1
|
Suzich JB, Cuddy SR, Baidas H, Dochnal S, Ke E, Schinlever AR, Babnis A, Boutell C, Cliffe AR. PML-NB-dependent type I interferon memory results in a restricted form of HSV latency. EMBO Rep 2021; 22:e52547. [PMID: 34197022 PMCID: PMC8419685 DOI: 10.15252/embr.202152547] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 01/23/2023] Open
Abstract
Herpes simplex virus (HSV) establishes latent infection in long-lived neurons. During initial infection, neurons are exposed to multiple inflammatory cytokines but the effects of immune signaling on the nature of HSV latency are unknown. We show that initial infection of primary murine neurons in the presence of type I interferon (IFN) results in a form of latency that is restricted for reactivation. We also find that the subnuclear condensates, promyelocytic leukemia nuclear bodies (PML-NBs), are absent from primary sympathetic and sensory neurons but form with type I IFN treatment and persist even when IFN signaling resolves. HSV-1 genomes colocalize with PML-NBs throughout a latent infection of neurons only when type I IFN is present during initial infection. Depletion of PML prior to or following infection does not impact the establishment latency; however, it does rescue the ability of HSV to reactivate from IFN-treated neurons. This study demonstrates that viral genomes possess a memory of the IFN response during de novo infection, which results in differential subnuclear positioning and ultimately restricts the ability of genomes to reactivate.
Collapse
Affiliation(s)
- Jon B Suzich
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Sean R Cuddy
- Neuroscience Graduate ProgramUniversity of VirginiaCharlottesvilleVAUSA
| | - Hiam Baidas
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Sara Dochnal
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Eugene Ke
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Austin R Schinlever
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Aleksandra Babnis
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Chris Boutell
- MRC‐University of Glasgow Centre for Virus Research (CVR)GlasgowUK
| | - Anna R Cliffe
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| |
Collapse
|
2
|
Zhang Y, Shao AW, Tang J, Geng Y. PML-II recruits ataxin-3 to PML-NBs and inhibits its deubiquitinating activity. Biochem Biophys Res Commun 2021; 554:186-192. [PMID: 33798946 DOI: 10.1016/j.bbrc.2021.03.098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/18/2021] [Indexed: 12/28/2022]
Abstract
Promyelocytic leukemia protein (PML) nuclear bodies (NBs) are dynamic and multiprotein complexes implicated in a variety of important biochemical events. Due to alternative mRNA splicing, PML has at least six nuclear isoforms that share a common N-terminus but differ in their C-terminal regions. However, the unique role of each PML isoform is not clear. Here, we report the characterization of the deubiquitinase ataxin-3 as a specific binding partner of PML isoform II (PML-II). Ataxin-3 was identified as a potential binding protein of PML-II in a yeast-hybrid screen employing the unique C-terminal region of PML-II as bait. Ataxin-3 only binds to the C-terminal region of PML-II and not that of other PML isoforms. The interaction between ataxin-3 and PML-II was confirmed by co-immunoprecipition assays, and immunofluorescent microscopy revealed that PML-II and ataxin-3 were co-localized in PML-NBs. In addition, PML-II not only interacts with ataxin-3 with a normal range of poly-Q repeats (13Q), but also with a pathological form of ataxin-3 with extended poly-Q repeats (79Q). Importantly, the deubiquitinase activity of ataxin-3 was inhibited by PML-II. Our results suggest that PML-II may be a negative regulator of ataxin-3.
Collapse
Affiliation(s)
- Ying Zhang
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - An-Wen Shao
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jun Tang
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Yunyun Geng
- Hebei University of Chinese Medicine, And Heibei Key Laboratory of Chinese Medicine Research on Cardiocerebrovascular Disease, Shijiazhuang, Hebei, 050000, China.
| |
Collapse
|
3
|
Epstein I, Finkbeiner S. The Arc of cognition: Signaling cascades regulating Arc and implications for cognitive function and disease. Semin Cell Dev Biol 2018; 77:63-72. [PMID: 29559111 DOI: 10.1016/j.semcdb.2017.09.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022]
Abstract
The activity-regulated cytoskeletal (Arc) gene is implicated in numerous synaptic plasticity paradigms, including long-term potentiation and depression and homeostatic plasticity, and is critical for consolidating memory. How Arc facilitates these forms of plasticity is not fully understood. Unlike other neuronal immediate-early genes, Arc encodes a protein that shuttles between the somatodendritic and nuclear compartments to regulate synaptic plasticity. Little attention has been paid to Arc's role in the nucleus. Here, we highlight the regulatory elements and signaling cascades required to induce Arc transcription and discuss the significance of Arc nuclear localization for synaptic plasticity and scaling. We integrate these findings into the context of cognitive function and disease and propose a model in which Arc mediates an effect on memory as a "chaser" of synaptic activity through homeostatic scaling.
Collapse
Affiliation(s)
- Irina Epstein
- Gladstone Institutes,1650 Owens Street, San Francisco, CA 94158, USA.
| | - Steven Finkbeiner
- Gladstone Institutes,1650 Owens Street, San Francisco, CA 94158, USA; Departments of Neurology and Physiology, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
4
|
Nakano Y, Takahashi-Fujigasaki J, Sengoku R, Kanemaru K, Arai T, Kanda T, Murayama S. PML Nuclear Bodies Are Altered in Adult-Onset Neuronal Intranuclear Hyaline Inclusion Disease. J Neuropathol Exp Neurol 2017; 76:585-594. [PMID: 28863453 DOI: 10.1093/jnen/nlx039] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Neuronal intranuclear hyaline inclusion disease (NIHID) is a neurodegenerative disorder characterized by the presence of eosinophilic nuclear inclusions (NIs) in diverse cell lines in systemic organs. Adult-onset NIHID typically manifests with dementia associated with leukoencephalopathy. The detection of NIs in skin biopsies is useful for an antemortem diagnosis. A previous analysis suggested that NIs in NIHID originated from nuclear bodies (NBs), an important nuclear domain related to the ubiquitin-p62-mediated protein degradation system. In this study, we analyzed skin samples from 5 NIHID and 5 control cases immunohistochemically and electron microscopically. In the control cases, small but significant amounts of ubiquitin- and p62-positive intranuclear structures were found. These structures were consistently colocalized with promyelocytic leukemia protein (PML), an essential component of NBs, in particular when activated. The p62- and PML-positive structures were more frequently found in NIHID cases. Activated NBs, having a core and a shell, were observed by electron microscopy in control but not in NIHID cases. Instead, immature and mature filamentous NIs were found only in the NIHID cases. Our results indicate that NBs could not be normally activated in the NIHID, and an abnormal alteration of NBs might be related to the pathogenesis of NIHID.
Collapse
Affiliation(s)
- Yuta Nakano
- Brain Bank for Aging Research, Department of Neuropathology, Department of Neurology, and Department of Pathology; Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology, Tokyo, Japan; Department of Neurology and Clinical Neuroscience; and Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Junko Takahashi-Fujigasaki
- Brain Bank for Aging Research, Department of Neuropathology, Department of Neurology, and Department of Pathology; Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology, Tokyo, Japan; Department of Neurology and Clinical Neuroscience; and Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Renpei Sengoku
- Brain Bank for Aging Research, Department of Neuropathology, Department of Neurology, and Department of Pathology; Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology, Tokyo, Japan; Department of Neurology and Clinical Neuroscience; and Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Kazutomi Kanemaru
- Brain Bank for Aging Research, Department of Neuropathology, Department of Neurology, and Department of Pathology; Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology, Tokyo, Japan; Department of Neurology and Clinical Neuroscience; and Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Tomio Arai
- Brain Bank for Aging Research, Department of Neuropathology, Department of Neurology, and Department of Pathology; Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology, Tokyo, Japan; Department of Neurology and Clinical Neuroscience; and Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Takashi Kanda
- Brain Bank for Aging Research, Department of Neuropathology, Department of Neurology, and Department of Pathology; Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology, Tokyo, Japan; Department of Neurology and Clinical Neuroscience; and Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Shigeo Murayama
- Brain Bank for Aging Research, Department of Neuropathology, Department of Neurology, and Department of Pathology; Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology, Tokyo, Japan; Department of Neurology and Clinical Neuroscience; and Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| |
Collapse
|
5
|
Takahashi-Fujigasaki J, Nakano Y, Uchino A, Murayama S. Adult-onset neuronal intranuclear hyaline inclusion disease is not rare in older adults. Geriatr Gerontol Int 2017; 16 Suppl 1:51-6. [PMID: 27018283 DOI: 10.1111/ggi.12725] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2015] [Indexed: 12/29/2022]
Abstract
Neuronal intranuclear hyaline inclusion disease (NIHID) is a rare neurodegenerative disorder pathologically characterized by localized neuronal loss, and the presence of eosinophilic intranuclear inclusions in neurons and glial cells. NIHID is a heterogeneous disease entity. It is divided into three clinical subgroups: infantile, juvenile and adult forms. Recently, reports of adult-onset cases have increased. Typical adult-onset NIHID consists of cognitive dysfunction with leukoencephalopathy. This type of adult-onset NIHID can be predicted by characteristic magnetic resonance images, high intensity areas on T2-weighted/fluid-attenuated inversion recovery images and persistent high intensity at the corticomedullary junction in diffusion-weighted images. When clinically suspected, the ante-mortem diagnosis can be made by biopsy. In adult-onset NIHID, nuclear inclusions are found more frequently in glial cells, and moderate to severe white matter degeneration is often associated. Although the underlying pathological mechanisms of NIHID are largely unknown, abnormal intranuclear accumulations of proteins and/or dysfunction of protein degradation systems might be related to the pathogenesis. To further clarify the characteristics of this disease entity, biological and pathological analysis of the patients is indispensable. As this disease entity becomes better known, diagnosed cases are expected to increase. Adult-onset NIHID might not be as extremely rare as previously thought.
