1
|
Zimyanin V, Dash BP, Großmann D, Simolka T, Glaß H, Verma R, Khatri V, Deppmann C, Zunder E, Redemann S, Hermann A. Axonal transcriptome reveals upregulation of PLK1 as a protective mechanism in response to increased DNA damage in FUS P525L spinal motor neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624439. [PMID: 39605661 PMCID: PMC11601502 DOI: 10.1101/2024.11.20.624439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Mutations in the gene FUSED IN SARCOMA ( FUS ) are among the most frequently occurring genetic forms of amyotrophic lateral sclerosis (ALS). Early pathogenesis of FUS -ALS involves impaired DNA damage response and axonal degeneration. However, it is still poorly understood how these gene mutations lead to selective spinal motor neuron (MN) degeneration and how nuclear and axonal phenotypes are linked. To specifically address this, we applied a compartment specific RNA-sequencing approach using microfluidic chambers to generate axonal as well as somatodendritic compartment-specific profiles from isogenic induced pluripotent stem cells (iPSCs)-derived MNs. We demonstrate high purity of axonal and soma fractions and show that the axonal transcriptome is unique and distinct from that of somas including significantly fewer number of transcripts. Functional enrichment analysis revealed that differentially expressed genes (DEGs) in axons were mainly enriched in key pathways like RNA metabolism and DNA damage, complementing our knowledge of early phenotypes in ALS pathogenesis and known functions of FUS. In addition, we demonstrate a strong enrichment for cell cycle associated genes including significant upregulation of polo-like kinase 1 (PLK1) in FUS P525L mutant MNs. PLK1 was increased upon DNA damage induction and PLK1 inhibition further increased the number of DNA damage foci in etoposide-treated cells, an effect that was diminished in case of FUS mutant MNs. In contrast, inhibition of PLK1 increased late apoptotic or necrosis-induced neuronal cell death in mutant neurons. Taken together, our findings provide insights into compartment-specific transcriptomics in human FUS -ALS MNs and we propose that specific upregulation of PLK1 might represent an early event in the pathogenesis of ALS, possibly modulating DNA damage response and other associated pathways.
Collapse
|
2
|
Wang MD, Yi L, Li Y, Xu R, Hu J, Hou DY, Liu C, Wang H. Homologous Peptide Foldamer Promotes FUS Aggregation and Triggers Cancer Cell Death. J Am Chem Soc 2024; 146:28669-28676. [PMID: 39403745 DOI: 10.1021/jacs.4c03420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Fused in sarcoma (FUS), a multifunctional deoxyribonucleic acid (DNA)/ribonucleic acid (RNA)-binding protein, has been implicated in various cancer types, including sarcoma and leukemia. Despite its association with these diseases, there has been limited exploration of FUS as a cancer therapy target, primarily because its dynamic nature makes it difficult to target specifically. In this study, we explored a kind of β-sheet peptide foldamer, named β4-TAT, to influence FUS aggregation by targeting its RNA recognition motifs (RRM). This approach leverages the noncovalent interaction characteristics of peptide self-assembly processes. The β4 sequence, derived from the FUS RRM β-sheet, in combination with TAT, a peptide known for its nuclear targeting capability, enables β4-TAT to bind specifically to the analogous β4 sequence within FUS. Notably, β4-TAT effectively induces FUS aggregation within cells, leading to the death of cancer cells. Our work developed a novel peptide foldamer-based strategy for inducing protein aggregation, paving the way for innovative therapeutic approaches in targeting FUS-associated cancers.
Collapse
Affiliation(s)
- Man-Di Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190 Beijing, China
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19A Yuquan Road, 100049 Beijing, China
| | - Li Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190 Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19A Yuquan Road, 100049 Beijing, China
| | - Yanying Li
- Department of Medical Cell Biology Science for Life Laboratory, Uppsala University, Uppsala SE-75124, Sweden
| | - Ruiwen Xu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Jiaojiao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210 Shanghai, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210 Shanghai, China
| | - Da-Yong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190 Beijing, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210 Shanghai, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210 Shanghai, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190 Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19A Yuquan Road, 100049 Beijing, China
| |
Collapse
|
3
|
Kyung J, Jeong D, Eom H, Kim J, Kim JS, Lee I. C-TERMINAL DOMAIN PHOSPHATASE-LIKE 1 promotes flowering with TAF15b by repressing the floral repressor gene FLOWERING LOCUS C. Mol Cells 2024; 47:100114. [PMID: 39293741 DOI: 10.1016/j.mocell.2024.100114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024] Open
Abstract
Arabidopsis TATA-BINDING PROTEIN-ASSOCIATED FACTOR15b (TAF15b) is a plant-specific component of the transcription factor IID complex. TAF15b is involved in the autonomous pathway for flowering and represses the transcription of FLOWERING LOCUS C (FLC), a major floral repressor in Arabidopsis. While components of the autonomous flowering pathway have been extensively studied, scant attention has been directed toward elucidating the direct transcriptional regulators responsible for repressing FLC transcription. Here, we demonstrate that C-TERMINAL DOMAIN PHOSPHATASE-LIKE 1 (CPL1) is a physical and functional partner of TAF15b, playing a role in FLC repression. CPL1 is a protein phosphatase that dephosphorylates the C-terminal domain of RNA polymerase II (Pol II). Through the immunoprecipitation and mass spectrometry technique, we identified CPL1 as an interacting partner of TAF15b. Similar to taf15b, the cpl1 mutant showed a late-flowering phenotype caused by an increase in FLC levels. Additionally, the increase in cpl1 was correlated with the enrichment of phosphorylated Pol II in the FLC chromatin, as expected. We also discovered that CPL1 and TAF15b share additional common target genes through transcriptome analysis. These results suggest that TAF15b and CPL1 cooperatively repress transcription through the dephosphorylation of Pol II, especially at the FLC locus.
Collapse
Affiliation(s)
- Jinseul Kyung
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea; Research Center for Plant Plasticity, Seoul National University, Seoul 08826, Korea
| | - Daesong Jeong
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea; Research Center for Plant Plasticity, Seoul National University, Seoul 08826, Korea
| | - Hyunjoo Eom
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Jeesoo Kim
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea; Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea
| | - Jong-Seo Kim
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea; Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea
| | - Ilha Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea; Research Center for Plant Plasticity, Seoul National University, Seoul 08826, Korea; Plant Genomics and Breeding Institute, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
4
|
Chen L, Chen G, Zhang M, Zhang X. Modeling sporadic juvenile ALS in iPSC-derived motor neurons explores the pathogenesis of FUS R503fs mutation. Front Cell Neurosci 2024; 18:1364164. [PMID: 38711616 PMCID: PMC11070534 DOI: 10.3389/fncel.2024.1364164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/26/2024] [Indexed: 05/08/2024] Open
Abstract
Introduction Fused in sarcoma (FUS) mutations represent the most common genetic etiology of juvenile amyotrophic lateral sclerosis (JALS), for which effective treatments are lacking. In a prior report, we identified a novel FUS mutation, c.1509dupA: p. R503fs (FUSR503fs), in a sporadic JALS patient. Methods The physicochemical properties and structure of FUSR503fs protein were analyzed by software: Multi-electrode array (MEA) assay, calcium activity imaging assay and transcriptome analysis were used to explore the pathophysiological mechanism of iPSC derived motor neurons. Results Structural analysis and predictions regarding physical and chemical properties of this mutation suggest that the reduction of phosphorylation and glycosylation sites, along with alterations in the amino acid sequence, may contribute to abnormal FUS accumulation within the cytoplasm and nucleus of induced pluripotent stem cell- derived motor neurons (MNs). Multi-electrode array and calcium activity imaging indicate diminished spontaneous electrical and calcium activity signals in MNs harboring the FUSR503fs mutation. Transcriptomic analysis reveals upregulation of genes associated with viral infection and downregulation of genes involved in neural function maintenance, such as the ATP6V1C2 gene. Treatment with ropinirole marginally mitigates the electrophysiological decline in FUSR503fs MNs, suggesting the utility of this cell model for mechanistic exploration and drug screening. Discussion iPSCs-derived motor neurons from JALS patients are promising tools for drug screening. The pathological changes in motor neurons of FUSR503fs may occur earlier than in other known mutation types that have been reported.
