1
|
Sirtori R, Gregoire M, Collins A, Santangelo S, Chatragadda B, Cullen R, Ratti A, Fallini C. Altered nuclear envelope homeostasis is a key pathogenic event in C9ORF72-linked ALS/FTD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578318. [PMID: 38352403 PMCID: PMC10862841 DOI: 10.1101/2024.02.01.578318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
ALS and FTD are complex neurodegenerative disorders that primarily affects motor neurons in the brain and spinal cord, and cortical neurons in the frontal lobe. Although the pathogenesis of ALS/FTD is unclear, recent research spotlights nucleocytoplasmic transport impairment, DNA damage, and nuclear abnormalities as drivers of neuronal death. In this study, we show that loss of nuclear envelope (NE) integrity is a key pathology associated with nuclear pore complex (NPC) injury in C9ORF72 mutant neurons. Importantly, we show that mechanical stresses generated by cytoskeletal forces on the NE can lead to NPC injury, loss of nuclear integrity, and accumulation of DNA damage. Importantly, we demonstrate that restoring NE tensional homeostasis, by disconnecting the nucleus from the cytoskeleton, can rescue NPC injury and reduce DNA damage in C9ORF72 mutant cells. Together, our data suggest that modulation of NE homeostasis and repair may represent a novel and promising therapeutic target for ALS/FTD.
Collapse
|
2
|
Giannattasio T, Testa E, Palombo R, Chellini L, Franceschini F, Crevenna Á, Petkov PM, Paronetto MP, Barchi M. The RNA-binding protein FUS/TLS interacts with SPO11 and PRDM9 and localize at meiotic recombination hotspots. Cell Mol Life Sci 2023; 80:107. [PMID: 36967403 PMCID: PMC10040399 DOI: 10.1007/s00018-023-04744-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/29/2023]
Abstract
In mammals, meiotic recombination is initiated by the introduction of DNA double strand breaks (DSBs) into narrow segments of the genome, defined as hotspots, which is carried out by the SPO11/TOPOVIBL complex. A major player in the specification of hotspots is PRDM9, a histone methyltransferase that, following sequence-specific DNA binding, generates trimethylation on lysine 4 (H3K4me3) and lysine 36 (H3K36me3) of histone H3, thus defining the hotspots. PRDM9 activity is key to successful meiosis, since in its absence DSBs are redirected to functional sites and synapsis between homologous chromosomes fails. One protein factor recently implicated in guiding PRDM9 activity at hotspots is EWS, a member of the FET family of proteins that also includes TAF15 and FUS/TLS. Here, we demonstrate that FUS/TLS partially colocalizes with PRDM9 on the meiotic chromosome axes, marked by the synaptonemal complex component SYCP3, and physically interacts with PRDM9. Furthermore, we show that FUS/TLS also interacts with REC114, one of the axis-bound SPO11-auxiliary factors essential for DSB formation. This finding suggests that FUS/TLS is a component of the protein complex that promotes the initiation of meiotic recombination. Accordingly, we document that FUS/TLS coimmunoprecipitates with SPO11 in vitro and in vivo. The interaction occurs with both SPO11β and SPO11α splice isoforms, which are believed to play distinct functions in the formation of DSBs in autosomes and male sex chromosomes, respectively. Finally, using chromatin immunoprecipitation experiments, we show that FUS/TLS is localized at H3K4me3-marked hotspots in autosomes and in the pseudo-autosomal region, the site of genetic exchange between the XY chromosomes.
Collapse
Affiliation(s)
- Teresa Giannattasio
- University of Rome "Tor Vergata", Section of Anatomy, Via Montpellier, 1, 00133, Rome, Italy
| | - Erika Testa
- University of Rome "Tor Vergata", Section of Anatomy, Via Montpellier, 1, 00133, Rome, Italy
| | - Ramona Palombo
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143, Rome, Italy
| | - Lidia Chellini
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143, Rome, Italy
| | - Flavia Franceschini
- University of Rome "Tor Vergata", Section of Anatomy, Via Montpellier, 1, 00133, Rome, Italy
| | - Álvaro Crevenna
- European Molecular Biology Laboratory, Neurobiology and Epigenetics Unit, Monterotondo, Italy
| | | | - Maria Paola Paronetto
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143, Rome, Italy.
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 6, 00135, Rome, Italy.
| | - Marco Barchi
- University of Rome "Tor Vergata", Section of Anatomy, Via Montpellier, 1, 00133, Rome, Italy.
| |
Collapse
|
3
|
Gleixner AM, Verdone BM, Otte CG, Anderson EN, Ramesh N, Shapiro OR, Gale JR, Mauna JC, Mann JR, Copley KE, Daley EL, Ortega JA, Cicardi ME, Kiskinis E, Kofler J, Pandey UB, Trotti D, Donnelly CJ. NUP62 localizes to ALS/FTLD pathological assemblies and contributes to TDP-43 insolubility. Nat Commun 2022; 13:3380. [PMID: 35697676 PMCID: PMC9192689 DOI: 10.1038/s41467-022-31098-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 06/03/2022] [Indexed: 01/12/2023] Open
Abstract
A G4C2 hexanucleotide repeat expansion in the C9orf72 gene is the most common genetic cause of ALS and FTLD (C9-ALS/FTLD) with cytoplasmic TDP-43 inclusions observed in regions of neurodegeneration. The accumulation of repetitive RNAs and dipeptide repeat protein (DPR) are two proposed mechanisms of toxicity in C9-ALS/FTLD and linked to impaired nucleocytoplasmic transport. Nucleocytoplasmic transport is regulated by the phenylalanine-glycine nucleoporins (FG nups) that comprise the nuclear pore complex (NPC) permeability barrier. However, the relationship between FG nups and TDP-43 pathology remains elusive. Our studies show that nuclear depletion and cytoplasmic mislocalization of one FG nup, NUP62, is linked to TDP-43 mislocalization in C9-ALS/FTLD iPSC neurons. Poly-glycine arginine (GR) DPR accumulation initiates the formation of cytoplasmic RNA granules that recruit NUP62 and TDP-43. Cytoplasmic NUP62 and TDP-43 interactions promotes their insolubility and NUP62:TDP-43 inclusions are frequently found in C9orf72 ALS/FTLD as well as sporadic ALS/FTLD postmortem CNS tissue. Our findings indicate NUP62 cytoplasmic mislocalization contributes to TDP-43 proteinopathy in ALS/FTLD.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
| | - Brandie Morris Verdone
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Charlton G Otte
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
- Physician Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric N Anderson
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Nandini Ramesh
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Olivia R Shapiro
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
| | - Jenna R Gale
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
| | - Jocelyn C Mauna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
| | - Jacob R Mann
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katie E Copley
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
| | - Elizabeth L Daley
- The Ken & Ruth Davee Department of Neurology, Northwestern University of Feinberg School of Medicine, Chicago, IL, USA
| | - Juan A Ortega
- The Ken & Ruth Davee Department of Neurology, Northwestern University of Feinberg School of Medicine, Chicago, IL, USA
| | - Maria Elena Cicardi
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Northwestern University of Feinberg School of Medicine, Chicago, IL, USA
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Julia Kofler
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Udai B Pandey
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Davide Trotti
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christopher J Donnelly
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Baron DM, Fenton AR, Saez-Atienzar S, Giampetruzzi A, Sreeram A, Shankaracharya, Keagle PJ, Doocy VR, Smith NJ, Danielson EW, Andresano M, McCormack MC, Garcia J, Bercier V, Van Den Bosch L, Brent JR, Fallini C, Traynor BJ, Holzbaur ELF, Landers JE. ALS-associated KIF5A mutations abolish autoinhibition resulting in a toxic gain of function. Cell Rep 2022; 39:110598. [PMID: 35385738 PMCID: PMC9134378 DOI: 10.1016/j.celrep.2022.110598] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/02/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Understanding the pathogenic mechanisms of disease mutations is critical to advancing treatments. ALS-associated mutations in the gene encoding the microtubule motor KIF5A result in skipping of exon 27 (KIF5AΔExon27) and the encoding of a protein with a novel 39 amino acid residue C-terminal sequence. Here, we report that expression of ALS-linked mutant KIF5A results in dysregulated motor activity, cellular mislocalization, altered axonal transport, and decreased neuronal survival. Single-molecule analysis revealed that the altered C terminus of mutant KIF5A results in a constitutively active state. Furthermore, mutant KIF5A possesses altered protein and RNA interactions and its expression results in altered gene expression/splicing. Taken together, our data support the hypothesis that causative ALS mutations result in a toxic gain of function in the intracellular motor KIF5A that disrupts intracellular trafficking and neuronal homeostasis.
Collapse
Affiliation(s)
- Desiree M Baron
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Adam R Fenton
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sara Saez-Atienzar
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Anthony Giampetruzzi
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Aparna Sreeram
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Shankaracharya
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Pamela J Keagle
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Victoria R Doocy
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Nathan J Smith
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Eric W Danielson
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Megan Andresano
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Mary C McCormack
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jaqueline Garcia
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Valérie Bercier
- KU Leuven-University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven-University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Jonathan R Brent
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Claudia Fallini
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA; Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA; Therapeutic Development Branch, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
5
|
Herbette M, Wei X, Chang CH, Larracuente AM, Loppin B, Dubruille R. Distinct spermiogenic phenotypes underlie sperm elimination in the Segregation Distorter meiotic drive system. PLoS Genet 2021; 17:e1009662. [PMID: 34228705 PMCID: PMC8284685 DOI: 10.1371/journal.pgen.1009662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/16/2021] [Accepted: 06/10/2021] [Indexed: 12/28/2022] Open
Abstract
Segregation Distorter (SD) is a male meiotic drive system in Drosophila melanogaster. Males heterozygous for a selfish SD chromosome rarely transmit the homologous SD+ chromosome. It is well established that distortion results from an interaction between Sd, the primary distorting locus on the SD chromosome and its target, a satellite DNA called Rsp, on the SD+ chromosome. However, the molecular and cellular mechanisms leading to post-meiotic SD+ sperm elimination remain unclear. Here we show that SD/SD+ males of different genotypes but with similarly strong degrees of distortion have distinct spermiogenic phenotypes. In some genotypes, SD+ spermatids fail to fully incorporate protamines after the removal of histones, and degenerate during the individualization stage of spermiogenesis. In contrast, in other SD/SD+ genotypes, protamine incorporation appears less disturbed, yet spermatid nuclei are abnormally compacted, and mature sperm nuclei are eventually released in the seminal vesicle. Our analyses of different SD+ chromosomes suggest that the severity of the spermiogenic defects associates with the copy number of the Rsp satellite. We propose that when Rsp copy number is very high (> 2000), spermatid nuclear compaction defects reach a threshold that triggers a checkpoint controlling sperm chromatin quality to eliminate abnormal spermatids during individualization.