Collapse
Affiliation(s)
- Junko Takahashi-Fujigasaki
- Department of Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yuta Nakano
- Bioresource Center for Aging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Akiko Uchino
- Department of Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shigeo Murayama
- Department of Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Bioresource Center for Aging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Neurology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
6
|
Abstract
Cajal is commonly regarded as the father of modern neuroscience in recognition of his fundamental work on the structure of the nervous system. But Cajal also made seminal contributions to the knowledge of nuclear structure in the early 1900s, including the discovery of the "accessory body" later renamed "Cajal body" (CB). This important nuclear structure has emerged as a center for the assembly of ribonucleoproteins (RNPs) required for splicing, ribosome biogenesis and telomere maintenance. The modern era of CB research started in the 1990s with the discovery of coilin, now known as a scaffold protein of CBs, and specific probes for small nuclear RNAs (snRNAs). In this review, we summarize what we have learned in the recent decades concerning CBs in post-mitotic neurons, thereby ruling out dynamic changes in CB functions during the cell cycle. We show that CBs are particularly prominent in neurons, where they frequently associate with the nucleolus. Neuronal CBs are transcription-dependent nuclear organelles. Indeed, their number dynamically accommodates to support the high neuronal demand for splicing and ribosome biogenesis required for sustaining metabolic and bioelectrical activity. Mature neurons have canonical CBs enriched in coilin, survival motor neuron protein and snRNPs. Disruption and loss of neuronal CBs associate with severe neuronal dysfunctions in several neurological disorders such as motor neuron diseases. In particular, CB depletion in motor neurons seems to reflect a perturbation of transcription and splicing in spinal muscular atrophy, the most common genetic cause of infant mortality.
Collapse
Affiliation(s)
- Miguel Lafarga
- a Departamento de Anatomía y Biología Celular and "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)" , Universidad de Cantabria-IDIVAL , Santander , Spain
| | - Olga Tapia
- a Departamento de Anatomía y Biología Celular and "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)" , Universidad de Cantabria-IDIVAL , Santander , Spain
| | - Ana M Romero
- a Departamento de Anatomía y Biología Celular and "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)" , Universidad de Cantabria-IDIVAL , Santander , Spain
| | - Maria T Berciano
- a Departamento de Anatomía y Biología Celular and "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)" , Universidad de Cantabria-IDIVAL , Santander , Spain
| |
Collapse
|
7
|
Abstract
In aging societies increasing cases of neurodegenerative protein deposit diseases urge for the identification of the underlying mechanisms. Expectations are that in 2050 the percentage of population over age 60 is 42% in Japan, 34% in China, and 27% in the US. The cell nucleus is a major target of amyloid-like protein fibrillation in a variety of disorders that are characterized by widespread aggregation of proteins with instable homopolymeric amino acid repeats, ubiquitin, and other proteinaceous components. Additionally, accumulation of insoluble, SDS-resistant proteins has been identified as an intrinsic property of organismal aging. This review collects current knowledge about the composition and function of insoluble, nuclear protein inclusions from the protein homeostasis perspective. It discusses the occurrence and role of nuclear amyloid in the diseased as well as the healthy cell. Features of nuclear inclusions such as protein composition and locally active protein degradation may predict neural fitness and survival in a variety of health or disease settings.
Collapse
Affiliation(s)
- Anna von Mikecz
- a IUF - Leibniz Research Institute for Environmental Medicine at Heinrich-Heine-University; Duesseldorf, Germany
| |
Collapse
|
8
|
Singh R, Brewer MK, Mashburn CB, Lou D, Bondada V, Graham B, Geddes JW. Calpain 5 is highly expressed in the central nervous system (CNS), carries dual nuclear localization signals, and is associated with nuclear promyelocytic leukemia protein bodies. J Biol Chem 2014; 289:19383-94. [PMID: 24838245 PMCID: PMC4094050 DOI: 10.1074/jbc.m114.575159] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/14/2014] [Indexed: 12/20/2022] Open
Abstract
Calpain 5 (CAPN5) is a non-classical member of the calpain family. It lacks the EF hand motif characteristic of classical calpains but retains catalytic and Ca(2+) binding domains, and it contains a unique C-terminal domain. TRA-3, an ortholog of CAPN5, has been shown to be involved in necrotic cell death in Caenorhabditis elegans. CAPN5 is expressed throughout the CNS, but its expression relative to other calpains and subcellular distribution has not been investigated previously. Based on relative mRNA levels, Capn5 is the second most highly expressed calpain in the rat CNS, with Capn2 mRNA being the most abundant. Unlike classical calpains, CAPN5 is a non-cytosolic protein localized to the nucleus and extra-nuclear locations. CAPN5 possesses two nuclear localization signals (NLS): an N-terminal monopartite NLS and a unique bipartite NLS closer to the C terminus. The C-terminal NLS contains a SUMO-interacting motif that contributes to nuclear localization, and mutation or deletion of both NLS renders CAPN5 exclusively cytosolic. Dual NLS motifs are common among transcription factors. Interestingly, CAPN5 is found in punctate domains associated with promyelocytic leukemia (PML) protein within the nucleus. PML nuclear bodies are implicated in transcriptional regulation, cell differentiation, cellular response to stress, viral defense, apoptosis, and cell senescence as well as protein sequestration, modification, and degradation. The roles of nuclear CAPN5 remain to be determined.
Collapse
Affiliation(s)
- Ranjana Singh
- From the Spinal Cord and Brain Injury Research Center and the Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky 40536
| | | | | | - Dingyuan Lou
- From the Spinal Cord and Brain Injury Research Center and
| | - Vimala Bondada
- From the Spinal Cord and Brain Injury Research Center and
| | | | - James W Geddes
- From the Spinal Cord and Brain Injury Research Center and the Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
9
|
Miki Y, Mori F, Kon T, Tanji K, Toyoshima Y, Yoshida M, Sasaki H, Kakita A, Takahashi H, Wakabayashi K. Accumulation of the sigma-1 receptor is common to neuronal nuclear inclusions in various neurodegenerative diseases. Neuropathology 2014; 34:148-58. [PMID: 24313828 DOI: 10.1111/neup.12080] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/23/2013] [Accepted: 10/28/2013] [Indexed: 01/02/2023]
Abstract
The sigma-1 receptor (SIGMAR1) is now known to be one of the endoplasmic reticulum (ER) chaperones, which participate in the degradation of misfolded proteins in cells via the ER-related degradation machinery linked to the ubiquitin-proteasome pathway. Mutations of the SIGMAR1 gene are implicated in the pathogenesis of familial frontotemporal lobar degeneration and motor neuron disease. Involvement of ER dysfunction in the formation of inclusion bodies in various neurodegenerative diseases has also become evident. We performed immunohistochemical staining to clarify the localization of SIGMAR1 in the brains of patients with neurodegenerative disorders, including trans-activation response DNA protein 43 (TDP-43) proteinopathy, tauopathy, α-synucleinopathy, polyglutamine disease and intranuclear inclusion body disease (INIBD). Double-immunocytofluorescence and Western blot analyses of cultured cells were also performed to investigate the role of SIGMAR1 using a specific exportin 1 inhibitor, leptomycin B and an ER stress inducer, thapsigargin. SIGMAR1 was consistently shown to be co-localized with neuronal nuclear inclusions in TDP-43 proteinopathy, five polyglutamine diseases and INIBD, as well as in intranuclear Marinesco bodies in aged normal controls. Cytoplasmic inclusions in neurons and glial cells were unreactive for SIGMAR1. In cultured cells, immunocytofluorescent study showed that leptomycin B and thapsigargin were shown to sequester SIGMAR1 within the nucleus, acting together with p62. This finding was also supported by immunoblot analysis. These results indicate that SIGMAR1 might shuttle between the nucleus and the cytoplasm. Neurodegenerative diseases characterized by neuronal nuclear inclusions might utilize the ER-related degradation machinery as a common pathway for the degradation of aberrant proteins.
Collapse
Affiliation(s)
- Yasuo Miki
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Korb E, Finkbeiner S. PML in the Brain: From Development to Degeneration. Front Oncol 2013; 3:242. [PMID: 24062991 PMCID: PMC3775456 DOI: 10.3389/fonc.2013.00242] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/02/2013] [Indexed: 11/28/2022] Open
Abstract
The promyelocytic leukemia (PML) protein is the main component of PML nuclear bodies, which have many functions in a wide range of cell types. Until recently, PML was not known to have a function in the nervous system or even be expressed in the brain. However, recent reports have changed that view. PML is found in neurons and functions in many aspects of the nervous system, including brain development, circadian rhythms, plasticity, and the response to proteins that cause neurodegenerative disorders. While the investigation of PML in the brain is still in its infancy, it promises to be a fascinating subject that will contribute to our understanding of the brain. Here we summarize what is known about PML expression and function in the brain and highlight both discrepancies in the field and areas that are particularly important to future research.
Collapse
Affiliation(s)
- Erica Korb
- Gladstone Institutes of Neurological Disease , San Francisco, CA , USA ; Neuroscience Graduate Program, University of California , San Francisco, CA , USA
| | | |
Collapse
|
11
|
Rivera-Molina YA, Martínez FP, Tang Q. Nuclear domain 10 of the viral aspect. World J Virol 2013; 2:110-122. [PMID: 24255882 PMCID: PMC3832855 DOI: 10.5501/wjv.v2.i3.110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 05/31/2013] [Accepted: 07/11/2013] [Indexed: 02/05/2023] Open
Abstract
Nuclear domain 10 (ND10) are spherical bodies distributed throughout the nucleoplasm and measuring around 0.2-1.0 μm. First observed under an electron microscope, they were originally described as dense bodies found in the nucleus. They are known by a number of other names, including Promyelocytic Leukemia bodies (PML bodies), Kremer bodies, and PML oncogenic domains. ND10 are frequently associated with Cajal bodies and cleavage bodies. It has been suggested that they play a role in regulating gene transcription. ND10 were originally characterized using human autoantisera, which recognizes Speckled Protein of 100 kDa, from patients with primary biliary cirrhosis. At the immunohistochemical level, ND10 appear as nuclear punctate structures, with 10 indicating the approximate number of dots per nucleus observed. ND10 do not colocalize with kinetochores, centromeres, sites of mRNA processing, or chromosomes. Resistance of ND10 antigens to nuclease digestion and salt extraction suggest that ND10 are associated with the nuclear matrix. They are often identified by immunofluorescent assay using specific antibodies against PML, Death domain-associated protein, nuclear dot protein (NDP55), and so on. The role of ND10 has long been the subject of investigation, with the specific connection of ND10 and viral infection having been a particular focus for almost 20 years. This review summarizes the relationship of ND10 and viral infection. Some future study directions are also discussed.