Collapse
Affiliation(s)
- Li Chen
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Guojie Chen
- Hunan YoBon Biotechnology Limited Company, Changsha, Hunan, China
| | - Mengting Zhang
- College of Integrated Chinese and Western Medicine (School of Life Sciences), Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiaojie Zhang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| |
Collapse
|
5
|
Chiusa M, Lee YA, Zhang MZ, Harris RC, Sherrill T, Lindner V, Brooks CR, Yu G, Fogo AB, Flynn CR, Zienkiewicz J, Hawiger J, Zent R, Pozzi A. Cytoplasmic retention of the DNA/RNA-binding protein FUS ameliorates organ fibrosis in mice. J Clin Invest 2024; 134:e175158. [PMID: 38488009 PMCID: PMC10940094 DOI: 10.1172/jci175158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/17/2024] [Indexed: 03/18/2024] Open
Abstract
Uncontrolled accumulation of extracellular matrix leads to tissue fibrosis and loss of organ function. We previously demonstrated in vitro that the DNA/RNA-binding protein fused in sarcoma (FUS) promotes fibrotic responses by translocating to the nucleus, where it initiates collagen gene transcription. However, it is still not known whether FUS is profibrotic in vivo and whether preventing its nuclear translocation might inhibit development of fibrosis following injury. We now demonstrate that levels of nuclear FUS are significantly increased in mouse models of kidney and liver fibrosis. To evaluate the direct role of FUS nuclear translocation in fibrosis, we used mice that carry a mutation in the FUS nuclear localization sequence (FUSR521G) and the cell-penetrating peptide CP-FUS-NLS that we previously showed inhibits FUS nuclear translocation in vitro. We provide evidence that FUSR521G mice or CP-FUS-NLS-treated mice showed reduced nuclear FUS and fibrosis following injury. Finally, differential gene expression analysis and immunohistochemistry of tissues from individuals with focal segmental glomerulosclerosis or nonalcoholic steatohepatitis revealed significant upregulation of FUS and/or collagen genes and FUS protein nuclear localization in diseased organs. These results demonstrate that injury-induced nuclear translocation of FUS contributes to fibrosis and highlight CP-FUS-NLS as a promising therapeutic option for organ fibrosis.
Collapse
Affiliation(s)
- Manuel Chiusa
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Youngmin A. Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ming-Zhi Zhang
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Raymond C. Harris
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Veterans Affairs, Nashville, Tennessee, USA
| | - Taylor Sherrill
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Volkhard Lindner
- Center for Molecular Medicine, Maine Health Institute for Research, Scarborough, Maine, USA
| | - Craig R. Brooks
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Gang Yu
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Agnes B. Fogo
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Charles R. Flynn
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jozef Zienkiewicz
- Department of Veterans Affairs, Nashville, Tennessee, USA
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jacek Hawiger
- Department of Veterans Affairs, Nashville, Tennessee, USA
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Veterans Affairs, Nashville, Tennessee, USA
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Veterans Affairs, Nashville, Tennessee, USA
| |
Collapse
|
6
|
Aksoy YA, Cole AJ, Deng W, Hesselson D. Zebrafish CCNF and FUS Mediate Stress-Specific Motor Responses. Cells 2024; 13:372. [PMID: 38474336 DOI: 10.3390/cells13050372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 03/14/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the degeneration of motor neurons. Mutations in the cyclin F (CCNF) and fused in sarcoma (FUS) genes have been associated with ALS pathology. In this study, we aimed to investigate the functional role of CCNF and FUS in ALS by using genome editing techniques to generate zebrafish models with genetic disruptions in these genes. Sequence comparisons showed significant homology between human and zebrafish CCNF and FUS proteins. We used CRISPR/Cas9 and TALEN-mediated genome editing to generate targeted disruptions in the zebrafish ccnf and fus genes. Ccnf-deficient zebrafish exhibited abnormal motor neuron development and axonal outgrowth, whereas Fus-deficient zebrafish did not exhibit developmental abnormalities or axonopathies in primary motor neurons. However, Fus-deficient zebrafish displayed motor impairments in response to oxidative and endoplasmic reticulum stress. The Ccnf-deficient zebrafish were only sensitized to endoplasmic reticulum stress, indicating that ALS genes have overlapping as well as unique cellular functions. These zebrafish models provide valuable platforms for studying the functional consequences of CCNF and FUS mutations in ALS pathogenesis. Furthermore, these zebrafish models expand the drug screening toolkit used to evaluate possible ALS treatments.
Collapse
Affiliation(s)
- Yagiz Alp Aksoy
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
| | - Alexander J Cole
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Daniel Hesselson
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
7
|
Zhu Y, Burg T, Neyrinck K, Vervliet T, Nami F, Vervoort E, Ahuja K, Sassano ML, Chai YC, Tharkeshwar AK, De Smedt J, Hu H, Bultynck G, Agostinis P, Swinnen JV, Van Den Bosch L, da Costa RFM, Verfaillie C. Disruption of MAM integrity in mutant FUS oligodendroglial progenitors from hiPSCs. Acta Neuropathol 2024; 147:6. [PMID: 38170217 PMCID: PMC10764485 DOI: 10.1007/s00401-023-02666-x] [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: 07/26/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal neurodegenerative disorder, characterized by selective loss of motor neurons (MNs). A number of causative genetic mutations underlie the disease, including mutations in the fused in sarcoma (FUS) gene, which can lead to both juvenile and late-onset ALS. Although ALS results from MN death, there is evidence that dysfunctional glial cells, including oligodendroglia, contribute to neurodegeneration. Here, we used human induced pluripotent stem cells (hiPSCs) with a R521H or a P525L mutation in FUS and their isogenic controls to generate oligodendrocyte progenitor cells (OPCs) by inducing SOX10 expression from a TET-On SOX10 cassette. Mutant and control iPSCs differentiated efficiently into OPCs. RNA sequencing identified a myelin sheath-related phenotype in mutant OPCs. Lipidomic studies demonstrated defects in myelin-related lipids, with a reduction of glycerophospholipids in mutant OPCs. Interestingly, FUSR521H OPCs displayed a decrease in the phosphatidylcholine/phosphatidylethanolamine ratio, known to be associated with maintaining membrane integrity. A proximity ligation assay further indicated that mitochondria-associated endoplasmic reticulum membranes (MAM) were diminished in both mutant FUS OPCs. Moreover, both mutant FUS OPCs displayed increased susceptibility to ER stress when exposed to thapsigargin, and exhibited impaired mitochondrial respiration and reduced Ca2+ signaling from ER Ca2+ stores. Taken together, these results demonstrate a pathological role of mutant FUS in OPCs, causing defects in lipid metabolism associated with MAM disruption manifested by impaired mitochondrial metabolism with increased susceptibility to ER stress and with suppressed physiological Ca2+ signaling. As such, further exploration of the role of oligodendrocyte dysfunction in the demise of MNs is crucial and will provide new insights into the complex cellular mechanisms underlying ALS.
Collapse
Affiliation(s)
- Yingli Zhu
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium.
| | - Thibaut Burg
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | - Katrien Neyrinck
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Tim Vervliet
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Fatemeharefeh Nami
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Ellen Vervoort
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Karan Ahuja
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
- Animal Physiology and Neurobiology Section, Department of Biology, Neural Circuit Development and Regeneration Research Group, 3000, Leuven, Belgium
| | - Maria Livia Sassano
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Yoke Chin Chai
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Arun Kumar Tharkeshwar
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | - Jonathan De Smedt
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Haibo Hu
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, 3000, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | | | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| |
Collapse
|
8
|
Collette D, Dunlap D, Finzi L. Macromolecular Crowding and DNA: Bridging the Gap between In Vitro and In Vivo. Int J Mol Sci 2023; 24:17502. [PMID: 38139331 PMCID: PMC10744201 DOI: 10.3390/ijms242417502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The cellular environment is highly crowded, with up to 40% of the volume fraction of the cell occupied by various macromolecules. Most laboratory experiments take place in dilute buffer solutions; by adding various synthetic or organic macromolecules, researchers have begun to bridge the gap between in vitro and in vivo measurements. This is a review of the reported effects of macromolecular crowding on the compaction and extension of DNA, the effect of macromolecular crowding on DNA kinetics, and protein-DNA interactions. Theoretical models related to macromolecular crowding and DNA are briefly reviewed. Gaps in the literature, including the use of biologically relevant crowders, simultaneous use of multi-sized crowders, empirical connections between macromolecular crowding and liquid-liquid phase separation of nucleic materials are discussed.