Collapse
Affiliation(s)
- Marion Herbette
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS UMR 5239, École Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Xiaolu Wei
- University of Rochester Medical Center, Department of Biomedical Genetics, Rochester, New York, United States of America
| | - Ching-Ho Chang
- University of Rochester Department of Biology, Rochester, New York, United States of America
| | - Amanda M. Larracuente
- University of Rochester Department of Biology, Rochester, New York, United States of America
| | - Benjamin Loppin
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS UMR 5239, École Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Raphaëlle Dubruille
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS UMR 5239, École Normale Supérieure de Lyon, University of Lyon, Lyon, France
| |
Collapse
|
6
|
Zhang M, Yang C, Zhu M, Qian L, Luo Y, Cheng H, Geng R, Xu X, Qian C, Liu Y. Saturated fatty acids entrap PDX1 in stress granules and impede islet beta cell function. Diabetologia 2021; 64:1144-1157. [PMID: 33569632 DOI: 10.1007/s00125-021-05389-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/16/2020] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Failure of pancreatic and duodenal homeobox factor 1 (PDX1) to localise in the nucleus of islet beta cells under high-fat diet (HFD) conditions may be an early functional defect that contributes to beta cell failure in type 2 diabetes; however, the mechanism of PDX1 intracellular mislocalisation is unclear. Stress granules (SGs) are membrane-less cytoplasmic structures formed under stress that impair nucleocytoplasmic transport by sequestering nucleocytoplasmic transport factors and components of the nuclear pore complex. In this study, we investigated the stimulators that trigger SG formation in islet beta cells and the effects of SGs on PDX1 localisation and beta cell function. METHODS The effect of palmitic acid (PA) on nucleocytoplasmic transport was investigated by using two reporters, S-tdTomato and S-GFP. SG assembly in rat insulinoma cell line INS1 cells, human islets under PA stress, and the pancreas of diet-induced obese mice was analysed using immunofluorescence and immunoblotting. SG protein components were identified through mass spectrometry. SG formation was blocked by specific inhibitors or genetic deletion of essential SG proteins, and then PDX1 localisation and beta cell function were investigated in vitro and in vivo. RESULTS We showed that saturated fatty acids (SFAs) are endogenous stressors that disrupted nucleocytoplasmic transport and stimulated SG formation in pancreatic beta cells. Using mass spectrometry approaches, we revealed that several nucleocytoplasmic transport factors and PDX1 were localised to SGs after SFA treatment, which inhibited glucose-induced insulin secretion. Furthermore, we found that SFAs induced SG formation in a phosphoinositide 3-kinase (PI3K)/eukaryotic translation initiation factor 2α (EIF2α) dependent manner. Disruption of SG assembly by PI3K/EIF2α inhibitors or genetic deletion of T cell restricted intracellular antigen 1 (TIA1) in pancreatic beta cells effectively suppressed PA-induced PDX1 mislocalisation and ameliorated HFD-mediated beta cell dysfunction. CONCLUSIONS/INTERPRETATION Our findings suggest a link between SG formation and beta cell dysfunction in the presence of SFAs. Preventing SG formation may be a potential therapeutic strategy for treating obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Mu Zhang
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Chunjie Yang
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Meng Zhu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Li Qian
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Luo
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Huimin Cheng
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Rong Geng
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaojun Xu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| | - Cheng Qian
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| | - Yu Liu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
7
|
Lee J, Park J, Kim JH, Lee G, Park TE, Yoon KJ, Kim YK, Lim C. LSM12-EPAC1 defines a neuroprotective pathway that sustains the nucleocytoplasmic RAN gradient. PLoS Biol 2020; 18:e3001002. [PMID: 33362237 PMCID: PMC7757817 DOI: 10.1371/journal.pbio.3001002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Nucleocytoplasmic transport (NCT) defects have been implicated in neurodegenerative diseases such as C9ORF72-associated amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). Here, we identify a neuroprotective pathway of like-Sm protein 12 (LSM12) and exchange protein directly activated by cyclic AMP 1 (EPAC1) that sustains the nucleocytoplasmic RAN gradient and thereby suppresses NCT dysfunction by the C9ORF72-derived poly(glycine-arginine) protein. LSM12 depletion in human neuroblastoma cells aggravated poly(GR)-induced impairment of NCT and nuclear integrity while promoting the nuclear accumulation of poly(GR) granules. In fact, LSM12 posttranscriptionally up-regulated EPAC1 expression, whereas EPAC1 overexpression rescued the RAN gradient and NCT defects in LSM12-deleted cells. C9-ALS patient-derived neurons differentiated from induced pluripotent stem cells (C9-ALS iPSNs) displayed low expression of LSM12 and EPAC1. Lentiviral overexpression of LSM12 or EPAC1 indeed restored the RAN gradient, mitigated the pathogenic mislocalization of TDP-43, and suppressed caspase-3 activation for apoptosis in C9-ALS iPSNs. EPAC1 depletion biochemically dissociated RAN-importin β1 from the cytoplasmic nuclear pore complex, thereby dissipating the nucleocytoplasmic RAN gradient essential for NCT. These findings define the LSM12-EPAC1 pathway as an important suppressor of the NCT-related pathologies in C9-ALS/FTD. A post-transcriptional circuit comprising LSM12 and EPAC1 suppresses neurodegenerative pathologies in C9ORF72-associated amyotrophic lateral sclerosis by establishing the RAN gradient and sustaining nucleocytoplasmic transport.
Collapse
Affiliation(s)
- Jongbo Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jumin Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Ji-hyung Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Giwook Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Tae-Eun Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yoon Ki Kim
- Creative Research Initiatives Center for Molecular Biology of Translation, Korea University, Seoul, Republic of Korea
- Division of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Chunghun Lim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- * E-mail:
| |
Collapse
|
8
|
Chang CC, Hsia KC. More than a zip code: global modulation of cellular function by nuclear localization signals. FEBS J 2020; 288:5569-5585. [PMID: 33296547 DOI: 10.1111/febs.15659] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/27/2022]
Abstract
Extensive structural and functional studies have been carried out in the field of nucleocytoplasmic transport. Nuclear transport factors, such as Importin-α/-β, recognize nuclear localization signals (NLSs) on cargo, and together with the small GTPase Ran, facilitate their nuclear localization. However, it is now emerging that binding of nuclear transport factors to NLSs not only mediates nuclear transport but also contributes to a variety of cellular functions in eukaryotes. Here, we describe recent advances that reveal how NLSs facilitate diverse cellular functions beyond nuclear transport activity. We review separately NLS-mediated regulatory mechanisms at different levels of biological organization, including (a) assembly of higher-order structures; (b) cellular organelle dynamics; and (c) modulation of cellular stress responses and viral infections. Finally, we provide mechanistic insights into how NLSs can regulate such a broad range of functions via their structural and biochemical properties.
Collapse
Affiliation(s)
- Chih-Chia Chang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Kuo-Chiang Hsia
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.,Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
9
|
Colón-Bolea P, García-Gómez R, Shackleton S, Crespo P, Bustelo XR, Casar B. RAC1 induces nuclear alterations through the LINC complex to enhance melanoma invasiveness. Mol Biol Cell 2020; 31:2768-2778. [PMID: 33026942 PMCID: PMC7851868 DOI: 10.1091/mbc.e20-02-0127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
RHO GTPases are key regulators of the cytoskeletal architecture, which impact a broad range of biological processes in malignant cells including motility, invasion, and metastasis, thereby affecting tumor progression. One of the constraints during cell migration is the diameter of the pores through which cells pass. In this respect, the size and shape of the nucleus pose a major limitation. Therefore, enhanced nuclear plasticity can promote cell migration. Nuclear morphology is determined in part through the cytoskeleton, which connects to the nucleoskeleton through the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex. Here, we unravel the role of RAC1 as an orchestrator of nuclear morphology in melanoma cells. We demonstrate that activated RAC1 promotes nuclear alterations through its effector PAK1 and the tubulin cytoskeleton, thereby enhancing migration and intravasation of melanoma cells. Disruption of the LINC complex prevented RAC1-induced nuclear alterations and the invasive properties of melanoma cells. Thus, RAC1 induces nuclear morphology alterations through microtubules and the LINC complex to promote an invasive phenotype in melanoma cells.
Collapse
Affiliation(s)
- Paula Colón-Bolea
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Cantabria, Santander 39011, Spain
| | - Rocío García-Gómez
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Cantabria, Santander 39011, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Sue Shackleton
- Department of Biochemistry, University of Leicester, Leicester LE1 9HM, UK
| | - Piero Crespo
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Cantabria, Santander 39011, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Xosé R Bustelo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid 28029, Spain.,Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer (CSIC), Universidad de Salamanca, Salamanca 37007, Spain
| | - Berta Casar
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Cantabria, Santander 39011, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
10
|
Keegan SE, Hughes SC. Role of nuclear-cytoplasmic protein localization during Drosophila neuroblast development. Genome 2020; 64:75-85. [PMID: 32526151 DOI: 10.1139/gen-2020-0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nuclear-cytoplasmic localization is an efficient way to regulate transcription factors and chromatin remodelers. Altering the location of existing protein pools also facilitates a more rapid response to changes in cell activity or extracellular signals. There are several examples of proteins that are regulated by nucleo-cytoplasmic shuttling, which are required for Drosophila neuroblast development. Disruption of the localization of homologs of these proteins has also been linked to several neurodegenerative disorders in humans. Drosophila has been used extensively to model the neurodegenerative disorders caused by aberrant nucleo-cytoplasmic localization. Here, we focus on the role of alternative nucleo-cytoplasmic protein localization in regulating proliferation and cell fate decisions in the Drosophila neuroblast and in neurodegenerative disorders. We also explore the analogous role of RNA binding proteins and mRNA localization in the context of regulation of nucleo-cytoplasmic localization during neural development and a role in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sophie E Keegan
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Sarah C Hughes
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Boudhraa Z, Carmona E, Provencher D, Mes-Masson AM. Ran GTPase: A Key Player in Tumor Progression and Metastasis. Front Cell Dev Biol 2020; 8:345. [PMID: 32528950 PMCID: PMC7264121 DOI: 10.3389/fcell.2020.00345] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Ran (Ras-related nuclear protein) GTPase is a member of the Ras superfamily. Like all the GTPases, Ran cycles between an active (GTP-bound) and inactive (GDP-bound) state. However, Ran lacks the CAAX motif at its C-terminus, a feature of other small GTPases that ensures a plasma membrane localization, and largely traffics between the nucleus and the cytoplasm. Ran regulates nucleo-cytoplasmic transport of molecules through the nuclear pore complex and controls cell cycle progression through the regulation of microtubule polymerization and mitotic spindle formation. The disruption of Ran expression has been linked to cancer at different levels - from cancer initiation to metastasis. In the present review, we discuss the contribution of Ran in the acquisition of three hallmarks of cancer, namely, proliferative signaling, resistance to apoptosis, and invasion/metastasis, and highlight its prognostic value in cancer patients. In addition, we discuss the use of this GTPase as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Zied Boudhraa
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du Cancer de Montréal (ICM), Montreal, QC, Canada
| | - Euridice Carmona
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du Cancer de Montréal (ICM), Montreal, QC, Canada
| | - Diane Provencher
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du Cancer de Montréal (ICM), Montreal, QC, Canada.,Division of Gynecologic Oncology, Université de Montréal, Montreal, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Institut du Cancer de Montréal (ICM), Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
12
|
Fallini C, Khalil B, Smith CL, Rossoll W. Traffic jam at the nuclear pore: All roads lead to nucleocytoplasmic transport defects in ALS/FTD. Neurobiol Dis 2020; 140:104835. [PMID: 32179176 DOI: 10.1016/j.nbd.2020.104835] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/25/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal late-onset neurodegenerative disease that specifically affects the function and survival of spinal and cortical motor neurons. ALS shares many genetic, clinical, and pathological characteristics with frontotemporal dementia (FTD), and these diseases are now recognized as presentations of a disease spectrum known as ALS/FTD. The molecular determinants of neuronal loss in ALS/FTD are still debated, but the recent discovery of nucleocytoplasmic transport defects as a common denominator of most if not all forms of ALS/FTD has dramatically changed our understanding of the pathogenic mechanisms of this disease. Loss of nuclear pores and nucleoporin aggregation, altered nuclear morphology, and impaired nuclear transport are some of the most prominent features that have been identified using a variety of animal, cellular, and human models of disease. Here, we review the experimental evidence linking nucleocytoplasmic transport defects to the pathogenesis of ALS/FTD and propose a unifying view on how these defects may lead to a vicious cycle that eventually causes neuronal death.