Collapse
|
12
|
Korb E, Wilkinson CL, Delgado RN, Lovero KL, Finkbeiner S. Arc in the nucleus regulates PML-dependent GluA1 transcription and homeostatic plasticity. Nat Neurosci 2013; 16:874-83. [PMID: 23749147 PMCID: PMC3703835 DOI: 10.1038/nn.3429] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 05/12/2013] [Indexed: 02/06/2023]
Abstract
The activity-regulated cytoskeletal protein Arc (also known as Arg3.1) is required for long-term memory formation and synaptic plasticity. Arc expression is robustly induced by activity, and Arc protein localizes to both active synapses and the nucleus. Whereas its synaptic function has been examined, it is not clear why or how Arc is localized to the nucleus. We found that murine Arc nuclear expression is regulated by synaptic activity in vivo and in vitro. We identified distinct regions of Arc that control its localization, including a nuclear localization signal, a nuclear retention domain and a nuclear export signal. Arc localization to the nucleus promotes an activity-induced increase in the expression of promyelocytic leukemia nuclear bodies, which decreases GluA1 (also called Gria1) transcription and synaptic strength. We further show that Arc nuclear localization regulates homeostatic plasticity. Thus, Arc mediates the homeostatic response to increased activity by translocating to the nucleus, increasing promyelocytic leukemia protein expression and decreasing GluA1 transcription, ultimately downscaling synaptic strength.
Collapse
Affiliation(s)
- Erica Korb
- Gladstone Institutes of Neurological Disease, San Francisco, California, USA
| | | | | | | | | |
Collapse
|
13
|
Lee J, Hwang YJ, Shin JY, Lee WC, Wie J, Kim KY, Lee MY, Hwang D, Ratan RR, Pae AN, Kowall NW, So I, Kim JI, Ryu H. Epigenetic regulation of cholinergic receptor M1 (CHRM1) by histone H3K9me3 impairs Ca(2+) signaling in Huntington's disease. Acta Neuropathol 2013; 125:727-39. [PMID: 23455440 DOI: 10.1007/s00401-013-1103-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/29/2013] [Accepted: 02/22/2013] [Indexed: 12/20/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by an expanded trinucleotide CAG repeat in the gene coding for huntingtin. Deregulation of chromatin remodeling is linked to the pathogenesis of HD but the mechanism remains elusive. To identify what genes are deregulated by trimethylated histone H3K9 (H3K9me3)-dependent heterochromatin, we performed H3K9me3-ChIP genome-wide sequencing combined with RNA sequencing followed by platform integration analysis in stable striatal HD cell lines (STHdhQ7/7 and STHdhQ111/111) cells. We found that genes involving neuronal synaptic transmission including cholinergic receptor M1 (CHRM1), cell motility, and neuronal differentiation pathways are downregulated while their promoter regions are highly occupied with H3K9me3 in HD. Moreover, we found that repression of CHRM1 gene expression by H3K9me3 impairs Ca(2+)-dependent neuronal signal transduction in stable cell lines expressing mutant HD protein. Thus, our data indicate that the epigenetic modifications, such as aberrant H3K9me3-dependent heterochromatin plasticity, directly contribute to the pathogenesis of HD.
Collapse
|
14
|
Chort A, Alves S, Marinello M, Dufresnois B, Dornbierer JG, Tesson C, Latouche M, Baker DP, Barkats M, El Hachimi KH, Ruberg M, Janer A, Stevanin G, Brice A, Sittler A. Interferon beta induces clearance of mutant ataxin 7 and improves locomotion in SCA7 knock-in mice. Brain 2013; 136:1732-45. [DOI: 10.1093/brain/awt061] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
15
|
Jeon GS, Kim KY, Hwang YJ, Jung MK, An S, Ouchi M, Ouchi T, Kowall N, Lee J, Ryu H. Deregulation of BRCA1 leads to impaired spatiotemporal dynamics of γ-H2AX and DNA damage responses in Huntington's disease. Mol Neurobiol 2012; 45:550-63. [PMID: 22580959 DOI: 10.1007/s12035-012-8274-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 04/26/2012] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder of mid-life onset characterized by involuntary movements and progressive cognitive decline caused by a CAG repeat expansion in exon 1 of the Huntingtin (Htt) gene. Neuronal DNA damage is one of the major features of neurodegeneration in HD, but it is not known how it arises or relates to the triplet repeat expansion mutation in the Htt gene. Herein, we found that imbalanced levels of non-phosphorylated and phosphorylated BRCA1 contribute to the DNA damage response in HD. Notably, nuclear foci of γ-H2AX, the molecular component that recruits various DNA damage repair factors to damage sites including BRCA1, were deregulated when DNA was damaged in HD cell lines. BRCA1 specifically interacted with γ-H2AX via the BRCT domain, and this association was reduced in HD. BRCA1 overexpression restored γ-H2AX level in the nucleus of HD cells, while BRCA1 knockdown reduced the spatiotemporal propagation of γ-H2AX foci to the nucleoplasm. The deregulation of BRCA1 correlated with an abnormal nuclear distribution of γ-H2AX in striatal neurons of HD transgenic (R6/2) mice and BRCA1(+/-) mice. Our data indicate that BRCA1 is required for the efficient focal recruitment of γ-H2AX to the sites of neuronal DNA damage. Taken together, our results show that BRCA1 directly modulates the spatiotemporal dynamics of γ-H2AX upon genotoxic stress and serves as a molecular maker for neuronal DNA damage response in HD.
Collapse
Affiliation(s)
- Gye Sun Jeon
- WCU Neurocytomics Group, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Shishido-Hara Y, Ichinose S, Uchihara T. JC virus intranuclear inclusions associated with PML-NBs: analysis by electron microscopy and structured illumination microscopy. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1095-1106. [PMID: 22266251 DOI: 10.1016/j.ajpath.2011.11.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 11/03/2011] [Accepted: 11/29/2011] [Indexed: 01/08/2023]
Abstract
Progressive multifocal leukoencephalopathy is a fatal demyelinating disorder caused by JC virus infection. JC virus was recently found to target promyelocytic leukemia nuclear bodies (PML-NBs), punctuate domains in the nuclei. Thus, the virus progenies cluster in dots as intranuclear inclusions (ie, as dot-shaped inclusions). In the present study, both the viral major and minor capsid proteins were expressed from polycistronic expression vectors with a powerful promoter, and formation into virus-like particles (VLPs) was examined by electron microscopy. When the upstream regulatory sequence including the agnogene (nt 275 to 490) was present, capsid protein expression was suppressed, but numerous VLPs were efficiently formed with restricted accumulation to PML-NBs. VLPs were uniform, and the cells were severely degraded. In contrast, when the 5' terminus of the agnogene (nt 275 to 409; 135 bp) was deleted, capsid protein expression was markedly enhanced, but VLPs were more randomly produced in the nucleus outside of PML-NBs. VLPs were pleomorphic, and cell degradation was minimal. JC virus association with PML-NBs was confirmed in human brain tissues by structured illumination microscopy. PML-NBs were shaped in spherical shells, with viral capsid proteins circumscribing the surface. These findings indicate that PML-NBs are intranuclear locations for pathogenic JC virus proliferation. Either the agnogene or its product likely supports efficient progeny production at PML-NBs, leading to subsequent degeneration of host glial cells.
Collapse
Affiliation(s)
| | - Shizuko Ichinose
- Research Center for Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshiki Uchihara
- Laboratory of Structural Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
17
|
Hinz J, Lehnhardt L, Zakrzewski S, Zhang G, Ignatova Z. Polyglutamine expansion alters the dynamics and molecular architecture of aggregates in dentatorubropallidoluysian atrophy. J Biol Chem 2011; 287:2068-78. [PMID: 22134925 DOI: 10.1074/jbc.m111.318915] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Preferential accumulation of mutant proteins in the nucleus has been suggested to be the molecular culprit that confers cellular toxicity in the neurodegenerative disorders caused by polyglutamine (polyQ) expansion. Here, we use dynamic imaging approaches, orthogonal cross-seeding, and composition analysis to examine the dynamics and structure of nuclear and cytoplasmic inclusions of atrophin-1, implicated in dentatorubropallidoluysian atrophy, a polyQ-based disease with complex clinical features. Our results reveal a large heterogeneity in the dynamics of the nuclear inclusions compared with the compact and immobile cytoplasmic aggregates. At least two types of inclusions of expanded atrophin-1 with different mobility of the molecular species and ability to exchange with the surrounding monomer pool coexist in the nucleus. Intriguingly, the enrichment of nuclear inclusions with slow dynamics parallels changes in the aggregate core architecture that are dominated by the polyQ stretch. We propose that the observed complexity in the dynamics of the nuclear inclusions provides a molecular explanation for the enhanced cellular toxicity of the nuclear aggregates in polyQ-based neurodegeneration.
Collapse
Affiliation(s)
- Justyna Hinz
- Institute of Biochemistry and Biology, University of Potsdam, 14467 Potsdam, Germany
| | | | | | | | | |
Collapse
|
18
|
Odagiri S, Tanji K, Mori F, Kakita A, Takahashi H, Kamitani T, Wakabayashi K. Immunohistochemical analysis of Marinesco bodies, using antibodies against proteins implicated in the ubiquitin-proteasome system, autophagy and aggresome formation. Neuropathology 2011; 32:261-6. [PMID: 22118216 DOI: 10.1111/j.1440-1789.2011.01267.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Marinesco bodies (MBs) are spherical eosinophilic intranuclear inclusions in pigmented neurons in the substantia nigra and locus ceruleus. Previous immunohistochemical studies have shown that MBs are positive for ubiquitin, p62 and SUMO-1, suggesting the involvement of ubiquitination and related proteins in the formation or disaggregation of MBs. However, the involvement is not thoroughly understood. Therefore, we immunohistochemically examined the midbrain from five control subjects ranged from 53 to 84 years old. MBs were positive for various proteins implicated in the ubiquitin-proteasome system (ubiquitin, p62, EDD1, NEDD8, NUB1, SUMO-1 and SUMO-2), aggresome formation (HDAC6) and autophagy (ubiquitin, p62, LC3, GABARAP and GATE-16). These findings suggest that proteins related to ubiquitination, proteasomal degradation and autophagy are involved in the formation or disaggregation of MBs.