Collapse
Affiliation(s)
| | | | - Laura Finzi
- Department of Physics, College of Arts & Sciences, Emory University, Atlanta, GA 30322, USA; (D.C.); (D.D.)
| |
Collapse
|
9
|
Swerdlow RH, Jawdat O, Swint-Kruse L, Townley R. FUS G559A Mutation in a Patient with a Frontotemporal Dementia-Motor Neuron Disease Compatible Syndrome: A Case Report. J Alzheimers Dis Rep 2023; 7:1121-1126. [PMID: 37849626 PMCID: PMC10578331 DOI: 10.3233/adr-230103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/13/2023] [Indexed: 10/19/2023] Open
Abstract
Fused in sarcoma (FUS) mutations cause frontotemporal dementia (FTD) and motor neuron disease (MND). Here, we describe a 43-year-old man with progressive behavioral and cognitive change, myelopathy, clinical and electrophysiologic evidence of MND, and a FUS variant of unknown significance (VUS). This VUS, a heterozygous G559A transition (Gly187Ser), was previously reported in a patient with sporadic MND and affects important FUS biophysical properties. While this rare variant's presence in a second patient with a related neurodegenerative syndrome does not establish pathogenicity, it raises the question of whether its association with our patient is coincidental and increases the possibility that FUS G559A is pathogenic.
Collapse
Affiliation(s)
- Russell H. Swerdlow
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Omar Jawdat
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Liskin Swint-Kruse
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ryan Townley
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
10
|
Oppong D, Schiff W, Shivamadhu MC, Ahn YH. Chemistry and biology of enzymes in protein glutathionylation. Curr Opin Chem Biol 2023; 75:102326. [PMID: 37245422 PMCID: PMC10524987 DOI: 10.1016/j.cbpa.2023.102326] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/30/2023]
Abstract
Protein S-glutathionylation is emerging as a central oxidation that regulates redox signaling and biological processes linked to diseases. In recent years, the field of protein S-glutathionylation has expanded by developing biochemical tools for the identification and functional analyses of S-glutathionylation, investigating knockout mouse models, and developing and evaluating chemical inhibitors for enzymes involved in glutathionylation. This review will highlight recent studies of two enzymes, glutathione transferase omega 1 (GSTO1) and glutaredoxin 1 (Grx1), especially introducing their glutathionylation substrates associated with inflammation, cancer, and neurodegeneration and showcasing the advancement of their chemical inhibitors. Lastly, we will feature protein substrates and chemical inducers of LanC-like protein (LanCL), the first enzyme in protein C-glutathionylation.
Collapse
Affiliation(s)
- Daniel Oppong
- Department of Chemistry, Drexel University, Philadelphia, PA 19104, USA
| | - William Schiff
- Department of Chemistry, Drexel University, Philadelphia, PA 19104, USA
| | | | - Young-Hoon Ahn
- Department of Chemistry, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Xiang L, Wang Y, Liu S, Liu B, Jin X, Cao X. Targeting Protein Aggregates with Natural Products: An Optional Strategy for Neurodegenerative Diseases. Int J Mol Sci 2023; 24:11275. [PMID: 37511037 PMCID: PMC10379780 DOI: 10.3390/ijms241411275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Protein aggregation is one of the hallmarks of aging and aging-related diseases, especially for the neurodegenerative diseases (NDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Amyotrophic lateral sclerosis (ALS), and others. In these diseases, many pathogenic proteins, such as amyloid-β, tau, α-Syn, Htt, and FUS, form aggregates that disrupt the normal physiological function of cells and lead to associated neuronal lesions. Protein aggregates in NDs are widely recognized as one of the important targets for the treatment of these diseases. Natural products, with their diverse biological activities and rich medical history, represent a great treasure trove for the development of therapeutic strategies to combat disease. A number of in vitro and in vivo studies have shown that natural products, by virtue of their complex molecular scaffolds that specifically bind to pathogenic proteins and their aggregates, can inhibit the formation of aggregates, disrupt the structure of aggregates and destabilize them, thereby alleviating conditions associated with NDs. Here, we systematically reviewed studies using natural products to improve disease-related symptoms by reducing or inhibiting the formation of five pathogenic protein aggregates associated with NDs. This information should provide valuable insights into new directions and ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lingzhi Xiang
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Yanan Wang
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Shenkui Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Beidong Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Xuejiao Jin
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Xiuling Cao
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| |
Collapse
|
12
|
Wong ET, Rosenberg H, Dawood O, Hertan L, Vega RA, Anderson M, Uhlmann EJ. Lewy body disease as a potential negative outcome modifier of glioblastoma treatment: a case report. BMC Neurol 2023; 23:257. [PMID: 37403078 DOI: 10.1186/s12883-023-03313-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/16/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Elderly patients with glioblastoma are particularly susceptible to the adverse effects of ionizing radiation to the brain. This population also has an increasing prevalence of dementia in the successive seventh, eighth and nineth decade of life, and dementia with Lewy bodies is characterized by pathologic α-synucleins, proteins that take part in neuronal DNA damage repair. CASE PRESENTATION We report a 77-year-old man, with a history of coronary artery disease and mild cognitive impairment, who experienced subacute behavioral changes over 3 months with wording-finding difficulty, memory loss, confusion, perseveration, and irritable mood. Neuroimaging studies disclosed a 2.5 × 2.4 × 2.7 cm cystic enhancing mass with central necrosis in the left temporal lobe of the brain. Gross total resection of the tumor revealed IDH-1 wild-type glioblastoma. After treatment with radiation and temozolomide chemotherapy, his cognitive status deteriorated rapidly, and he died from unexpected sudden death 2 months after radiation. Autopsy of his brain revealed (i) tumor cells with atypical nuclei and small lymphocytes, (ii) neuronal cytoplasmic inclusions and Lewy bodies that were positive for α-synuclein in the midbrain, pons, amygdala, putamen and globus pallidus, and (iii) no amyloid plaques and only rare neurofibrillary tangles near the hippocampi. CONCLUSIONS This patient most likely had pre-clinical limbic subtype of dementia with Lewy bodies prior to his diagnosis of glioblastoma. The radiation and temozolomide that was used to treat his tumor may have accelerated neuronal damage due to induction of DNA breakage when his brain was already compromised by pathologic α-synucleins. α-Synucleinopathy could be a negative outcome modifier in glioblastoma patients.
Collapse
Affiliation(s)
- Eric T Wong
- Brain Tumor Center & Neuro-Oncology Unit, Department of Neurology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, United States.
- Department of Neurology, Medicine (Division of Hematology/Oncology), Neurosurgery & Radiation Oncology, Rhode Island Hospital, 593 Eddy St, Providence, 02903, United States.
| | - Harry Rosenberg
- Division of Neuropathology, Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, United States
| | - Olivia Dawood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, United States
| | - Lauren Hertan
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, United States
| | - Rafael A Vega
- Division of Neurosurgery, Department of Surgery, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, United States
| | - Matthew Anderson
- Division of Neuropathology, Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, United States
- Present Address: Regeneron Pharmaceutical Company, 777 Old Saw Mill Rive Road, Tarrytown, NY, 10591, United States
| | - Erik J Uhlmann
- Brain Tumor Center & Neuro-Oncology Unit, Department of Neurology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, 02215, United States
| |
Collapse
|
13
|
Cortés Mancera EA, Sinisterra Solis FA, Romero-Castellanos FR, Diaz-Meneses IE, Kerik-Rotenberg NE. 18F-FDG PET/CT as a molecular biomarker in the diagnosis of amyotrophic lateral sclerosis associated with prostate cancer and progressive supranuclear palsy: A case report. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1137875. [PMID: 39355053 PMCID: PMC11440934 DOI: 10.3389/fnume.2023.1137875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/23/2023] [Indexed: 10/03/2024]
Abstract
Introduction Amyotrophic lateral sclerosis (ALS) is a neurodegenerative, multisystem disorder. Its clinical presentation typically consists of progressive focal muscle atrophy and weakness. In addition to motor disorders, the association between ALS and cancer has been researched, such as frontotemporal dementia and progressive supranuclear palsy. The diagnosis is based primarily on the clinical history, physical examination, electrodiagnostic tests (with an EMG needle), and neuroimaging, such as MRI and 18F-FDG PET/CT. Presentation of the case A 67-year-old male patient was diagnosed with prostate adenocarcinoma with a clinical picture of muscle weakness in the lower limbs that caused falls and was associated with fasciculations in the thighs and arms, alterations in the tone of voice, poor memory, and difficulty articulating words. In the neurological assessment, he described walking supported by a walker with decreased strength in both lower limbs and sensitivity without alterations. The diagnoses of upper and lower motor neuron disease and probable ALS were integrated. Furthermore, the probable coexistence of frontotemporal dementia/disorder (FDD) with ALS was considered. The main findings in the 18F-FDG PET/CT study was hypometabolism in the cortex of the bilateral motor and premotor areas, the anterior cingulate, both caudate and putamen, a metabolic pattern compatible with ALS, and progressive supranuclear palsy. Conclusion Through the PET/CT studies, we demonstrated a case in which ALS, prostate cancer and progressive supranuclear palsy coexisted molecularly; it was clinically difficult to diagnose. Molecular imaging has potential in the diagnostic and prognostic evaluation of ALS. It is crucial to identify the disease early and reliably through metabolic patterns that allow us to confirm the disease or differentiate it from other pathologies.