Collapse
Affiliation(s)
- Claudia Fallini
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA; Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA.
| | - Bilal Khalil
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Courtney L Smith
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Wilfried Rossoll
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
13
|
González-Velasco Ó, De Las Rivas J, Lacal J. Proteomic and Transcriptomic Profiling Identifies Early Developmentally Regulated Proteins in Dictyostelium Discoideum. Cells 2019; 8:cells8101187. [PMID: 31581556 PMCID: PMC6830349 DOI: 10.3390/cells8101187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023] Open
Abstract
Cyclic AMP acts as a secondary messenger involving different cellular functions in eukaryotes. Here, proteomic and transcriptomic profiling has been combined to identify novel early developmentally regulated proteins in eukaryote cells. These proteomic and transcriptomic experiments were performed in Dictyostelium discoideum given the unique advantages that this organism offers as a eukaryotic model for cell motility and as a nonmammalian model of human disease. By comparing whole-cell proteome analysis of developed (cAMP-pulsed) wild-type AX2 cells and an independent transcriptomic analysis of developed wild-type AX4 cells, our results show that up to 70% of the identified proteins overlap in the two independent studies. Among them, we have found 26 proteins previously related to cAMP signaling and identified 110 novel proteins involved in calcium signaling, adhesion, actin cytoskeleton, the ubiquitin-proteasome pathway, metabolism, and proteins that previously lacked any annotation. Our study validates previous findings, mostly for the canonical cAMP-pathway, and also generates further insight into the complexity of the transcriptomic changes during early development. This article also compares proteomic data between parental and cells lacking glkA, a GSK-3 kinase implicated in substrate adhesion and chemotaxis in Dictyostelium. This analysis reveals a set of proteins that show differences in expression in the two strains as well as overlapping protein level changes independent of GlkA.
Collapse
Affiliation(s)
- Óscar González-Velasco
- Bioinformatics and Functional Genomics Research Group. Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), 37007 Salamanca, Spain.
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Research Group. Cancer Research Center (CIC-IBMCC, CSIC/USAL/IBSAL), 37007 Salamanca, Spain.
| | - Jesus Lacal
- Department of Microbiology and Genetics, Faculty of Biology, University of Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
14
|
Hawiger J, Zienkiewicz J. Decoding inflammation, its causes, genomic responses, and emerging countermeasures. Scand J Immunol 2019; 90:e12812. [PMID: 31378956 PMCID: PMC6883124 DOI: 10.1111/sji.12812] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/03/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022]
Abstract
Inflammation is the mechanism of diseases caused by microbial, autoimmune, allergic, metabolic and physical insults that produce distinct types of inflammatory responses. This aetiologic view of inflammation informs its classification based on a cause‐dependent mechanism as well as a cause‐directed therapy and prevention. The genomic era ushered in a new understanding of inflammation by highlighting the cell's nucleus as the centre of the inflammatory response. Exogenous or endogenous inflammatory insults evoke genomic responses in immune and non‐immune cells. These genomic responses depend on transcription factors, which switch on and off a myriad of inflammatory genes through their regulatory networks. We discuss the transcriptional paradigm of inflammation based on denying transcription factors’ access to the nucleus. We present two approaches that control proinflammatory signalling to the nucleus. The first approach constitutes a novel intracellular protein therapy with bioengineered physiologic suppressors of cytokine signalling. The second approach entails control of proinflammatory transcriptional cascades by targeting nuclear transport with a cell‐penetrating peptide that inhibits the expression of 23 out of the 26 mediators of inflammation along with the nine genes required for metabolic responses. We compare these emerging anti‐inflammatory countermeasures to current therapies. The transcriptional paradigm of inflammation offers nucleocentric strategies for microbial, autoimmune, metabolic, physical and other types of inflammation afflicting millions of people worldwide.
Collapse
Affiliation(s)
- Jacek Hawiger
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jozef Zienkiewicz
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA
| |
Collapse
|
15
|
Nuclear formation induced by DNA-conjugated beads in living fertilised mouse egg. Sci Rep 2019; 9:8461. [PMID: 31186495 PMCID: PMC6560220 DOI: 10.1038/s41598-019-44941-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/17/2019] [Indexed: 11/08/2022] Open
Abstract
Reformation of a functional nucleus at the end of mitosis is crucial for normal cellular activity. Reconstitution approaches using artificial beads in frog egg extracts have clarified the molecules required for nuclear formation in vitro. However, the spatiotemporal regulation of these components, which is required for the formation of a functional nucleus in living embryos, remains unknown. Here we demonstrate that exogenous DNA introduced in the form of DNA-conjugated beads induces the assembly of an artificial nucleus in living mouse cleavage-stage embryos. Live-cell imaging and immunofluorescence studies revealed that core histones and regulator of chromosome condensation 1 (RCC1) assembled on the DNA, suggesting that nucleosomes were formed. Electron microscopy showed that double-membrane structures, partly extended from annulate lamellae, formed around the beads. Nuclear pore complex-like structures indistinguishable from those of native nuclei were also formed, suggesting that this membranous structure resembled the normal nuclear envelope (NE). However, the reconstituted NE had no nuclear import activity, probably because of the absence of Ras-related nuclear protein (Ran). Thus, DNA is necessary for NE reassembly in mouse embryos but is insufficient to form a functional nucleus. This approach provides a new tool to examine factors of interest and their spatiotemporal regulation in nuclear formation.
Collapse
|
16
|
Chu J, Chen Z. Molecular identification of histone acetyltransferases and deacetylases in lower plant Marchantia polymorpha. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2018; 132:612-622. [PMID: 30336381 DOI: 10.1016/j.plaphy.2018.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/18/2018] [Accepted: 10/10/2018] [Indexed: 06/08/2023]
Abstract
Histone is the core component of nucleosome and modification of amino acid residues on histone tails is one of the most pivotal epigenetic regulatory mechanisms. Histone acetylation or deacetylation is carried out by two groups of proteins: histone acetyltransferases (HATs) or histone deacetylases (HDACs), and has been proven to be tightly linked to regulation of gene expression in animals and vascular plants. The biological functions of HATs and HDACs in non-flowering plants remain largely unknown. We found that there are seven MpHAT genes and twelve MpHDAC genes present in the Marchantia genome, and the comprehensive protein sequence analysis of the HAT and HDAC families was introduced to investigate their potential functions. On the basis of the functional domain analysis, eight MpHATs and twelve MpHDACs contain the conserved functional domains as the defining feature of each family. Phylogenetic trees of all families of MpHATs and MpHDACs along with their homologs from different plant and green algae species were constructed to illustrate evolutionary relationship of HAT and HDAC proteins. We found both SIR2 family and RPD3/HDA1 superfamily possess lower plant-specific proteins indicating the potential unknown functions of HATs and HDACs in Marchantia and other lower plant or algae species. Subcellular localization prediction suggests that MpHATs and MpHDACs are likely functioning in various organelles. Expression analysis shows that all MpHAT and MpHDAC genes are expressed in all tissues with differences at the transcriptional level. In addition, their expression patterns were altered in response to various treatments with plant hormones and environmental stress. We concluded that all MpHATs and MpHDACs are functional proteins in Marchantia and involved in various signaling pathways. Marchantia could have developed a complex histone acetylation epigenetic mechanism to regulate growth and development, as well as responses to environment.
Collapse
Affiliation(s)
- Jiashu Chu
- Natural Sciences and Science Education, National Institute of Education, Nanyang Technological University, 1 Nanyang Walk, Singapore, 637616, Singapore
| | - Zhong Chen
- Natural Sciences and Science Education, National Institute of Education, Nanyang Technological University, 1 Nanyang Walk, Singapore, 637616, Singapore.
| |
Collapse
|
17
|
Drosophila nucleostemin 3 is required to maintain larval neuroblast proliferation. Dev Biol 2018; 440:1-12. [PMID: 29679561 PMCID: PMC6278609 DOI: 10.1016/j.ydbio.2018.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/14/2018] [Accepted: 04/17/2018] [Indexed: 02/03/2023]
Abstract
Stem cells must maintain proliferation during tissue development, repair and homeostasis, yet avoid tumor formation. In Drosophila, neural stem cells (neuroblasts) maintain proliferation during embryonic and larval development and terminate cell cycle during metamorphosis. An important question for understanding how tissues are generated and maintained is: what regulates stem cell proliferation versus differentiation? We performed a genetic screen which identified nucleostemin 3 (ns3) as a gene required to maintain neuroblast proliferation. ns3 is evolutionarily conserved with yeast and human Lsg1, which encode putative GTPases and are essential for organism growth and viability. We found NS3 is cytoplasmic and it is required to retain the cell cycle repressor Prospero in neuroblast cytoplasm via a Ran-independent pathway. NS3 is also required for proper neuroblast cell polarity and asymmetric cell division. Structure-function analysis further shows that the GTP-binding domain and acidic domain are required for NS3 function in neuroblast proliferation. We conclude NS3 has novel roles in regulating neuroblast cell polarity and proliferation.
