1
|
Giudice J, Jiang H. Splicing regulation through biomolecular condensates and membraneless organelles. Nat Rev Mol Cell Biol 2024; 25:683-700. [PMID: 38773325 DOI: 10.1038/s41580-024-00739-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 05/23/2024]
Abstract
Biomolecular condensates, sometimes also known as membraneless organelles (MLOs), can form through weak multivalent intermolecular interactions of proteins and nucleic acids, a process often associated with liquid-liquid phase separation. Biomolecular condensates are emerging as sites and regulatory platforms of vital cellular functions, including transcription and RNA processing. In the first part of this Review, we comprehensively discuss how alternative splicing regulates the formation and properties of condensates, and conversely the roles of biomolecular condensates in splicing regulation. In the second part, we focus on the spatial connection between splicing regulation and nuclear MLOs such as transcriptional condensates, splicing condensates and nuclear speckles. We then discuss key studies showing how splicing regulation through biomolecular condensates is implicated in human pathologies such as neurodegenerative diseases, different types of cancer, developmental disorders and cardiomyopathies, and conclude with a discussion of outstanding questions pertaining to the roles of condensates and MLOs in splicing regulation and how to experimentally study them.
Collapse
Affiliation(s)
- Jimena Giudice
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- McAllister Heart Institute, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Hao Jiang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
2
|
Cryo-EM structure of hnRNPDL-2 fibrils, a functional amyloid associated with limb-girdle muscular dystrophy D3. Nat Commun 2023; 14:239. [PMID: 36646699 PMCID: PMC9842712 DOI: 10.1038/s41467-023-35854-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
hnRNPDL is a ribonucleoprotein (RNP) involved in transcription and RNA-processing that hosts missense mutations causing limb-girdle muscular dystrophy D3 (LGMD D3). Mammalian-specific alternative splicing (AS) renders three natural isoforms, hnRNPDL-2 being predominant in humans. We present the cryo-electron microscopy structure of full-length hnRNPDL-2 amyloid fibrils, which are stable, non-toxic, and bind nucleic acids. The high-resolution amyloid core consists of a single Gly/Tyr-rich and highly hydrophilic filament containing internal water channels. The RNA binding domains are located as a solenoidal coat around the core. The architecture and activity of hnRNPDL-2 fibrils are reminiscent of functional amyloids, our results suggesting that LGMD D3 might be a loss-of-function disease associated with impaired fibrillation. Strikingly, the fibril core matches exon 6, absent in the soluble hnRNPDL-3 isoform. This provides structural evidence for AS controlling hnRNPDL assembly by precisely including/skipping an amyloid exon, a mechanism that holds the potential to generate functional diversity in RNPs.
Collapse
|
3
|
Marklein B, Jenning M, Konthur Z, Häupl T, Welzel F, Nonhoff U, Krobitsch S, Mulder DM, Koenders MI, Joshua V, Cope AP, Shlomchik MJ, Anders HJ, Burmester GR, Hensvold A, Catrina AI, Rönnelid J, Steiner G, Skriner K. The citrullinated/native index of autoantibodies against hnRNP-DL predicts an individual "window of treatment success" in RA patients. Arthritis Res Ther 2021; 23:239. [PMID: 34521462 PMCID: PMC8439038 DOI: 10.1186/s13075-021-02603-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There is a need for biomarker to identify patients "at risk" for rheumatoid arthritis (risk-RA) and to better predict the therapeutic response and in this study we tested the hypothesis that novel native and citrullinated heterogeneous nuclear ribonucleoprotein (hnRNP)-DL autoantibodies could be possible biomarkers. METHODS Using protein macroarray and ELISA, epitope recognition against hnRNP-DL was analysed in sera from different developed RA disease and diagnosed SLE patients. Toll-like receptor (TLR) 7/9 and myeloid differentiation primary response gene 88 (MyD88)-dependency were studied in sera from murine disease models. HnRNP-DL expression in cultivated cells and synovial tissue was analysed by indirect immunofluorescence, immunoblot and immunohistochemistry. RESULTS HnRNP-DL was highly expressed in stress granules, citrullinated in the rheumatoid joint and targeted by autoantibodies either as native or citrullinated proteins in patient subsets with different developed RA disease. Structural citrullination dependent epitopes (SCEs) of hnRNP-DL were detected in 58% of the SLE patients although 98% of these sera were α-CCP-2-negative. To obtain a specific citrullinated signal value, we subtracted the native antibody value from the citrullinated signal. The citrullinated/native index of autoantibodies against hnRNP-DL (CNDL-Index) was identified as a new value for an "individual window of treatment success" in early RA and for the detection of RF IgM/α-CCP-2 seronegative RA patients (24-46%). Negative CNDL-index was found in SLE patients, risk-RA and early RA cohorts such as EIRA where the majority of these patients are DAS28-responders to methotrexate (MTX) treatment (87%). High positive CNDL-values were associated with more severe RA, shared epitope and parenchymal changes in the lung. Specifically, native α-hnRNP-DL is TLR7/9-dependent, associated with pain and ROC analysis revealed an association to initial MTX or etanercept treatment response, especially in seronegative RA patients. CONCLUSION CNDL-index defines people at risk to develop RA and the "window of treatment success" thereby closing the sensitivity gap in RA.
Collapse
Affiliation(s)
- Bianka Marklein
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Charite Campus Mitte, Rheumatologisches Forschungslabor - AG Skriner, Chariteplatz 1 (intern Virchowweg 11, 5.OG, R011), 10117, Berlin, Germany
| | - Madeleine Jenning
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Charite Campus Mitte, Rheumatologisches Forschungslabor - AG Skriner, Chariteplatz 1 (intern Virchowweg 11, 5.OG, R011), 10117, Berlin, Germany
- German Rheumatism Research Centre, Leibniz Institute, 10117, Berlin, Germany
| | - Zoltán Konthur
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Analytical Chemistry (Dpt.1), Bundesanstalt für Materialforschung und-prüfung (BAM), Berlin, Germany
| | - Thomas Häupl
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Charite Campus Mitte, Rheumatologisches Forschungslabor - AG Skriner, Chariteplatz 1 (intern Virchowweg 11, 5.OG, R011), 10117, Berlin, Germany
| | | | - Ute Nonhoff
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Debbie M Mulder
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marije I Koenders
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vijay Joshua
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Andrew P Cope
- Centre for Rheumatic Diseases, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hans-Joachim Anders
- Medical Clinic and Policlinic IV, Nephrological Center, Ludwig-Maximilian-University Hospital, Munich, Germany
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Charite Campus Mitte, Rheumatologisches Forschungslabor - AG Skriner, Chariteplatz 1 (intern Virchowweg 11, 5.OG, R011), 10117, Berlin, Germany
| | - Aase Hensvold
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Academic Specialist Center, Center for Rheumatology, Stockholm Health Region, Stockholm, Sweden
| | - Anca I Catrina
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Rönnelid
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Günter Steiner
- Division of Rheumatology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Cluster for Arthritis and Rehabilitation, Vienna, Austria
| | - Karl Skriner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Charite Campus Mitte, Rheumatologisches Forschungslabor - AG Skriner, Chariteplatz 1 (intern Virchowweg 11, 5.OG, R011), 10117, Berlin, Germany.
- German Rheumatism Research Centre, Leibniz Institute, 10117, Berlin, Germany.
| |
Collapse
|
4
|
Zhang Y, Zhao H, Liu B, Li L, Zhang L, Bao M, Ji X, He X, Yi J, Chen P, Lu C, Lu A. Low Level Antibodies Against Alpha-Tropomyosin Are Associated With Increased Risk of Coronary Heart Disease. Front Pharmacol 2020; 11:195. [PMID: 32174839 PMCID: PMC7056748 DOI: 10.3389/fphar.2020.00195] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
Objective Natural autoantibodies have been implicated to play a key role in the pathogenesis of coronary heart disease (CHD) because they augment autoimmune activation. The aim of this study was to identify novel specific autoantibodies of CHD, and analyze the relationship between their levels and CHD risk indicators. Approach and Results First, clinical data and sera from CHD patients were collected. Then, one protein microarray containing 37 proteins that represent candidate autoantigens was developed. The arrays were used to profile autoantibodies in randomly selected sera from 35 samples (20 CHD patients, and 15 healthy controls). After that, microarray data were analyzed and autoantibodies for CHD were screened out. Then, ELISA detection was conducted to validate the differentiable autoantibodies using larger numbers of serum samples (131 CHD patients, and 131 healthy controls). Finally, the associations of antibodies with CHD risk indicator parameters were assessed. Inter-group comparison by microarray indicated that three CHD novel autoantibodies, including glucose-6-phosphate isomerase (G6PI), alpha-tropomyosin (TPM1), and heterogeneous nuclear ribonucleoprotein D-like (HnRNPDL), were significantly (P < 0.05) increased when compared with the healthy controls. Moreover, a significant increase of IgG autoantibodies for these three autoantigens was confirmed in CHD patients by ELISA (P < 0.0001). The correction analysis revealed a negative correlation of anti-TPM1 antibody levels and total cholesterol (P = 0.0034), and low-density lipoprotein cholesterol (P = 0.0086), respectively. Conclusion G6PI, TPM1, and HnRNPDL were CHD natural autoantigens, and serum anti-TPM1 antibody could be used as a potential marker to predict the risk for CHD patients.
Collapse
Affiliation(s)
- Yin Zhang
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Heru Zhao
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lulu Zhang
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mei Bao
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyu Ji
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianfeng Yi
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
5
|
Batlle C, Yang P, Coughlin M, Messing J, Pesarrodona M, Szulc E, Salvatella X, Kim HJ, Taylor JP, Ventura S. hnRNPDL Phase Separation Is Regulated by Alternative Splicing and Disease-Causing Mutations Accelerate Its Aggregation. Cell Rep 2020; 30:1117-1128.e5. [PMID: 31995753 PMCID: PMC6996132 DOI: 10.1016/j.celrep.2019.12.080] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/12/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022] Open
Abstract
Prion-like proteins form multivalent assemblies and phase separate into membraneless organelles. Heterogeneous ribonucleoprotein D-like (hnRNPDL) is a RNA-processing prion-like protein with three alternative splicing (AS) isoforms, which lack none, one, or both of its two disordered domains. It has been suggested that AS might regulate the assembly properties of RNA-processing proteins by controlling the incorporation of multivalent disordered regions in the isoforms. This, in turn, would modulate their activity in the downstream splicing program. Here, we demonstrate that AS controls the phase separation of hnRNPDL, as well as the size and dynamics of its nuclear complexes, its nucleus-cytoplasm shuttling, and amyloidogenicity. Mutation of the highly conserved D378 in the disordered C-terminal prion-like domain of hnRNPDL causes limb-girdle muscular dystrophy 1G. We show that D378H/N disease mutations impact hnRNPDL assembly properties, accelerating aggregation and dramatically reducing the protein solubility in the muscle of Drosophila, suggesting a genetic loss-of-function mechanism for this muscular disorder.
Collapse
Affiliation(s)
- Cristina Batlle
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autónoma de Barcelona, Bellaterra 08193, Spain
| | - Peiguo Yang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Maura Coughlin
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - James Messing
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD 201815, USA
| | - Mireia Pesarrodona
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain; Joint BSC-IRB Research Programme in Computational Biology, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Elzbieta Szulc
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain; Joint BSC-IRB Research Programme in Computational Biology, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Xavier Salvatella
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain; Joint BSC-IRB Research Programme in Computational Biology, Baldiri Reixac 10, 08028 Barcelona, Spain; ICREA, Passeig Lluís Companys 23, 08010 Barcelona, Spain
| | - Hong Joo Kim
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - J Paul Taylor
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD 201815, USA.