Collapse
Affiliation(s)
- Emilly A Cortés Mancera
- PET/CT Molecular Imaging Unit. National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Fabio A Sinisterra Solis
- PET/CT Molecular Imaging Unit. National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | | | - Ivan E Diaz-Meneses
- PET/CT Molecular Imaging Unit. National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Nora E Kerik-Rotenberg
- PET/CT Molecular Imaging Unit. National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| |
Collapse
|
14
|
Kakuo M, Horii T, Tonomura N, Sato R, Ogawa M, Okajima T, Kamemura K. Evidence that only EWS among the FET proteins acquires a low partitioning property for the hyperosmotic stress response by O-GlcNAc glycosylation on its low-complexity domain. Exp Cell Res 2023; 424:113504. [PMID: 36736606 DOI: 10.1016/j.yexcr.2023.113504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
FET proteins (FUS, EWS, and TAF15) share a common domain organization, bind RNA/DNA, and perform similarly multifunctional roles in the regulation of gene expression. Of the FET proteins, however, only EWS appears to have a distinct property in the cellular stress response. Therefore, we focused on the relationship between hyperosmotic stress response and post-translational modifications of the FET proteins. We confirmed that the hyperosmotic stress-dependent translocation from the nucleus to the cytoplasm and the cellular granule formation of FET proteins, and that EWS is less likely to partition into cellular granules in the cytoplasm than FUS or TAF15. The domain involved in the less partitioning property of EWS was found to be its low-complexity domain (LCD). Chemoenzymatic labeling analysis of O-linked β-N-acetylglucosamine (O-GlcNAc) residues revealed that O-GlcNAc glycosylation occurs frequently in the LCD of EWS. A correlation was observed between the glycosylation of EWS and the less partitioning property under the hyperosmotic stress. These results suggest that among the FET proteins, only EWS has acquired the unique property through O-GlcNAc glycosylation. The glycosylation may play an essential role in regulating physiological functions of EWS, such as transcriptional activity, in addition to the property in cellular stress response.
Collapse
Affiliation(s)
- Manami Kakuo
- Department of Medical Bioscience, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Takeshi Horii
- Department of Medical Bioscience, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Naoto Tonomura
- Department of Medical Bioscience, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Runa Sato
- Department of Medical Bioscience, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan
| | - Mitsutaka Ogawa
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Tetsuya Okajima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Kazuo Kamemura
- Department of Medical Bioscience, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan.
| |
Collapse
|
15
|
Gawade K, Plewka P, Häfner SJ, Lund AH, Marchand V, Motorin Y, Szczesniak MW, Raczynska KD. FUS regulates a subset of snoRNA expression and modulates the level of rRNA modifications. Sci Rep 2023; 13:2974. [PMID: 36806717 PMCID: PMC9941101 DOI: 10.1038/s41598-023-30068-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
FUS is a multifunctional protein involved in many aspects of RNA metabolism, including transcription, splicing, translation, miRNA processing, and replication-dependent histone gene expression. In this work, we show that FUS depletion results in the differential expression of numerous small nucleolar RNAs (snoRNAs) that guide 2'-O methylation (2'-O-Me) and pseudouridylation of specific positions in ribosomal RNAs (rRNAs) and small nuclear RNAs (snRNAs). Using RiboMeth-seq and HydraPsiSeq for the profiling of 2'-O-Me and pseudouridylation status of rRNA species, we demonstrated considerable hypermodification at several sites in HEK293T and SH-SY5Y cells with FUS knockout (FUS KO) compared to wild-type cells. We observed a similar direction of changes in rRNA modification in differentiated SH-SY5Y cells with the FUS mutation (R495X) related to the severe disease phenotype of amyotrophic lateral sclerosis (ALS). Furthermore, the pattern of modification of some rRNA positions was correlated with the abundance of corresponding guide snoRNAs in FUS KO and FUS R495X cells. Our findings reveal a new role for FUS in modulating the modification pattern of rRNA molecules, that in turn might generate ribosome heterogeneity and constitute a fine-tuning mechanism for translation efficiency/fidelity. Therefore, we suggest that increased levels of 2'-O-Me and pseudouridylation at particular positions in rRNAs from cells with the ALS-linked FUS mutation may represent a possible new translation-related mechanism that underlies disease development and progression.
Collapse
Affiliation(s)
- Kishor Gawade
- Laboratory of RNA Processing, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University in Poznan, 61-614, Poznan, Poland
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, 61-614, Poznan, Poland
| | - Patrycja Plewka
- Laboratory of RNA Processing, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University in Poznan, 61-614, Poznan, Poland
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, 61-614, Poznan, Poland
| | - Sophia J Häfner
- Biotech Research and Innovation Centre, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Anders H Lund
- Biotech Research and Innovation Centre, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Virginie Marchand
- Université de Lorraine, UAR2008/US40 IBSLor CNRS-INSERM and UMR7365 IMoPA CNRS, Nancy, France
| | - Yuri Motorin
- Université de Lorraine, UAR2008/US40 IBSLor CNRS-INSERM and UMR7365 IMoPA CNRS, Nancy, France
| | - Michal W Szczesniak
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University in Poznan, 61-614, Poznan, Poland
| | - Katarzyna D Raczynska
- Laboratory of RNA Processing, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University in Poznan, 61-614, Poznan, Poland.
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, 61-614, Poznan, Poland.
| |
Collapse
|
16
|
Wang YW, Dong HZ, Tan YX, Bao X, Su YM, Li X, Jiang F, Liang J, Huang ZC, Ren YL, Xu YL, Su Q. HIF-1α-regulated lncRNA-TUG1 promotes mitochondrial dysfunction and pyroptosis by directly binding to FUS in myocardial infarction. Cell Death Dis 2022; 8:178. [PMID: 35396503 PMCID: PMC8993815 DOI: 10.1038/s41420-022-00969-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/02/2022]
Abstract
Myocardial infarction (MI) is a fatal heart disease that affects millions of lives worldwide each year. This study investigated the roles of HIF-1α/lncRNA-TUG1 in mitochondrial dysfunction and pyroptosis in MI. CCK-8, DHE, lactate dehydrogenase (LDH) assays, and JC-1 staining were performed to measure proliferation, reactive oxygen species (ROS), LDH leakage, and mitochondrial damage in hypoxia/reoxygenation (H/R)-treated cardiomyocytes. Enzyme-linked immunoassay (ELISA) and flow cytometry were used to detect LDH, creatine kinase (CK), and its isoenzyme (CK-MB) levels and caspase-1 activity. Chromatin immunoprecipitation (ChIP), luciferase assay, and RNA-immunoprecipitation (RIP) were used to assess the interaction between HIF-1α, TUG1, and FUS. Quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, and immunohistochemistry were used to measure HIF-1α, TUG1 and pyroptosis-related molecules. Hematoxylin and eosin (HE), 2,3,5-triphenyltetrazolium chloride (TTC), and terminal deoxynucleotidyl transferase dUTP risk end labelling (TUNEL) staining were employed to examine the morphology, infarction area, and myocardial injury in the MI mouse model. Mitochondrial dysfunction and pyroptosis were induced in H/R-treated cardiomyocytes, accompanied by an increase in the expression of HIF-α and TUG1. HIF-1α promoted TUG1 expression by directly binding to the TUG1 promoter. TUG1 silencing inhibited H/R-induced ROS production, mitochondrial injury and the expression of the pyroptosis-related proteins NLRP3, caspase-1 and GSDMD. Additionally, H/R elevated FUS levels in cardiomyocytes, which were directly inhibited by TUG1 silencing. Fused in sarcoma (FUS) overexpression reversed the effect of TUG1 silencing on mitochondrial damage and caspase-1 activation. However, the ROS inhibitor N-acetylcysteine (NAC) promoted the protective effect of TUG1 knockdown on H/R-induced cardiomyocyte damage. The in vivo MI model showed increased infarction, myocardial injury, ROS levels and pyroptosis, which were inhibited by TUG1 silencing. HIF-1α targeting upregulated TUG1 promotes mitochondrial damage and cardiomyocyte pyroptosis by combining with FUS, thereby promoting the occurrence of MI. HIF-1α/TUG1/FUS may serve as a potential treatment target for MI.