Collapse
|
18
|
Ceballos-Laita L, Gutierrez-Carbonell E, Imai H, Abadía A, Uemura M, Abadía J, López-Millán AF. Effects of manganese toxicity on the protein profile of tomato ( Solanum lycopersicum ) roots as revealed by two complementary proteomic approaches, two-dimensional electrophoresis and shotgun analysis. J Proteomics 2018; 185:51-63. [DOI: 10.1016/j.jprot.2018.06.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/11/2018] [Accepted: 06/19/2018] [Indexed: 12/31/2022]
|
19
|
Jiang M, Chu Z. Comparative analysis of plant MKK gene family reveals novel expansion mechanism of the members and sheds new light on functional conservation. BMC Genomics 2018; 19:407. [PMID: 29843611 PMCID: PMC5975520 DOI: 10.1186/s12864-018-4793-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 05/14/2018] [Indexed: 12/15/2022] Open
Abstract
Background Mitogen-activated protein kinase (MAPK) cascades play critical functions in almost every aspect of plant growth and development, which regulates many physiological and biochemical processes. As a middle nodal point of the MAPK cascades, although evolutionary analysis of MKK from individual plant families had some reports, their evolutionary history in entire plants is still not clear. Results To better understand the evolution and function of plant MKKs, we performed systematical molecular evolutionary analysis of the MAPKK gene family and also surveyed their gene organizations, sequence features and expression patterns in different subfamilies. Phylogenetic analysis showed that plant MAPKK fall into five different groups (Group A–E). Majority orthology groups seemed to be a single or low-copy genes in all plant species analyzed in Group B, C and D, whereas group A MKKs undergo several duplication events, generating multiple gene copies. Further analysis showed that these duplication events were on account of whole genome duplications (WGDs) in plants and the duplicate genes maybe have undergone functional divergence. We also found that group E MKKs had mutation with one change of serine or theronine might lead to inactivity originated through the ancient tandem duplicates in monocots. Moreover, we also identified MKK3 integrated NTF2 domain that might have gradually lost the cytoplasmic-nuclear trafficking activity, which suggests that they may involve with the gene function more and more sophistication in the evolutionary process. Moreover, expression analyses indicated that plant MKK genes play probable roles in UV-B signaling. Conclusion In general, ancient gene and genome duplications are significantly conducive to the expansion of the plant MKK gene family. Our study reveals two distinct evolutionary patterns for plant MKK proteins and sheds new light on the functional evolution of this gene family. Electronic supplementary material The online version of this article (10.1186/s12864-018-4793-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Jiang
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai, China.,Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, China
| | - Zhaoqing Chu
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai, China. .,Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
20
|
Zhang K, Daigle JG, Cunningham KM, Coyne AN, Ruan K, Grima JC, Bowen KE, Wadhwa H, Yang P, Rigo F, Taylor JP, Gitler AD, Rothstein JD, Lloyd TE. Stress Granule Assembly Disrupts Nucleocytoplasmic Transport. Cell 2018; 173:958-971.e17. [PMID: 29628143 DOI: 10.1016/j.cell.2018.03.025] [Citation(s) in RCA: 250] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/08/2018] [Accepted: 03/06/2018] [Indexed: 01/21/2023]
Abstract
Defects in nucleocytoplasmic transport have been identified as a key pathogenic event in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) mediated by a GGGGCC hexanucleotide repeat expansion in C9ORF72, the most common genetic cause of ALS/FTD. Furthermore, nucleocytoplasmic transport disruption has also been implicated in other neurodegenerative diseases with protein aggregation, suggesting a shared mechanism by which protein stress disrupts nucleocytoplasmic transport. Here, we show that cellular stress disrupts nucleocytoplasmic transport by localizing critical nucleocytoplasmic transport factors into stress granules, RNA/protein complexes that play a crucial role in ALS pathogenesis. Importantly, inhibiting stress granule assembly, such as by knocking down Ataxin-2, suppresses nucleocytoplasmic transport defects as well as neurodegeneration in C9ORF72-mediated ALS/FTD. Our findings identify a link between stress granule assembly and nucleocytoplasmic transport, two fundamental cellular processes implicated in the pathogenesis of C9ORF72-mediated ALS/FTD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - J Gavin Daigle
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kathleen M Cunningham
- Cellular and Molecular Medicine Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Alyssa N Coyne
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kai Ruan
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jonathan C Grima
- Brain Science Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kelly E Bowen
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Harsh Wadhwa
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Peiguo Yang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010, USA
| | - J Paul Taylor
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, Howard Hughes Medical Institute, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Aaron D Gitler
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Jeffrey D Rothstein
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Cellular and Molecular Medicine Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Thomas E Lloyd
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Cellular and Molecular Medicine Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
Dubessay P, Larroche C, Fontanille P. Cloning and Characterization of the Gene Encoding Alpha-Pinene Oxide Lyase Enzyme (Prα-POL) from Pseudomonas rhodesiae CIP 107491 and Production of the Recombinant Protein in Escherichia coli. Appl Biochem Biotechnol 2017; 185:676-690. [PMID: 29285675 DOI: 10.1007/s12010-017-2685-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/19/2017] [Indexed: 12/01/2022]
Abstract
The alpha-pinene oxide lyase (Prα-POL) from Pseudomonas rhodesiae CIP107491 belongs to catabolic alpha-pinene degradation pathway. In this study, the gene encoding Prα-POL has been identified using mapping approach combined to inverse PCR (iPCR) strategy. The Prα-POL gene included a 609-bp open reading frame encoding 202 amino acids and giving rise to a 23.7 kDa protein, with a theoretical isoelectric point (pI) of 5.23. The amino acids sequence analysis showed homologies with those of proteins with unknown function from GammaProteobacteria group. Identification of a conserved domain in amino acid in positions 18 to 190 permitted to classify Prα-POL among the nuclear transport factor 2 (NTF2) protein superfamily. Heterologous expression of Prα-POL, both under its native form and with a histidin tag, was successfully performed in Escherichia coli, and enzymatic kinetics were analyzed. Bioconversion assay using recombinant E. coli strain allowed to reach a rate of isonovalal production per gramme of biomass about 40-fold higher than the rate obtained with P. rhodesiae.
Collapse
Affiliation(s)
- Pascal Dubessay
- Institut Pascal UMR CNRS 6602, Polytech Clermont-Ferrand, Université Clermont Auvergne (UCA), F-63178, Aubière, France.
| | - Christian Larroche
- Institut Pascal UMR CNRS 6602, Polytech Clermont-Ferrand, Université Clermont Auvergne (UCA), F-63178, Aubière, France
| | - Pierre Fontanille
- Institut Pascal UMR CNRS 6602, Polytech Clermont-Ferrand, Université Clermont Auvergne (UCA), F-63178, Aubière, France
| |
Collapse
|
22
|
Demyanenko SV, Uzdensky AB. The Focal-Focal Preconditioning Effect of Photothrombotic Impact on the Signaling Protein Profile in the Penumbra Surrounding the Ischemic Core Induced by Another Photothrombotic Impact. Mol Neurobiol 2017; 55:229-248. [DOI: 10.1007/s12035-017-0736-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Benhamman R, Bai F, Drory SB, Loubert-Hudon A, Ellis B, Matton DP. The Arabidopsis Mitogen-Activated Protein Kinase Kinase Kinase 20 (MKKK20) Acts Upstream of MKK3 and MPK18 in Two Separate Signaling Pathways Involved in Root Microtubule Functions. FRONTIERS IN PLANT SCIENCE 2017; 8:1352. [PMID: 28848569 PMCID: PMC5550695 DOI: 10.3389/fpls.2017.01352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/19/2017] [Indexed: 05/04/2023]
Abstract
Mitogen-activated protein kinase (MAPK) signaling networks represent important means of signal transduction in plants and other eukaryotes, controlling intracellular signaling by linking perception of environmental or developmental cues to downstream targets. In the Arabidopsis MEKK subfamily, the MKKK19, 20, and 21 form a highly supported clade with the Solanaceous Fertilization-Related Kinases. In Arabidopsis, little is known about this group, except for MKKK20, which is involved in osmotic stress. Using a directed MKKK-MKK yeast two-hybrid (Y2H) screen, MKKK20 was found to interact only with MKK3, while a MKKK20 large-scale Y2H screen retrieved MPK18 as a direct interactant. In vitro phosphorylation assays showed that MKKK20 phosphorylates both MKK3 and MPK18. However, when all three kinases are combined, no synergistic effect is observed on MPK18 phosphorylation, suggesting a direct access to MPK18, consistent with the absence of interaction between MKK3 and MPK18 in protein-protein interaction assays. Since mpk18 mutant plants were previously shown to be defective in microtubule-related functions, phenotypes of mkkk20 single and mkkk20/mpk18 double mutants were investigated to determine if MKKK20 acts upstream of MPK18. This was the case, as mkkk20 root length was shorter than WT in media containing microtubule-disrupting drugs as previously observed for mpk18 plants. Surprisingly, mkk3 plants were also similarly affected, suggesting the presence of two non-complementary pathways involved in Arabidopsis cortical microtubule function, the first including MKKK20, MKK3 and an unknown MPK; the second, a non-canonical MAPK cascade made of MKKK20 and MPK18 that bypasses the need for an MKK intermediate.
Collapse
Affiliation(s)
- Rachid Benhamman
- Institut de Recherche en Biologie Végétale, Département de Sciences Biologiques, Université de Montréal, MontréalQC, Canada
| | - Fangwen Bai
- Institut de Recherche en Biologie Végétale, Département de Sciences Biologiques, Université de Montréal, MontréalQC, Canada
| | - Samuel B. Drory
- Institut de Recherche en Biologie Végétale, Département de Sciences Biologiques, Université de Montréal, MontréalQC, Canada
| | - Audrey Loubert-Hudon
- Institut de Recherche en Biologie Végétale, Département de Sciences Biologiques, Université de Montréal, MontréalQC, Canada
| | - Brian Ellis
- Michael Smith Laboratories, University of British Columbia, VancouverBC, Canada
| | - Daniel P. Matton
- Institut de Recherche en Biologie Végétale, Département de Sciences Biologiques, Université de Montréal, MontréalQC, Canada
- *Correspondence: Daniel P. Matton,
| |
Collapse
|
24
|
Parvanov ED, Tian H, Billings T, Saxl RL, Spruce C, Aithal R, Krejci L, Paigen K, Petkov PM. PRDM9 interactions with other proteins provide a link between recombination hotspots and the chromosomal axis in meiosis. Mol Biol Cell 2016; 28:488-499. [PMID: 27932493 PMCID: PMC5341731 DOI: 10.1091/mbc.e16-09-0686] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/28/2016] [Accepted: 11/30/2016] [Indexed: 11/11/2022] Open
Abstract
In mammals, meiotic recombination occurs at 1- to 2-kb genomic regions termed hotspots, whose positions and activities are determined by PRDM9, a DNA-binding histone methyltransferase. We show that the KRAB domain of PRDM9 forms complexes with additional proteins to allow hotspots to proceed into the next phase of recombination. By a combination of yeast-two hybrid assay, in vitro binding, and coimmunoprecipitation from mouse spermatocytes, we identified four proteins that directly interact with PRDM9's KRAB domain, namely CXXC1, EWSR1, EHMT2, and CDYL. These proteins are coexpressed in spermatocytes at the early stages of meiotic prophase I, the limited period when PRDM9 is expressed. We also detected association of PRDM9-bound complexes with the meiotic cohesin REC8 and the synaptonemal complex proteins SYCP3 and SYCP1. Our results suggest a model in which PRDM9-bound hotspot DNA is brought to the chromosomal axis by the action of these proteins, ensuring the proper chromatin and spatial environment for subsequent recombination events.
