1
|
Zhang XH, Li JX, Wu XX, Zhang Q, Tian M, Yang SQ, Liu D, Yang XQ. PABPN1 functions as a downstream gene of CREB to inhibit the proliferation of preadipocytes. Anim Biosci 2025; 38:41-53. [PMID: 39210800 PMCID: PMC11725739 DOI: 10.5713/ab.24.0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/03/2023] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE This study was conducted to reveal the role of nuclear poly(A) binding protein 1 (PABPN1) in the proliferation of preadipocytes, and to reveal the relationship between PABPN1 and cAMP response element (CRE)-binding protein (CREB) in the regulation of preadipocyte proliferation. METHODS Vectors overexpressing and siRNAs against PABPN1/CREB were transiently transfected into both porcine preadipocytes and mouse 3T3-L1 cells. Preadipocyte proliferation was measured with cell counting kit-8, 5-ethynyl-2'-deoxyuridine, real-time quantitative polymerase chain reaction, Western blotting, and flow cytometry analyses. Additionally, the transcriptional regulation of CREB on PABPN1 were analyzed with dual-luciferase reporter gene and electrophoretic mobility shift assay. RESULTS Overexpression of PABPN1 inhibits, and knockdown of PABPN1 promotes, the proliferation of both porcine preadipocytes and 3T3-L1 cell lines. PABPN1 overexpression increased, while knockdown decreased, the cell population in the G0/G1 phase. These indicates that PABPN1 repressed preadipocyte proliferation by inhibiting cell cycle progress. Additionally, it was revealed that CREB regulated the expression of PABPN1 through binding to the promoter and that CREB inhibited preadipocyte proliferation by repressed cell cycle progress. Furthermore, we showed that PABPN1 functions as a downstream gene of CREB to regulate the proliferation of preadipocytes. CONCLUSION PABPN1 inhibits preadipocyte proliferation by suppressing the cell cycle. We also found that CREB could promote PABPN1 expression by binding to a motif in the promoter. Further analysis confirmed that PABPN1 functions as a downstream gene of CREB to regulate the proliferation of preadipocytes. These results suggest that the CREB/PABPN1 axis plays a role in the regulation of preadipocyte proliferation, which will contribute to further revealing the mechanism of fat accumulation.
Collapse
Affiliation(s)
- Xiao-Han Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Jia-Xin Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Xiao-Xu Wu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Qian Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Ming Tian
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086,
China
| | - Si-Qi Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086,
China
| | - Xiu-Qin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| |
Collapse
|
2
|
Gordon J, Phizicky D, Schärfen L, Brown C, Arias Escayola D, Kanyo J, Lam T, Simon M, Neugebauer K. Phosphorylation of the nuclear poly(A) binding protein (PABPN1) during mitosis protects mRNA from hyperadenylation and maintains transcriptome dynamics. Nucleic Acids Res 2024; 52:9886-9903. [PMID: 38943343 PMCID: PMC11381358 DOI: 10.1093/nar/gkae562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/23/2024] [Accepted: 06/28/2024] [Indexed: 07/01/2024] Open
Abstract
Polyadenylation controls mRNA biogenesis, nucleo-cytoplasmic export, translation and decay. These processes are interdependent and coordinately regulated by poly(A)-binding proteins (PABPs), yet how PABPs are themselves regulated is not fully understood. Here, we report the discovery that human nuclear PABPN1 is phosphorylated by mitotic kinases at four specific sites during mitosis, a time when nucleoplasm and cytoplasm mix. To understand the functional consequences of phosphorylation, we generated a panel of stable cell lines inducibly over-expressing PABPN1 with point mutations at these sites. Phospho-inhibitory mutations decreased cell proliferation, highlighting the importance of PABPN1 phosphorylation in cycling cells. Dynamic regulation of poly(A) tail length and RNA stability have emerged as important modes of gene regulation. We therefore employed long-read sequencing to determine how PABPN1 phospho-site mutants affected poly(A) tails lengths and TimeLapse-seq to monitor mRNA synthesis and decay. Widespread poly(A) tail lengthening was observed for phospho-inhibitory PABPN1 mutants. In contrast, expression of phospho-mimetic PABPN1 resulted in shorter poly(A) tails with increased non-A nucleotides, in addition to increased transcription and reduced stability of a distinct cohort of mRNAs. Taken together, PABPN1 phosphorylation remodels poly(A) tails and increases mRNA turnover, supporting the model that enhanced transcriptome dynamics reset gene expression programs across the cell cycle.