Collapse
Affiliation(s)
- Yong-Wang Wang
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Hong-Zhi Dong
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, 300222, P. R. China
| | - Yong-Xing Tan
- Department of Intensive Care Unit, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Xu Bao
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Ying-Man Su
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Xin Li
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Fang Jiang
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Jing Liang
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Zhen-Cai Huang
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Yan-Ling Ren
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Yu-Li Xu
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China
| | - Qiang Su
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, Zhuang Autonomous Region, P. R. China.
| |
Collapse
|
17
|
Mechanistic Insights of Mitochondrial Dysfunction in Amyotrophic Lateral Sclerosis: An Update on a Lasting Relationship. Metabolites 2022; 12:metabo12030233. [PMID: 35323676 PMCID: PMC8951432 DOI: 10.3390/metabo12030233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive loss of the upper and lower motor neurons. Despite the increasing effort in understanding the etiopathology of ALS, it still remains an obscure disease, and no therapies are currently available to halt its progression. Following the discovery of the first gene associated with familial forms of ALS, Cu–Zn superoxide dismutase, it appeared evident that mitochondria were key elements in the onset of the pathology. However, as more and more ALS-related genes were discovered, the attention shifted from mitochondria impairment to other biological functions such as protein aggregation and RNA metabolism. In recent years, mitochondria have again earned central, mechanistic roles in the pathology, due to accumulating evidence of their derangement in ALS animal models and patients, often resulting in the dysregulation of the energetic metabolism. In this review, we first provide an update of the last lustrum on the molecular mechanisms by which the most well-known ALS-related proteins affect mitochondrial functions and cellular bioenergetics. Next, we focus on evidence gathered from human specimens and advance the concept of a cellular-specific mitochondrial “metabolic threshold”, which may appear pivotal in ALS pathogenesis.
Collapse
|
18
|
Nicolas G, Sévigny M, Lecoquierre F, Marguet F, Deschênes A, del Pelaez MC, Feuillette S, Audebrand A, Lecourtois M, Rousseau S, Richard AC, Cassinari K, Deramecourt V, Duyckaerts C, Boland A, Deleuze JF, Meyer V, Clarimon Echavarria J, Gelpi E, Akiyama H, Hasegawa M, Kawakami I, Wong TH, Van Rooij JGJ, Van Swieten JC, Campion D, Dutchak PA, Wallon D, Lavoie-Cardinal F, Laquerrière A, Rovelet-Lecrux A, Sephton CF. A postzygotic de novo NCDN mutation identified in a sporadic FTLD patient results in neurochondrin haploinsufficiency and altered FUS granule dynamics. Acta Neuropathol Commun 2022; 10:20. [PMID: 35151370 PMCID: PMC8841087 DOI: 10.1186/s40478-022-01314-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/18/2022] [Indexed: 12/19/2022] Open
Abstract
Frontotemporal dementia (FTD) is a heterogeneous clinical disorder characterized by progressive abnormalities in behavior, executive functions, personality, language and/or motricity. A neuropathological subtype of FTD, frontotemporal lobar degeneration (FTLD)-FET, is characterized by protein aggregates consisting of the RNA-binding protein fused in sarcoma (FUS). The cause of FTLD-FET is not well understood and there is a lack of genetic evidence to aid in the investigation of mechanisms of the disease. The goal of this study was to identify genetic variants contributing to FTLD-FET and to investigate their effects on FUS pathology. We performed whole-exome sequencing on a 50-year-old FTLD patient with ubiquitin and FUS-positive neuronal inclusions and unaffected parents, and identified a de novo postzygotic nonsense variant in the NCDN gene encoding Neurochondrin (NCDN), NM_014284.3:c.1206G > A, p.(Trp402*). The variant was associated with a ~ 31% reduction in full-length protein levels in the patient’s brain, suggesting that this mutation leads to NCDN haploinsufficiency. We examined the effects of NCDN haploinsufficiency on FUS and found that depleting primary cortical neurons of NCDN causes a reduction in the total number of FUS-positive cytoplasmic granules. Moreover, we found that these granules were significantly larger and more highly enriched with FUS. We then examined the effects of a loss of FUS function on NCDN in neurons and found that depleting cells of FUS leads to a decrease in NCDN protein and mRNA levels. Our study identifies the NCDN protein as a likely contributor of FTLD-FET pathophysiology. Moreover, we provide evidence for a negative feedback loop of toxicity between NCDN and FUS, where loss of NCDN alters FUS cytoplasmic dynamics, which in turn has an impact on NCDN expression.
Collapse
|
19
|
A novel promoter-associated non-coding small RNA paGLI1 recruits FUS/P65 to transactivate GLI1 gene expression and promotes infiltrating glioma progression. Cancer Lett 2022; 530:68-84. [DOI: 10.1016/j.canlet.2022.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/29/2021] [Accepted: 01/13/2022] [Indexed: 11/17/2022]
|
20
|
Merighi A, Gionchiglia N, Granato A, Lossi L. The Phosphorylated Form of the Histone H2AX (γH2AX) in the Brain from Embryonic Life to Old Age. Molecules 2021; 26:7198. [PMID: 34885784 PMCID: PMC8659122 DOI: 10.3390/molecules26237198] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
The γ phosphorylated form of the histone H2AX (γH2AX) was described more than 40 years ago and it was demonstrated that phosphorylation of H2AX was one of the first cellular responses to DNA damage. Since then, γH2AX has been implicated in diverse cellular functions in normal and pathological cells. In the first part of this review, we will briefly describe the intervention of H2AX in the DNA damage response (DDR) and its role in some pivotal cellular events, such as regulation of cell cycle checkpoints, genomic instability, cell growth, mitosis, embryogenesis, and apoptosis. Then, in the main part of this contribution, we will discuss the involvement of γH2AX in the normal and pathological central nervous system, with particular attention to the differences in the DDR between immature and mature neurons, and to the significance of H2AX phosphorylation in neurogenesis and neuronal cell death. The emerging picture is that H2AX is a pleiotropic molecule with an array of yet not fully understood functions in the brain, from embryonic life to old age.
Collapse
Affiliation(s)
| | | | | | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, I-10095 Grugliasco, Italy; (A.M.); (N.G.); (A.G.)
| |
Collapse
|
21
|
Peggion C, Massimino ML, Bonadio RS, Lia F, Lopreiato R, Cagnin S, Calì T, Bertoli A. Regulation of Endoplasmic Reticulum-Mitochondria Tethering and Ca 2+ Fluxes by TDP-43 via GSK3β. Int J Mol Sci 2021; 22:11853. [PMID: 34769284 PMCID: PMC8584823 DOI: 10.3390/ijms222111853] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Mitochondria-ER contacts (MERCs), tightly regulated by numerous tethering proteins that act as molecular and functional connections between the two organelles, are essential to maintain a variety of cellular functions. Such contacts are often compromised in the early stages of many neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). TDP-43, a nuclear protein mainly involved in RNA metabolism, has been repeatedly associated with ALS pathogenesis and other neurodegenerative diseases. Although TDP-43 neuropathological mechanisms are still unclear, the accumulation of the protein in cytoplasmic inclusions may underlie a protein loss-of-function effect. Accordingly, we investigated the impact of siRNA-mediated TDP-43 silencing on MERCs and the related cellular parameters in HeLa cells using GFP-based probes for MERCs quantification and aequorin-based probes for local Ca2+ measurements, combined with targeted protein and mRNA profiling. Our results demonstrated that TDP-43 down-regulation decreases MERCs density, thereby remarkably reducing mitochondria Ca2+ uptake after ER Ca2+ release. Thorough mRNA and protein analyses did not highlight altered expression of proteins involved in MERCs assembly or Ca2+-mediated ER-mitochondria cross-talk, nor alterations of mitochondrial density and morphology were observed by confocal microscopy. Further mechanistic inspections, however, suggested that the observed cellular alterations are correlated to increased expression/activity of GSK3β, previously associated with MERCs disruption.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (F.L.); (R.L.); (T.C.)
| | | | - Raphael Severino Bonadio
- Department of Biology, CRIBI Biotechnology Center, University of Padova, 35131 Padova, Italy; (R.S.B.); (S.C.)
| | - Federica Lia
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (F.L.); (R.L.); (T.C.)
| | - Raffaele Lopreiato
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (F.L.); (R.L.); (T.C.)
| | - Stefano Cagnin
- Department of Biology, CRIBI Biotechnology Center, University of Padova, 35131 Padova, Italy; (R.S.B.); (S.C.)