Collapse
Affiliation(s)
- Emil D Parvanov
- Center for Genome Dynamics, Jackson Laboratory, Bar Harbor, ME 04609.,Department of Biology, Masaryk University, Brno, Czech Republic 625 00
| | - Hui Tian
- Center for Genome Dynamics, Jackson Laboratory, Bar Harbor, ME 04609
| | - Timothy Billings
- Center for Genome Dynamics, Jackson Laboratory, Bar Harbor, ME 04609
| | - Ruth L Saxl
- Center for Genome Dynamics, Jackson Laboratory, Bar Harbor, ME 04609
| | - Catrina Spruce
- Center for Genome Dynamics, Jackson Laboratory, Bar Harbor, ME 04609
| | - Rakesh Aithal
- Department of Biology, Masaryk University, Brno, Czech Republic 625 00
| | - Lumir Krejci
- Department of Biology, Masaryk University, Brno, Czech Republic 625 00 .,National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic 625 00
| | - Kenneth Paigen
- Center for Genome Dynamics, Jackson Laboratory, Bar Harbor, ME 04609
| | - Petko M Petkov
- Center for Genome Dynamics, Jackson Laboratory, Bar Harbor, ME 04609
| |
Collapse
|
25
|
The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 2015; 525:56-61. [PMID: 26308891 DOI: 10.1038/nature14973] [Citation(s) in RCA: 744] [Impact Index Per Article: 82.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 07/24/2015] [Indexed: 12/14/2022]
Abstract
The hexanucleotide repeat expansion (HRE) GGGGCC (G4C2) in C9orf72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Recent studies support an HRE RNA gain-of-function mechanism of neurotoxicity, and we previously identified protein interactors for the G4C2 RNA including RanGAP1. A candidate-based genetic screen in Drosophila expressing 30 G4C2 repeats identified RanGAP (Drosophila orthologue of human RanGAP1), a key regulator of nucleocytoplasmic transport, as a potent suppressor of neurodegeneration. Enhancing nuclear import or suppressing nuclear export of proteins also suppresses neurodegeneration. RanGAP physically interacts with HRE RNA and is mislocalized in HRE-expressing flies, neurons from C9orf72 ALS patient-derived induced pluripotent stem cells (iPSC-derived neurons), and in C9orf72 ALS patient brain tissue. Nuclear import is impaired as a result of HRE expression in the fly model and in C9orf72 iPSC-derived neurons, and these deficits are rescued by small molecules and antisense oligonucleotides targeting the HRE G-quadruplexes. Nucleocytoplasmic transport defects may be a fundamental pathway for ALS and FTD that is amenable to pharmacotherapeutic intervention.
Collapse
|
26
|
Chen P, Zhang X, Zhao T, Li Y, Gai J. Genome-wide identification and characterization of RBR ubiquitin ligase genes in soybean. PLoS One 2014; 9:e87282. [PMID: 24489889 PMCID: PMC3904995 DOI: 10.1371/journal.pone.0087282] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/20/2013] [Indexed: 11/18/2022] Open
Abstract
RBR (RING1-IBR-RING2) proteins play an important role in protein ubiquitination and are involved in many cellular processes. Recent studies showed plant RBR genes were induced by abiotic and biotic stresses. However, detailed studies on RBR genes in the important oil crop, soybean (Glycine max (L.) Merr.), is still lacking. Here we performed a genome-wide search and identified 24 RBR domain-containing genes from the soybean genome sequence and cloned 11 of them. Most soybean RBR proteins contain a highly conserved RBR supra-domain. Phylogenetic analyses indicated all 24 soybean RBR proteins are most related to the RBR proteins from Phaseolus vulgaris, and could be classified into seven groups including Ariadne A, Ariadne B, ARA54, Plant IIA, Plant IIB, Plant IIC, and Helicase. Tandem duplication and block duplication were found among the Ariadne B and Plant IIC group of soybean RBR genes. Despite the conserved RBR supra-domain, there are extensive variations in the additional protein motifs and exon-intron structures between different groups, which indicate they might have diverse functions. Most soybean RBR proteins are predicted to localize in nucleus, and four of them were experimentally confirmed by GFP fusion proteins. Soybean RBR genes are broadly expressed in many tissue types with a little more abundant in the roots and flowers than leaves, stems, and seeds. The expression of GmRTRTP3 (Plant IIB) and GmRTRTP5 (Plant IIC) are induced by NaCl treatment, which suggests these RBR genes might be involved in soybean response to abiotic stresses.
Collapse
Affiliation(s)
- Pei Chen
- Soybean Research Institute, National Center for Soybean Improvement, MOA Key Laboratory for Biology and Genetic Improvement of Soybean (General), National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xiaolian Zhang
- Soybean Research Institute, National Center for Soybean Improvement, MOA Key Laboratory for Biology and Genetic Improvement of Soybean (General), National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Tuanjie Zhao
- Soybean Research Institute, National Center for Soybean Improvement, MOA Key Laboratory for Biology and Genetic Improvement of Soybean (General), National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yan Li
- Soybean Research Institute, National Center for Soybean Improvement, MOA Key Laboratory for Biology and Genetic Improvement of Soybean (General), National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Junyi Gai
- Soybean Research Institute, National Center for Soybean Improvement, MOA Key Laboratory for Biology and Genetic Improvement of Soybean (General), National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing, Jiangsu, China
| |
Collapse
|
27
|
Kinoshita Y, Kalir T, Dottino P, Kohtz DS. Nuclear distributions of NUP62 and NUP214 suggest architectural diversity and spatial patterning among nuclear pore complexes. PLoS One 2012; 7:e36137. [PMID: 22558357 PMCID: PMC3338603 DOI: 10.1371/journal.pone.0036137] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 04/02/2012] [Indexed: 11/19/2022] Open
Abstract
The shape of nuclei in many adherent cultured cells approximates an oblate ellipsoid, with contralateral flattened surfaces facing the culture plate or the medium. Observations of cultured cell nuclei from orthogonal perspectives revealed that nucleoporin p62 (NUP62) and nucleoporin 214 (NUP214) are differentially distributed between nuclear pore complexes on the flattened surfaces and peripheral rim of the nucleus. High resolution stimulated emission depletion (STED) immunofluorescence microscopy resolved individual NPCs, and suggested both heterogeneity and microheterogeneity in NUP62 and NUP214 immunolabeling among in NPC populations. Similar to nuclear domains and interphase chromosome territories, architectural diversity and spatial patterning of NPCs may be an intrinsic property of the nucleus that is linked to the functions and organization of underlying chromatin.
Collapse
Affiliation(s)
- Yayoi Kinoshita
- Department of Pathology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Tamara Kalir
- Department of Pathology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Peter Dottino
- Obstetrics, Gynecology, and Reproductive Science, Mount Sinai School of Medicine, New York, New York, United States of America
| | - D. Stave Kohtz
- Department of Pathology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| |
Collapse
|
28
|
Kiyan Y, Limbourg A, Kiyan R, Tkachuk S, Limbourg FP, Ovsianikov A, Chichkov BN, Haller H, Dumler I. Urokinase receptor associates with myocardin to control vascular smooth muscle cells phenotype in vascular disease. Arterioscler Thromb Vasc Biol 2011; 32:110-22. [PMID: 22075245 DOI: 10.1161/atvbaha.111.234369] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The urokinase-type plasminogen activator (uPA) and its specific receptor (uPAR) are a potent multifunctional system involved in vascular remodeling. The goal of the study was to unravel the mechanisms of uPA/uPAR-directed vascular smooth muscle cell (VSMC) differentiation. METHODS AND RESULTS Using cultured human primary VSMCs, we identified a new molecular mechanism controlling phenotypic modulation in vitro and in vivo. We found that the urokinase-type plasminogen activator receptor (uPAR) acts together with the transcriptional coactivator myocardin to regulate the VSMC phenotype. uPAR, a glycosylphosphatidylinositol-anchored cell-surface receptor family member, undergoes ligand-induced internalization and nuclear transport in VSMCs. Platelet-derived growth factor receptor β and SUMOylated RanGAP1 mediate this trafficking. Nuclear uPAR associates with myocardin, which is then recruited from the promoters of serum response factor target genes and undergoes proteasomal degradation. This chain of events initiates the synthetic VSMC phenotype. Using mouse carotid artery ligation model, we show that this mechanism contributes to adverse vascular remodeling after injury in vivo. We then cultured cells on a microstructured biomaterial and found that substrate topography induced uPAR-mediated VSMC differentiation. CONCLUSIONS These findings reveal the transcriptional activity of uPAR, controlling the differentiation of VSMCs in a vascular disease model. They also suggest a new role for uPAR as a therapeutic target and as a marker for VSMC phenotyping on prosthetic biomaterials.
Collapse
Affiliation(s)
- Yulia Kiyan
- Nephrology Department, Hannover Medical School, Carl-Neuberg Str 1, 30625 Hannover, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Dormann D, Haass C. TDP-43 and FUS: a nuclear affair. Trends Neurosci 2011; 34:339-48. [PMID: 21700347 DOI: 10.1016/j.tins.2011.05.002] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 05/05/2011] [Accepted: 05/09/2011] [Indexed: 12/12/2022]
Abstract
Misfolded TAR DNA binding protein 43 (TDP-43) and Fused-In-Sarcoma (FUS) protein have recently been identified as pathological hallmarks of the neurodegenerative disorders amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) characterized by the presence of ubiquitin-positive inclusions (FTLD-U). Although TDP-43 and FUS are normally located predominantly in the nucleus, pathological TDP-43 and FUS inclusions are mostly found in the cytosol. Cytosolic deposition is paralleled by a striking nuclear depletion of either protein. Based on a number of recent findings, we postulate that defects in nuclear import are an important step towards TDP-43 and FUS dysfunction. Failure of nuclear transport can arise from mutations within a nuclear localization signal or from age-related decline of nuclear import mechanisms. We propose that nuclear import defects in combination with additional hits, for example cellular stress and genetic risk factors, may be a central underlying cause of ALS and FTLD-U pathology.