Collapse
Affiliation(s)
- Jackson M Gordon
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - David V Phizicky
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Leonard Schärfen
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Courtney L Brown
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Dahyana Arias Escayola
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Jean Kanyo
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT 06520, USA
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT 06520, USA
| | - Matthew D Simon
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Karla M Neugebauer
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
3
|
Boulinguiez A, Roth F, Mouigni HR, Butler-Browne G, Mouly V, Trollet C. [Nuclear aggregates in oculopharyngeal muscular dystrophy]. Med Sci (Paris) 2022; 38 Hors série n° 1:13-16. [PMID: 36649629 DOI: 10.1051/medsci/2022175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is one of the diseases related to pathological expansions of trinucleotides. Its pathogenesis remains unclear although the presence of aggregates within the nuclei of the muscle fiber seems to play an important role. The basic research studies presented here help understand their composition and their deleterious role. These elements may result in new therapeutic avenues.
Collapse
Affiliation(s)
- Alexis Boulinguiez
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Fany Roth
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Hadidja Rose Mouigni
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Vincent Mouly
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| | - Capucine Trollet
- Sorbonne Université-Inserm, Centre de Recherche en Myologie, Institut de Myologie, Paris, France
| |
Collapse
|
4
|
Serra-Cardona A, Duan S, Yu C, Zhang Z. H3K4me3 recognition by the COMPASS complex facilitates the restoration of this histone mark following DNA replication. SCIENCE ADVANCES 2022; 8:eabm6246. [PMID: 35544640 PMCID: PMC9075808 DOI: 10.1126/sciadv.abm6246] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/21/2022] [Indexed: 06/15/2023]
Abstract
During DNA replication, parental H3-H4 marked by H3K4me3 are transferred almost equally onto leading and lagging strands of DNA replication forks. Mutations in replicative helicase subunit, Mcm2 (Mcm2-3A), and leading strand DNA polymerase subunit, Dpb3 (dpb3∆), result in asymmetric distributions of H3K4me3 at replicating DNA strands immediately following DNA replication. Here, we show that mcm2-3A and dpb3∆ mutant cells markedly reduce the asymmetric distribution of H3K4me3 during cell cycle progression before mitosis. Furthermore, the restoration of a more symmetric distribution of H3K4me3 at replicating DNA strands in these mutant cells is driven by methylating nucleosomes without H3K4me3 by the H3K4 methyltransferase complex, COMPASS. Last, both gene transcription machinery and the binding of parental H3K4me3 by Spp1 subunit of the COMPASS complex help recruit the enzyme to chromatin for the restoration of the H3K4me3-marked state following DNA replication, shedding light on inheritance of this mark following DNA replication.