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autónoma de Barcelona, Bellaterra 08193, Spain.
| |
Collapse
|
6
|
Oncogenic heterogeneous nuclear ribonucleoprotein D-like modulates the growth and imatinib response of human chronic myeloid leukemia CD34 + cells via pre-B-cell leukemia homeobox 1. Oncogene 2019; 39:443-453. [PMID: 31488872 DOI: 10.1038/s41388-019-0998-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 08/09/2019] [Indexed: 12/20/2022]
Abstract
Chronic myeloid leukemia (CML) originates from normal hematopoietic stem cells acquiring BCR-ABL fusion gene, specific BCR-ABL inhibitors (e.g., imatinib mesylate, IM) have greatly improved patient management. However, some patients are still suffering from relapse and drug resistance, which urges better understanding of the growth/survival mechanisms of CML stem/progenitor cells. In the present study, the role and its underlying mechanism of heterogeneous nuclear ribonucleoprotein D-like (HNRPDL) in CML cells were investigated. Firstly, overexpression of HNRPDL promoted the growth of murine BaF3 cells in vitro and induced leukemia in vivo, which was enhanced by co-expression of BCR-ABL. Conversely, HNRPDL silencing inhibited colony-forming cell (CFC) production of CML CD34+ cells and attenuated BCR-ABL induced leukemia. In addition, HNRPDL modulated imatinib response of K562 cells and HNRPDL silencing sensitized CML CD34+ cells to imatinib treatment. Mechanistically, we found the stability of pre-B-cell leukemia homeobox 1 (PBX1) mRNA was sustained by HNRPDL through its binding to a specific motif (ACUAGC) in 3'-untranslated region (3'-UTR) of PBX1. The expression of PBX1 was significantly higher in CML CD34+ cells than that in control cells and PBX silencing inhibited the growth of CML cells and sensitized them to imatinib treatment. In contrast, overexpression of PBX1 elevated the CFC production of normal hematopoietic CD34+ cells and "rescued" HNRPDL silencing induced growth inhibition and imatinib sensitization. Taken together, our data have demonstrated that HNRPDL transforms hematopoietic cells and a novel HNRPDL/PBX1 axis plays an important role in human CML CD34+ cells.
Collapse
|
7
|
Berardo A, Lornage X, Johari M, Evangelista T, Cejas C, Barroso F, Dubrovsky A, Bui MT, Brochier G, Saccoliti M, Bohm J, Udd B, Laporte J, Romero NB, Taratuto AL. HNRNPDL-related muscular dystrophy: expanding the clinical, morphological and MRI phenotypes. J Neurol 2019; 266:2524-2534. [PMID: 31267206 DOI: 10.1007/s00415-019-09437-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 11/25/2022]
Abstract
Autosomal dominant limb girdle muscular dystrophy D3 HNRNPDL-related is a rare dominant myopathy caused by mutations in HNRNPDL. Only three unrelated families have been described worldwide, a Brazilian and a Chinese carrying the mutation c.1132G>A p.(Asp378Asn), and one Uruguayan with the mutation c.1132G>C p. (Asp378His), both mutations occurring in the same codon. The present study enlarges the clinical, morphological and muscle MRI spectrum of AD-HNRNPDL-related myopathies demonstrating the significant particularities of the disease. We describe two new unrelated Argentinean families, carrying the previously reported c.1132G>C p.(Asp378His) HNRNPDL mutation. There was a wide phenotypic spectrum including oligo-symptomatic cases, pure limb girdle muscle involvement or distal lower limb muscle weakness. Scapular winging was the most common finding, observed in all patients. Muscle MRIs of the thigh, at different stages of the disease, showed particular involvement of adductor magnus and vastus besides a constant preservation of the rectus femoris and the adductor longus muscles, defining a novel MRI pattern. Muscle biopsy findings were characterized by the presence of numerous rimmed vacuoles, cytoplasmic bodies, and abundant autophagic material at the histochemistry and ultrastructural levels. HNRNPDL-related LGMD D3 results in a wide range of clinical phenotypes from the classic proximal form of LGMD to a more distal phenotype. Thigh MRI suggests a specific pattern. Codon 378 of HNRNPDL gene can be considered a mutation hotspot for HNRNPDL-related myopathy. Pathologically, the disease can be classified among the autophagic rimmed vacuolar myopathies as with the other multisystem proteinopathies.
Collapse
Affiliation(s)
- Andrés Berardo
- Neuropsychiatry Center Valencia Positiva, Córdoba, Argentina
| | - Xavière Lornage
- Department of Translational Medicine, Institut de Génétique Et de Biologie Moléculaire Et Cellulaire (IGBMC)INSERM U1258, UMR7104, Strasbourg University, Illkirch, France
| | - Mridul Johari
- Department of Medical Genetics, University of Helsinki, Medicum, Finland.,Folkhälsan Research Center, Helsinki, Finland
| | - Teresinha Evangelista
- Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, 75013, NeuromuscularParis, France.,Sorbonne Université, AP-HP, INSERM, Centre de référence Des Maladies Neuromusculaires Nord/Est, Ile de France, Paris, France
| | - Claudia Cejas
- Instituto de Investigaciones Neurológicas FLENI, Buenos Aires, Argentina
| | - Fabio Barroso
- Instituto de Investigaciones Neurológicas FLENI, Buenos Aires, Argentina
| | | | - Mai Thao Bui
- Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, 75013, NeuromuscularParis, France
| | - Guy Brochier
- Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, 75013, NeuromuscularParis, France.,Sorbonne Université, AP-HP, INSERM, Centre de référence Des Maladies Neuromusculaires Nord/Est, Ile de France, Paris, France
| | - Maria Saccoliti
- Neuropathology and Neuromuscular Diseases Laboratory, Buenos Aires, Argentina
| | - Johann Bohm
- Department of Translational Medicine, Institut de Génétique Et de Biologie Moléculaire Et Cellulaire (IGBMC)INSERM U1258, UMR7104, Strasbourg University, Illkirch, France
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland.,Neuromuscular Research Center, Tampere University and University Hospital, Tampere, Finland
| | - Jocelyn Laporte
- Department of Translational Medicine, Institut de Génétique Et de Biologie Moléculaire Et Cellulaire (IGBMC)INSERM U1258, UMR7104, Strasbourg University, Illkirch, France
| | - Norma Beatriz Romero
- Morphology Unit, Myology Institute, GHU Pitié-Salpêtrière, 75013, NeuromuscularParis, France. .,Sorbonne Université, AP-HP, INSERM, Centre de référence Des Maladies Neuromusculaires Nord/Est, Ile de France, Paris, France.
| | - Ana Lia Taratuto
- Neuropathology and Neuromuscular Diseases Laboratory, Buenos Aires, Argentina
| |
Collapse
|
8
|
Limb girdle muscular dystrophy D3 HNRNPDL related in a Chinese family with distal muscle weakness caused by a mutation in the prion-like domain. J Neurol 2019; 266:498-506. [PMID: 30604053 DOI: 10.1007/s00415-018-9165-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022]
Abstract
Limb-girdle muscular dystrophies (LGMD) are a group of clinically and genetically heterogeneous diseases characterized by weakness and wasting of the pelvic and shoulder girdle muscles. Twenty-four recessive LGMD (types R1-R24) and five dominant LGMD (types D1-D5) have been identified with characterization of mutations in various genes. To date, LGMD D3 (previously known as LGMD1G) has been characterized in only two families with Brazilian or Uruguayan origin. Each was caused by a distinct mutation at codon 378 in the prion-like domain of HNRNPDL encoding heterogeneous nuclear ribonucleoprotein D like (HNRNPDL), an RNA processing protein. Our study characterized eight patients suffering from LGMD D3 in a Chinese family spanning three generations. Muscle biopsy specimens from two patients showed a myopathy with rimmed vacuoles. Sequencing analysis revealed a heterozygous c.1132G > A (p.D378N) mutation in HNRNPDL that co-segregated with disease phenotype in the family. The same mutation has been identified previously in the Brazilian family with LGMD D3. However, most patients in the current family showed distal as well as proximal limb weakness rather than weakness of toe and finger flexor muscles that were typical features in the other two LGMD D3 families reported previously. The present study indicates that the same mutation in HNRNPDL results in various phenotypes of LGMD D3. That all mutations in three unrelated families with different ethnic background occur at the same position in codon 378 of HNRNPDL gene suggests a mutation hotspot. Acceleration of intrinsic self-aggregation of HNRNPDL caused by mutation of the prior-like domain may contribute to the pathogenesis of the disease.
Collapse
|
9
|
hnRNPDL extensively regulates transcription and alternative splicing. Gene 2018; 687:125-134. [PMID: 30447347 DOI: 10.1016/j.gene.2018.11.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 08/30/2018] [Accepted: 11/08/2018] [Indexed: 01/31/2023]
Abstract
RNA binding proteins (RBPs) are key players of genome regulation. Here we report the transcriptome study of HnRNP D-Like protein, which belongs to the hnRNP family. We used RNA-seq to analyze the global transcript level and alternative splicing on hnRNPDL shRNA-treated cells and control. Sh-hnRNPDL extensively increased in the expression of genes involved in female pregnancy, cell apoptosis, cell proliferation and cell migration. HnRNPDL regulated alternative splicing of hundreds of genes enriched in transcription regulation and signaling pathways including NOD-like receptor signaling, Notch signaling, and TNF signaling. This study provides the first transcriptome-wide analysis of hnRNPDL regulation of gene expression, which adds to the understanding of critical hnRNPDL functions.
Collapse
|
10
|
Zhang P, Ji D, Hu X, Ni H, Ma W, Zhang X, Liao S, Zeng Z, Zhao Y, Zhou H. Oncogenic heterogeneous nuclear ribonucleoprotein D-like promotes the growth of human colon cancer SW620 cells via its regulation of cell-cycle. Acta Biochim Biophys Sin (Shanghai) 2018; 50:880-887. [PMID: 30052712 DOI: 10.1093/abbs/gmy085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Indexed: 01/01/2023] Open
Abstract
Heterogeneous nuclear ribonucleoproteins (hnRNPs) represent a large family of RNA-binding proteins. Heterogeneous nuclear ribonucleoprotein D-like (HNRPDL) is a member of this family. Though aberrant expression of HNRPDL has been reported in a few cancers, whether HNRPDL is deregulated in colon cancer patients and what role this protein plays in these cells are not known yet. In this study, we found that HNRPDL was significantly up-regulated in colon cancer specimens than control. We also demonstrated that HNRPDL silencing inhibited the growth of SW620 cells both in vitro and in vivo. Conversely, we constructed a retroviral vector to deliver HNRPDL into non-malignant NIH-3T3 cells and injected these cells into nude mice. HNRPDL-overexpressing NIH-3T3 cells generated tumors in nude mice but not the control cells. Mechanistically, HNRPDL promoted cell-cycle progression associated with enhanced expressions of cyclin D3 and Ki-67 but decreased expressions of p53 and p21. Taken together, our data demonstrate that HNRPDL is aberrantly expressed in colon cancer cells, which promotes the growth of these cells by activating cell-cycle progression.