- CIR-Myo Myology Center, University of Padova, 35131 Padova, Italy
| | - Tito Calì
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (F.L.); (R.L.); (T.C.)
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (F.L.); (R.L.); (T.C.)
- CNR—Neuroscience Institute, 35131 Padova, Italy;
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
| |
Collapse
|
22
|
Choi HJ, Lee JY, Cha SJ, Han YJ, Yoon JH, Kim HJ, Kim K. FUS-induced neurotoxicity is prevented by inhibiting GSK-3β in a drosophila model of amyotrophic lateral sclerosis. Hum Mol Genet 2021; 31:850-862. [PMID: 34605896 DOI: 10.1093/hmg/ddab290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/12/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS)-linked mutations in fused in sarcoma (FUS) lead to the formation of cytoplasmic aggregates in neurons. They are believed play a critical role in the pathogenesis of FUS-associated ALS. Therefore, the clearance and degradation of cytoplasmic FUS aggregates in neurons may be considered a therapeutic strategy for ALS. However, the molecular pathogenic mechanisms behind FUS-associated ALS remain poorly understood. Here, we report GSK-3β as a potential modulator of FUS-induced toxicity. We demonstrated that RNAi-mediated knockdown of Drosophila ortholog Shaggy in FUS-expressing flies suppresses defective phenotypes, including retinal degeneration, motor defects, motor neuron degeneration, and mitochondrial dysfunction. Furthermore, we found that cytoplasmic FUS aggregates were significantly reduced by Shaggy knockdown. In addition, we found that the levels of FUS proteins were significantly reduced by co-overexpression of Slimb, a F-box protein, in FUS-expressing flies, indicating that Slimb is critical for the suppressive effect of Shaggy/GSK-3β inhibition on FUS-induced toxicity in Drosophila. These findings revealed a novel mechanism of neuronal protective effect through SCFSlimb-mediated FUS degradation via GSK-3β inhibition, and provided in vivo evidence of the potential for modulating FUS-induced ALS progression using GSK-3β inhibitors.
Collapse
Affiliation(s)
- Hyun-Jun Choi
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan 31151, Korea.,Department of Integrated Biomedical Sciences, Soonchunhyang University, Cheonan 31151, Korea
| | - Ji Young Lee
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31538, Korea
| | - Sun Joo Cha
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea
| | - Yeo Jeong Han
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31538, Korea.,Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea
| | - Ja Hoon Yoon
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31538, Korea
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu 41068, Korea
| | - Kiyoung Kim
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31538, Korea.,Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea
| |
Collapse
|
23
|
Laneve P, Tollis P, Caffarelli E. RNA Deregulation in Amyotrophic Lateral Sclerosis: The Noncoding Perspective. Int J Mol Sci 2021; 22:10285. [PMID: 34638636 PMCID: PMC8508793 DOI: 10.3390/ijms221910285] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022] Open
Abstract
RNA metabolism is central to cellular physiopathology. Almost all the molecular pathways underpinning biological processes are affected by the events governing the RNA life cycle, ranging from transcription to degradation. The deregulation of these processes contributes to the onset and progression of human diseases. In recent decades, considerable efforts have been devoted to the characterization of noncoding RNAs (ncRNAs) and to the study of their role in the homeostasis of the nervous system (NS), where they are highly enriched. Acting as major regulators of gene expression, ncRNAs orchestrate all the steps of the differentiation programs, participate in the mechanisms underlying neural functions, and are crucially implicated in the development of neuronal pathologies, among which are neurodegenerative diseases. This review aims to explore the link between ncRNA dysregulation and amyotrophic lateral sclerosis (ALS), the most frequent motoneuron (MN) disorder in adults. Notably, defective RNA metabolism is known to be largely associated with this pathology, which is often regarded as an RNA disease. We also discuss the potential role that these transcripts may play as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Pietro Laneve
- Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy
| | - Paolo Tollis
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy;
| | - Elisa Caffarelli
- Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy
| |
Collapse
|
24
|
Arenas A, Chen J, Kuang L, Barnett KR, Kasarskis EJ, Gal J, Zhu H. Lysine acetylation regulates the RNA binding, subcellular localization and inclusion formation of FUS. Hum Mol Genet 2021; 29:2684-2697. [PMID: 32691043 DOI: 10.1093/hmg/ddaa159] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/17/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the preferential death of motor neurons. Approximately 10% of ALS cases are familial and 90% are sporadic. Fused in sarcoma (FUS) is a ubiquitously expressed RNA-binding protein implicated in familial ALS and frontotemporal dementia (FTD). The physiological function and pathological mechanism of FUS are not well understood, particularly whether post-translational modifications play a role in regulating FUS function. In this study, we discovered that FUS was acetylated at lysine-315/316 (K315/K316) and lysine-510 (K510) residues in two distinct domains. Located in the nuclear localization sequence, K510 acetylation disrupted the interaction between FUS and Transportin-1, resulting in the mislocalization of FUS in the cytoplasm and formation of stress granule-like inclusions. Located in the RNA recognition motif, K315/K316 acetylation reduced RNA binding to FUS and decreased the formation of cytoplasmic inclusions. Treatment with deacetylase inhibitors also significantly reduced the inclusion formation in cells expressing ALS mutation P525L. More interestingly, familial ALS patient fibroblasts showed higher levels of FUS K510 acetylation as compared with healthy controls. Lastly, CREB-binding protein/p300 acetylated FUS, whereas both sirtuins and histone deacetylases families of lysine deacetylases contributed to FUS deacetylation. These findings demonstrate that FUS acetylation regulates the RNA binding, subcellular localization and inclusion formation of FUS, implicating a potential role of acetylation in the pathophysiological process leading to FUS-mediated ALS/FTD.
Collapse
Affiliation(s)
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry
| | - Lisha Kuang
- Department of Molecular and Cellular Biochemistry
| | | | - Edward J Kasarskis
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jozsef Gal
- Department of Molecular and Cellular Biochemistry
| | - Haining Zhu
- Department of Toxicology and Cancer Biology.,Department of Molecular and Cellular Biochemistry.,Lexington VA Medical Center, Research and Development, Lexington, KY 40502, USA
| |
Collapse
|
25
|
Tejido C, Pakravan D, Bosch LVD. Potential Therapeutic Role of HDAC Inhibitors in FUS-ALS. Front Mol Neurosci 2021; 14:686995. [PMID: 34434087 PMCID: PMC8380926 DOI: 10.3389/fnmol.2021.686995] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/29/2021] [Indexed: 01/05/2023] Open
Abstract
Mutations in the FUS gene cause amyotrophic lateral sclerosis (ALS-FUS). However, the exact pathogenic mechanism of mutant fused in sarcoma (FUS) protein is not completely understood. FUS is an RNA binding protein (RBP) localized predominantly in the nucleus, but ALS-linked FUS mutations can affect its nuclear localization signal impairing its import into the nucleus. This mislocalization to the cytoplasm facilitates FUS aggregation in cytoplasmic inclusions. Therapies targeting post translational modifications are rising as new treatments for ALS, in particular acetylation which could have a role in the dynamics of RBPs. Research using histone deacetylase (HDAC) inhibitors in FUS-ALS models showed that HDACs can influence cytoplasmic FUS localization. Inhibition of HDACs could promote acetylation of the FUS RNA binding domain (RRM) and altering its RNA interactions resulting in FUS maintenance in the nucleus. In addition, acetylation of FUS RRMs might also favor or disfavor its incorporation into pathological inclusions. In this review, we summarize and discuss the evidence for the potential role of HDACs in the context of FUS-ALS and we propose a new hypothesis based on this overview.