Collapse
Affiliation(s)
- Saori Odagiri
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan.
| | | | | | | | | | | | | |
Collapse
|
19
|
Dysregulation of upstream binding factor-1 acetylation at K352 is linked to impaired ribosomal DNA transcription in Huntington's disease. Cell Death Differ 2011; 18:1726-35. [PMID: 21546905 DOI: 10.1038/cdd.2011.38] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Huntington's disease (HD) is an autosomal-dominant neurological disorder caused by expanded CAG repeats in the Huntingtin (Htt) gene, but it is not known how this mutation causes neurodegeneration. Herein, we found that dysfunction of upstream binding factor-1 (UBF-1) is linked to reduced ribosomal DNA (rDNA) transcription in HD. We identified that UBF1 acetylation at Lys (K) 352 by CREB binding protein (CBP) is crucial for the transcriptional activity of rDNA. UBF1 mutation (K352A, K352Q, and K352R) decreased rDNA transcriptional activity. Moreover, both CBP-dHAT mutant and knockdown of CBP by siRNA reduced acetylation of UBF1 and resulted in the decreased transcription of rDNA into rRNA. ChIP analysis showed a significant reduction of UBF1 occupancy in the promoter of rDNA in STHdh(Q111) cell line model of HD. These results demonstrate that abnormal activity of UBF1 and its acetylation by CBP are linked to impaired rDNA transcription in HD. This novel mechanism suggests that modulation of UBF-mediated rDNA synthesis by CBP may be a therapeutic target for improving neuronal rDNA transcription in HD.
Collapse
|
20
|
Reichelt M, Wang L, Sommer M, Perrino J, Nour AM, Sen N, Baiker A, Zerboni L, Arvin AM. Entrapment of viral capsids in nuclear PML cages is an intrinsic antiviral host defense against varicella-zoster virus. PLoS Pathog 2011; 7:e1001266. [PMID: 21304940 PMCID: PMC3033373 DOI: 10.1371/journal.ppat.1001266] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 12/30/2010] [Indexed: 12/24/2022] Open
Abstract
The herpesviruses, like most other DNA viruses, replicate in the host cell nucleus. Subnuclear domains known as promyelocytic leukemia protein nuclear bodies (PML-NBs), or ND10 bodies, have been implicated in restricting early herpesviral gene expression. These viruses have evolved countermeasures to disperse PML-NBs, as shown in cells infected in vitro, but information about the fate of PML-NBs and their functions in herpesvirus infected cells in vivo is limited. Varicella-zoster virus (VZV) is an alphaherpesvirus with tropism for skin, lymphocytes and sensory ganglia, where it establishes latency. Here, we identify large PML-NBs that sequester newly assembled nucleocapsids (NC) in neurons and satellite cells of human dorsal root ganglia (DRG) and skin cells infected with VZV in vivo. Quantitative immuno-electron microscopy revealed that these distinctive nuclear bodies consisted of PML fibers forming spherical cages that enclosed mature and immature VZV NCs. Of six PML isoforms, only PML IV promoted the sequestration of NCs. PML IV significantly inhibited viral infection and interacted with the ORF23 capsid surface protein, which was identified as a target for PML-mediated NC sequestration. The unique PML IV C-terminal domain was required for both capsid entrapment and antiviral activity. Similar large PML-NBs, termed clastosomes, sequester aberrant polyglutamine (polyQ) proteins, such as Huntingtin (Htt), in several neurodegenerative disorders. We found that PML IV cages co-sequester HttQ72 and ORF23 protein in VZV infected cells. Our data show that PML cages contribute to the intrinsic antiviral defense by sensing and entrapping VZV nucleocapsids, thereby preventing their nuclear egress and inhibiting formation of infectious virus particles. The efficient sequestration of virion capsids in PML cages appears to be the outcome of a basic cytoprotective function of this distinctive category of PML-NBs in sensing and safely containing nuclear aggregates of aberrant proteins.
Collapse
Affiliation(s)
- Mike Reichelt
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Salomoni P, Betts-Henderson J. The role of PML in the nervous system. Mol Neurobiol 2010; 43:114-23. [PMID: 21161613 DOI: 10.1007/s12035-010-8156-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2010] [Accepted: 11/24/2010] [Indexed: 11/25/2022]
Abstract
The promyeloctic leukemia protein PML is a tumor suppressor that was originally identified due to its involvement in the (15;17) translocation of acute promyelocytic leukemia. While the majority of early research has focused upon the role of PML in the pathogenesis of leukemia, more recent evidence has identified important roles for PML in tissues outside the hemopoietic system, including the central nervous system (CNS). Here, we review recent literature on the role of PML in the CNS, with particular focus on the processes of neurodevelopment and neurodegeneration, and propose new lines of investigation.
Collapse
Affiliation(s)
- Paolo Salomoni
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK.
| | | |
Collapse
|
22
|
Hands SL, Wyttenbach A. Neurotoxic protein oligomerisation associated with polyglutamine diseases. Acta Neuropathol 2010; 120:419-37. [PMID: 20514488 DOI: 10.1007/s00401-010-0703-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 05/20/2010] [Accepted: 05/23/2010] [Indexed: 02/07/2023]
Abstract
Polyglutamine (polyQ) diseases are associated with a CAG/polyQ expansion mutation in unrelated proteins. Upon elongation of the glutamine tract, disease proteins aggregate within cells, mainly in the central nervous system (CNS) and this aggregation process is associated with neurotoxicity. However, it remains unclear to what extent and how this aggregation causes neuronal dysfunction in the CNS. Aiming at preventing neuronal dysfunction, it will be crucial to determine the links between aggregation and cellular dysfunction, understand the folding pathway of polyQ proteins and discover the relative neurotoxicity of polyQ protein species formed along the aggregation pathway. Here, we review what is known about conformations of polyQ peptides and proteins in their monomeric state from experimental and modelling data, how conformational changes of polyQ proteins relate to their oligomerisation and morphology of aggregates and which cellular function are impaired by oligomers, in vitro and in vivo. We also summarise the key modulatory cellular mechanisms and co-factors, which could affect the folding pathway and kinetics of polyQ aggregation. Although many studies have investigated the relationship between polyQ aggregation and toxicity, these have mainly focussed on investigating changes in the formation of the classical hallmark of polyQ diseases, i.e. microscopically visible inclusion bodies. However, recent studies in which oligomeric species have been considered start to shed light on the identity of neurotoxic oligomeric species. Initial evidence suggests that conformational changes induced by polyQ expansions and their surrounding sequence lead to the formation of particular oligomeric intermediates that may differentially affect neurotoxicity.
Collapse
Affiliation(s)
- Sarah L Hands
- Southampton Neuroscience Group, School of Biological Sciences, University of Southampton, Southampton SO16 7PX, UK
| | | |
Collapse
|
23
|
Abstract
PML nuclear bodies are matrix-associated domains that recruit an astonishing variety of seemingly unrelated proteins. Since their discovery in the early 1960s, PML bodies have fascinated cell biologists because of their beauty and their tight association with cellular disorders. The identification of PML, a gene involved in an oncogenic chromosomal translocation, as the key organizer of these domains drew instant interest onto them. The multiple levels of PML body regulation by a specific posttranslational modification, sumoylation, have raised several unsolved issues. Functionally, PML bodies may sequester, modify or degrade partner proteins, but in many ways, PML bodies still constitute an enigma.
Collapse
Affiliation(s)
- Valérie Lallemand-Breitenbach
- INSERM/CNRS/Université Paris Diderot/Institut Universitaire Hématologie U944/ UMR7212, Laboratoire associé de la Ligue Nationale contre le Cancer, Hôpital St. Louis, 1, Av. C. Vellefaux 75475 Paris Cedex 10, France
| | | |
Collapse
|
24
|
Moran DM, Shen H, Maki CG. Puromycin-based vectors promote a ROS-dependent recruitment of PML to nuclear inclusions enriched with HSP70 and Proteasomes. BMC Cell Biol 2009; 10:32. [PMID: 19409099 PMCID: PMC2685373 DOI: 10.1186/1471-2121-10-32] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Accepted: 05/01/2009] [Indexed: 11/25/2022] Open
Abstract
Background Promyelocytic Leukemia (PML) protein can interact with a multitude of cellular factors and has been implicated in the regulation of various processes, including protein sequestration, cell cycle regulation and DNA damage responses. Previous studies reported that misfolded proteins or proteins containing polyglutamine tracts form aggregates with PML, chaperones, and components of the proteasome, supporting a role for PML in misfolded protein degradation. Results In the current study, we have identified a reactive oxygen species (ROS) dependent aggregation of PML, small ubiquitin-like modifier 1 (SUMO-1), heat shock protein 70 (HSP70) and 20S proteasomes in human cell lines that have been transiently transfected with vectors expressing the puromycin resistance gene, puromycin n-acetyl transferase (pac). Immunofluorescent studies demonstrated that PML, SUMO-1, HSP70 and 20S proteasomes aggregated to form nuclear inclusions in multiple cell lines transfected with vectors expressing puromycin (puro) resistance in regions distinct from nucleoli. This effect does not occur in cells transfected with identical vectors expressing other antibiotic resistance genes or with vectors from which the pac sequence has been deleted. Furthermore, ROS scavengers were shown to ablate the effect of puro vectors on protein aggregation in transfected cells demonstrating a dependency of this effect on the redox state of transfected cells. Conclusion Taken together we propose that puromycin vectors may elicit an unexpected misfolded protein response, associated with the formation of nuclear aggresome like structures in human cell lines. This effect has broad implications for cellular behavior and experimental design.
Collapse
Affiliation(s)
- Diarmuid M Moran
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois, USA.
| | | | | |
Collapse
|
25
|
Woulfe J. Nuclear bodies in neurodegenerative disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:2195-206. [PMID: 18539152 DOI: 10.1016/j.bbamcr.2008.05.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 03/31/2008] [Accepted: 05/08/2008] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are characterized by a relentlessly progressive loss of the functional and structural integrity of the central nervous system. In many cases, these diseases arise sporadically and the causes are unknown. The abnormal aggregation of protein within the cytoplasm or the nucleus of brain cells represents a unifying pathological feature of these diseases. There is increasing evidence for nuclear dysfunction in neurodegenerative diseases. How this relates to protein aggregation in the context of "cause and effect" remains to be determined in most cases. Co-ordinated nuclear function is predicated on the activity of distinct nuclear subdomains, or nuclear bodies, each responsible for a specific function. If nuclear dysfunction represents an important etiopathological feature in neurodegenerative disease, then this should be reflected by functional and/or morphological alterations in this nuclear compartmentalization. For most neurodegenerative diseases, evidence for nuclear dysfunction, with attendant consequences for nuclear architecture, is only beginning to emerge. In this review, I will discuss neurodegenerative diseases in the context of nuclear dysfunction and, more specifically, alterations in nuclear bodies. Although research in this field is in its infancy, identifying alterations in the nucleus in neurodegenerative disease has potentially profound implications for elucidating the pathogenesis of these disorders.
Collapse
Affiliation(s)
- John Woulfe
- Department of Pathology, The Ottawa Hospital, Civic Campus, 1053 Carling Avenue, Ottawa, Ontario, Canada K1Y 4E9.
| |
Collapse
|
26
|
Promyelocytic Leukemia Nuclear Bodies Provide a Scaffold for Human Polyomavirus JC Replication and Are Disrupted After Development of Viral Inclusions in Progressive Multifocal Leukoencephalopathy. J Neuropathol Exp Neurol 2008; 67:299-308. [DOI: 10.1097/nen.0b013e31816a1dd3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
27
|
Villagra NT, Bengoechea R, Vaqué JP, Llorca J, Berciano MT, Lafarga M. Nuclear compartmentalization and dynamics of the poly(A)-binding protein nuclear 1 (PABPN1) inclusions in supraoptic neurons under physiological and osmotic stress conditions. Mol Cell Neurosci 2008; 37:622-33. [DOI: 10.1016/j.mcn.2007.12.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 11/14/2007] [Accepted: 12/06/2007] [Indexed: 11/26/2022] Open
|
28
|
Yamada M, Sato T, Tsuji S, Takahashi H. CAG repeat disorder models and human neuropathology: similarities and differences. Acta Neuropathol 2008; 115:71-86. [PMID: 17786457 DOI: 10.1007/s00401-007-0287-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Revised: 08/15/2007] [Accepted: 08/16/2007] [Indexed: 11/24/2022]
Abstract
CAG repeat diseases are hereditary neurodegenerative disorders caused by expansion of a polyglutamine tract in each respective disease protein. They include at least nine disorders, including Huntington's disease (HD), dentatorubral pallidoluysian atrophy (DRPLA), spinal and bulbar muscular atrophy (SBMA), and the spinocerebellar ataxias SCA1, SCA2, SCA3 (also known as Machado-Joseph disease), SCA6, SCA7, and SCA17. It is thought that a gain of toxic function resulting from the protein mutation plays important and common roles in the pathogenesis of these diseases. Recent studies have disclosed that, in addition to the presence of clinical phenotypes and conventional neuropathology in each disease, human brains affected by CAG repeat diseases share several polyglutamine-related changes in their neuronal nuclei and cytoplasm including the formation of intranuclear inclusions. Although these novel pathologic changes also show a distribution pattern characteristic to each disease, they are generally present beyond the lesion distribution of neuronal loss, suggesting that neurons are affected much more widely than has been recognized previously. Various mouse models of CAG repeat diseases have revealed that CAG repeat lengths, which are responsible for polyglutamine diseases in humans, are not sufficient for creating the conditions characteristic of each disease in mice. Although high expression of mutant proteins in mice results in the successful generation of polyglutamine-related changes in the brain, there are still some differences from human pathology in the lesion distribution or cell types that are affected. In addition, no model has yet successfully reproduced the specific neuronal loss observed in humans. Although there are no models that fully represent the neuropathologic changes present in humans, the data obtained have provided evidence that clinical onset is not clearly associated with neuronal cell death, but depends on intranuclear accumulation of mutant proteins in neurons.
Collapse
Affiliation(s)
- Mitsunori Yamada
- Department of Pathology, Brain Research Institute, Niigata University, 1 Asahimachi, Niigata, 951-8585, Japan.
| | | | | | | |
Collapse
|
29
|
Sun J, Xu H, Negi S, Subramony SH, Hebert MD. Differential effects of polyglutamine proteins on nuclear organization and artificial reporter splicing. J Neurosci Res 2007; 85:2306-17. [PMID: 17526020 DOI: 10.1002/jnr.21369] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Nuclear inclusions formed by proteins with expanded polyglutamine tracts are found in several neurodegenerative diseases. The effect of nuclear inclusions formed by these disease proteins on the functional organization of the nucleus is only partially understood. In particular, it is not known whether polyglutamine disease proteins disrupt the function of Cajal bodies, which are subnuclear domains that play a role in the biogenesis of small nuclear ribonucleoproteins (snRNPs). snRNPs are an integral part of the pre-mRNA splicing machinery, so it is possible that mutant proteins that alter Cajal body activity indirectly affect pre-mRNA splicing. Here, we evaluate three different polyglutamine disease proteins--ataxin-1, ataxin-3, and huntingtin--for their ability to disrupt Cajal body localization and reduce the splicing of an artificial reporter in HeLa cells. Consistent with previous observations, ataxin-1 inclusions do not drastically alter the localization of Cajal bodies. In contrast, ataxin-3 inclusions associate with this structure. Inclusions formed by a fragment of the huntingtin protein do not associate with Cajal bodies or PML bodies, another subnuclear domain. Among the three disease proteins, only ataxin-3 significantly decreases the splicing of an artificial reporter. These results support the hypothesis that different mutant proteins vary in their ability to disrupt nuclear organization and function.
Collapse
Affiliation(s)
- June Sun
- Department of Neurology, The University of Mississippi Medical Center, Jackson, Mississippi 39216-4505, USA
| | | | | | | | | |
Collapse
|
30
|
The nuclear RhoA exchange factor Net1 interacts with proteins of the Dlg family, affects their localization, and influences their tumor suppressor activity. Mol Cell Biol 2007; 27:8683-97. [PMID: 17938206 DOI: 10.1128/mcb.00157-07] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Net1 is a RhoA-specific guanine nucleotide exchange factor which localizes to the nucleus at steady state. A deletion in its N terminus redistributes the protein to the cytosol, where it activates RhoA and can promote transformation. Net1 contains a PDZ-binding motif at the C terminus which is essential for its transformation properties. Here, we found that Net1 interacts through its PDZ-binding motif with tumor suppressor proteins of the Dlg family, including Dlg1/SAP97, SAP102, and PSD95. The interaction between Net1 and its PDZ partners promotes the translocation of the PDZ proteins to nuclear subdomains associated with PML bodies. Interestingly, the oncogenic mutant of Net1 is unable to shuttle the PDZ proteins to the nucleus, although these proteins still associate as clusters in the cytosol. Our results suggest that the ability of oncogenic Net1 to transform cells may be in part related to its ability to sequester tumor suppressor proteins like Dlg1 in the cytosol, thereby interfering with their normal cellular function. In agreement with this, the transformation potential of oncogenic Net1 is reduced when it is coexpressed with Dlg1 or SAP102. Together, our results suggest that the interaction between Net1 and Dlg1 may contribute to the mechanism of Net1-mediated transformation.
Collapse
|
31
|
Bloomer WAC, VanDongen HMA, VanDongen AMJ. Activity-regulated cytoskeleton-associated protein Arc/Arg3.1 binds to spectrin and associates with nuclear promyelocytic leukemia (PML) bodies. Brain Res 2007; 1153:20-33. [PMID: 17466953 DOI: 10.1016/j.brainres.2007.03.079] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 03/13/2007] [Accepted: 03/19/2007] [Indexed: 01/23/2023]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) is an immediate early gene, whose expression in the central nervous system is induced by specific patterns of synaptic activity. Arc is required for the late-phase of long-term potentiation (LTP) and memory consolidation, and has been implicated in AMPA receptor trafficking. Since Arc's molecular function remains incompletely understood, we have determined its subcellular localization in cultured hippocampal neurons and HEK 293T cells. Fluorescence microscopy experiments revealed that both endogenous and exogenous Arc protein was primarily found in the nucleus, where it concentrated in puncta associated with promyelocytic leukemia (PML) bodies, proposed sites of transcriptional regulation. Arc co-localized and interacted with the betaIV spectrin splice variant betaSpIVSigma5, a nuclear spectrin isoform associated with PML bodies and the nuclear matrix. A small region of Arc containing the coiled-coil domain is also restricted to beta-spectrin-positive puncta, while the isolated spectrin homology domain is diffusely localized. Finally, Arc and betaSpIVSigma5 synergistically increased the number of PML bodies. These results suggest that Arc functions as a spectrin-binding protein, forming a complex that may provide a role at sites of transcriptional regulation within the nucleus.