Collapse
Affiliation(s)
- Albert Serra-Cardona
- Institute for Cancer Genetics, Department of Pediatrics and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Shoufu Duan
- Institute for Cancer Genetics, Department of Pediatrics and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Chuanhe Yu
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Zhiguo Zhang
- Institute for Cancer Genetics, Department of Pediatrics and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
5
|
Abu-Baker A, Parker A, Ramalingam S, Laganiere J, Brais B, Neri C, Dion P, Rouleau G. Valproic acid is protective in cellular and worm models of oculopharyngeal muscular dystrophy. Neurology 2018; 91:e551-e561. [PMID: 30006409 PMCID: PMC6105050 DOI: 10.1212/wnl.0000000000005942] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/08/2018] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE To explore valproic acid (VPA) as a potentially beneficial drug in cellular and worm models of oculopharyngeal muscular dystrophy (OPMD). METHODS Using a combination of live cell imaging and biochemical measures, we evaluated the potential protective effect of VPA in a stable C2C12 muscle cell model of OPMD, in lymphoblastoid cell lines derived from patients with OPMD and in a transgenic Caenorhabditis elegans OPMD model expressing human mutant PABPN1. RESULTS We demonstrated that VPA protects against the toxicity of mutant PABPN1. Of note, we found that VPA confers its long-term protective effects on C2C12 cell survival, proliferation, and differentiation by increasing the acetylated level of histones. Furthermore, VPA enhances the level of histone acetylation in lymphoblastoid cell lines derived from patients with OPMD. Moreover, treatment of nematodes with moderate concentrations of VPA significantly improved the motility of the PABPN-13 Alanines worms. CONCLUSIONS Our results suggest that VPA helps to counteract OPMD-related phenotypes in the cellular and C elegans disease models.
Collapse
Affiliation(s)
- Aida Abu-Baker
- From the Montreal Neurological Institute and Hospital (A.A.-B., P.D., G.R.), Ingram School of Nursing, Faculty of Medicine (S.R.), and Department of Neurology and Neurosurgery (G.R.), McGill University, Montreal; CHUM Research Center (A.P.), Montreal; Department of Neuroscience (A.P.), and Ophthalmology Research Hôpital Maisonneuve Rosemont, Laboratoire de Isabelle Brunette (J.L.), University of Montreal; Neuromuscular Group (B.B.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and Brain C-lab (C.N.), Institute of Biology Paris-Seine, CNRS UMR 8256 Biology of Adaptation & Aging, University Pierre and Marie Curie, Paris, France
| | - Alex Parker
- From the Montreal Neurological Institute and Hospital (A.A.-B., P.D., G.R.), Ingram School of Nursing, Faculty of Medicine (S.R.), and Department of Neurology and Neurosurgery (G.R.), McGill University, Montreal; CHUM Research Center (A.P.), Montreal; Department of Neuroscience (A.P.), and Ophthalmology Research Hôpital Maisonneuve Rosemont, Laboratoire de Isabelle Brunette (J.L.), University of Montreal; Neuromuscular Group (B.B.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and Brain C-lab (C.N.), Institute of Biology Paris-Seine, CNRS UMR 8256 Biology of Adaptation & Aging, University Pierre and Marie Curie, Paris, France
| | - Siriram Ramalingam
- From the Montreal Neurological Institute and Hospital (A.A.-B., P.D., G.R.), Ingram School of Nursing, Faculty of Medicine (S.R.), and Department of Neurology and Neurosurgery (G.R.), McGill University, Montreal; CHUM Research Center (A.P.), Montreal; Department of Neuroscience (A.P.), and Ophthalmology Research Hôpital Maisonneuve Rosemont, Laboratoire de Isabelle Brunette (J.L.), University of Montreal; Neuromuscular Group (B.B.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and Brain C-lab (C.N.), Institute of Biology Paris-Seine, CNRS UMR 8256 Biology of Adaptation & Aging, University Pierre and Marie Curie, Paris, France
| | - Janet Laganiere
- From the Montreal Neurological Institute and Hospital (A.A.-B., P.D., G.R.), Ingram School of Nursing, Faculty of Medicine (S.R.), and Department of Neurology and Neurosurgery (G.R.), McGill University, Montreal; CHUM Research Center (A.P.), Montreal; Department of Neuroscience (A.P.), and Ophthalmology Research Hôpital Maisonneuve Rosemont, Laboratoire de Isabelle Brunette (J.L.), University of Montreal; Neuromuscular Group (B.B.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and Brain C-lab (C.N.), Institute of Biology Paris-Seine, CNRS UMR 8256 Biology of Adaptation & Aging, University Pierre and Marie Curie, Paris, France