Collapse
Affiliation(s)
- Clara Tejido
- Vlaams Instituut voor Biotechnologie (VIB), Center for Brain and Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Donya Pakravan
- Vlaams Instituut voor Biotechnologie (VIB), Center for Brain and Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurosciences, Experimental Neurology, Leuven Brain Institute, Katholieke Universiteit Leuven (KU Leuven)-University of Leuven, Leuven, Belgium
| | - Ludo Van Den Bosch
- Vlaams Instituut voor Biotechnologie (VIB), Center for Brain and Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurosciences, Experimental Neurology, Leuven Brain Institute, Katholieke Universiteit Leuven (KU Leuven)-University of Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Borgese N, Iacomino N, Colombo SF, Navone F. The Link between VAPB Loss of Function and Amyotrophic Lateral Sclerosis. Cells 2021; 10:1865. [PMID: 34440634 PMCID: PMC8392409 DOI: 10.3390/cells10081865] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
The VAP proteins are integral adaptor proteins of the endoplasmic reticulum (ER) membrane that recruit a myriad of interacting partners to the ER surface. Through these interactions, the VAPs mediate a large number of processes, notably the generation of membrane contact sites between the ER and essentially all other cellular membranes. In 2004, it was discovered that a mutation (p.P56S) in the VAPB paralogue causes a rare form of dominantly inherited familial amyotrophic lateral sclerosis (ALS8). The mutant protein is aggregation-prone, non-functional and unstable, and its expression from a single allele appears to be insufficient to support toxic gain-of-function effects within motor neurons. Instead, loss-of-function of the single wild-type allele is required for pathological effects, and VAPB haploinsufficiency may be the main driver of the disease. In this article, we review the studies on the effects of VAPB deficit in cellular and animal models. Several basic cell physiological processes are affected by downregulation or complete depletion of VAPB, impinging on phosphoinositide homeostasis, Ca2+ signalling, ion transport, neurite extension, and ER stress. In the future, the distinction between the roles of the two VAP paralogues (A and B), as well as studies on motor neurons generated from induced pluripotent stem cells (iPSC) of ALS8 patients will further elucidate the pathogenic basis of p.P56S familial ALS, as well as of other more common forms of the disease.
Collapse
Affiliation(s)
- Nica Borgese
- CNR Institute of Neuroscience, Via Follereau 3, Bldg U28, 20854 Vedano al Lambro, Italy; (N.I.); (S.F.C.)
| | | | | | - Francesca Navone
- CNR Institute of Neuroscience, Via Follereau 3, Bldg U28, 20854 Vedano al Lambro, Italy; (N.I.); (S.F.C.)
| |
Collapse
|
27
|
Mukherjee S, Biswas D, Epari S, Shetty P, Moiyadi A, Ball GR, Srivastava S. Comprehensive proteomic analysis reveals distinct functional modules associated with skull base and supratentorial meningiomas and perturbations in collagen pathway components. J Proteomics 2021; 246:104303. [PMID: 34174477 DOI: 10.1016/j.jprot.2021.104303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/31/2021] [Accepted: 06/05/2021] [Indexed: 12/18/2022]
Abstract
Meningiomas are brain tumors that originate from the meninges and has been primarily classified into three grades by the current WHO guidelines. Although widely prevalent and can be managed by surgery there are instances when the tumors are located in difficult regions. This results in considerable challenges for complete surgical resection and further clinical management. While the genetic signature of the skull base tumors is now known to be different from the non-skull base tumors, there is a lack of information at the functional aspects of these tumors at the proteomic level. Thus, the current study thereby aims to obtain mechanistic insights between the two radiologically distinct groups of meningiomas, namely the skull base & supratentorial (non-skull base-NSB) regions. We have employed a comprehensive mass spectrometry-based label-free quantitative proteomic analysis in Skull base and supratentorial meningiomas. Further, we have used an Artificial Neural Networking employing a sparse Multilayer perceptron (MLP) architecture to predict protein concordance. A patient-derived spectral library has been employed for a novel peptide-level validation of proteins that are specific to the radiological regions using the SRM assay based targeted proteomics approach. The comprehensive proteomics enabled the identification of nearly 4000 proteins with high confidence (1%FDR ≥ 2 unique peptides) among which 170 proteins were differentially abundant in Skull base vs Supratentorial tumors (p-value ≤0.05). In silico analysis enabled mapping of the major alterations and hinted towards an overall perturbation of extracellular matrix and collagen biosynthesis components in the non-skull base meningiomas and a prominent perturbation of molecular trafficking in the skull base meningiomas. Therefore, this study has yielded novel insights into the functional association of the proteins that are differentially abundant in the two radiological subgroups. SIGNIFICANCE: In the current study, we have performed label-free proteomic analysis on fresh frozen tissue of 14 Supratentorial (NSB) and 7 Skull base meningiomas to assess perturbations in the global proteome, we have further employed an in-depth in silico analysis to map the pathways that have enabled functional mapping of the differentially abundant proteins in the Skull base and Supratentorial tumors. The findings from the above were also subjected to a machine learning-based neural networking to find out the proteins that have the most concordance of occurrence to determine the most influential proteins of the network. We further validated the differential abundance of identified protein markers in a larger patient cohort of Skull base and Supratentorial employing targeted proteomics approach to validate key protein candidates emerging from ours and other recent studies. The previous studies that have explored the skull base and convexity meningiomas have been able to reveal alterations in the genetic mutations in these tumor types. However, there are not many studies that have explored the functional aspects of these tumors, especially at the proteome level. We have attempted for the first time to map the functional modules associated with altered proteins in these tumors and have been able to identify that there is a possibility that the Skull base meningiomas to be considerably different from the Non-skull base (NSB) tumors in terms of the perturbed pathways. Our study employed global as well as targeted proteomics to examine the proteomic alterations in these two tumor groups. The study indicates that proteins that were more abundant in Skull base tumors were part of molecular transport components, non-skull base proteins majorly mapped to the components of extracellular matrix remodeling pathways. In conclusion, this study substantiates the distinction in the proteomic signatures in the skull base and supratentorial meningiomas paving way for further investigation of the identified markers for determining if some of these proteins can be used for therapeutic interventions for cases that pose considerable challenges for complete resection.
Collapse
Affiliation(s)
- Shuvolina Mukherjee
- Proteomics Lab, Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, 400076, Maharashtra, India; Department of Immunotechnology, Lund University, Medicon Village, 22100 Lund, Sweden
| | - Deeptarup Biswas
- Proteomics Lab, Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, 400076, Maharashtra, India
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Centre, Mumbai, Dr. E Borges Road, Parel, Mumbai 400 012, India
| | - Prakash Shetty
- Department of Neurosurgery, Tata Memorial Centre, Mumbai, Dr. E Borges Road, Parel, Mumbai 400 012, India
| | - Aliasgar Moiyadi
- Department of Neurosurgery, Tata Memorial Centre, Mumbai, Dr. E Borges Road, Parel, Mumbai 400 012, India
| | - Graham Roy Ball
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Sanjeeva Srivastava
- Proteomics Lab, Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, 400076, Maharashtra, India.
| |
Collapse
|
28
|
ALS-linked FUS mutants affect the localization of U7 snRNP and replication-dependent histone gene expression in human cells. Sci Rep 2021; 11:11868. [PMID: 34088960 PMCID: PMC8178370 DOI: 10.1038/s41598-021-91453-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/18/2021] [Indexed: 11/24/2022] Open
Abstract
Genes encoding replication-dependent histones lack introns, and the mRNAs produced are a unique class of RNA polymerase II transcripts in eukaryotic cells that do not end in a polyadenylated tail. Mature mRNAs are thus formed by a single endonucleolytic cleavage that releases the pre-mRNA from the DNA and is the only processing event necessary. U7 snRNP is one of the key factors that determines the cleavage site within the 3ʹUTR of replication-dependent histone pre-mRNAs. We have previously showed that the FUS protein interacts with U7 snRNA/snRNP and regulates the expression of histone genes by stimulating transcription and 3ʹ end maturation. Mutations in the FUS gene first identified in patients with amyotrophic lateral sclerosis (ALS) lead to the accumulation of the FUS protein in cytoplasmic inclusions. Here, we report that mutations in FUS lead to disruption of the transcriptional activity of FUS and mislocalization of U7 snRNA/snRNP in cytoplasmic aggregates in cellular models and primary neurons. As a consequence, decreased transcriptional efficiency and aberrant 3ʹ end processing of histone pre-mRNAs were observed. This study highlights for the first time the deregulation of replication-dependent histone gene expression and its involvement in ALS.
Collapse
|
29
|
Scekic-Zahirovic J, Sanjuan-Ruiz I, Kan V, Megat S, De Rossi P, Dieterlé S, Cassel R, Jamet M, Kessler P, Wiesner D, Tzeplaeff L, Demais V, Sahadevan S, Hembach KM, Muller HP, Picchiarelli G, Mishra N, Antonucci S, Dirrig-Grosch S, Kassubek J, Rasche V, Ludolph A, Boutillier AL, Roselli F, Polymenidou M, Lagier-Tourenne C, Liebscher S, Dupuis L. Cytoplasmic FUS triggers early behavioral alterations linked to cortical neuronal hyperactivity and inhibitory synaptic defects. Nat Commun 2021; 12:3028. [PMID: 34021132 PMCID: PMC8140148 DOI: 10.1038/s41467-021-23187-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Gene mutations causing cytoplasmic mislocalization of the RNA-binding protein FUS lead to severe forms of amyotrophic lateral sclerosis (ALS). Cytoplasmic accumulation of FUS is also observed in other diseases, with unknown consequences. Here, we show that cytoplasmic mislocalization of FUS drives behavioral abnormalities in knock-in mice, including locomotor hyperactivity and alterations in social interactions, in the absence of widespread neuronal loss. Mechanistically, we identified a progressive increase in neuronal activity in the frontal cortex of Fus knock-in mice in vivo, associated with altered synaptic gene expression. Synaptic ultrastructural and morphological defects were more pronounced in inhibitory than excitatory synapses and associated with increased synaptosomal levels of FUS and its RNA targets. Thus, cytoplasmic FUS triggers synaptic deficits, which is leading to increased neuronal activity in frontal cortex and causing related behavioral phenotypes. These results indicate that FUS mislocalization may trigger deleterious phenotypes beyond motor neuron impairment in ALS, likely relevant also for other neurodegenerative diseases characterized by FUS mislocalization.