Collapse
Affiliation(s)
- Wendy A C Bloomer
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
32
|
Navascués J, Bengoechea R, Tapia O, Vaqué JP, Lafarga M, Berciano MT. Characterization of a new SUMO-1 nuclear body (SNB) enriched in pCREB, CBP, c-Jun in neuron-like UR61 cells. Chromosoma 2007; 116:441-51. [PMID: 17549507 DOI: 10.1007/s00412-007-0107-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Revised: 03/22/2007] [Accepted: 03/30/2007] [Indexed: 12/29/2022]
Abstract
The neuron-like UR61 cell is a stable PC12 subline that contains a mouse N-ras oncogene. Dexamethasone (Dex) treatment induces a neuron-like differentiation, which is associated with neuritogenesis and nuclear expression of the glucocorticoid receptor and c-Jun. In differentiated UR61 cells, small ubiquitin-like modifiers 1 (SUMO-1) is concentrated in a new category of SUMO-1 nuclear bodies (SNBs) distinct from promyelocytic leukemia (PML) bodies by their large size and absence of PML protein. SNBs are 1 to 3 mum in diameter and exhibit a fine granular texture by electron microscopy. They are free of splicing factors and transcription foci and show spatial associations with Cajal bodies. In addition to SUMO-1 and the E2-conjugating enzyme Ubc9, which is essential for sumoylation, SNBs concentrate the transcriptional regulators CBP, CREB, and c-Jun. Moreover, transfection experiments demonstrate that SNBs accumulate the active conjugating form of SUMO-1 but not the conjugation defective variant of SUMO-1, supporting that SNBs are sites of sumoylation. Our results suggest that SNBs play a role in the control of the nucleoplasmic concentration of transcription regulators involved in neuroprotection and survival of the UR61 cells.
Collapse
Affiliation(s)
- Joaquín Navascués
- Division of Gene Regulation and Expression, Wellcome Trust Biocentre, University of Dundee, DD1 5EH, Dundee, UK
| | | | | | | | | | | |
Collapse
|
33
|
Valero J, Berciano MT, Weruaga E, Lafarga M, Alonso JR. Pre-neurodegeneration of mitral cells in the pcd mutant mouse is associated with DNA damage, transcriptional repression, and reorganization of nuclear speckles and Cajal bodies. Mol Cell Neurosci 2006; 33:283-95. [PMID: 16978877 DOI: 10.1016/j.mcn.2006.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Revised: 07/04/2006] [Accepted: 08/01/2006] [Indexed: 01/01/2023] Open
Abstract
DNA damage and impairment of its repair underlie several neurodegenerative diseases. The Purkinje cell degeneration (pcd) mutation causes the loss of Nna1 expression and is associated with a selective and progressive degeneration of specific neuronal populations, including mitral cells in the olfactory bulb. Using an in situ transcription assay, molecular markers for both nuclear compartments and components of the DNA damage/repair pathway, and ultrastructural analysis, here we demonstrate that the pcd mutation induces the formation of DNA damage/repair foci in mitral cells. Furthermore, this effect is associated with transcriptional inhibition, heterochromatinization, nucleolar segregation and the reorganization of nuclear speckles of splicing factors and Cajal bodies. The most significant cytoplasmic alteration observed was a partial replacement of rough endoplasmic reticulum cisternae by a larger amount of free ribosomes, while other organelles were structurally preserved. The tools employed in this work may be of use for the early detection of predegenerative processes in neurodegenerative disorders and for validating rescue strategies.
Collapse
Affiliation(s)
- Jorge Valero
- Laboratorio de Plasticidad Neuronal y Neurorreparación, Instituto de Neurociencias de Castilla y León, Universidad de Salamanca. Avd. Alfonso X el Sabio s/n, E-37007 Salamanca, Spain
| | | | | | | | | |
Collapse
|
34
|
Yamada M, Shimohata M, Sato T, Tsuji S, Takahashi H. Polyglutamine disease: Recent advances in the neuropathology of dentatorubral-pallidoluysian atrophy. Neuropathology 2006; 26:346-51. [PMID: 16961072 DOI: 10.1111/j.1440-1789.2006.00670.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Polyglutamine diseases are hereditary neurodegenerative disorders that are caused by the expansion of a CAG repeat in the causative genes. They comprise at least nine disorders, including DRPLA, HD, and Machado-Joseph disease. Initially, the discovery of neuronal intranuclear inclusions (NIIs) in human brains and in a murine model of HD provided a plausible hypothesis that the expression of expanded polyglutamine stretches leads to NII formation, resulting in neuronal cell death in selective brain regions characteristic to each disease. Recent studies, however, suggest that nuclear dysfunction, especially transcriptional abnormalities caused by the diffuse intranuclear accumulation of mutant proteins, plays a pivotal role in the development and progression of clinical symptoms. Polyglutamine diseases have a similarity with neuronal storage disease, and this pathological process might become a target for the establishment of an effective therapy for these diseases.
Collapse
Affiliation(s)
- Mitsunori Yamada
- Department of Pathology, Brain Research Institute, Niigata University, Japan.
| | | | | | | | | |
Collapse
|
35
|
Takahashi-Fujigasaki J, Arai K, Funata N, Fujigasaki H. SUMOylation substrates in neuronal intranuclear inclusion disease. Neuropathol Appl Neurobiol 2006; 32:92-100. [PMID: 16409557 DOI: 10.1111/j.1365-2990.2005.00705.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neuronal intranuclear inclusion disease (NIID) is a rare neurodegenerative disorder characterized pathologically by the presence of ubiquitinated intranuclear inclusions (NII) in neuronal cells. We demonstrate that NIIs in both sporadic and familial NIID contained the small ubiquitin modifier-1 (SUMO-1) and the SUMOylation substrates promyelocytic leukaemia protein (PML) and histone deacetylase 4 (HDAC4). Both PML and SUMO-1 are major components of nuclear bodies (NBs), suggesting that the NIIs in NIID, as well as the intranuclear inclusions in polyglutamine diseases, might derive from these intranuclear functional domains that serve as sites for ubiquitin-related protein degradation. HDAC4 was also a major component of the NIIs. HDACs are transcriptional corepressors that regulate histone remodelling, and NBs are thought to be sites at which the level of histone acetylation is controlled. The presence of PML, SUMO-1 and HDAC4 in NIIs suggests that transcriptional activity regulated by histone acetylation might contribute to the disease process in NIID. In addition, we showed that another SUMOylation substrate, RanGAP1 is associated with NIIs only in the familial NIID patient. This might be explained by different pathogenetic mechanisms underlying subcategories of NIID, which is very heterogeneous.
Collapse
Affiliation(s)
- J Takahashi-Fujigasaki
- Division of Neuropathology, The Jikei University School of Medicine, Tokyo, 105-8461, Japan.
| | | | | | | |
Collapse
|
36
|
Abu-Baker A, Laganiere S, Fan X, Laganiere J, Brais B, Rouleau GA. Cytoplasmic targeting of mutant poly(A)-binding protein nuclear 1 suppresses protein aggregation and toxicity in oculopharyngeal muscular dystrophy. Traffic 2005; 6:766-79. [PMID: 16101680 DOI: 10.1111/j.1600-0854.2005.00315.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is an adult-onset disorder characterized by progressive eyelid drooping, swallowing difficulties and proximal limb weakness. The autosomal dominant form of this disease is caused by a polyalanine expansion from 10 to 12-17 residues, located at the N-terminus of the poly(A)-binding protein nuclear 1 (PABPN1). A distinct pathological hallmark of OPMD is the presence of filamentous intranuclear aggregates in patients' skeletal muscle cells. Wildtype PABPN1 protein is expressed ubiquitously and was shown to be mostly concentrated in discrete nuclear domains called 'speckles'. Using an established cell- culture model, we show that most mutant PABPN1- positive (alanine expanded form) intranuclear aggregates are structures distinct from intranuclear speckles. In contrast, the promyelocytic leukaemia protein, a major component of nuclear bodies, strongly colocalized to intranuclear aggregates of mutant PABPN1. Wildtype PABPN1 can freely shuttle between the nucleus and cytoplasm. We determined whether the nuclear environment is necessary for mutant PABPN1 inclusion formation and cellular toxicity. This was achieved by inactivating the mutant PABPN1 nuclear localization signal and by generating full-length mutant PABPN1 fused to a strong nuclear export sequence. A green fluorescence protein tag inserted at the N-terminus of both wildtype PABPN1 (ala10) and mutant PABPN1 (ala17) proteins allowed us to visualize their subcellular localization. Targeting mutant PABPN1 to the cytoplasm resulted in a significant suppression of both intranuclear aggregates formation and cellular toxicity, two histological consequences of OPMD. Our results indicate that the nuclear localization of mutant PABPN1 is crucial to OPMD pathogenesis.
Collapse
Affiliation(s)
- Aida Abu-Baker
- Center for Research in Neuroscience, McGill University, and the McGill University Health Center, 1650 Cedar Avenue, Montreal, Quebec, Canada H3G 1A4
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
This review surveys what is known about the structure and function of the subnuclear domains called Cajal bodies (CBs). The major focus is on CBs in mammalian cells but we provide an overview of homologous CB structures in other organisms. We discuss the protein and RNA components of CBs, including factors recently found to associate in a cell cycle-dependent fashion or under specific metabolic or stress conditions. We also consider the dynamic properties of both CBs and their molecular components, based largely on recent data obtained thanks to the advent of improved in vivo detection and imaging methods. We discuss how these data contribute to an understanding of CB functions and highlight major questions that remain to be answered. Finally, we consider the interesting links that have emerged between CBs and alterations in nuclear structure apparent in a range of human pathologies, including cancer and inherited neurodegenerative diseases. We speculate on the relationship between CB function and molecular disease.
Collapse
Affiliation(s)
- Mario Cioce
- IRBM (Merck Research Laboratories Rome), Rome, Italy.
| | | |
Collapse
|
38
|
Sun J, Xu H, Subramony SH, Hebert MD. Interactions between coilin and PIASy partially link Cajal bodies to PML bodies. J Cell Sci 2005; 118:4995-5003. [PMID: 16219678 DOI: 10.1242/jcs.02613] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The eukaryotic nucleus contains a variety of dynamic structures, yet studies into the functional relationship of one type of subnuclear domain to another have been limited. For example, PML bodies and Cajal bodies associate, but the functional consequence of this association and the mediating factors are unknown. Here we report that an associated PML body/Cajal body can co-localize to an snRNA gene locus, with the Cajal body invariably situated between the PML body and the snRNA locus. Binding studies demonstrate that coilin (a Cajal body protein) directly interacts with PIASy (a PML body protein). Cell biological experiments using coilin and PIASy knockout cell lines demonstrate that interactions between coilin and PIASy account in part for the observed association of Cajal bodies with PML bodies. When the PIASy interaction region on coilin is deleted, the frequency of the association between Cajal bodies and PML bodies is reduced. These studies provide another example of coilin's role in the functional organization of the nucleus.