| | - Bernard Brais
- From the Montreal Neurological Institute and Hospital (A.A.-B., P.D., G.R.), Ingram School of Nursing, Faculty of Medicine (S.R.), and Department of Neurology and Neurosurgery (G.R.), McGill University, Montreal; CHUM Research Center (A.P.), Montreal; Department of Neuroscience (A.P.), and Ophthalmology Research Hôpital Maisonneuve Rosemont, Laboratoire de Isabelle Brunette (J.L.), University of Montreal; Neuromuscular Group (B.B.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and Brain C-lab (C.N.), Institute of Biology Paris-Seine, CNRS UMR 8256 Biology of Adaptation & Aging, University Pierre and Marie Curie, Paris, France
| | - Christian Neri
- From the Montreal Neurological Institute and Hospital (A.A.-B., P.D., G.R.), Ingram School of Nursing, Faculty of Medicine (S.R.), and Department of Neurology and Neurosurgery (G.R.), McGill University, Montreal; CHUM Research Center (A.P.), Montreal; Department of Neuroscience (A.P.), and Ophthalmology Research Hôpital Maisonneuve Rosemont, Laboratoire de Isabelle Brunette (J.L.), University of Montreal; Neuromuscular Group (B.B.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and Brain C-lab (C.N.), Institute of Biology Paris-Seine, CNRS UMR 8256 Biology of Adaptation & Aging, University Pierre and Marie Curie, Paris, France
| | - Patrick Dion
- From the Montreal Neurological Institute and Hospital (A.A.-B., P.D., G.R.), Ingram School of Nursing, Faculty of Medicine (S.R.), and Department of Neurology and Neurosurgery (G.R.), McGill University, Montreal; CHUM Research Center (A.P.), Montreal; Department of Neuroscience (A.P.), and Ophthalmology Research Hôpital Maisonneuve Rosemont, Laboratoire de Isabelle Brunette (J.L.), University of Montreal; Neuromuscular Group (B.B.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and Brain C-lab (C.N.), Institute of Biology Paris-Seine, CNRS UMR 8256 Biology of Adaptation & Aging, University Pierre and Marie Curie, Paris, France
| | - Guy Rouleau
- From the Montreal Neurological Institute and Hospital (A.A.-B., P.D., G.R.), Ingram School of Nursing, Faculty of Medicine (S.R.), and Department of Neurology and Neurosurgery (G.R.), McGill University, Montreal; CHUM Research Center (A.P.), Montreal; Department of Neuroscience (A.P.), and Ophthalmology Research Hôpital Maisonneuve Rosemont, Laboratoire de Isabelle Brunette (J.L.), University of Montreal; Neuromuscular Group (B.B.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and Brain C-lab (C.N.), Institute of Biology Paris-Seine, CNRS UMR 8256 Biology of Adaptation & Aging, University Pierre and Marie Curie, Paris, France.
| |
Collapse
|
6
|
Richard P, Roth F, Stojkovic T, Trollet C. Distrofia muscolare oculofaringea. Neurologia 2017. [DOI: 10.1016/s1634-7072(16)81777-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
7
|
Banerjee A, Apponi LH, Pavlath GK, Corbett AH. PABPN1: molecular function and muscle disease. FEBS J 2013; 280:4230-50. [PMID: 23601051 DOI: 10.1111/febs.12294] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 04/03/2013] [Accepted: 04/11/2013] [Indexed: 12/17/2022]
Abstract
The polyadenosine RNA binding protein polyadenylate-binding nuclear protein 1 (PABPN1) plays key roles in post-transcriptional processing of RNA. Although PABPN1 is ubiquitously expressed and presumably contributes to control of gene expression in all tissues, mutation of the PABPN1 gene causes the disease oculopharyngeal muscular dystrophy (OPMD), in which a limited set of skeletal muscles are affected. A major goal in the field of OPMD research is to understand why mutation of a ubiquitously expressed gene leads to a muscle-specific disease. PABPN1 plays a well-documented role in controlling the poly(A) tail length of RNA transcripts but new functions are emerging through studies that exploit a variety of unbiased screens as well as model organisms. This review addresses (a) the molecular function of PABPN1 incorporating recent findings that reveal novel cellular functions for PABPN1 and (b) the approaches that are being used to understand the molecular defects that stem from expression of mutant PABPN1. The long-term goal in this field of research is to understand the key molecular functions of PABPN1 in muscle as well as the mechanisms that underlie the pathological consequences of mutant PABPN1. Armed with this information, researchers can seek to develop therapeutic approaches to enhance the quality of life for patients afflicted with OPMD.