Collapse
Affiliation(s)
- Pengshan Zhang
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Dehuan Ji
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Xiaohui Hu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hengli Ni
- Department of Pathology, Medical College of Soochow University, Suzhou, China
| | - Wenjuan Ma
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Xiuyan Zhang
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Shibing Liao
- Department of Oncology, The Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Zheng Zeng
- Department of Oncology, The Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Yun Zhao
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- The Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haixia Zhou
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
11
|
Khan SM, Min A, Gora S, Houranieh GM, Campden R, Robitaille M, Trieu P, Pétrin D, Jacobi AM, Behlke MA, Angers S, Hébert TE. Gβ 4 γ 1 as a modulator of M3 muscarinic receptor signalling and novel roles of Gβ 1 subunits in the modulation of cellular signalling. Cell Signal 2015; 27:1597-608. [DOI: 10.1016/j.cellsig.2015.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 04/16/2015] [Accepted: 04/17/2015] [Indexed: 01/01/2023]
|
12
|
Dufoo-Hurtado MD, Huerta-Ocampo JÁ, Barrera-Pacheco A, Barba de la Rosa AP, Mercado-Silva EM. Low temperature conditioning of garlic (Allium sativum L.) "seed" cloves induces alterations in sprouts proteome. FRONTIERS IN PLANT SCIENCE 2015; 6:332. [PMID: 26029231 PMCID: PMC4429546 DOI: 10.3389/fpls.2015.00332] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/27/2015] [Indexed: 06/04/2023]
Abstract
Low-temperature conditioning of garlic "seed" cloves substitutes the initial climatic requirements of the crop and accelerates the cycle. We have reported that "seed" bulbs from "Coreano" variety conditioned at 5°C for 5 weeks reduces growth and plant weight as well as the crop yields and increases the synthesis of phenolic compounds and anthocyanins. Therefore, this treatment suggests a cold stress. Plant acclimation to stress is associated with deep changes in proteome composition. Since proteins are directly involved in plant stress response, proteomics studies can significantly contribute to unravel the possible relationships between protein abundance and plant stress acclimation. The aim of this work was to study the changes in the protein profiles of garlic "seed" cloves subjected to conditioning at low-temperature using proteomics approach. Two sets of garlic bulbs were used, one set was stored at room temperature (23°C), and the other was conditioned at low temperature (5°C) for 5 weeks. Total soluble proteins were extracted from sprouts of cloves and separated by two-dimensional gel electrophoresis. Protein spots showing statistically significant changes in abundance were analyzed by LC-ESI-MS/MS and identified by database search analysis using the Mascot search engine. The results revealed that low-temperature conditioning of garlic "seed" cloves causes alterations in the accumulation of proteins involved in different physiological processes such as cellular growth, antioxidative/oxidative state, macromolecules transport, protein folding and transcription regulation process. The metabolic pathways affected include protein biosynthesis and quality control system, photosynthesis, photorespiration, energy production, and carbohydrate and nucleotide metabolism. These processes can work cooperatively to establish a new cellular homeostasis that might be related with the physiological and biochemical changes observed in previous studies.
Collapse
Affiliation(s)
- Miguel D. Dufoo-Hurtado
- Laboratorio de Fisiología y Bioquímica Poscosecha de Frutas y Hortalizas, Departamento de Investigación y Posgrado, Facultad de Química, Universidad Autónoma de QuerétaroQuerétaro, Mexico
| | - José Á. Huerta-Ocampo
- Laboratorio de Proteómica y Biomedicina Molecular, División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica A.C.San Luis Potosí, Mexico
| | - Alberto Barrera-Pacheco
- Laboratorio de Proteómica y Biomedicina Molecular, División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica A.C.San Luis Potosí, Mexico
| | - Ana P. Barba de la Rosa
- Laboratorio de Proteómica y Biomedicina Molecular, División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica A.C.San Luis Potosí, Mexico
| | - Edmundo M. Mercado-Silva
- Laboratorio de Fisiología y Bioquímica Poscosecha de Frutas y Hortalizas, Departamento de Investigación y Posgrado, Facultad de Química, Universidad Autónoma de QuerétaroQuerétaro, Mexico
| |
Collapse
|
13
|
Vecsey CG, Park AJ, Khatib N, Abel T. Effects of sleep deprivation and aging on long-term and remote memory in mice. ACTA ACUST UNITED AC 2015; 22:197-202. [PMID: 25776037 PMCID: PMC4371173 DOI: 10.1101/lm.036590.114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Sleep deprivation (SD) following hippocampus-dependent learning in young mice impairs memory when tested the following day. Here, we examined the effects of SD on remote memory in both young and aged mice. In young mice, we found that memory is still impaired 1 mo after training. SD also impaired memory in aged mice 1 d after training, but, by a month after training, sleep-deprived and control aged animals performed similarly, primarily due to remote memory decay in the control aged animals. Gene expression analysis supported the finding that SD has similar effects on the hippocampus in young and aged mice.
Collapse
Affiliation(s)
- Christopher G Vecsey
- Department of Biology, Swarthmore College, Swarthmore, Pennsylvania, USA Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alan J Park
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nora Khatib
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Lenarcic EM, Ziehr BJ, Moorman NJ. An unbiased proteomics approach to identify human cytomegalovirus RNA-associated proteins. Virology 2015; 481:13-23. [PMID: 25765003 DOI: 10.1016/j.virol.2015.02.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/18/2014] [Accepted: 02/06/2015] [Indexed: 12/13/2022]
Abstract
Post-transcriptional events regulate herpesvirus gene expression, yet few herpesvirus RNA-binding proteins have been identified. We used an unbiased approach coupling oligo(dT) affinity capture with proteomics to identify viral RNA-associated proteins during infection. Using this approach, we identified and confirmed changes in the abundance or activity of two host RNA-associated proteins, DHX9 and DDX3, in cells infected with human cytomegalovirus (HCMV). We also identified and confirmed previously unreported activities for the HCMV US22 and pp71 proteins as RNA-associated viral proteins and confirmed that a known viral RNA-binding protein, pTRS1, associates with RNA in infected cells. Further, we found that HCMV pp71 co-sedimented with polysomes, associated with host and viral RNAs, and stimulated the overall rate of protein synthesis. These results demonstrate that oligo(dT) affinity capture coupled with proteomics provides a rapid and straightforward means to identify RNA-associated viral proteins during infection that may participate in the post-transcriptional control of gene expression.
Collapse
Affiliation(s)
- Erik M Lenarcic
- Department of Microbiology & Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Benjamin J Ziehr
- Department of Microbiology & Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Nathaniel J Moorman
- Department of Microbiology & Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
15
|
Cellular STAT3 functions via PCBP2 to restrain Epstein-Barr Virus lytic activation in B lymphocytes. J Virol 2015; 89:5002-11. [PMID: 25717101 DOI: 10.1128/jvi.00121-15] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/13/2015] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED A major hurdle to killing Epstein-Barr virus (EBV)-infected tumor cells using oncolytic therapy is the presence of a substantial fraction of EBV-infected cells that does not support the lytic phase of EBV despite exposure to lytic cycle-promoting agents. To determine the mechanism(s) underlying this refractory state, we developed a strategy to separate lytic from refractory EBV-positive (EBV(+)) cells. By examining the cellular transcriptome in separated cells, we previously discovered that high levels of host STAT3 (signal transducer and activator of transcription 3) curtail the susceptibility of latently infected cells to lytic cycle activation signals. The goals of the present study were 2-fold: (i) to determine the mechanism of STAT3-mediated resistance to lytic activation and (ii) to exploit our findings to enhance susceptibility to lytic activation. We therefore analyzed our microarray data set, cellular proteomes of separated lytic and refractory cells, and a publically available STAT3 chromatin immunoprecipitation sequencing (ChIP-Seq) data set to identify cellular PCBP2 [poly(C)-binding protein 2], an RNA-binding protein, as a transcriptional target of STAT3 in refractory cells. Using Burkitt lymphoma cells and EBV(+) cell lines from patients with hypomorphic STAT3 mutations, we demonstrate that single cells expressing high levels of PCBP2 are refractory to spontaneous and induced EBV lytic activation, STAT3 functions via cellular PCBP2 to regulate lytic susceptibility, and suppression of PCBP2 levels is sufficient to increase the number of EBV lytic cells. We expect that these findings and the genome-wide resources that they provide will accelerate our understanding of a longstanding mystery in EBV biology and guide efforts to improve oncolytic therapy for EBV-associated cancers. IMPORTANCE Most humans are infected with Epstein-Barr virus (EBV), a cancer-causing virus. While EBV generally persists silently in B lymphocytes, periodic lytic (re)activation of latent virus is central to its life cycle and to most EBV-related diseases. However, a substantial fraction of EBV-infected B cells and tumor cells in a population is refractory to lytic activation. This resistance to lytic activation directly and profoundly impacts viral persistence and the effectiveness of oncolytic therapy for EBV(+) cancers. To identify the mechanisms that underlie susceptibility to EBV lytic activation, we used host gene and protein expression profiling of separated lytic and refractory cells. We find that STAT3, a transcription factor overactive in many cancers, regulates PCBP2, a protein important in RNA biogenesis, to regulate susceptibility to lytic cycle activation signals. These findings advance our understanding of EBV persistence and provide important leads on devising methods to improve viral oncolytic therapies.
Collapse
|
16
|
Wu S, Lin L, Zhao W, Li X, Wang Y, Si X, Wang T, Wu H, Zhai X, Zhong X, Gao S, Tong L, Xu Z, Zhong Z. AUF1 is recruited to the stress granules induced by coxsackievirus B3. Virus Res 2014; 192:52-61. [PMID: 25148713 DOI: 10.1016/j.virusres.2014.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 08/05/2014] [Accepted: 08/11/2014] [Indexed: 11/18/2022]
Abstract
Stress granules (SGs) are cytoplasmic granules that are formed in cells when stress occurs. In this study, we found that SGs formed in cells infected with coxsackievirus B3 (CVB3), evidenced with the co-localization of some accepted SG markers in the viral infection-induced granules. We further discovered that adenosine-uridine (AU)-rich element RNA binding factor 1 (AUF1), which can bind to mRNAs and regulate their translation, was recruited to the SGs in response to high dose of CVB3 by detecting the co-localization of AUF1 with SG markers. Similar results were also observed in the enterovirus 71 (EV71)-infected cells. Finally, we demonstrated that AUF1 was also recruited to arsenite-induced SGs, suggesting that the recruitment of AUF1 to SG is not a specific response to viral infection. In summary, our data indicate that both CVB3 and EV71 infections can induce SG formation, and AUF1 is a novel SG component upon the viral infections. Our findings may shed light on understanding the picornavirus-host interaction.
Collapse
Affiliation(s)
- Shuo Wu
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Lexun Lin
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Wenran Zhao
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin 150081, China
| | - Yan Wang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Xiaoning Si
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Tianying Wang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Heng Wu
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Xia Zhai
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Xiaoyan Zhong
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Shuoyang Gao
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Lei Tong
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Zhikai Xu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhaohua Zhong
- Department of Microbiology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
17
|
Twyffels L, Gueydan C, Kruys V. Transportin-1 and Transportin-2: protein nuclear import and beyond. FEBS Lett 2014; 588:1857-68. [PMID: 24780099 DOI: 10.1016/j.febslet.2014.04.023] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 04/12/2014] [Accepted: 04/16/2014] [Indexed: 12/22/2022]
Abstract
Nearly 20 years after its identification as a new β-karyopherin mediating the nuclear import of the RNA-binding protein hnRNP A1, Transportin-1 is still commonly overlooked in comparison with its best known cousin, Importin-β. Transportin-1 is nonetheless a considerable player in nucleo-cytoplasmic transport. Over the past few years, significant progress has been made in the characterization of the nuclear localization signals (NLSs) that Transportin-1 recognizes, thereby providing the molecular basis of its diversified repertoire of cargoes. The recent discovery that mutations in the Transportin-dependent NLS of FUS cause mislocalization of this protein and result in amyotrophic lateral sclerosis illustrates the importance of Transportin-dependent import for human health. Besides, new functions of Transportin-1 are emerging in processes other than nuclear import. Here, we summarize what is known about Transportin-1 and the related β-karyopherin Transportin-2.