Collapse
Affiliation(s)
- Jun Sun
- Department of Neurology, The University of Mississippi Medical Center, Jackson, MS 39216-4505, USA
| | | | | | | |
Collapse
|
39
|
Fu L, Gao YS, Tousson A, Shah A, Chen TLL, Vertel BM, Sztul E. Nuclear aggresomes form by fusion of PML-associated aggregates. Mol Biol Cell 2005; 16:4905-17. [PMID: 16055507 PMCID: PMC1237092 DOI: 10.1091/mbc.e05-01-0019] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Revised: 07/08/2005] [Accepted: 07/18/2005] [Indexed: 12/28/2022] Open
Abstract
Nuclear aggregates formed by proteins containing expanded poly-glutamine (poly-Q) tracts have been linked to the pathogenesis of poly-Q neurodegenerative diseases. Here, we show that a protein (GFP170*) lacking poly-Q tracts forms nuclear aggregates that share characteristics of poly-Q aggregates. GFP170* aggregates recruit cellular chaperones and proteasomes, and alter the organization of nuclear domains containing the promyelocytic leukemia (PML) protein. These results suggest that the formation of nuclear aggregates and their effects on nuclear architecture are not specific to poly-Q proteins. Using GFP170* as a model substrate, we explored the mechanistic details of nuclear aggregate formation. Fluorescence recovery after photobleaching and fluorescence loss in photobleaching analyses show that GFP170* molecules exchange rapidly between aggregates and a soluble pool of GFP170*, indicating that the aggregates are dynamic accumulations of GFP170*. The formation of cytoplasmic and nuclear GFP170* aggregates is microtubule-dependent. We show that within the nucleus, GFP170* initially deposits in small aggregates at or adjacent to PML bodies. Time-lapse imaging of live cells shows that small aggregates move toward each other and fuse to form larger aggregates. The coalescence of the aggregates is accompanied by spatial rearrangements of the PML bodies. Significantly, we find that the larger nuclear aggregates have complex internal substructures that reposition extensively during fusion of the aggregates. These studies suggest that nuclear aggregates may be viewed as dynamic multidomain inclusions that continuously remodel their components.
Collapse
Affiliation(s)
- Lianwu Fu
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Villagra NT, Navascues J, Casafont I, Val-Bernal JF, Lafarga M, Berciano MT. The PML-nuclear inclusion of human supraoptic neurons: a new compartment with SUMO-1- and ubiquitin-proteasome-associated domains. Neurobiol Dis 2005; 21:181-93. [PMID: 16125395 DOI: 10.1016/j.nbd.2005.07.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 06/30/2005] [Accepted: 07/06/2005] [Indexed: 12/12/2022] Open
Abstract
It is well known that the cell nucleus is organized in structural and functional compartments involved in transcription, RNA processing and protein modifications such as conjugation with SUMO-1 and proteolysis. Promyelocytic leukaemia (PML) bodies are dynamic nuclear structures that concentrate PML protein, SUMO-1 and several sumoylated and non-sumoylated protein regulators of nuclear functions. PML bodies and their associated CBP has been involved in neuronal survival. By light and electron microscopy immunocytochemistry and in situ hybridization we reported the presence, in non-pathological conditions, of a large PML-nuclear inclusion (PML-NI) in human supraoptic neurons. This inclusion appears as a single nuclear structure composed of a capsule enriched in PML, SUMO-1 and CBP proteins and a central lattice of filaments immunoreactive for class III beta-tubulin, ubiquitinated proteins and proteasomes. Furthermore, the PML-NI concentrates the SUMO-conjugating enzyme E2 (UBC9). The PML-NI may be considered a nuclear factory involved in sumoylation and proteolysis via ubiquitin-proteasome system, two nuclear pathways engaged in the control of the nucleoplasmic concentration of active transcriptional regulators. Interestingly, the structural and molecular organization of the PML-NI is related to the Marinesco bodies, age-associated ubiquitinated intranuclear inclusions, and to the intranuclear rodlets enriched in class III beta-tubulin, which are nuclear structures markedly decreased in Alzheimer's disease.
Collapse
Affiliation(s)
- Nuria T Villagra
- Department of Anatomic Pathology, Marqués de Valdecilla University Hospital, University of Cantabria, Santander, Spain
| | | | | | | | | | | |
Collapse
|
41
|
Dentatorubral-pallidoluysian atrophy (DRPLA): model for Huntington's disease and other polyglutamine diseases. NEURODEGENER DIS 2005. [DOI: 10.1017/cbo9780511544873.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
42
|
Wang A, Ikura T, Eto K, Ota MS. Dynamic interaction of p220(NPAT) and CBP/p300 promotes S-phase entry. Biochem Biophys Res Commun 2005; 325:1509-16. [PMID: 15555599 DOI: 10.1016/j.bbrc.2004.10.198] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2004] [Indexed: 11/24/2022]
Abstract
Cajal bodies contain cyclin E/cdk2 and the substrate p220(NPAT) to regulate the transcription of histones, which is essential for cell proliferation, however, recent mouse knockout studies indicate that cyclin E and cdk2 are dispensable for these events. Because the CBP/p300 histone acetyltransferase are also known to be involved in cell proliferation, we examined the molecular and functional interactions of p220(NPAT) with the CBP/p300 at the G1/S boundary as cell cycle regulators. The subnuclear localization of p220(NPAT) and CBP/p300 proteins showed that their foci partially overlapped in a cell cycle dependent manner. Overexpression of p220(NPAT) and CBP/p300 cooperatively enhanced G1/S transition and DNA synthesis even without cdk2 phosphorylation site. Finally, molecular alignment analysis indicated that p220(NPAT) contains several potential substrate sites for CBP/p300. Overall, our findings demonstrate that p220(NPAT) and CBP/p300 form a transient complex at the G1/S boundary to play cooperative roles to promote the S-phase entry.
Collapse
Affiliation(s)
- Aiyan Wang
- Section of Molecular Craniofacial Embryology, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8549, Japan
| | | | | | | |
Collapse
|
43
|
Woulfe J, Gray D, Prichett-Pejic W, Munoz DG, Chretien M. Intranuclear Rodlets in the Substantia Nigra: Interactions with Marinesco Bodies, Ubiquitin, and Promyelocytic Leukemia Protein. J Neuropathol Exp Neurol 2004; 63:1200-7. [PMID: 15581187 DOI: 10.1093/jnen/63.11.1200] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
There is growing appreciation that the nucleus is organized into an array of discrete structural domains, each subserving a specific function. These functional nuclear bodies are to be distinguished from pathological intranuclear inclusions which have been described in a variety of neurodegenerative diseases. Marinesco bodies (MBs) are eosinophilic ubiquitinated intranuclear inclusions found in pigmented neurons of the human substantia nigra and locus coeruleus. Traditionally considered non-pathological entities, more recent studies have indicated that MBs are associated with the age-associated degenerative changes in the substantia nigra and striatal loss of dopaminergic terminals. In the present morphological study of the human substantia nigra, we demonstrate colocalization, contiguity, and focally shared immunoreactivity between MBs and neuronal intranuclear rodlets (INRs). The latter nuclear structures of uncertain function are markedly decreased in the cortex of Alzheimer's disease, but not dementia with Lewy bodies. In addition, we demonstrate an interaction between INRs and promyelocytic leukemia (PML) protein, the signature protein of PML nuclear bodies. These results suggest that structures which subserve the functional compartmentalization of the neuronal nucleus may be relevant to elucidating cellular mechanisms of age-related motor dysfunction.
Collapse
Affiliation(s)
- John Woulfe
- The Ottawa Hospital, Ottawa, Ontario, Canada.
| | | | | | | | | |
Collapse
|
44
|
Zimber A, Nguyen QD, Gespach C. Nuclear bodies and compartments: functional roles and cellular signalling in health and disease. Cell Signal 2004; 16:1085-104. [PMID: 15240004 DOI: 10.1016/j.cellsig.2004.03.020] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2004] [Accepted: 03/17/2004] [Indexed: 01/08/2023]
Abstract
There is much interest in recent years in the possible role of different nuclear compartments and subnuclear domains in the regulation of gene expression, signalling, and cellular functions. The nucleus contains inositol phosphates, actin and actin-binding proteins and myosin isoforms, multiple protein kinases and phosphatases targeting Cdk-1 and Cdk-2, MAPK/SAPK, and Src-related kinases and their substrates, suggesting the implication of several signalling pathways in the intranuclear organization and function of nuclear bodies (NBs). NBs include the well-characterized Cajal bodies (CBs; or coiled bodies), the nucleolus, perinucleolar and perichromatin regions, additional NBs best illustrated by the promyelocytic leukemia nuclear bodies [PML-NBs, also named PML oncogenic dots (PODs), ND10, Kr-bodies] and similar intranuclear foci containing multi-molecular complexes with major role in DNA replication, surveillance, and repair, as well as messenger RNA and ribosomal RNA synthesis and assembly. Chromatin modifying proteins, such as the CBP acetyltransferase and type I histone deacetylase, accumulate at PML-NBs. PML-NBs and Cajal bodies are very dynamic and mobile within the nuclear space and are regulated by cellular stress (heat shock, apoptosis, senescence, heavy metal exposure, viral infection, and DNA damage responses). NBs strongly interact, using signalling mechanisms for the directional and ordered traffic of essential molecular components. NBs organize the delivery and storage of essential RNAs and proteins that play a role in transcription, pre-mRNA biosynthesis and splicing, and the sequestration and/or degradation of regulatory proteins, such as heterogenous nuclear ribonuclear proteins (hnRNPs), p53, Rb1, CBP, STAT3, and others. The objective of this review is to summarize some aspects of these nuclear structures/bodies/domains, including their proposed roles in cellular signalling and in human diseases, mainly neurodegenerative disorders and cancer.