Collapse
Affiliation(s)
- Jelena Scekic-Zahirovic
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Inmaculada Sanjuan-Ruiz
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Vanessa Kan
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
- BioMedical Center, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Salim Megat
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Pierre De Rossi
- Department of Quantitative Biomedicine, University of Zurich, Zürich, Switzerland
| | - Stéphane Dieterlé
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Raphaelle Cassel
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
- Université de Strasbourg, UMR 7364 CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Strasbourg, France
| | - Marguerite Jamet
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Pascal Kessler
- Université de Strasbourg, Inserm, Unité mixte de service du CRBS, UMS 038, Strasbourg, France
| | - Diana Wiesner
- Department of Neurology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Laura Tzeplaeff
- Université de Strasbourg, UMR 7364 CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Strasbourg, France
| | - Valérie Demais
- Plateforme Imagerie In Vitro, CNRS UPS-3156, NeuroPôle, Strasbourg, France
| | - Sonu Sahadevan
- Department of Quantitative Biomedicine, University of Zurich, Zürich, Switzerland
| | - Katharina M Hembach
- Department of Quantitative Biomedicine, University of Zurich, Zürich, Switzerland
| | | | - Gina Picchiarelli
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Nibha Mishra
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Stefano Antonucci
- Department of Neurology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Sylvie Dirrig-Grosch
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Jan Kassubek
- Department of Neurology, Ulm University, Ulm, Germany
| | - Volker Rasche
- Ulm University Medical Center, Department of Internal Medicine II, Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Anne-Laurence Boutillier
- Université de Strasbourg, UMR 7364 CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Strasbourg, France
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | | | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Sabine Liebscher
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany.
- BioMedical Center, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Luc Dupuis
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France.
| |
Collapse
|
30
|
Levone BR, Lenzken SC, Antonaci M, Maiser A, Rapp A, Conte F, Reber S, Mechtersheimer J, Ronchi AE, Mühlemann O, Leonhardt H, Cardoso MC, Ruepp MD, Barabino SM. FUS-dependent liquid-liquid phase separation is important for DNA repair initiation. J Cell Biol 2021; 220:e202008030. [PMID: 33704371 PMCID: PMC7953258 DOI: 10.1083/jcb.202008030] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 01/17/2021] [Accepted: 02/04/2021] [Indexed: 12/17/2022] Open
Abstract
RNA-binding proteins (RBPs) are emerging as important effectors of the cellular DNA damage response (DDR). The RBP FUS is implicated in RNA metabolism and DNA repair, and it undergoes reversible liquid-liquid phase separation (LLPS) in vitro. Here, we demonstrate that FUS-dependent LLPS is necessary for the initiation of the DDR. Using laser microirradiation in FUS-knockout cells, we show that FUS is required for the recruitment to DNA damage sites of the DDR factors KU80, NBS1, and 53BP1 and of SFPQ, another RBP implicated in the DDR. The relocation of KU80, NBS1, and SFPQ is similarly impaired by LLPS inhibitors, or LLPS-deficient FUS variants. We also show that LLPS is necessary for efficient γH2AX foci formation. Finally, using superresolution structured illumination microscopy, we demonstrate that the absence of FUS impairs the proper arrangement of γH2AX nanofoci into higher-order clusters. These findings demonstrate the early requirement for FUS-dependent LLPS in the activation of the DDR and the proper assembly of DSB repair complexes.
Collapse
Affiliation(s)
- Brunno R. Levone
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Silvia C. Lenzken
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Marco Antonaci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Andreas Maiser
- Department of Biology II, Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Planegg-Martinsried, Germany
| | - Alexander Rapp
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Francesca Conte
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Stefan Reber
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Jonas Mechtersheimer
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Antonella E. Ronchi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Oliver Mühlemann
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Heinrich Leonhardt
- Department of Biology II, Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Planegg-Martinsried, Germany
| | - M. Cristina Cardoso
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Marc-David Ruepp
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Silvia M.L. Barabino
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
31
|
Hachiya N, Sochocka M, Brzecka A, Shimizu T, Gąsiorowski K, Szczechowiak K, Leszek J. Nuclear Envelope and Nuclear Pore Complexes in Neurodegenerative Diseases-New Perspectives for Therapeutic Interventions. Mol Neurobiol 2021; 58:983-995. [PMID: 33067781 PMCID: PMC7878205 DOI: 10.1007/s12035-020-02168-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Transport of proteins, transcription factors, and other signaling molecules between the nucleus and cytoplasm is necessary for signal transduction. The study of these transport phenomena is particularly challenging in neurons because of their highly polarized structure. The bidirectional exchange of molecular cargoes across the nuclear envelope (NE) occurs through nuclear pore complexes (NPCs), which are aqueous channels embedded in the nuclear envelope. The NE and NPCs regulate nuclear transport but are also emerging as relevant regulators of chromatin organization and gene expression. The alterations in nuclear transport are regularly identified in affected neurons associated with human neurodegenerative diseases. This review presents insights into the roles played by nuclear transport defects in neurodegenerative disease, focusing primarily on NE proteins and NPCs. The subcellular mislocalization of proteins might be a very desirable means of therapeutic intervention in neurodegenerative disorders.
Collapse
Affiliation(s)
- Naomi Hachiya
- Tokyo Metropolitan Industrial Technology Research Institute, Tokyo, Japan
| | - Marta Sochocka
- Laboratory of Virology, Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Anna Brzecka
- Department of Pulmonology and Lung Cancer, Wroclaw Medical University, Wroclaw, Poland
| | - Takuto Shimizu
- Tokyo Metropolitan Industrial Technology Research Institute, Tokyo, Japan
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | | | | | - Jerzy Leszek
- Department of Psychiatry, Wroclaw Medical University, Wybrzeże L. Pasteura 10, 50-367, Wroclaw, Poland.
| |
Collapse
|
32
|
Buratti E. Trends in Understanding the Pathological Roles of TDP-43 and FUS Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:243-267. [PMID: 33433879 DOI: 10.1007/978-3-030-51140-1_15] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Following the discovery of TDP-43 and FUS involvement in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia (FTLD), the major challenge in the field has been to understand their physiological functions, both in normal and disease conditions. The hope is that this knowledge will improve our understanding of disease and lead to the development of effective therapeutic options. Initially, the focus has been directed at characterizing the role of these proteins in the control of RNA metabolism, because the main function of TDP-43 and FUS is to bind coding and noncoding RNAs to regulate their life cycle within cells. As a result, we now have an in-depth picture of the alterations that occur in RNA metabolism following their aggregation in various ALS/FTLD models and, to a somewhat lesser extent, in patients' brains. In parallel, progress has been made with regard to understanding how aggregation of these proteins occurs in neurons, how it can spread in different brain regions, and how these changes affect various metabolic cellular pathways to result in neuronal death. The aim of this chapter will be to provide a general overview of the trending topics in TDP-43 and FUS investigations and to highlight what might represent the most promising avenues of research in the years to come.
Collapse
Affiliation(s)
- Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.
| |
Collapse
|
33
|
Sévigny M, Bourdeau Julien I, Venkatasubramani JP, Hui JB, Dutchak PA, Sephton CF. FUS contributes to mTOR-dependent inhibition of translation. J Biol Chem 2020; 295:18459-18473. [PMID: 33082139 PMCID: PMC7939483 DOI: 10.1074/jbc.ra120.013801] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 10/15/2020] [Indexed: 12/13/2022] Open
Abstract
The amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD)-linked RNA-binding protein called FUS (fused in sarcoma) has been implicated in several aspects of RNA regulation, including mRNA translation. The mechanism by which FUS affects the translation of polyribosomes has not been established. Here we show that FUS can associate with stalled polyribosomes and that this association is sensitive to mTOR (mammalian target of rapamycin) kinase activity. