a postnatal retrograde dying-back degeneration of LMNs in SMN-mutated SMA.
Collapse
Affiliation(s)
- Yasushi Ito
- Department of Pediatrics, School of Medicine, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
22
|
Chen MJ, Peng ZF, Manikandan J, Melendez AJ, Tan GS, Chung CM, Li QT, Tan TM, Deng LW, Whiteman M, Beart PM, Moore PK, Cheung NS. Gene profiling reveals hydrogen sulphide recruits death signaling via the N-methyl-D-aspartate receptor identifying commonalities with excitotoxicity. J Cell Physiol 2011; 226:1308-22. [DOI: 10.1002/jcp.22459] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
23
|
Proteasome and oxidative phoshorylation changes may explain why aging is a risk factor for neurodegenerative disorders. J Proteomics 2010; 73:2230-8. [DOI: 10.1016/j.jprot.2010.08.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 08/24/2010] [Accepted: 08/25/2010] [Indexed: 01/17/2023]
|