Collapse
Affiliation(s)
- Laure Twyffels
- Laboratoire de Biologie moléculaire du gène (CP300), Faculté des Sciences, Université Libre de Bruxelles (ULB), Belgium; Center for Microscopy and Molecular Imaging (CMMI), 6041 Gosselies, Belgium.
| | - Cyril Gueydan
- Laboratoire de Biologie moléculaire du gène (CP300), Faculté des Sciences, Université Libre de Bruxelles (ULB), Belgium
| | - Véronique Kruys
- Laboratoire de Biologie moléculaire du gène (CP300), Faculté des Sciences, Université Libre de Bruxelles (ULB), Belgium; Center for Microscopy and Molecular Imaging (CMMI), 6041 Gosselies, Belgium
| |
Collapse
|
18
|
Vieira NM, Naslavsky MS, Licinio L, Kok F, Schlesinger D, Vainzof M, Sanchez N, Kitajima JP, Gal L, Cavaçana N, Serafini PR, Chuartzman S, Vasquez C, Mimbacas A, Nigro V, Pavanello RC, Schuldiner M, Kunkel LM, Zatz M. A defect in the RNA-processing protein HNRPDL causes limb-girdle muscular dystrophy 1G (LGMD1G). Hum Mol Genet 2014; 23:4103-10. [PMID: 24647604 DOI: 10.1093/hmg/ddu127] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Limb-girdle muscular dystrophies (LGMD) are a heterogeneous group of genetically determined muscle disorders with a primary or predominant involvement of the pelvic or shoulder girdle musculature. More than 20 genes with autosomal recessive (LGMD2A to LGMD2Q) and autosomal dominant inheritance (LGMD1A to LGMD1H) have been mapped/identified to date. Mutations are known for six among the eight mapped autosomal dominant forms: LGMD1A (myotilin), LGMD1B (lamin A/C), LGMD1C (caveolin-3), LGMD1D (desmin), LGMD1E (DNAJB6), and more recently for LGMD1F (transportin-3). Our group previously mapped the LGMD1G gene at 4q21 in a Caucasian-Brazilian family. We now mapped a Uruguayan family with patients displaying a similar LGMD1G phenotype at the same locus. Whole genome sequencing identified, in both families, mutations in the HNRPDL gene. HNRPDL is a heterogeneous ribonucleoprotein family member, which participates in mRNA biogenesis and metabolism. Functional studies performed in S. cerevisiae showed that the loss of HRP1 (yeast orthologue) had pronounced effects on both protein levels and cell localizations, and yeast proteome revealed dramatic reorganization of proteins involved in RNA-processing pathways. In vivo analysis showed that hnrpdl is important for muscle development in zebrafish, causing a myopathic phenotype when knocked down. The present study presents a novel association between a muscular disorder and a RNA-related gene and reinforces the importance of RNA binding/processing proteins in muscle development and muscle disease. Understanding the role of these proteins in muscle might open new therapeutic approaches for muscular dystrophies.
Collapse
Affiliation(s)
- Natássia M Vieira
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil Program in Genomics, Department of Pediatrics and The Manton Center for Orphan Disease Research, Children's Hospital Boston, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Michel S Naslavsky
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Luciana Licinio
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Fernando Kok
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil Department of Neurology, University of São Paulo Medical School, São Paulo, Brazil Mendelics Análise Genômica, São Paulo, Brazil
| | - David Schlesinger
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil Mendelics Análise Genômica, São Paulo, Brazil Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Mariz Vainzof
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Nury Sanchez
- Hospital de Clínicas, Montevideu, Montevideo, Uruguay
| | - João Paulo Kitajima
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil Mendelics Análise Genômica, São Paulo, Brazil
| | - Lihi Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Natale Cavaçana
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Peter R Serafini
- Program in Genomics, Department of Pediatrics and The Manton Center for Orphan Disease Research, Children's Hospital Boston, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Silvia Chuartzman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Adriana Mimbacas
- Department of Genetics, Clemente Estabele Institute, Montevideo, Uruguay
| | - Vincenzo Nigro
- TIGEM (Telethon Institute of Genetics and Medicine), Napoli, Italy
| | - Rita C Pavanello
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Louis M Kunkel
- Program in Genomics, Department of Pediatrics and The Manton Center for Orphan Disease Research, Children's Hospital Boston, Boston, MA, USA Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Mayana Zatz
- Human Genome and Stem Cell Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Regulated transport into the nucleus of herpesviridae DNA replication core proteins. Viruses 2013; 5:2210-34. [PMID: 24064794 PMCID: PMC3798897 DOI: 10.3390/v5092210] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 12/11/2022] Open
Abstract
The Herpesvirdae family comprises several major human pathogens belonging to three distinct subfamilies. Their double stranded DNA genome is replicated in the nuclei of infected cells by a number of host and viral products. Among the latter the viral replication complex, whose activity is strictly required for viral replication, is composed of six different polypeptides, including a two-subunit DNA polymerase holoenzyme, a trimeric primase/helicase complex and a single stranded DNA binding protein. The study of herpesviral DNA replication machinery is extremely important, both because it provides an excellent model to understand processes related to eukaryotic DNA replication and it has important implications for the development of highly needed antiviral agents. Even though all known herpesviruses utilize very similar mechanisms for amplification of their genomes, the nuclear import of the replication complex components appears to be a heterogeneous and highly regulated process to ensure the correct spatiotemporal localization of each protein. The nuclear transport process of these enzymes is controlled by three mechanisms, typifying the main processes through which protein nuclear import is generally regulated in eukaryotic cells. These include cargo post-translational modification-based recognition by the intracellular transporters, piggy-back events allowing coordinated nuclear import of multimeric holoenzymes, and chaperone-assisted nuclear import of specific subunits. In this review we summarize these mechanisms and discuss potential implications for the development of antiviral compounds aimed at inhibiting the Herpesvirus life cycle by targeting nuclear import of the Herpesvirus DNA replicating enzymes.
Collapse
|
20
|
Proteomic analyses and identification of arginine methylated proteins differentially recognized by autosera from anti-Sm positive SLE patients. J Biomed Sci 2013; 20:27. [PMID: 23642268 PMCID: PMC3663782 DOI: 10.1186/1423-0127-20-27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 04/25/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antibodies against spliceosome Sm proteins (anti-Sm autoantibodies) are specific to the autoimmune disease systemic lupus erythematosus (SLE). Anti-Sm autosera have been reported to specifically recognize Sm D1 and D3 with symmetric di-methylarginines (sDMA). We investigated if anti-Sm sera from local SLE patients can differentially recognize Sm proteins or any other proteins due to their methylation states. RESULTS We prepared HeLa cell proteins at normal or hypomethylation states (treated with an indirect methyltransferase inhibitor adenosine dialdehyde, AdOx). A few signals detected by the anti-Sm positive sera from typical SLE patients decreased consistently in the immunoblots of hypomethylated cell extracts. The differentially detected signals by one serum (Sm1) were pinpointed by two-dimensional electrophoresis and identified by mass spectrometry. Three identified proteins: splicing factor, proline- and glutamine-rich (SFPQ), heterogeneous nuclear ribonucleoprotein D-like (hnRNP DL) and cellular nucleic acid binding protein (CNBP) are known to contain methylarginines in their glycine and arginine rich (GAR) sequences. We showed that recombinant hnRNP DL and CNBP expressed in Escherichia coli can be detected by all anti-Sm positive sera we tested. As CNBP appeared to be differentially detected by the SLE sera in the pilot study, differential recognition of arginine methylated CNBP protein by the anti-Sm positive sera were further examined. Hypomethylated FLAG-CNBP protein immunopurified from AdOx-treated HeLa cells was less recognized by Sm1 compared to the CNBP protein expressed in untreated cells. Two of 20 other anti-Sm positive sera specifically differentiated the FLAG-CNBP protein expressed in HeLa cells due to the methylation. We also observed deferential recognition of methylated recombinant CNBP proteins expressed from E. coli by some of the autosera. CONCLUSION Our study showed that hnRNP DL and CNBP are novel antigens for SLE patients and the recognition of CNBP might be differentiated dependent on the level of arginine methylation.
Collapse
|
21
|
Soreq L, Bergman H, Goll Y, Greenberg DS, Israel Z, Soreq H. Deep brain stimulation induces rapidly reversible transcript changes in Parkinson's leucocytes. J Cell Mol Med 2012; 16:1496-507. [PMID: 21910823 PMCID: PMC3823218 DOI: 10.1111/j.1582-4934.2011.01444.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Subthalamic deep brain stimulation (DBS) reversibly modulates Parkinson's disease (PD) motor symptoms, providing an unusual opportunity to compare leucocyte transcripts in the same individuals before and after neurosurgery and 1 hr after stimulus cessation (ON- and OFF-stimulus). Here, we report DBS-induced reversibility and OFF-stimulus restoration in 12 of 16 molecular functions and 3 of 4 biological processes shown in exon microarrays to be differentially expressed between PD patients and controls, post-DBS from pre-DBS and OFF from ON states. Intriguingly, 6 of 18 inflammation and immune-related functions exhibited reversibility, and the extent of stimulus-induced changes correlated with the neurological DBS efficacy, suggesting mechanistic implications. A minimal list of 29 transcripts that changed in all three comparisons between states discriminated pre-surgery and OFF states from post-surgery and controls. Six of these transcripts were found to be able to distinguish between PD patients and both healthy controls and patients with other neurological diseases in a previously published whole blood 3’ array data study of early PD patients. Our findings support the future use of this approach for identifying targets for therapeutic intervention and assessing the efficacy of current and new treatments in this and other neurological diseases.
Collapse
Affiliation(s)
- Lilach Soreq
- Department of Medical Neurobiology (Physiology), IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
22
|
Kimura M, Kose S, Okumura N, Imai K, Furuta M, Sakiyama N, Tomii K, Horton P, Takao T, Imamoto N. Identification of cargo proteins specific for the nucleocytoplasmic transport carrier transportin by combination of an in vitro transport system and stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics. Mol Cell Proteomics 2012; 12:145-57. [PMID: 23087160 DOI: 10.1074/mcp.m112.019414] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The human importin-β family consists of 21 nucleocytoplasmic transport carrier proteins that carry proteins and RNAs across the nuclear envelope through nuclear pores in specific directions. These transport carriers are responsible for the nucleocytoplasmic transport of thousands of proteins, but the cargo allocation of each carrier, which is necessary information if one wishes to understand the physiological context of transport, is poorly characterized. To address this issue, we developed a high-throughput method to identify the cargoes of transport carriers by applying stable isotope labeling by amino acids in cell culture to construct an in vitro transport system. Our method can be outlined in three steps. (1) Cells are cultured in a medium containing a stable isotope. (2) The cell membranes of the labeled cells are permeabilized, and proteins extracted from unlabeled cells are transported into the nuclei of the permeabilized cells. In this step, the reaction system is first depleted of all importin-β family carriers and then supplemented with a particular importin-β family carrier of interest. (3) Proteins in the nuclei are extracted and analyzed quantitatively via LC-MS/MS. As an important test case, we used this method to identify cargo proteins of transportin, a representative member of the importin-β family. As expected, the identified candidate cargo proteins included previously reported transportin cargoes as well as new potential cargoes, which we corroborated via in vitro binding assays. The identified cargoes are predominately RNA-interacting proteins, affirming that cargoes allotted to the same carrier share functional characteristics. Finally, we found that the transportin cargoes possessed at least two classes of signal sequences: the well characterized PY-nuclear localization signals specific for transportin, and Lys/Arg-rich segments capable of binding to both transportin and importin-β. Thus, our method will be useful for linking a carrier to features shared among its cargoes and to specific nuclear localization signals.