Collapse
Affiliation(s)
- Ayan Banerjee
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
8
|
Poly(A) binding protein C1 is essential for efficient L1 retrotransposition and affects L1 RNP formation. Mol Cell Biol 2012; 32:4323-36. [PMID: 22907758 DOI: 10.1128/mcb.06785-11] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Poly(A) binding proteins (PABPs) specifically bind the polyadenosine tail of mRNA and have been shown to be important for RNA polyadenylation, translation initiation, and mRNA stability. Using a modified L1 retrotransposition vector, we examined the effects of two PABPs (encoded by PABPN1 and PABPC1) on the retrotransposition activity of the L1 non-long-terminal-repeat (non-LTR) retrotransposon in both HeLa and HEK293T cells. We demonstrated that knockdown of these two genes by RNA interference (RNAi) effectively reduced L1 retrotransposition by 70 to 80% without significantly changing L1 transcription or translation or the status of the poly(A) tail. We identified that both poly(A) binding proteins were associated with the L1 ribonucleoprotein complex, presumably through L1 mRNA. Depletion of PABPC1 caused a defect in L1 RNP formation. Knockdown of the PABPC1 inhibitor PAIP2 increased L1 retrotransposition up to 2-fold. Low levels of exogenous overexpression of PABPN1 and PABPC1 increased L1 retrotransposition, whereas unregulated overexpression of these two proteins caused pleiotropic effects, such as hypersensitivity to puromycin and decreased L1 activity. Our data suggest that PABPC1 is essential for the formation of L1 RNA-protein complexes and may play a role in L1 RNP translocation in the host cell.
Collapse
|
9
|
Raz V, Abraham T, van Zwet EW, Dirks RW, Tanke HJ, van der Maarel SM. Reversible aggregation of PABPN1 pre-inclusion structures. Nucleus 2012; 2:208-18. [PMID: 21818414 DOI: 10.4161/nucl.2.3.15736] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 03/08/2011] [Accepted: 04/05/2011] [Indexed: 11/19/2022] Open
Abstract
Increased aggregation of misfolded proteins is associated with aging, and characterizes a number of neurodegenerative disorders caused by homopolymeric amino acid expansion mutations. PABPN1 is an aggregation-prone nuclear protein. Natural aggregation of wild-type (WT) PABPN1 is not known to be disease-associated, but alanine-expanded PABPN1 (expPABPN1) accumulates in insoluble intranuclear inclusions in muscle of patients with oculopharyngeal muscular dystrophy (OPMD). We applied microscopic image quantification to study PABPN1 aggregation process in living cells. We identified transitional pre-inclusion foci and demonstrate that these structures significantly differ between WT- and expPABPN1-expressing cells, while inclusions of these proteins are indistinguishable. In addition to the immobile PABPN1 in inclusions, in the nucleoplasm of expPABPN1 expressing cells we also found a fraction of immobile proteins, representing pre-aggregated species. We found that pre-aggregated and pre-inclusion structures are reverted by a PABPN1 specific affinity binder while inclusion structures are not. Together our results demonstrate that the aggregation process of WT- and expPABPN1 differs in steps preceding inclusion formation, suggesting that pre-aggregated protein species could represent the cytotoxic structures.
Collapse
Affiliation(s)
- Vered Raz
- Department of Human Genetics, Leiden University Medical Centre, The Netherlands.
| | | | | | | | | | | |
Collapse
|
10
|
Bengoechea R, Tapia O, Casafont I, Berciano J, Lafarga M, Berciano MT. Nuclear speckles are involved in nuclear aggregation of PABPN1 and in the pathophysiology of oculopharyngeal muscular dystrophy. Neurobiol Dis 2012; 46:118-29. [PMID: 22249111 DOI: 10.1016/j.nbd.2011.12.052] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 12/19/2011] [Accepted: 12/31/2011] [Indexed: 10/14/2022] Open
Abstract
Nuclear speckles are essential nuclear compartments involved in the assembly, delivery and recycling of pre-mRNA processing factors, and in the post-transcriptional processing of pre-mRNAs. Oculopharyngeal muscular dystrophy (OPMD) is caused by a small expansion of the polyalanine tract in the poly(A)-binding protein nuclear 1 (PABPN1). Aggregation of expanded PABPN1 into intranuclear inclusions (INIs) in skeletal muscle fibers is the pathological hallmark of OPMD. In this study what we have analyzed in muscle fibers of OPMD patients and in primary cultures of human myoblasts are the relationships between nuclear speckles and INIs, and the contribution of the former to the biogenesis of the latter. While nuclear speckles concentrate snRNP splicing factors and PABPN1 in control muscle fibers, they are depleted of PABPN1 and appear closely associated with INIs in muscle fibers of OPMD patients. The induction of INI formation in human myoblasts expressing either wild type GFP-PABPN1 or expanded GFP-PABPN1-17ala demonstrates that the initial aggregation of PABPN1 proteins and their subsequent growth in INIs occurs at the edges of the nuclear speckles. Moreover, the growing of INIs gradually depletes PABPN1 proteins and poly(A) RNA from nuclear speckles, although the existence of these nuclear compartments is preserved. Time-lapse experiments in cultured myoblasts confirm nuclear speckles as biogenesis sites of PABPN1 inclusions. Given the functional importance of nuclear speckles in the post-transcriptional processing of pre-mRNAs, the INI-dependent molecular reorganization of these nuclear compartments in muscle fibers may cause a severe dysfunction in nuclear trafficking and processing of polyadenylated mRNAs, thereby contributing to the molecular pathophysiology of OPMD. Our results emphasize the potential importance of nuclear speckles as nuclear targets of neuromuscular disorders.
Collapse
Affiliation(s)
- Rocío Bengoechea
- Department of Anatomy and Cell Biology and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, University of Cantabria, Santander, Spain
| | | | | | | | | | | |
Collapse
|
11
|
Trollet C, Anvar SY, Venema A, Hargreaves IP, Foster K, Vignaud A, Ferry A, Negroni E, Hourde C, Baraibar MA, 't Hoen PAC, Davies JE, Rubinsztein DC, Heales SJ, Mouly V, van der Maarel SM, Butler-Browne G, Raz V, Dickson G. Molecular and phenotypic characterization of a mouse model of oculopharyngeal muscular dystrophy reveals severe muscular atrophy restricted to fast glycolytic fibres. Hum Mol Genet 2010; 19:2191-207. [PMID: 20207626 DOI: 10.1093/hmg/ddq098] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is an adult-onset disorder characterized by ptosis, dysphagia and proximal limb weakness. Autosomal-dominant OPMD is caused by a short (GCG)(8-13) expansions within the first exon of the poly(A)-binding protein nuclear 1 gene (PABPN1), leading to an expanded polyalanine tract in the mutated protein. Expanded PABPN1 forms insoluble aggregates in the nuclei of skeletal muscle fibres. In order to gain insight into the different physiological processes affected in OPMD muscles, we have used a transgenic mouse model of OPMD (A17.1) and performed transcriptomic studies combined with a detailed phenotypic characterization of this model at three time points. The transcriptomic analysis revealed a massive gene deregulation in the A17.1 mice, among which we identified a significant deregulation of pathways associated with muscle atrophy. Using a mathematical model for progression, we have identified that one-third of the progressive genes were also associated with muscle atrophy. Functional and histological analysis of the skeletal muscle of this mouse model confirmed a severe and progressive muscular atrophy associated with a reduction in muscle strength. Moreover, muscle atrophy in the A17.1 mice was restricted to fast glycolytic fibres, containing a large number of intranuclear inclusions (INIs). The soleus muscle and, in particular, oxidative fibres were spared, even though they contained INIs albeit to a lesser degree. These results demonstrate a fibre-type specificity of muscle atrophy in this OPMD model. This study improves our understanding of the biological pathways modified in OPMD to identify potential biomarkers and new therapeutic targets.
Collapse
|
12
|
Klein AF, Ebihara M, Alexander C, Dicaire MJ, Sasseville AMJ, Langelier Y, Rouleau GA, Brais B. PABPN1 polyalanine tract deletion and long expansions modify its aggregation pattern and expression. Exp Cell Res 2008; 314:1652-66. [PMID: 18367172 DOI: 10.1016/j.yexcr.2008.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 02/07/2008] [Accepted: 02/07/2008] [Indexed: 10/22/2022]
Abstract
Expansions of a (GCN)10/polyalanine tract in the Poly(A) Binding Protein Nuclear 1 (PABPN1) cause autosomal dominant oculopharyngeal muscular dystrophy (OPMD). In OPMD muscles, as in models, PABPN1 accumulates in intranuclear inclusions (INIs) whereas in other diseases caused by similar polyalanine expansions, the mutated proteins have been shown to abnormally accumulate in the cytoplasm. This study presents the impact on the subcellular localization of PABPN1 produced by large expansions or deletion of its polyalanine tract. Large tracts of more than 24 alanines result in the nuclear accumulation of PABPN1 in SFRS2-positive functional speckles and a significant decline in cell survival. These large expansions do not cause INIs formation nor do they lead to cytoplasmic accumulation. Deletion of the polyalanine tract induces the formation of aggregates that are located on either side and cross the nuclear membrane, highlighting the possible role of the N-terminal polyalanine tract in PABPN1 nucleo-cytoplasmic transport. We also show that even though five other proteins with polyalanine tracts tend to aggregate when over-expressed they do not co-aggregate with PABPN1 INIs. This study presents the first experimental evidence that there may be a relative loss of function in OPMD by decreasing the availability of PABPN1 through an INI-independent mechanism.
Collapse
Affiliation(s)
- Arnaud F Klein
- Laboratory of neurogenetics of motion, Centre d'excellence en neuromique de l'Université de Montréal, CRCHUM, Université de Montréal, Montréal, Canada
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Oma Y, Kino Y, Toriumi K, Sasagawa N, Ishiura S. Interactions between homopolymeric amino acids (HPAAs). Protein Sci 2007; 16:2195-204. [PMID: 17766374 PMCID: PMC2204140 DOI: 10.1110/ps.072955307] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Many human proteins contain consecutive amino acid repeats, known as homopolymeric amino acid (HPAA) tracts. Some inherited diseases are caused by proteins in which HPAAs are expanded to an excessive length. To this day, nine polyglutamine-related diseases and nine polyalanine-related diseases have been reported, including Huntington's disease and oculopharyngeal muscular dystrophy. In this study, potential HPAA-HPAA interactions were examined by yeast two-hybrid assays using HPAAs of approximately 30 residues in length. The results indicate that hydrophobic HPAAs interact with themselves and with other hydrophobic HPAAs. Previously, we reported that hydrophobic HPAAs formed large aggregates in COS-7 cells. Here, those HPAAs were shown to have significant interactions with each other, suggesting that hydrophobicity plays an important role in aggregation. Among the observed HPAA-HPAA interactions, the Ala28-Ala29 interaction was notable because polyalanine tracts of these lengths have been established to be pathogenic in several polyalanine-related diseases. By testing several constructs of different lengths, we clarified that polyalanine self-interacts at longer lengths (>23 residues) but not at shorter lengths (six to approximately 23 residues) in a yeast two-hybrid assay and a GST pulldown assay. This self-interaction was found to be SDS sensitive in SDS-PAGE and native-PAGE assays. Moreover, the intracellular localization of these long polyalanine tracts was also observed to be disturbed. Our results suggest that long tracts of polyalanine acquire SDS-sensitive self-association properties, which may be a prerequisite event for their abnormal folding. The misfolding of these tracts is thought to be a common molecular aspect underlying the pathogenesis of polyalanine-related diseases.
Collapse
Affiliation(s)
- Yoko Oma
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
14
|
Messaed C, Dion PA, Abu-Baker A, Rochefort D, Laganiere J, Brais B, Rouleau GA. Soluble expanded PABPN1 promotes cell death in oculopharyngeal muscular dystrophy. Neurobiol Dis 2007; 26:546-57. [PMID: 17418585 DOI: 10.1016/j.nbd.2007.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2006] [Revised: 01/19/2007] [Accepted: 02/04/2007] [Indexed: 11/23/2022] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is an autosomal dominant disease caused by the expansion of a polyalanine repeat (GCG)(8-13) in exon 1 of the PABPN1 gene. Skeletal muscle fibers nuclei from OPMD patients contain insoluble polyalanine expanded PABPN1 (expPABPN1) nuclear aggregates that sequester different cellular components. Whether these aggregates are pathogenic, or the consequence of a molecular defense mechanism, remains controversial in the field of neurodegenerative disorders and OPMD. Our cellular model shows that interfering with the formation of expPABPN1-induced large nuclear aggregates increases the availability of nuclear expPABPN1 and significantly exacerbates cell death. Live microscopy reveals that cells harboring an increased amount of the soluble forms of expPABPN1 are significantly more prone to toxicity than those with nuclear aggregates. This is the first report directly indicating that nuclear aggregation in OPMD may reflect an active process by which cells sequester and inactivate the soluble toxic form of expPABPN1.
Collapse
Affiliation(s)
- Christiane Messaed
- Center for Study of Brain Disease, CHUM Research Center, Notre-Dame Hospital, J.A. De-Sève Pavilion Y-3633, 1560 Sherbrooke East, Montreal (Québec), Canada H2L 4M1
| | | | | | | | | | | | | |
Collapse
|
15
|
Abu-Baker A, Rouleau GA. Oculopharyngeal muscular dystrophy: Recent advances in the understanding of the molecular pathogenic mechanisms and treatment strategies. Biochim Biophys Acta Mol Basis Dis 2007; 1772:173-85. [PMID: 17110089 DOI: 10.1016/j.bbadis.2006.10.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2006] [Revised: 10/05/2006] [Accepted: 10/06/2006] [Indexed: 12/24/2022]
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is an adult-onset disorder characterized by progressive eyelid drooping, swallowing difficulties and proximal limb weakness. OPMD is caused by a small expansion of a short polyalanine tract in the poly (A) binding protein nuclear 1 protein (PABPN1). The mechanism by which the polyalanine expansion mutation in PABPN1 causes disease is unclear. PABPN1 is a nuclear multi-functional protein which is involved in pre-mRNA polyadenylation, transcription regulation, and mRNA nucleocytoplasmic transport. The distinct pathological hallmark of OPMD is the presence of filamentous intranuclear inclusions (INIs) in patient's skeletal muscle cells. The exact relationship between mutant PABPN1 intranuclear aggregates and pathology is not clear. OPMD is a unique disease sharing common pathogenic features with other polyalanine disorders, as well as with polyglutamine and dystrophic disorders. This chapter aims to review the rapidly growing body of knowledge concerning OPMD. First, we outline the background of OPMD. Second, we compare OPMD with other trinucleotide repeat disorders. Third, we discuss the recent advances in the understanding of the molecular mechanisms underlying OPMD pathogenesis. Finally, we review recent therapeutic strategies for OPMD.
Collapse
Affiliation(s)
- Aida Abu-Baker
- Center for the Study of Brain Diseases, CHUM Research Center-Notre Dame Hospital, J.A. de Sève Pavillion, Room Y-3633, 1560, Sherbrooke Street East, Montreal, QC, Canada H2L 4M1
| | | |
Collapse
|