Collapse
Affiliation(s)
- Dorothee Dormann
- Adolf-Butenandt-Institute, Biochemistry, Ludwig-Maximilians-University and German Center for Neurodegenerative Diseases (DZNE) Munich, Schillerstr. 44, 80336 Munich, Germany
| | | |
Collapse
|
30
|
Kattygnarath D, Maugenre S, Neyroud N, Balse E, Ichai C, Denjoy I, Dilanian G, Martins RP, Fressart V, Berthet M, Schott JJ, Leenhardt A, Probst V, Le Marec H, Hainque B, Coulombe A, Hatem SN, Guicheney P. MOG1: a new susceptibility gene for Brugada syndrome. ACTA ACUST UNITED AC 2011; 4:261-8. [PMID: 21447824 DOI: 10.1161/circgenetics.110.959130] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Brugada syndrome (BrS) is caused mainly by mutations in the SCN5A gene, which encodes the α-subunit of the cardiac sodium channel Na(v)1.5. However, ≈ 20% of probands have SCN5A mutations, suggesting the implication of other genes. MOG1 recently was described as a new partner of Na(v)1.5, playing a potential role in the regulation of its expression and trafficking. We investigated whether mutations in MOG1 could cause BrS. METHODS AND RESULTS MOG1 was screened by direct sequencing in patients with BrS and idiopathic ventricular fibrillation. A missense mutation p.Glu83Asp (E83D) was detected in a symptomatic female patient with a type-1 BrS ECG but not in 281 controls. Wild type (WT)- and mutant E83D-MOG1 were expressed in HEK Na(v)1.5 stable cells and studied using patch-clamp assays. Overexpression of WT-MOG1 alone doubled sodium current (I(Na)) density compared to control conditions (P<0.01). In contrast, overexpression of mutant E83D alone or E83D+WT failed to increase I(Na) (P<0.05), demonstrating the dominant-negative effect of the mutant. Microscopy revealed that Na(v)1.5 channels failed to properly traffic to the cell membrane in the presence of the mutant. Silencing endogenous MOG1 demonstrated a 54% decrease in I(Na) density. CONCLUSIONS Our results support the hypothesis that dominant-negative mutations in MOG1 can impair the trafficking of Na(v)1.5 to the membrane, leading to I(Na) reduction and clinical manifestation of BrS. Moreover, silencing MOG1 reduced I(Na), demonstrating that MOG1 is likely to be important in the surface expression of Na(v)1.5 channels. All together, our data support MOG1 as a new susceptibility gene for BrS.
Collapse
|
31
|
Bao J, Wu Q, Song R, Jie Z, Zheng H, Xu C, Yan W. RANBP17 is localized to the XY body of spermatocytes and interacts with SPEM1 on the manchette of elongating spermatids. Mol Cell Endocrinol 2011; 333:134-42. [PMID: 21184802 PMCID: PMC3039071 DOI: 10.1016/j.mce.2010.12.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Revised: 12/04/2010] [Accepted: 12/14/2010] [Indexed: 11/28/2022]
Abstract
We identified Ran-binding protein 17 (RANBP17) as one of the interacting partners of sperm maturation 1 (SPEM1) using yeast 2-hybrid screening and immunoprecipitation assays. Expression profiling analyses suggested that RANBP17 was preferentially expressed in the testis. Immunofluorescent confocal microscopy revealed a dynamic localization pattern of RANBP17 during spermatogenesis. In primary spermatocytes RANBP17 was mainly localized to the XY body. In the subsequent spermiogenesis, RANBP17 was first observed in the nuclei of round spermatids (steps 1-7) and then confined to the manchette of elongating spermatids (steps 8-14) together with its interacting partner SPEM1. In the Spem1-null testes, levels of RANBP17 were significantly elevated. As a member of a large protein family involved in the nucleocytoplasmic transport, RANBP17 may have a role in sex chromosome inactivation during the meiotic phase of spermatogenesis, and also in the intramanchette transport during spermiogenesis. Interactions between RANBP17 and SPEM1, for the first time, point to a potential function of SPEM1 in the RANBP17-mediated nucleocytoplasmic transport.
Collapse
Affiliation(s)
- Jianqiang Bao
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
- Department of Embryology and Histology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Reproductive Medicine, Shanghai, China
| | - Qiuxia Wu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Rui Song
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Zhang Jie
- Department of Biochemistry, China Medical University, Shenyang, China
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Chen Xu
- Department of Embryology and Histology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Reproductive Medicine, Shanghai, China
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
- Corresponding author: Wei Yan MD, PhD Associate Professor Department of Physiology and Cell Biology University of Nevada School of Medicine Anderson Biomedical Science Building 105C/111 1664 North Virginia Street, MS 352 Reno, NV 89557 Tel: 775 784 7765 (Office) 775 784 4688 (Lab) Fax: 775 784 6903 URL: http://www.medicine.nevada.edu/physio/facyan.html
| |
Collapse
|
32
|
Kapon R, Naim B, Zbaida D, Nevo R, Tsabari O, Reich Z. Permeating the nuclear pore complex. Nucleus 2010; 1:475-80. [PMID: 21327089 PMCID: PMC3027049 DOI: 10.4161/nucl.1.6.13112] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 06/22/2010] [Accepted: 07/22/2010] [Indexed: 11/19/2022] Open
Abstract
The extensive and multifaceted traffic between nucleus and cytoplasm is handled by a single type of macromolecular assembly called the nuclear pore complex (NPC). While being readily accessible to ions and metabolites, the NPC imposes stringent selectivity on the passage of proteins and RNA, tightly regulating their traffic between the two major cellular compartments. Here we discuss how shuttling carriers, which mediate the transport of macromolecules through NPCs, cross its permeability barrier. We also discuss the co-existence of receptor-mediated macromolecular transport with the passive diffusion of small molecules in the context of the various models suggested for the permeability barrier of the NPC. Finally, we speculate on how nuclear transport receptors negotiate the dependence of their NPC-permeating abilities on hydrophobic interactions with the necessity of avoiding these promiscuous interactions in the cytoplasm and nucleus.
Collapse
Affiliation(s)
- Ruti Kapon
- Department of Biological Chemistry; Weizmann Institute of Science; Rehovot Israel
| | - Bracha Naim
- Department of Biological Chemistry; Weizmann Institute of Science; Rehovot Israel
| | - David Zbaida
- Department of Materials and Interfaces; Weizmann Institute of Science; Rehovot Israel
| | - Reinat Nevo
- Department of Biological Chemistry; Weizmann Institute of Science; Rehovot Israel
| | - Onie Tsabari
- Department of Biological Chemistry; Weizmann Institute of Science; Rehovot Israel
| | - Ziv Reich
- Department of Biological Chemistry; Weizmann Institute of Science; Rehovot Israel
| |
Collapse
|
33
|
Rodriguez MCS, Petersen M, Mundy J. Mitogen-activated protein kinase signaling in plants. ANNUAL REVIEW OF PLANT BIOLOGY 2010; 61:621-49. [PMID: 20441529 DOI: 10.1146/annurev-arplant-042809-112252] [Citation(s) in RCA: 685] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Eukaryotic mitogen-activated protein kinase (MAPK) cascades have evolved to transduce environmental and developmental signals into adaptive and programmed responses. MAPK cascades relay and amplify signals via three types of reversibly phosphorylated kinases leading to the phosphorylation of substrate proteins, whose altered activities mediate a wide array of responses, including changes in gene expression. Cascades may share kinase components, but their signaling specificity is maintained by spaciotemporal constraints and dynamic protein-protein interactions and by mechanisms that include crossinhibition, feedback control, and scaffolding. Plant MAPK cascades regulate numerous processes, including stress and hormonal responses, innate immunity, and developmental programs. Genetic analyses have uncovered several predominant MAPK components shared by several of these processes including the Arabidopsis thaliana MAPKs MPK3, 4, and 6 and MAP2Ks MKK1, 2, 4, and 5. Future work needs to focus on identifying substrates of MAPKs, and on understanding how specificity is achieved among MAPK signaling pathways.
Collapse
|
34
|
Abstract
In eukaryotic cells, the physical separation of the genetic material in the nucleus from the translation and signaling machinery in the cytoplasm by the nuclear envelope creates a requirement for a mechanism through which macromolecules can enter or exit the nucleus as necessary. Nucleocytoplasmic transport involves the specific recognition of cargo molecules by transport receptors in one compartment followed by the physical relocation of that cargo into the other compartment through regulated pores that perforate the nuclear envelope. The recognition of protein cargoes by their transport receptors occurs via amino acid sequences in cargo proteins called nuclear targeting signals. Both nuclear import and export of proteins are highly regulated processes that control, not only what cargo can enter and/or exit the nucleus, but also when in the cell cycle and in what cell type, the cargo can be transported. Deregulation of the nuclear transport of specific cargoes has been linked to numerous cancers and developmental disorders highlighting the importance of understanding the mechanisms underlying nucleocytoplasmic transport and particularly the modulation of the specific interactions between transporter receptors and nuclear targeting signals within target cargo proteins.
Collapse
Affiliation(s)
- Laura M McLane
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | |
Collapse
|
35
|
Ploski JE, Topisirovic I, Park KW, Borden KLB, Radu A. A mechanism of nucleocytoplasmic trafficking for the homeodomain protein PRH. Mol Cell Biochem 2009; 332:173-81. [PMID: 19588232 DOI: 10.1007/s11010-009-0188-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Accepted: 06/25/2009] [Indexed: 11/24/2022]
Abstract
Proline-rich homeodomain (PRH)/hematopoietically expressed homeodomain (Hex) is a homeodomain protein that plays an important role in early embryonic patterning and hematopoiesis. PRH can act as either a tumor suppressor or an oncogene and its expression is dysregulated in certain types of lymphoid and myeloid leukemias. Aberrant exclusion of PRH from the nuclei has been associated with thyroid and breast cancers and a subset of myeloid leukemias. Accordingly, nuclear localization of PRH was found to be necessary for the inhibition of eIF4E-dependent transformation. Since PRH's nuclear-cytoplasmic localization has been associated with neoplastic transformation we sought to better understand how PRH is transported to the nuclear compartment. Here, we report an essential element that controls the mechanism of PRH nucleocytoplasmic trafficking, namely that it is imported into the nuclei by Karyopherin/Importin 7. Kap7 was identified as a binding partner for PRH in a GST-pull down from a HeLa cell protein lysate, followed by mass-spectrometry. The Kap7-PRH complex is dissociated in the presence of RanGTP, as expected for a nuclear import complex. Kap7 can bind directly to PRH in a GST-pull down assay with purified proteins, as well as mediates the transport of PRH to the nuclear compartment in a digitonin permeabilized cells assay. Finally, in vivo depletion of Kap7 dramatically reduces accumulation of PRH in the nucleus. Our data open the way for investigations of the mechanism of perturbed PRH localization in tumors and possible therapeutic interventions.
Collapse
|
36
|
Ruan Y, Sensen CW, van der Hoorn FA. A novel group of multi-GAP-domain proteins. Mol Reprod Dev 2008; 75:1578-89. [PMID: 18363199 DOI: 10.1002/mrd.20896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Rho GTPase-activating proteins (RhoGAPs) play an essential role in regulating various cellular processes. Rat tGAP1 is the first reported protein that has multiple GAP domains. It is exclusively expressed in male germ cells. However, tGAP1 does not possess GAP activities in vitro. No tGAP1 homology has been identified in other species. In this study, we searched the genomic databases and identified many genes whose protein products possess 2-4 GAP domains in rat, mouse and dog. These genes all showed sequence similarity to tGAP1. The rat tGAP gene loci all locate on chromosome 2 and are all expressed in testes in RT-PCR analysis. The mouse tGAP gene loci also clustered on chromosome 3 but RT-PCR analysis showed most are pseudogene loci. Multiple sequence alignment showed that many conserved residues of the "arginine finger" motif within the GAP domains of predicted tGAP proteins have mutated, suggesting that tGAP proteins do not possess GAP activity. We also elucidated the evolutionary relations among the rat tGAP genes. Based on the phylogenetic analysis data, we proposed that tGAP genes and Arhgap20 genes have a common ancestor.
Collapse
Affiliation(s)
- Yibing Ruan
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | | | | |
Collapse
|
37
|
Walczak CE, Heald R. Mechanisms of mitotic spindle assembly and function. INTERNATIONAL REVIEW OF CYTOLOGY 2008; 265:111-58. [PMID: 18275887 DOI: 10.1016/s0074-7696(07)65003-7] [Citation(s) in RCA: 280] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The mitotic spindle is the macromolecular machine that segregates chromosomes to two daughter cells during mitosis. The major structural elements of the spindle are microtubule polymers, whose intrinsic polarity and dynamic properties are critical for bipolar spindle organization and function. In most cell types, spindle microtubule nucleation occurs primarily at two centrosomes, which define the spindle poles, but microtubules can also be generated by the chromosomes and within the spindle itself. Many associated factors help organize the spindle, including molecular motors and regulators of microtubule dynamics. The past decade has provided a wealth of information on the molecular players that are critical for spindle assembly as well as a high-resolution view of the intricate movements and dynamics of the spindle microtubules and the chromosomes. In this chapter we provide a historical account of the key observations leading to current models of spindle assembly, as well as an up-to-date status report on this exciting field.
Collapse
Affiliation(s)
- Claire E Walczak
- Medical Sciences Program, Indiana University, Bloomington, Indiana 47405, USA
| | | |
Collapse
|
38
|
Krichevsky A, Kozlovsky SV, Tian GW, Chen MH, Zaltsman A, Citovsky V. How pollen tubes grow. Dev Biol 2007; 303:405-20. [PMID: 17214979 DOI: 10.1016/j.ydbio.2006.12.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2006] [Revised: 11/16/2006] [Accepted: 12/02/2006] [Indexed: 10/23/2022]
Abstract
Sexual reproduction of flowering plants depends on delivery of the sperm to the egg, which occurs through a long, polarized projection of a pollen cell, called the pollen tube. The pollen tube grows exclusively at its tip, and this growth is distinguished by very fast rates and reaches extended lengths. Thus, one of the most fascinating aspects of pollen biology is the question of how enough cell wall material is produced to accommodate such rapid extension of pollen tube, and how the cell wall deposition and structure are regulated to allow for rapid changes in the direction of growth. This review discusses recent advances in our understanding of the mechanism of pollen tube growth, focusing on such basic cellular processes as control of cell shape and growth by a network of cell wall-modifying enzymes, molecular motor-mediated vesicular transport, and intracellular signaling by localized gradients of second messengers.
Collapse
Affiliation(s)
- Alexander Krichevsky
- Department of Biochemistry and Cell Biology, State University of New York, Stony Brook, NY 11794-5215, USA.
| | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Hearps A, Jans D. HIV-1 integrase is capable of targeting DNA to the nucleus via an importin alpha/beta-dependent mechanism. Biochem J 2006; 398:475-84. [PMID: 16716146 PMCID: PMC1559465 DOI: 10.1042/bj20060466] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In addition to its well-documented role in integration of the viral genome, the HIV-1 enzyme IN (integrase) is thought to be involved in the preceding step of importing the viral cDNA into the nucleus. The ability of HIV to transport its cDNA through an intact nuclear envelope allows HIV-1 to infect non-dividing cells, which is thought to be crucial for the persistent nature of HIV/AIDS. Despite this, the mechanism utilized by HIV-1 to import its cDNA into the nucleus, and the viral proteins involved, remains ill-defined. In the present study we utilize in vitro techniques to assess the nuclear import properties of the IN protein, and show that IN interacts with members of the Imp (Importin) family of nuclear transport proteins with high affinity and exhibits rapid nuclear accumulation within an in vitro assay, indicating that IN possesses potent nucleophilic potential. IN nuclear import appears to be dependent on the Imp alpha/beta heterodimer and Ran GTP (Ran in its GTP-bound state), but does not require ATP. Importantly, we show that IN is capable of binding DNA and facilitating its import into the nucleus of semi-intact cells via a process that involves basic residues within amino acids 186-188 of IN. These results confirm IN as an efficient mediator of DNA nuclear import in vitro and imply the potential for IN to fulfil such a role in vivo. These results may not only aid in highlighting potential therapeutic targets for impeding the progression of HIV/AIDS, but may also be relevant for non-viral gene delivery.
Collapse
Affiliation(s)
- Anna C. Hearps
- Nuclear Signalling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - David A. Jans
- Nuclear Signalling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
- To whom correspondence should be addressed (email )
| |
Collapse
|
41
|
Kortvely E, Burkovics P, Varszegi S, Gulya K. Cloning and characterization of rat importin 9: implication for its neuronal function. ACTA ACUST UNITED AC 2005; 139:103-14. [PMID: 15992958 DOI: 10.1016/j.molbrainres.2005.05.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2004] [Revised: 05/04/2005] [Accepted: 05/12/2005] [Indexed: 11/22/2022]
Abstract
We describe the structure of the rat importin 9 gene, together with its transcripts and the encoded protein with its putative functional domains. The importin 9 gene contains 24 exons in a genomic region spanning >52,000 bp. It is transcribed into two mRNAs, generated by means of alternative polyadenylation site usage arranged in tandem. Both transcripts possess the same noncanonical polyadenylation signal (AGUAAA) in rat, this hexamer being conserved in all vertebrates examined. Additionally, intron 8 is bordered by AT-AC dinucleotides. Importin 9 is expressed throughout adult rat tissues, but the 114-kDa Importin 9 protein was detected only in the brain. The localization of the Importin 9 protein was examined by immunohistochemistry in both adult rat tissues and primary hippocampal cell cultures. The strongest labeling was detected in vivo in areas populated by neurons in high density and also in the dendritic processes emanating from these cells. This protein was clearly concentrated in the nuclei of these cells, although their cytoplasms too were heavily labeled. Strong cytoplasmic and very strong nuclear staining was found in a vast majority of the cells with neuronal morphology in vitro. Cultured cells with glial morphology generally exhibited a weaker cytoplasmic labeling. In these cells, the signal decorated the nuclear envelope without nuclear staining and gradually dwindled toward the cell periphery. These results hint at the cell- or tissue-type specific functions of this type of importin protein.
Collapse
Affiliation(s)
- Elod Kortvely
- Department of Zoology and Cell Biology, University of Szeged, 2 Egyetem St., POB 659, Szeged H-6722, Hungary
| | | | | | | |
Collapse
|
42
|
Jiang SY, Ramachandran S. Comparative and evolutionary analysis of genes encoding small GTPases and their activating proteins in eukaryotic genomes. Physiol Genomics 2005; 24:235-51. [PMID: 16332933 DOI: 10.1152/physiolgenomics.00210.2005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Both small GTPase and its activating protein (GAP) superfamilies exist in various eukaryotes. The small GTPases regulate a wide variety of cellular processes by cycling between active GTP- and inactive GAP-bound conformations. The GAPs promote GTPase inactivation by stimulating the GTP hydrolysis. In this study, we identified 111 small GTPases and 85 GAPs in rice, 65 GAPs in Arabidopsis, 90 small GTPases in Drosophila melanogaster, and 35 GAPs in Saccharomyces cerevisiaeby genome-wide analysis. We then analyzed and compared a total of 498 small GTPases and 422 GAPs from these four eukaryotic and human genomes. Both animals and yeast genomes contained five families of small GTPases and their GAPs. However, plants had only four of these five families because of a lack of the Ras and RasGAP genes. Small GTPases were conserved with common motifs, but GAPs exhibited higher and much more rapid divergence. On the basis of phylogenetic analysis of all small GTPases and GAPs in five eukaryotic organisms, we estimated that their ancestors had small sizes of small GTPases and GAPs and their large-scale expansions occurred after the divergence from their ancestors. Further investigation showed that genome duplications represented the major mechanism for such expansions. Nonsynonymous substitutions per site (Ka) and synonymous substitutions per site (Ks) analyses showed that most of the divergence due to a positive selection occurred in common ancestors, suggesting a major functional divergence in an ancient era.
Collapse
Affiliation(s)
- Shu-Ye Jiang
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore
| | | |
Collapse
|
43
|
Trougakos IP, Saridaki A, Panayotou G, Gonos ES. Identification of differentially expressed proteins in senescent human embryonic fibroblasts. Mech Ageing Dev 2005; 127:88-92. [PMID: 16213575 DOI: 10.1016/j.mad.2005.08.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Accepted: 08/24/2005] [Indexed: 11/20/2022]
Abstract
Normal human fibroblasts undergo a limited number of divisions in culture, a process known as replicative senescence (RS). Although several senescence-specific genes have been identified, analysis at the level of protein expression can provide additional insights into the mechanisms that regulate RS. We have performed a proteomic comparison between young and replicative senescent human embryonic WI-38 fibroblasts and we have identified 13 proteins, which are differentially expressed in senescent cells. Some of the identified proteins are components of the cellular cytoskeleton, while others are implicated in key cellular functions including metabolism and energy production, Ca(2+) signalling, nucleo-cytoplasmic trafficking and telomerase activity regulation. In summary, our analysis contributes to the list of senescence-associated proteins by identifying new biomarkers and provides novel information on functional protein networks that are perturbed during replicative senescence of human fibroblast cultures.
Collapse
Affiliation(s)
- Ioannis P Trougakos
- Laboratory of Molecular & Cellular Ageing, Institute of Biological Research & Biotechnology, National Hellenic Research Foundation, 48 Vas. Constantinou Ave., Athens 11635, Greece
| | | | | | | |
Collapse
|
44
|
Kronfeld K, Hochleitner E, Mendler S, Goldschmidt J, Lichtenfels R, Lottspeich F, Abken H, Seliger B. B7/CD28 costimulation of T cells induces a distinct proteome pattern. Mol Cell Proteomics 2005; 4:1876-87. [PMID: 16113399 DOI: 10.1074/mcp.m500194-mcp200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Effective immune strategies for the eradication of human tumors require a detailed understanding of the interaction of tumor cells with the immune system, which might lead to an optimization of T cell responses. To understand the impact of B7-mediated costimulation on T cell activation comprehensive proteome analysis of B7-primed T cell populations were performed. Using this approach we identified different classes of proteins in T cells whose expression is either elevated or reduced upon B7-1- or B7-2-mediated CD28 costimulation. The altered proteins include regulators of the cell cycle and cell proliferation, signal transducers, components of the antigen processing machinery, transporters, cytoskeletal proteins, and metabolic enzymes. A number of differentially expressed proteins are further modified by phosphorylation. Our results provide novel insights into the complexity of the CD28 costimulatory pathway of T cells and will help to identify potential targets of therapeutic interventions for modulating anti-tumor T cell activation.
Collapse
Affiliation(s)
- Kai Kronfeld
- IIIrd Department of Internal Medicine, Johannes Gutenberg University, 55131 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Geyer JP, Döker R, Kremer W, Zhao X, Kuhlmann J, Kalbitzer HR. Solution structure of the Ran-binding domain 2 of RanBP2 and its interaction with the C terminus of Ran. J Mol Biol 2005; 348:711-25. [PMID: 15826666 DOI: 10.1016/j.jmb.2005.02.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2004] [Revised: 02/15/2005] [Accepted: 02/16/2005] [Indexed: 10/25/2022]
Abstract
The termination of export processes from the nucleus to the cytoplasm in higher eukaryotes is mediated by binding of the small GTPase Ran as part of the export complexes to the Ran-binding domains (RanBD) of Ran-binding protein 2 (RanBP2) of the nuclear pore complex. So far, the structures of the first RanBD of RanBP2 and of RanBP1 in complexes with Ran have been known from X-ray crystallographic studies. Here we report the NMR solution structure of the uncomplexed second RanBD of RanBP2. The structure shows a pleckstrin homology (PH) fold featuring two almost orthogonal beta-sheets consisting of three and four strands and an alpha-helix sitting on top. This is in contrast to the RanBD in the crystal structure complexes in which one beta-strand is missing. That is probably due to the binding of the C-terminal alpha-helix of Ran to the RanBD in these complexes. To analyze the interaction between RanBD2 and the C terminus of Ran, NMR-titration studies with peptides comprising the six or 28 C-terminal residues of Ran were performed. While the six-residue peptide alone does not bind to RanBD2 in a specific manner, the 28-residue peptide, including the entire C-terminal helix of Ran, binds to RanBD2 in a manner analogous to the crystal structures. By solving the solution structure of the 28mer peptide alone, we confirmed that it adopts a stable alpha-helical structure like in native Ran and therefore serves as a valid model of the Ran C terminus. These results support current models that assume recognition of the transport complexes by the RanBDs through the Ran C terminus that is exposed in these complexes.
Collapse
Affiliation(s)
- J Peter Geyer
- Institut für Biophysik und physikalische Biochemie, Universität Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Huang H, Zhang B, Hartenstein PA, Chen JN, Lin S. NXT2 is required for embryonic heart development in zebrafish. BMC DEVELOPMENTAL BIOLOGY 2005; 5:7. [PMID: 15790397 PMCID: PMC1079804 DOI: 10.1186/1471-213x-5-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2004] [Accepted: 03/24/2005] [Indexed: 11/10/2022]
Abstract
Background NXT2 is a member of NXT family proteins that are generally involved in exporting nuclear RNA in eukaryotic cells. It is not known if NXT2 has any function in specific biological processes. Results A zebrafish mutant exhibiting specific heart defects during embryogenesis was generated by animal cloning-mediated retroviral insertions. Molecular analysis indicated that the mutant phenotype was caused by a disruption of NXT2. Whole-mount RNA in situ hybridization showed that NXT2 transcripts were clearly detectable in embryonic heart as well as other tissues. Further analysis revealed that expression level of one form of alternative splicing NXT2 mRNA transcripts was significantly reduced, resulting in deficient myocardial cell differentiation and the malformation of cardiac valve at the atrioventricular boundary. The defects could be reproduced by morpholino anti-sense oligo knockdown of NXT2. Conclusion NXT2 has a critical role in maintaining morphogenetic integrity of embryonic heart in vertebrate species.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Alternative Splicing
- Animals
- Cell Differentiation/genetics
- Cloning, Organism
- DNA Transposable Elements
- Edema, Cardiac/genetics
- Edema, Cardiac/pathology
- Heart/embryology
- Heart/physiology
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/pathology
- Heart Valves/pathology
- Myocardium/chemistry
- Myocardium/cytology
- Myocardium/pathology
- Nuclear Export Signals/genetics
- Nuclear Export Signals/physiology
- Phenotype
- RNA, Antisense
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription, Genetic
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish Proteins/genetics
- Zebrafish Proteins/physiology
Collapse
Affiliation(s)
- Haigen Huang
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095, USA
| | - Bo Zhang
- Center of Developmental Biology and Genetics, College of Life Sciences Peking University, Beijing 100871, P. R. CHINA
| | - Parvana A Hartenstein
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095, USA
| | - Jau-nian Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095, USA
| | - Shuo Lin
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095, USA
- Center of Developmental Biology and Genetics, College of Life Sciences Peking University, Beijing 100871, P. R. CHINA
| |
Collapse
|
47
|
Abstract
Importin beta, once thought to be exclusively a nuclear transport receptor, is emerging as a global regulator of diverse cellular functions. Importin beta acts positively in multiple interphase roles: in nuclear import, as a chaperone for highly charged nuclear proteins, and as a potential motor adaptor for movement along microtubules. In contrast, importin beta plays a negative regulatory role in mitotic spindle assembly, centrosome dynamics, nuclear membrane formation, and nuclear pore assembly. In most of these, importin beta is counteracted by its regulator, Ran-GTP. In light of this, the recent discovery of Ran's involvement in spindle checkpoint control suggested a potential new arena for importin beta action, although it is also possible that one of importin beta's relatives, the karyopherin family of proteins, manages this checkpoint. Lastly, importin beta plays a role in transducing damage signals from the axons of injured neurons back to the cell body.
Collapse
Affiliation(s)
- Amnon Harel
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California, San Diego, 9500 Gilman Drive, Room 2124A, Pacific Hall, La Jolla, CA 92093, USA
| | | |
Collapse
|
48
|
Abstract
We have entered an exciting era for androgen-receptor (AR) research that should provide a detailed description of how the AR functions as a ligand-regulated transcription factor. That AR activity is regulated by subcellular compartmentalization was first established a decade ago with the finding that binding of androgen to the AR induces its translocation from the cytoplasm to the nucleus. The contribution of compartmentalization to AR activity is, however, likely to extend beyond simple delivery to the nucleus. Defects in AR and coregulator compartmentalization in the nucleus have been demonstrated in prostate cancer, androgen-insensitivity syndrome, and spinal and bulbar muscular atrophy. A complete understanding of AR function and dysfunction in disease requires integrating transcription with the spatial and temporal regulation imposed by subnuclear organization and nuclear transport.
Collapse
Affiliation(s)
- Ben E Black
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
49
|
Belov GA, Lidsky PV, Mikitas OV, Egger D, Lukyanov KA, Bienz K, Agol VI. Bidirectional increase in permeability of nuclear envelope upon poliovirus infection and accompanying alterations of nuclear pores. J Virol 2004; 78:10166-77. [PMID: 15331749 PMCID: PMC514989 DOI: 10.1128/jvi.78.18.10166-10177.2004] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Poliovirus and some other picornaviruses trigger relocation of certain nuclear proteins into the cytoplasm. Here, by using a protein changing its fluorescence color with time and containing a nuclear localization signal (NLS), we demonstrate that the poliovirus-triggered relocation is largely due to the exit of presynthesized nuclear protein into the cytoplasm. The leakiness of the nuclear envelope was also documented by the inability of nuclei from digitonin-permeabilized, virus-infected (but not mock-infected) cells to retain an NLS-containing derivative of green fluorescent protein (GFP). The cytoplasm-to-nucleus traffic was also facilitated during infection, as evidenced by experiments with GAPDH (glyceraldehyde-3-phosphate dehydrogenase), cyclin B1, and an NLS-lacking derivative of GFP, which are predominantly cytoplasmic in uninfected cells. Electron microscopy demonstrated that a bar-like barrier structure in the channel of the nuclear pores, seen in uninfected cells, was missing in the infected cells, giving the impression of fully open pores. Transient expression of poliovirus 2A protease also resulted in relocation of the nuclear proteins. Lysates from poliovirus-infected or 2A-expressing cells induced efflux of 3xEGFP-NLS from the nuclei of permeabilized uninfected cells. This activity was inhibited by the elastase inhibitors elastatinal and N-(methoxysuccinyl)-L-alanyl-L-alanyl-L-prolyl-L-valine chloromethylketone (drugs known also to be inhibitors of poliovirus protease 2A), a caspase inhibitor zVAD(OMe), fmk, and some other protease inhibitors. These data suggest that 2A elicited nuclear efflux, possibly in cooperation with a zVAD(OMe).fmk-sensitive protease. However, poliovirus infection facilitated nuclear protein efflux also in cells deficient in caspase-3 and caspase-9, suggesting that the efflux may occur without the involvement of these enzymes. The biological relevance of nucleocytoplasmic traffic alterations in infected cells is discussed.
Collapse
Affiliation(s)
- George A Belov
- M. P. Chumakov Institute of Poliomyelitis and Viral Encephalitides, Russian Academy of Medical Sciences, Moscow
| | | | | | | | | | | | | |
Collapse
|
50
|
Harel A, Chan RC, Lachish-Zalait A, Zimmerman E, Elbaum M, Forbes DJ. Importin beta negatively regulates nuclear membrane fusion and nuclear pore complex assembly. Mol Biol Cell 2003; 14:4387-96. [PMID: 14551248 PMCID: PMC266759 DOI: 10.1091/mbc.e03-05-0275] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Assembly of a eukaryotic nucleus involves three distinct events: membrane recruitment, fusion to form a double nuclear membrane, and nuclear pore complex (NPC) assembly. We report that importin beta negatively regulates two of these events, membrane fusion and NPC assembly. When excess importin beta is added to a full Xenopus nuclear reconstitution reaction, vesicles are recruited to chromatin but their fusion is blocked. The importin beta down-regulation of membrane fusion is Ran-GTP reversible. Indeed, excess RanGTP (RanQ69L) alone stimulates excessive membrane fusion, leading to intranuclear membrane tubules and cytoplasmic annulate lamellae-like structures. We propose that a precise balance of importin beta to Ran is required to create a correct double nuclear membrane and simultaneously to repress undesirable fusion events. Interestingly, truncated importin beta 45-462 allows membrane fusion but produces nuclei lacking any NPCs. This reveals distinct importin beta-regulation of NPC assembly. Excess full-length importin beta and beta 45-462 act similarly when added to prefused nuclear intermediates, i.e., both block NPC assembly. The importin beta NPC block, which maps downstream of GTPgammaS and BAPTA-sensitive steps in NPC assembly, is reversible by cytosol. Remarkably, it is not reversible by 25 microM RanGTP, a concentration that easily reverses fusion inhibition. This report, using a full reconstitution system and natural chromatin substrates, significantly expands the repertoire of importin beta. Its roles now encompass negative regulation of two of the major events of nuclear assembly: membrane fusion and NPC assembly.
Collapse
Affiliation(s)
- Amnon Harel
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California, San Diego, La Jolla, California 92093-0347, USA
| | | | | | | | | | | |
Collapse
|