Collapse
Affiliation(s)
- Makoto Kimura
- Cellular Dynamics Laboratory, Advanced Science Institute, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Vecsey CG, Peixoto L, Choi JHK, Wimmer M, Jaganath D, Hernandez PJ, Blackwell J, Meda K, Park AJ, Hannenhalli S, Abel T. Genomic analysis of sleep deprivation reveals translational regulation in the hippocampus. Physiol Genomics 2012; 44:981-91. [PMID: 22930738 PMCID: PMC3472468 DOI: 10.1152/physiolgenomics.00084.2012] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 08/22/2012] [Indexed: 01/12/2023] Open
Abstract
Sleep deprivation is a common problem of considerable health and economic impact in today's society. Sleep loss is associated with deleterious effects on cognitive functions such as memory and has a high comorbidity with many neurodegenerative and neuropsychiatric disorders. Therefore, it is crucial to understand the molecular basis of the effect of sleep deprivation in the brain. In this study, we combined genome-wide and traditional molecular biological approaches to determine the cellular and molecular impacts of sleep deprivation in the mouse hippocampus, a brain area crucial for many forms of memory. Microarray analysis examining the effects of 5 h of sleep deprivation on gene expression in the mouse hippocampus found 533 genes with altered expression. Bioinformatic analysis revealed that a prominent effect of sleep deprivation was to downregulate translation, potentially mediated through components of the insulin signaling pathway such as the mammalian target of rapamycin (mTOR), a key regulator of protein synthesis. Consistent with this analysis, sleep deprivation reduced levels of total and phosphorylated mTOR, and levels returned to baseline after 2.5 h of recovery sleep. Our findings represent the first genome-wide analysis of the effects of sleep deprivation on the mouse hippocampus, and they suggest that the detrimental effects of sleep deprivation may be mediated by reductions in protein synthesis via downregulation of mTOR. Because protein synthesis and mTOR activation are required for long-term memory formation, our study improves our understanding of the molecular mechanisms underlying the memory impairments induced by sleep deprivation.
Collapse
|
24
|
Wang L, Li M, Cai M, Xing J, Wang S, Zheng C. A PY-nuclear localization signal is required for nuclear accumulation of HCMV UL79 protein. Med Microbiol Immunol 2012; 201:381-7. [PMID: 22628116 DOI: 10.1007/s00430-012-0243-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 05/08/2012] [Indexed: 01/19/2023]
Abstract
Human cytomegalovirus UL79 protein is recently reported to be required for transcription or efficient accumulation of late viral mRNAs during viral infection. An absolute nuclear distribution of UL79 proteins has been observed with immunofluorescence assay, both during the infection of Flag-tagged UL79 recombinant virus and in the HFFs expressing HA-tagged UL79, with or without virus infection. However, little is known about the nuclear import mechanism of UL79 protein. Here, by utilizing living cells fluorescent microscopy, a predominant nuclear localization of UL79 protein in living cells was detected. Furthermore, the nuclear import of UL79 protein was demonstrated to be dependent on the transportin-1-mediated pathway. Finally, a hydrophobic PY-nuclear localization signal (PY-NLS) was delineated between the amino acids 66-92 of UL79 protein. Collectively, we provide evidence that a PY-NLS, firstly described in viral proteins, is responsible for the nuclear accumulation of UL79 protein.
Collapse
Affiliation(s)
- Lin Wang
- Molecular Virology and Viral Immunology Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 44 Xiaohongshan, Wuchang, Wuhan, 430071, People's Republic of China
| | | | | | | | | | | |
Collapse
|
25
|
Hanel ML, Sun CYJ, Jones TI, Long SW, Zanotti S, Milner D, Jones PL. Facioscapulohumeral muscular dystrophy (FSHD) region gene 1 (FRG1) is a dynamic nuclear and sarcomeric protein. Differentiation 2011; 81:107-18. [PMID: 20970242 DOI: 10.1016/j.diff.2010.09.185] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 08/20/2010] [Accepted: 09/30/2010] [Indexed: 01/01/2023]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) region gene 1 (FRG1) is a candidate gene for mediating FSHD pathophysiology, however, very little is known about the endogenous FRG1 protein. This study uses immunocytochemistry (ICC) and histology to provide insight into FRG1's role in vertebrate muscle development and address its potential involvement in FSHD pathophysiology. In cell culture, primary myoblast/myotube cultures, and mouse and human muscle sections, FRG1 showed distinct nuclear and cytoplasmic localizations and nuclear shuttling assays indicated the subcellular pools of FRG1 are linked. During myoblast differentiation, FRG1's subcellular distribution changed dramatically with FRG1 eventually associating with the matured Z-discs. This Z-disc localization was confirmed using isolated mouse myofibers and found to be maintained in adult human skeletal muscle biopsies. Thus, FRG1 is not likely involved in the initial assembly and alignment of the Z-disc but may be involved in sarcomere maintenance or signaling. Further analysis of human tissue showed FRG1 is strongly expressed in arteries, veins, and capillaries, the other prominently affected tissue in FSHD. Overall, we show that in mammalian cells, FRG1 is a dynamic nuclear and cytoplasmic protein, however in muscle, FRG1 is also a developmentally regulated sarcomeric protein suggesting FRG1 may perform a muscle-specific function. Thus, FRG1 is the only FSHD candidate protein linked to the muscle contractile machinery and may address why the musculature and vasculature are specifically susceptible in FSHD.
Collapse
Affiliation(s)
- Meredith L Hanel
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, 601 S. Goodwin Ave, B107 Chemical and Life Sciences Laboratory, Urbana, IL 61801, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Sato M, Mizoro Y, Atobe Y, Fujimoto Y, Yamaguchi Y, Fustin JM, Doi M, Okamura H. Transportin 1 in the mouse brain: Appearance in regions of neurogenesis, cerebrospinal fluid production/sensing, and circadian clock. J Comp Neurol 2011; 519:1770-80. [DOI: 10.1002/cne.22600] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
27
|
JKTBP1 Is Involved in Stabilization and IRES-Dependent Translation of NRF mRNAs by Binding to 5′ and 3′ Untranslated Regions. J Mol Biol 2011; 407:492-504. [DOI: 10.1016/j.jmb.2011.01.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 01/26/2011] [Accepted: 01/27/2011] [Indexed: 11/15/2022]
|
28
|
Chook YM, Süel KE. Nuclear import by karyopherin-βs: recognition and inhibition. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:1593-606. [PMID: 21029754 DOI: 10.1016/j.bbamcr.2010.10.014] [Citation(s) in RCA: 309] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 10/06/2010] [Accepted: 10/19/2010] [Indexed: 01/24/2023]
Abstract
Proteins in the karyopherin-β family mediate the majority of macromolecular transport between the nucleus and the cytoplasm. Eleven of the 19 known human karyopherin-βs and 10 of the 14S. cerevisiae karyopherin-βs mediate nuclear import through recognition of nuclear localization signals or NLSs in their cargos. This receptor-mediated process is essential to cellular viability as proteins are translated in the cytoplasm but many have functional roles in the nucleus. Many known karyopherin-β-cargo interactions were discovered through studies of the individual cargos rather than the karyopherins, and this information is thus widely scattered in the literature. We consolidate information about cargos that are directly recognized by import-karyopherin-βs and review common characteristics or lack thereof among cargos of different import pathways. Knowledge of karyopherin-β-cargo interactions is also critical for the development of nuclear import inhibitors and the understanding of their mechanisms of inhibition. This article is part of a Special Issue entitled: Regulation of Signaling and Cellular Fate through Modulation of Nuclear Protein Import.
Collapse
Affiliation(s)
- Yuh Min Chook
- Department of Pharmacology, University of Texas Southerwestern Medical Center, Dallas, TX 75206, USA.
| | | |
Collapse
|
29
|
Xu D, Farmer A, Chook YM. Recognition of nuclear targeting signals by Karyopherin-β proteins. Curr Opin Struct Biol 2010; 20:782-90. [PMID: 20951026 DOI: 10.1016/j.sbi.2010.09.008] [Citation(s) in RCA: 171] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 09/10/2010] [Accepted: 09/15/2010] [Indexed: 12/22/2022]
Abstract
The Karyopherin-β family of nuclear transport factors mediates the majority of nucleocytoplasmic transport. Although each of the 19 Karyopherin-βs transports unique sets of cargos, only three classes of nuclear localization and export signals, or NLSs and NESs, have been characterized. The short basic classical-NLS was first discovered in the 1980s and their karyopherin-bound structures were first reported more than 10 years ago. More recently, structural and biophysical studies of Karyopherin-β2-cargo complexes led to definition of the complex and diverse PY-NLS. Structural knowledge of the leucine-rich NES is finally available more than 10 years after the discovery of its recognition by the exportin CRM1. We review recent findings relating to how these three classes of nuclear targeting signals are recognized by their Karyopherin-β nuclear transport factors.
Collapse
Affiliation(s)
- Darui Xu
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9041, USA
| | | | | |
Collapse
|
30
|
A glycine-rich domain of hnRNP H/F promotes nucleocytoplasmic shuttling and nuclear import through an interaction with transportin 1. Mol Cell Biol 2010; 30:2552-62. [PMID: 20308327 DOI: 10.1128/mcb.00230-09] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Heterogeneous nuclear ribonucleoprotein (hnRNP) H and F are members of a closely related subfamily of hnRNP proteins that are implicated in many aspects of RNA processing. hnRNP H and F are alternative splicing factors for numerous U2- and U12-dependent introns. The proteins have three RNA binding domains and two glycine-rich domains and localize to both the nucleus and cytoplasm, but little is known about which domains govern subcellular localization or splicing activity. We show here that the central glycine-tyrosine-arginine-rich (GYR) domain is responsible for nuclear localization, and a nonclassical nuclear localization signal (NLS) was mapped to a short, highly conserved sequence whose activity was compromised by point mutations. Glutathione S-transferase (GST) pulldown assays demonstrated that the hnRNP H NLS interacts with the import receptor transportin 1. Finally, we show that hnRNP H/F are transcription-dependent shuttling proteins. Collectively, the results suggest that hnRNP H and F are GYR domain-dependent shuttling proteins whose posttranslational modifications may alter nuclear localization and hence function.
Collapse
|
31
|
Kroll TT, Swenson LB, Hartland EI, Snedden DD, Goodson HV, Huber PW. Interactions of 40LoVe within the ribonucleoprotein complex that forms on the localization element of Xenopus Vg1 mRNA. Mech Dev 2009; 126:523-38. [PMID: 19345262 DOI: 10.1016/j.mod.2009.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 03/06/2009] [Accepted: 03/24/2009] [Indexed: 02/07/2023]
Abstract
Proline rich RNA-binding protein (Prrp), which associates with mRNAs that employ the late pathway for localization in Xenopus oocytes, was used as bait in a yeast two-hybrid screen of an expression library. Several independent clones were recovered that correspond to a paralog of 40LoVe, a factor required for proper localization of Vg1 mRNA to the vegetal cortex. 40LoVe is present in at least three alternatively spliced isoforms; however, only one, corresponding to the variant identified in the two-hybrid screen, can be crosslinked to Vg1 mRNA. In vitro binding assays revealed that 40LoVe has high affinity for RNA, but exhibits little binding specificity on its own. Nonetheless, it was only found associated with localized mRNAs in oocytes. 40LoVe also interacts directly with VgRBP71 and VgRBP60/hnRNP I; it is the latter factor that likely determines the binding specificity of 40LoVe. Initially, 40LoVe binds to Vg1 mRNA in the nucleus and remains with the RNA in the cytoplasm. Immunohistochemical staining of oocytes shows that the protein is distributed between the nucleus and cytoplasm, consistent with nucleocytoplasmic shuttling activity. 40LoVe is excluded from the mitochondrial cloud, which is used by RNAs that localize through the early (METRO) pathway in stage I oocytes; nonetheless, it is associated with at least some early pathway RNAs during later stages of oogenesis. A phylogenetic analysis of 2xRBD hnRNP proteins combined with other experimental evidence suggests that 40LoVe is a distant homolog of Drosophila Squid.