Collapse
Affiliation(s)
- Amazia Zimber
- Department of Animal Sciences, Faculty of Agriculture, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | | | | |
Collapse
|
45
|
Yamada M, Tan CF, Inenaga C, Tsuji S, Takahashi H. Sharing of polyglutamine localization by the neuronal nucleus and cytoplasm in CAG-repeat diseases. Neuropathol Appl Neurobiol 2004; 30:665-75. [PMID: 15541006 DOI: 10.1111/j.1365-2990.2004.00583.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The expansion of a trinucleotide cytosine adenine and guanine (CAG) repeat that codes for polyglutamine is a common gene mutation in the family of hereditary neurodegenerative diseases that includes Machado-Joseph disease (MJD) and dentatorubral-pallidoluysian atrophy (DRPLA). The presence of ubiquitinated neuronal intranuclear inclusions (NIIs) has been recognized as a neuropathological hallmark of these diseases, although the significance of NIIs in the pathogenesis remains a matter of controversy. In a previous study of DRPLA, we proposed that intranuclear diffuse accumulation of mutant proteins is another pathological characteristic of neurones, and that the variable prevalence of this characteristic may be relevant to the variation of clinical symptoms in patients with different repeat sizes. Recently, we also disclosed that polyglutamine tracts are localized in a subset of lysosomes in affected neurones. The present immunohistochemical study of autopsied MJD and DRPLA brains shows that the nucleus and cytoplasm of affected neurones share the subcellular distribution of expanded polyglutamine tracts, the pattern of distribution being specific to each diseased brain. The results suggest that in CAG-repeat diseases, mutant proteins are involved in both the ubiquitin/proteasome and endosomal/lysosomal pathways for protein degradation in different intraneuronal compartments, where their accumulation may exert distinct influences on neuronal physiology.
Collapse
Affiliation(s)
- M Yamada
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan.
| | | | | | | | | |
Collapse
|
46
|
Villagrá NT, Berciano J, Altable M, Navascués J, Casafont I, Lafarga M, Berciano MT. PML bodies in reactive sensory ganglion neurons of the Guillain–Barré syndrome. Neurobiol Dis 2004; 16:158-68. [PMID: 15207273 DOI: 10.1016/j.nbd.2004.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2003] [Revised: 01/28/2004] [Accepted: 02/18/2004] [Indexed: 11/16/2022] Open
Abstract
Acute inflammatory demyelinating polyneuropathy (AIDP) is a type of Guillain-Barré syndrome (GBS) characterized by primary nerve demyelination sometimes with secondary axonal degeneration. Studies on the fine structure of dorsal root ganglia in AIDP are lacking. Our aim was to investigate the cytology and nuclear organization of primary sensory neurons in AIDP with axonal injury using ultrastructural and immunohistochemical analysis. The light cytology of the L5 dorsal ganglion showed the characteristic findings of neuronal axonal reaction. The organization of chromatin, nucleolus, Cajal bodies, and nuclear pores corresponded to transcriptionally active neurons. However, the hallmark of the nuclear response to axonal injury was the formation of numerous nuclear bodies (NBs; 6.37 +/- 0.6, in the AIDP, vs. 2.53 +/- 0.2, in the control, mean +/- SDM), identified as promyelocytic leukemia (PML) bodies by the presence of the protein PML. In addition to PML protein, nuclear bodies contained SUMO-1 and the transcriptional regulators CREB-binding protein (CBP) and glucocorticoid receptor (GR). The presence of proteasome 19S was also detected in some nuclear bodies. We suggest that neuronal PML bodies could regulate the nuclear concentration of active proteins, a process mediated by protein interactions with PML and SUMO-1 proteins. In the AIDP case, the proliferation of PML bodies may result from the overexpression of some nuclear proteins due to changes in gene expression associated with axonal injury.
Collapse
Affiliation(s)
- Nuria T Villagrá
- Department of Anatomy and Cell Biology, University Hospital Marqués de Valdecilla, Santander, Spain
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Neuronal intranuclear hyaline inclusion disease (NIHID) is a rare neurodegenerative disorder characterized pathologically by the presence of eosinophilic intranuclear inclusions in neuronal and glial cells. It has been considered to be a heterogeneous disease entity because the clinical pictures of previously described cases were highly variable. In the present review, reported NIHID cases have been categorized into three clinical subgroups according to onset and disease duration, and the clinical phenotype of each subgroup is discussed. Neuronal intranuclear inclusions (NII) in NIHID are ubiquitinated and their prevalence is inversely correlated with neuronal loss, suggesting that NII formation is a protective mechanism involving the ubiquitin-proteasome-dependent proteolytic pathway. In several polyglutamine diseases, disease-related proteins containing abnormally expanded polyglutamine tracts aggregate in neuronal nuclei, resulting in NII formation. The similarity between NII in NIHID and polyglutamine diseases suggests that they are formed during a common proteolysis-related process that takes place in the nucleus. Although the pathogenetic mechanism underlying NIHID remains unknown, the data reviewed here suggest that it might be related to accumulation of as yet unidentified abnormal proteins or dysfunction of the intranuclear ubiquitin-proteasome pathway.
Collapse
|
48
|
Evert BO, Vogt IR, Vieira-Saecker AM, Ozimek L, de Vos RAI, Brunt ERP, Klockgether T, Wüllner U. Gene expression profiling in ataxin-3 expressing cell lines reveals distinct effects of normal and mutant ataxin-3. J Neuropathol Exp Neurol 2003; 62:1006-18. [PMID: 14575237 DOI: 10.1093/jnen/62.10.1006] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is a late-onset neurodegenerative disorder caused by the expansion of a polyglutamine tract within the gene product, ataxin-3. We have previously shown that mutant ataxin-3 causes upregulation of inflammatory genes in transgenic SCA3 cell lines and human SCA3 pontine neurons. We report here a complex pattern of transcriptional changes by microarray gene expression profiling and Northern blot analysis in a SCA3 cell model. Twenty-three differentially expressed genes involved in inflammatory reactions, nuclear transcription, and cell surface-associated processes were identified. The identified corresponding proteins were analyzed by immunohistochemistry in human disease and control brain tissue to evaluate their implication in SCA3 pathogenesis. In addition to several inflammatory mediators upregulated in mutant ataxin-3 expressing cell lines and pontine neurons of SCA3 patients, we identified a profound repression of genes encoding cell surface-associated proteins in cells overexpressing normal ataxin-3. Correspondingly, these genes were upregulated in mutant ataxin-3 expressing cell lines and in pontine neurons of SCA3 patients. These findings identify for the first time target genes transcriptionally regulated by normal ataxin-3 and support the hypothesis that both loss of normal ataxin-3 and gain of function through protein-protein interacting properties of mutant ataxin-3 contribute to SCA3 pathogenesis.
Collapse
Affiliation(s)
- Bernd O Evert
- Department of Neurology, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Michalik A, Van Broeckhoven C. Pathogenesis of polyglutamine disorders: aggregation revisited. Hum Mol Genet 2003; 12 Spec No 2:R173-86. [PMID: 14504263 DOI: 10.1093/hmg/ddg295] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Expansion of CAG trinucleotide repeats coding for polyglutamine in unrelated proteins causes at least nine late-onset progressive neurodegenerative disorders, including Huntington's disease and a number of spinocerebellar ataxias. Expanded polyglutamine provokes a dominant gain-of-function neurotoxicity, regardless of the specific protein context within which it resides. Nevertheless, the protein context does modulate polyglutamine toxicity, as evidenced by the distinct clinical and pathological features of the various disorders. Importantly, polyglutamine toxicity might derive from its ability to aggregate. Indeed, aggregation probably underlies some defining attributes of the polyglutamine disorders, such as their late onset, progressive nature, and the dependence of onset age on polyglutamine length. However, the central role of aggregation in polyglutamine pathogenesis has been challenged by several studies, which instead argued that the soluble form of the disease proteins is responsible for neuronal damage. Thus, the question whether polyglutamine aggregates are deleterious, harmless or protective remains the most passionately disputed issue in the study of these diseases. In this review, we attempt to reconcile some of these controversies.
Collapse
Affiliation(s)
- Andrej Michalik
- Department of Molecular Genetics, Flanders Interuniversity Institute of Biotechnology, University of Antwerp, Antwerpen, Belgium
| | | |
Collapse
|
50
|
Takahashi J, Fujigasaki H, Iwabuchi K, Bruni AC, Uchihara T, El Hachimi KH, Stevanin G, Dürr A, Lebre AS, Trottier Y, de Thé H, Tanaka J, Hauw JJ, Duyckaerts C, Brice A. PML nuclear bodies and neuronal intranuclear inclusion in polyglutamine diseases. Neurobiol Dis 2003; 13:230-7. [PMID: 12901837 DOI: 10.1016/s0969-9961(03)00080-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In polyglutamine diseases, accumulation in the nucleus of mutant proteins induces the formation of neuronal intranuclear inclusions (NIIs). The nucleus is compartmentalized into structural and functional domains, which are involved in NII formation. Promyelocytic leukemia protein (PML), a major component of nuclear bodies, and mSin3A, a component of the transcription co-repressor complex, were used to investigate how the intranuclear domains/sites relate to NII formation in SCA2, SCA3, SCA7, SCA17 and DRPLA brains. We demonstrate that the size of PML-positive intranuclear structures was larger in pathological brains than in control ones and that these structures contained mutant proteins. PML colocalized only with small NIIs, which maintained the ring-like structure of normal nuclear bodies. Enlarged ring-like PML-positive structures, devoid of mutant proteins, were also found and might represent structures where mutant polyglutamine proteins have been successfully processed. These data suggest that NIIs originate from nuclear bodies, where mutant proteins accumulate for degradation.
Collapse
Affiliation(s)
- Junko Takahashi
- Laboratoire de Neuropathologie Raymond Escourolle, Hôpital de la Salpêtrière, AP-HP, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|