Collapse
Affiliation(s)
- Todd T Kroll
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | | | | | |
Collapse
|
32
|
Li H, Chen W, Zhou Y, Abidi P, Sharpe O, Robinson WH, Kraemer FB, Liu J. Identification of mRNA binding proteins that regulate the stability of LDL receptor mRNA through AU-rich elements. J Lipid Res 2009; 50:820-31. [PMID: 19141871 DOI: 10.1194/jlr.m800375-jlr200] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The 3'untranslated region (UTR) of human LDL receptor (LDLR) mRNA contains three AU-rich elements (AREs) responsible for rapid mRNA turnover and mediates the stabilization induced by berberine (BBR). However, the identities of the specific RNA binding proteins involved in the regulation of LDLR mRNA stability at the steady state level or upon BBR treatment are unknown. By conducting small interfering RNA library screenings, biotinylated RNA pull-down, mass spectrometry analysis, and functional assays, we now identify heterogeneous nuclear ribonucleoprotein D (hnRNP D), hnRNP I, and KH-type splicing regulatory protein (KSRP) as key modulators of LDLR mRNA stability in liver cells. We show that hnRNP D, I, and KSRP interact with AREs of the LDLR 3'UTR with sequence specificity. Silencing the expression of these proteins increased LDLR mRNA and protein levels. We further demonstrate that BBR-induced mRNA stabilization involves hnRNP I and KSRP, as their cellular depletions abolished the BBR effect and BBR treatment reduced the binding of hnRNP I and KSRP to the LDLR mRNA 3'UTR. These new findings demonstrate that LDLR mRNA stability is controlled by a group of ARE binding proteins, including hnRNP D, hnRNP I, and KSRP. Our results suggest that interference with the ability of destabilizing ARE binding proteins to interact with LDLR-ARE motifs is likely a mechanism for regulating LDLR expression by compounds such as BBR and perhaps others.
Collapse
Affiliation(s)
- Hai Li
- Department of Veterans Affairs, Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Süel KE, Gu H, Chook YM. Modular organization and combinatorial energetics of proline-tyrosine nuclear localization signals. PLoS Biol 2008; 6:e137. [PMID: 18532879 PMCID: PMC2408616 DOI: 10.1371/journal.pbio.0060137] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Accepted: 04/23/2008] [Indexed: 01/21/2023] Open
Abstract
Proline–tyrosine nuclear localization signals (PY-NLSs) are recognized and transported into the nucleus by human Karyopherin (Kap) β2/Transportin and yeast Kap104p. Multipartite PY-NLSs are highly diverse in sequence and structure, share a common C-terminal R/H/KX2–5PY motif, and can be subdivided into hydrophobic and basic subclasses based on loose N-terminal sequence motifs. PY-NLS variability is consistent with weak consensus motifs, but such diversity potentially renders comprehensive genome-scale searches intractable. Here, we use yeast Kap104p as a model system to understand the energetic organization of this NLS. First, we show that Kap104p substrates contain PY-NLSs, demonstrating their generality across eukaryotes. Previously reported Kapβ2–NLS structures explain Kap104p specificity for the basic PY-NLS. More importantly, thermodynamic analyses revealed physical properties that govern PY-NLS binding affinity: (1) PY-NLSs contain three energetically significant linear epitopes, (2) each epitope accommodates substantial sequence diversity, within defined limits, (3) the epitopes are energetically quasi-independent, and (4) a given linear epitope can contribute differently to total binding energy in different PY-NLSs, amplifying signal diversity through combinatorial mixing of energetically weak and strong motifs. The modular organization of the PY-NLS coupled with its combinatorial energetics lays a path to decode this diverse and evolvable signal for future comprehensive genome-scale identification of nuclear import substrates. To travel between the cytoplasm and nucleus, proteins rely on a family of transport proteins known as the karyopherinβ family. Karyopherinβ2, the human version of a family member, recognizes cargo proteins containing a class of nuclear localization signal known as the PY-NLS. The yeast homolog of Karyopherinβ2, Kap104p, also recognizes PY-NLSs, indicating that this pathway has been conserved between evolutionarily distant species. We mutated residues in the PY-NLSs of two Kap104p cargo proteins and analyzed how tightly these mutants bound Kap104p. These experiments revealed three PY-NLS regions, or epitopes, that are important for binding Kap104p. Each epitope is composed of amino acids that vary between cargoes. The epitopes are energetically independent and bind Kap104p with varying strengths in different PY-NLSs, such that mutating the epitope of one PY-NLS may mistakenly direct cargo to the cytoplasm, while a similar mutation in a different PY-NLS has little effect on cargo localization. This flexible, energetically modular, and combinatorial architecture of PY-NLSs may confer higher tolerance to mutations, but it also allows greater sequence diversity, making prediction of new PY-NLSs difficult. The characteristics of PY-NLSs reported here will assist in the identification of new Kap104p cargoes. And the approach used may be applicable to other biological recognition pathways. PY-nuclear localization signals contain three binding regions that are not closely related in sequence and are energetically quasi-independent. These modular epitopes can contribute differently to the total binding energy in different signals, to tune their affinity for binding to the carrier protein Karyopherinβ2/Kap104p, and also to amplify signal diversity.
Collapse
Affiliation(s)
- Katherine E Süel
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Hongmei Gu
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Yuh Min Chook
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
34
|
Weitzdörfer R, Höger H, Burda G, Pollak A, Lubec G. Differences in Hippocampal Protein Expression at 3 Days, 3 Weeks, and 3 Months Following Induction of Perinatal Asphyxia in the Rat. J Proteome Res 2008; 7:1945-52. [DOI: 10.1021/pr700835y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Rachel Weitzdörfer
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| | - Harald Höger
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| | - Gudrun Burda
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| | - Arnold Pollak
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| | - Gert Lubec
- Department of Pediatrics, Medical University of Vienna, Waehringer Guertel 18, 1090 Vienna, Austria, and Division for Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, 2325 Himberg, Austria
| |
Collapse
|
35
|
Imasaki T, Shimizu T, Hashimoto H, Hidaka Y, Kose S, Imamoto N, Yamada M, Sato M. Structural basis for substrate recognition and dissociation by human transportin 1. Mol Cell 2008; 28:57-67. [PMID: 17936704 DOI: 10.1016/j.molcel.2007.08.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 06/22/2007] [Accepted: 08/06/2007] [Indexed: 01/29/2023]
Abstract
Transportin 1 (Trn1) is a transport receptor that transports substrates from the cytoplasm to the nucleus through nuclear pore complexes by recognizing nuclear localization signals (NLSs). Here we describe four crystal structures of human Trn1 in a substrate-free form as well as in the complex with three NLSs (hnRNP D, JKTBP, and TAP, respectively). Our data have revealed that (1) Trn1 has two sites for binding NLSs, one with high affinity (site A) and one with low affinity (site B), and NLS interaction at site B controls overall binding affinity for Trn1; (2) Trn1 recognizes the NLSs at site A followed by conformational change at site B to interact with the NLSs; and (3) a long flexible loop, characteristic of Trn1, interacts with site B, thereby displacing transport substrate in the nucleus. These studies provide deep understanding of substrate recognition and dissociation by Trn1 in import pathways.
Collapse
Affiliation(s)
- Tsuyoshi Imasaki
- Field of Supramolecular Biology, International Graduate School of Arts and Sciences, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | | | | | | | | | | | | | | |
Collapse
|
36
|
David PS, Tanveer R, Port JD. FRET-detectable interactions between the ARE binding proteins, HuR and p37AUF1. RNA (NEW YORK, N.Y.) 2007; 13:1453-68. [PMID: 17626845 PMCID: PMC1950754 DOI: 10.1261/rna.501707] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
A number of highly regulated gene classes are regulated post-transcriptionally at the level of mRNA stability. A central feature in these mRNAs is the presence of A+U-rich elements (ARE) within their 3' UTRs. Two ARE binding proteins, HuR and AUF1, are associated with mRNA stabilization and destabilization, respectively. Previous studies have demonstrated homomultimerization of each protein and the capacity to bind simultaneous or competitively to a single ARE. To investigate this possibility further, cell biological and biophysical approaches were undertaken. Protein-protein interaction was monitored by fluorescence resonance energy transfer (FRET) and by immunocytochemistry in live and fixed cells using fluorescently labeled CFP/YFP fusion proteins of HuR and p37AUF1. Strong nuclear FRET between HuR/HuR and AUF1/AUF1 homodimers as well as HuR/AUF1 heterodimers was observed. Treatment with the MAP kinase activator, anisomycin, which commonly stabilizes ARE-containing mRNAs, caused rapid nuclear to cytoplasmic shuttling of HuR. AUF1 also underwent shuttling, but on a longer time scale. After shuttling, HuR/HuR, AUF1/AUF1, and HuR/AUF1, FRET was also observed in the cytoplasm. In further studies, arsenite rapidly induced the formation of stress granules containing HuR and TIA-1 but not AUF1. The current studies demonstrate that two mRNA binding proteins, HuR and AUF1, are colocalized and are capable of functional interaction in both the nucleus and cytoplasm. FRET-based detection of AUF1/HuR interaction may serve as a basis of opening up new dimensions in delineating the functional interaction of mRNA binding proteins with RNA turnover.
Collapse
Affiliation(s)
- Pamela S David
- Department of Medicine, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | |
Collapse
|
37
|
Barrandon C, Bonnet F, Nguyen VT, Labas V, Bensaude O. The transcription-dependent dissociation of P-TEFb-HEXIM1-7SK RNA relies upon formation of hnRNP-7SK RNA complexes. Mol Cell Biol 2007; 27:6996-7006. [PMID: 17709395 PMCID: PMC2168891 DOI: 10.1128/mcb.00975-07] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The positive transcription elongation factor P-TEFb controls the elongation of transcription by RNA polymerase II. P-TEFb is inactivated upon binding to HEXIM1 or HEXIM2 proteins associated with a noncoding RNA, 7SK. In response to the inhibition of transcription, 7SK RNA, as well as HEXIM proteins, is released by an unknown mechanism and P-TEFb is activated. New partners of 7SK RNA were searched for as potential players in this feedback process. A subset of heterogeneous ribonuclear proteins, hnRNPs Q and R and hnRNPs A1 and A2, were thus identified as major 7SK RNA-associated proteins. The degree of association of 7SK RNA with these hnRNPs increased when P-TEFb-HEXIM1-7SK was dissociated following the inhibition of transcription or HEXIM1 knockdown. This finding suggested that 7SK RNA shuttles from HEXIM1-P-TEFb complexes to hnRNPs. The transcription-dependent dissociation of P-TEFb-HEXIM1-7SK complexes was attenuated when both hnRNPs A1 and A2 were knocked down by small interfering RNA. As hnRNPs are known to interact transiently with RNA while it is synthesized, hnRNPs released from nascent transcripts may trap 7SK RNA and thereby contribute to the activation of P-TEFb.
Collapse
Affiliation(s)
- Charlotte Barrandon
- UMR 8541 CNRS, Ecole Normale Supérieure, 46 rue d'Ulm, 75230 Paris Cedex 05, France
| | | | | | | | | |
Collapse
|
38
|
Reboll MR, Oumard A, Gazdag AC, Renger I, Ritter B, Schwarzer M, Hauser H, Wood M, Yamada M, Resch K, Nourbakhsh M. NRF IRES activity is mediated by RNA binding protein JKTBP1 and a 14-nt RNA element. RNA (NEW YORK, N.Y.) 2007; 13:1328-40. [PMID: 17592041 PMCID: PMC1924892 DOI: 10.1261/rna.545407] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The mRNA of human NF-kappaB repressing factor (NRF) contains a long 5'-untranslated region (UTR) that directs ribosomes to the downstream start codon by a cap-independent mechanism. Comparison of the nucleotide (nt) sequences of human and mouse NRF mRNAs reveals a high degree of identity throughout a fragment of 150 nt proximal to the start codon. Here, we show that this region constitutes a minimal internal ribosome entry segment (IRES) module. Enzymatic RNA structure analysis reveals a secondary structure model of the NRF IRES module. Point mutation analysis of the module determines a short, 14-nt RNA element (nt 640-653) as a mediator of IRES function. Purification of IRES binding cellular proteins and subsequent ESI/MS/MS sequence analysis led to identification of the RNA-binding protein, JKTBP1. EMSA experiments show that JKTBP1 binds upstream to the 14-nt RNA element in the NRF IRES module (nt 579-639). Over-expression of JKTBP1 significantly enhances activity of the NRF IRES module in dicistronic constructs. Moreover, siRNA experiments demonstrate that down-regulation of endogenous JKTBP1 decreases NRF IRES activity and the level of endogenous NRF protein. The data of this study show that JKTBP1 and the 14-nt element act independently to mediate NRF IRES activity.
Collapse
Affiliation(s)
- Marc René Reboll
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Gene function in early mouse embryonic stem cell differentiation. BMC Genomics 2007; 8:85. [PMID: 17394647 PMCID: PMC1851713 DOI: 10.1186/1471-2164-8-85] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Accepted: 03/29/2007] [Indexed: 12/20/2022] Open
Abstract
Background Little is known about the genes that drive embryonic stem cell differentiation. However, such knowledge is necessary if we are to exploit the therapeutic potential of stem cells. To uncover the genetic determinants of mouse embryonic stem cell (mESC) differentiation, we have generated and analyzed 11-point time-series of DNA microarray data for three biologically equivalent but genetically distinct mESC lines (R1, J1, and V6.5) undergoing undirected differentiation into embryoid bodies (EBs) over a period of two weeks. Results We identified the initial 12 hour period as reflecting the early stages of mESC differentiation and studied probe sets showing consistent changes of gene expression in that period. Gene function analysis indicated significant up-regulation of genes related to regulation of transcription and mRNA splicing, and down-regulation of genes related to intracellular signaling. Phylogenetic analysis indicated that the genes showing the largest expression changes were more likely to have originated in metazoans. The probe sets with the most consistent gene changes in the three cell lines represented 24 down-regulated and 12 up-regulated genes, all with closely related human homologues. Whereas some of these genes are known to be involved in embryonic developmental processes (e.g. Klf4, Otx2, Smn1, Socs3, Tagln, Tdgf1), our analysis points to others (such as transcription factor Phf21a, extracellular matrix related Lama1 and Cyr61, or endoplasmic reticulum related Sc4mol and Scd2) that have not been previously related to mESC function. The majority of identified functions were related to transcriptional regulation, intracellular signaling, and cytoskeleton. Genes involved in other cellular functions important in ESC differentiation such as chromatin remodeling and transmembrane receptors were not observed in this set. Conclusion Our analysis profiles for the first time gene expression at a very early stage of mESC differentiation, and identifies a functional and phylogenetic signature for the genes involved. The data generated constitute a valuable resource for further studies. All DNA microarray data used in this study are available in the StemBase database of stem cell gene expression data [1] and in the NCBI's GEO database.
Collapse
|
40
|
Lee BJ, Cansizoglu AE, Süel KE, Louis TH, Zhang Z, Chook YM. Rules for nuclear localization sequence recognition by karyopherin beta 2. Cell 2006; 126:543-58. [PMID: 16901787 PMCID: PMC3442361 DOI: 10.1016/j.cell.2006.05.049] [Citation(s) in RCA: 435] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Revised: 03/01/2006] [Accepted: 05/30/2006] [Indexed: 10/24/2022]
Abstract
Karyopherinbeta (Kapbeta) proteins bind nuclear localization and export signals (NLSs and NESs) to mediate nucleocytoplasmic trafficking, a process regulated by Ran GTPase through its nucleotide cycle. Diversity and complexity of signals recognized by Kap betas have prevented prediction of new Kap beta substrates. The structure of Kap beta 2 (also known as Transportin) bound to one of its substrates, the NLS of hnRNP A1, that we report here explains the mechanism of substrate displacement by Ran GTPase. Further analyses reveal three rules for NLS recognition by Kap beta 2: NLSs are structurally disordered in free substrates, have overall basic character, and possess a central hydrophobic or basic motif followed by a C-terminal R/H/KX(2-5)PY consensus sequence. We demonstrate the predictive nature of these rules by identifying NLSs in seven previously known Kap beta 2 substrates and uncovering 81 new candidate substrates, confirming five experimentally. These studies define and validate a new NLS that could not be predicted by primary sequence analysis alone.
Collapse
Affiliation(s)
- Brittany J. Lee
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park, Dallas, TX 75390, USA
| | - Ahmet E. Cansizoglu
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park, Dallas, TX 75390, USA
| | - Katherine E. Süel
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park, Dallas, TX 75390, USA
| | - Thomas H. Louis
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park, Dallas, TX 75390, USA
| | - Zichao Zhang
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park, Dallas, TX 75390, USA
| | - Yuh Min Chook
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park, Dallas, TX 75390, USA
- Contact:
| |
Collapse
|
41
|
Hare JF. Intracellular pathways of folded and misfolded amyloid precursor protein degradation. Arch Biochem Biophys 2006; 451:79-90. [PMID: 16764819 DOI: 10.1016/j.abb.2006.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Revised: 05/02/2006] [Accepted: 05/05/2006] [Indexed: 01/19/2023]
Abstract
A number of studies suggest that early events in the maturation of amyloid precursor protein (APP) are important in determining its entry into one of several alternative processing pathways, one of which leads to the toxic protein beta-amyloid (Abeta). In pulse-labeled APP expressing CHO cells two proteolytic systems can degrade newly translated APP: the proteosome and a cysteine protease. When N-glycosylation was inhibited by tunicamycin, the former system is the dominant mechanism of APP degradation. Without tunicamycin present, the cysteine protease is operational: cysteine protease inhibitors completely inhibit APP turnover in cells in which the secretory pathway is interrupted with brefeldin A or when alpha-secretase and endosomal degradation are also pharmacologically blocked. APP immunoprecipitated from cells extracted under mild conditions and labeled in the presence of tunicamycin exhibited greater sensitivity to endoproteinase glu-C (V8) or lys-C than from cells without drug. The V8 fragment missing in tunicamyin treated cells encompassed the KPI inhibitor insertion site but was distinct from the site of N-glycosylation. It is concluded that a conformational change caused by interrupted N-glycosylation shunts newly translated APP into the proteasomal degradation pathway. Pulse-labeled and chased cells showed an additional V8 fragment that was not present in pulsed-labeled cells and was not due to glycosylation since it was also present in cells labeled in the presence of brefeldin. This latter result indicates that an additional, delayed conformational alteration occurs in the endoplasmic reticulum.
Collapse
Affiliation(s)
- James F Hare
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97219, USA.
| |
Collapse
|
42
|
Westmark PR, Shin HC, Westmark CJ, Soltaninassab SR, Reinke EK, Malter JS. Decoy mRNAs reduce beta-amyloid precursor protein mRNA in neuronal cells. Neurobiol Aging 2006; 27:787-96. [PMID: 16672170 DOI: 10.1016/j.neurobiolaging.2006.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 02/26/2006] [Accepted: 03/09/2006] [Indexed: 11/30/2022]
Abstract
Overproduction of amyloid precursor protein (APP) and beta-amyloid likely contribute to neurodegeneration in Alzheimer's disease (AD). In an effort to understand neuronal APP gene regulation, we identified a 52 base element (52sce) immediately downstream from the stop codon that stabilizes APP mRNA. Deletion of this domain drastically destabilized APP mRNAs and reduced APP synthesis in vitro. Chimeric globin-APP mRNAs containing the globin coding sequence fused to the entire APP 3'-UTR, showed regulation similar to full-length APP mRNA. A variety of cytoplasmic lysates contain 52sce RNA binding activity, suggesting cis-trans interactions regulate the element's functionality. Finally, the overexpression of chimeric mRNAs, containing the GFP coding sequence and APP 3'-UTR, dramatically reduced endogenous APP steady-state levels in SH-SY5Y neuroblastoma cells and suggests a novel approach to reduce the amyloid burden in AD patients.
Collapse
Affiliation(s)
- Pamela R Westmark
- Department of Pathology and Laboratory Medicine, Neuroscience Training Program, Waisman Center for Developmental Disabilities and Institute on Aging, University of Wisconsin-Madison, 1500 Highland Avenue, Madison, WI 53705, USA
| | | | | | | | | | | |
Collapse
|
43
|
Czaplinski K, Mattaj IW. 40LoVe interacts with Vg1RBP/Vera and hnRNP I in binding the Vg1-localization element. RNA (NEW YORK, N.Y.) 2006; 12:213-22. [PMID: 16373488 PMCID: PMC1370901 DOI: 10.1261/rna.2820106] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Localizing mRNAs within the cytoplasm gives cells the ability to spatially restrict protein production, a powerful means to regulate gene expression. Localized mRNA is often visible in microscopically observable particles or granules, and the association of mRNA localization with these structures is an indication that particles or granules may be essential to the localization process. Understanding how such structures form will therefore be important for understanding the function of localization RNPs (L-RNPs). We previously identified a novel component of an L-RNP from the Vg1 mRNA from Xenopus oocytes called 40LoVe. 40LoVe interaction with the Vg1-localization element (Vg1LE) was previously shown to be dependent on the VM1 and E2 sequence motifs within the Vg1LE that cross-link to hnRNP I and Vg1RBP/Vera, respectively. We report interaction of these motif-binding proteins with 40LoVe and identify a 40LoVe-Xenopus hnRNP D/AUF1 interaction. We further demonstrate that titration of VM1 and E2 motif binding activity in vivo surprisingly suggests that the motif binding proteins have differing roles during Vg1LE-dependent mRNA localization.
Collapse
|
44
|
Iijima M, Suzuki M, Tanabe A, Nishimura A, Yamada M. Two motifs essential for nuclear import of the hnRNP A1 nucleocytoplasmic shuttling sequence M9 core. FEBS Lett 2006; 580:1365-70. [PMID: 16455081 DOI: 10.1016/j.febslet.2006.01.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2005] [Revised: 12/31/2005] [Accepted: 01/04/2006] [Indexed: 10/25/2022]
Abstract
Heterogeneous nuclear ribonucleoprotein (hnRNP) A1 regulates mRNA genesis. It shuttles between the nucleus and cytoplasm. Its shuttling signal is a 38-residue sequence M9. We studied the nuclear import and export of M9 by mutational analysis. Heterokaryon assay indicated that the 19-residue sequence SNFGPMKGGNFGGRSSGPY (M9 core) is necessary and sufficient for shuttling. Moreover, M9 core mutation revealed that in addition to the hitherto characterized N-terminal motif SNFGPMK, the C-terminal motif PY is crucial for nuclear import as well as for binding to transportin. Key residues of the motifs are conserved in the shuttling signals of hnRNP D and JKTBP.
Collapse
Affiliation(s)
- Megumi Iijima
- Graduate School of Integrated Science, Yokohama City University, 22-2, Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | | | | | | | | |
Collapse
|
45
|
Suzuki M, Iijima M, Nishimura A, Tomozoe Y, Kamei D, Yamada M. Two separate regions essential for nuclear import of the hnRNP D nucleocytoplasmic shuttling sequence. FEBS J 2005; 272:3975-87. [PMID: 16045768 DOI: 10.1111/j.1742-4658.2005.04820.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Heterogeneous nuclear ribonucleoprotein (hnRNP) D/AUF1 functions in mRNA genesis in the nucleus and modulates mRNA decay in the cytoplasm. Although it is primarily nuclear, it shuttles between the nucleus and cytoplasm. We studied the nuclear import and export of the last exon-encoding sequence common to all its isoforms by its expression as a green fluorescent protein-fusion protein in HeLa cells and by heterokaryon assay. The C-terminal 19-residue sequence (SGYGKVSRRGGHQNSYKPY) was identified as an hnRNP D nucleocytoplasmic shuttling sequence (DNS). In vitro nuclear transport using permeabilized cells indicated that nuclear import of DNS is mediated by transportin-1 (Trn-1). DNS accumulation in the nucleus was dependent on Trn-1, Ran, and energy in multiple rounds of nuclear transport. Use of DNS with deletions, alanine scanning mutagenesis and point mutations revealed that two separate regions (the N-terminal seven residues and the C-terminal two residues) are crucial for in vivo and in vitro transport as well as for interaction with Trn-1. The N- and C-terminal motifs are conserved in the shuttling sequences of hnRNP A1 and JKTBP.
Collapse
Affiliation(s)
- Maiko Suzuki
- Graduate School of Integrated Science, Yokohama City University, Yokohama, Japan
| | | | | | | | | | | |
Collapse
|
46
|
ABDUL-GHANI MOHAMMAD, HARTMAN KRISTINL, NGSEE JOHNNYK. Abstrakt interacts with and regulates the expression of sorting nexin-2. J Cell Physiol 2005; 204:210-8. [PMID: 15690390 PMCID: PMC2963638 DOI: 10.1002/jcp.20285] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Protein sorting through vesicular compartments is highly regulated to maintain the integrity and signaling of intracellular organelles in eukaryotic cells. Sorting Nexin-2 (SNX2) is involved in protein sorting in the trans-Golgi network, endosome, and/or lysosome compartments, with loss of function leading to defect in protein sorting and stress on organelles. To investigate the function of SNX2, we have identified the DEAD-box helicase Abstrakt (Abs) as an SNX2-interacting protein. The N-terminal domain of Abs interacts with the phox homology (PX) domain of SNX2 suggesting that PX domains may also participate in protein-protein interaction. Interestingly, both proteins undergo nucleocytoplasmic shuttling, and this process is responsive to serum withdrawal for Abs. Finally, expression of Abs reduced the cellular expression of SNX2 without altering its steady state mRNA levels. This unexpected interaction provides a novel mechanism whereby expression of proteins involved in membrane trafficking could be regulated by an RNA helicase.
Collapse
Affiliation(s)
| | | | - JOHNNY K. NGSEE
- Correspondence to: Johnny K. Ngsee, Ottawa Health Research Institute, 725 Parkdale Avenue, Ottawa, ON, Canada K1Y 4E9.
| |
Collapse
|
47
|
Czaplinski K, Köcher T, Schelder M, Segref A, Wilm M, Mattaj IW. Identification of 40LoVe, a Xenopus hnRNP D family protein involved in localizing a TGF-beta-related mRNA during oogenesis. Dev Cell 2005; 8:505-15. [PMID: 15809033 DOI: 10.1016/j.devcel.2005.01.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2004] [Revised: 01/13/2005] [Accepted: 01/19/2005] [Indexed: 01/10/2023]
Abstract
Asymmetric distribution of cellular components underlies many biological processes, and the localization of mRNAs within domains of the cytoplasm is one important mechanism of establishing and maintaining cellular asymmetry. mRNA localization often involves assembly of large ribonucleoproteins (RNPs) in the cytoplasm. Using an RNA affinity chromatography approach, we investigated localization RNP formation on the vegetal localization element (VLE) of the mRNA encoding Vg1, a Xenopus TGF-beta family member. We identified 40LoVe, an hnRNP D family protein, as a specific VLE binding protein from Xenopus oocytes. Interaction of 40LoVe with the VLE strictly correlates with the ability of the RNA to localize, and antibodies against 40LoVe inhibit vegetal localization in vivo in oocytes. Our results associate an hnRNP D protein with mRNA localization and have implications for several functions mediated by this important protein family.
Collapse
Affiliation(s)
- Kevin Czaplinski
- EMBL Gene Expression Programme, Meyerhofstrasse 1, Heidelberg 69117, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Many genes are expressed in mammalian liver in a sexually dimorphic manner. DNA microarray analysis has shown that growth hormone (GH) and its sex-dependent pattern of pituitary secretion play a major role in establishing the sexually dimorphic patterns of liver gene expression. However, GH may exert effects on protein post-translational modification and nuclear localization that are not reflected at the mRNA level. To investigate these potential effects of GH, we used two-dimensional gel electrophoresis followed by LC-MS/MS to: 1) identify rat liver nuclear proteins whose abundance or state of post-translational modification displays sex-dependent differences; and 2) determine the role of the plasma GH profile in establishing these differences. Nuclear extracts prepared from livers of individual male (n=9) and female (n=5) adult rats, and from males given GH by continuous infusion for 7 days to feminize liver gene expression (n=5 rats), were resolved by two-dimensional electrophoresis. Image analysis of SYPRO Ruby-stained gels revealed 165 sexually dimorphic protein spots that differ in normalized volume between male and female groups by >1.5-fold at p<0.05. Sixty of these proteins exhibited female-like changes in spot abundance following continuous GH treatment. Comparison of male and GH-treated male groups revealed 130 proteins that displayed >1.5-fold differences in abundance, with 60 of these GH-responsive spots being sexually dimorphic. Thus, GH plays an important role in establishing the sex-dependent differences in liver nuclear protein content. Twenty-eight of the sexually dimorphic and/or GH-regulated protein spots were identified by LC-MS/MS. Proteins identified include regucalcin, nuclear factor 45, and heterogeneous nuclear ribonucleoproteins A3, D-like, and K, in addition to proteins such as GST, normally associated with cytosolic extracts but also reported to be localized in the nucleus.
Collapse
Affiliation(s)
- Ekaterina V Laz
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, MA 02215, USA
| | | | | |
Collapse
|
49
|
O'Hagan HM, Ljungman M. Efficient NES-dependent protein nuclear export requires ongoing synthesis and export of mRNAs. Exp Cell Res 2004; 297:548-59. [PMID: 15212955 DOI: 10.1016/j.yexcr.2004.03.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2004] [Revised: 03/20/2004] [Indexed: 11/25/2022]
Abstract
The mechanisms regulating nuclear export of proteins are not fully understood. To investigate whether the efficiency of protein nuclear export may depend on ongoing RNA synthesis and/or mRNA nuclear export, we used a microinjection approach with a fluorescent reporter protein containing a nuclear export signal (NES) and scored protein export in human fibroblasts under conditions when the synthesis or export of mRNAs was inhibited. We show that inhibition of transcription significantly attenuated generic NES-dependent nuclear export. Furthermore, digestion of endogenous nuclear RNAs by co-microinjection of RNAse A inhibited NES-dependent nuclear export. Finally, nuclear export of the NES reporter protein was significantly inhibited in cells in which nuclear export of mRNAs had been specifically blocked by microinjection of anti-TAP antibodies or by expression of a dominant negative form of NUP160. These results demonstrate a novel role for ongoing synthesis and export of mRNAs in NES-dependent protein nuclear export.
Collapse
MESH Headings
- ATP-Binding Cassette Transporters
- Active Transport, Cell Nucleus/drug effects
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Cytoplasm/drug effects
- Cytoplasm/metabolism
- Dose-Response Relationship, Drug
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Fluorescent Dyes
- Glutathione Transferase/metabolism
- Green Fluorescent Proteins
- Histocompatibility Antigens Class I/metabolism
- Humans
- Karyopherins/metabolism
- Luminescent Proteins
- Microinjections
- Models, Biological
- Nuclear Localization Signals/metabolism
- Nuclear Pore Complex Proteins/metabolism
- Nuclear Proteins/metabolism
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/biosynthesis
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear
- Recombinant Proteins/metabolism
- Rhodamines
- Ribonuclease, Pancreatic/pharmacology
- Transcription, Genetic/drug effects
- Exportin 1 Protein
Collapse
Affiliation(s)
- Heather M O'Hagan
- Department of Radiation Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
50
|
Tsuchiya N, Fukuda H, Nakashima K, Nagao M, Sugimura T, Nakagama H. LRP130, a single-stranded DNA/RNA-binding protein, localizes at the outer nuclear and endoplasmic reticulum membrane, and interacts with mRNA in vivo. Biochem Biophys Res Commun 2004; 317:736-43. [PMID: 15081402 DOI: 10.1016/j.bbrc.2004.03.103] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Indexed: 11/23/2022]
Abstract
LRP130 (also known as a LRPPRC) is an RNA and single-stranded DNA-binding protein, and recently identified as a candidate gene responsible for the Leigh syndrome, a French-Canadian type cytochrome c oxidase deficiency. However, the biological function of LRP130 still remains largely unresolved. In the present study, we found that the C-terminal half of the mouse LRP130 located within a 120 amino acid sequence (a.a. 845-964) binds to synthetic RNA homopolymers, poly(G), poly(U), and poly(C), as well as r(CUGCC)(6). Assessment of the subcellular localization indicated both nuclear/endoplasmic reticulum (ER) and mitochondrial fractions to be positive. To further analyze the subcellular localization of LRP130, a nuclear/ER fraction was fractionated into the nucleoplasm (NP) and nuclear envelope (NE)/ER, and the latter was further separated into outer nuclear membrane (ONM)/ER and inner nuclear membrane (INM) by treatment with Triton X-100. LRP130 was detectable in all three fractions, and the distribution pattern was in good accordance with that known for ONM/ER proteins. Interestingly, immunostaining of HeLa cells demonstrated nuclear rim staining of LRP130, specifically at the outside of the NE and also at ER, and association of LRP130 with poly(A)(+) RNA was restricted only to the ONM/ER fraction. Overexpression of full-length mouse LRP130 fused with EGFP resulted in nuclear accumulation of poly(A)(+) RNA in HeLa cells. Taking all these results together, it is suggested that LRP130, a novel type of RNA-binding protein, associates with mRNA/mRNP complexes at the outside of NE and ER, and plays a role in control of mRNA metabolisms.
Collapse
Affiliation(s)
- Naoto Tsuchiya
- Biochemistry Division, National Cancer Center Research Institute, 5-1-1 Tsukiji Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | |
Collapse
|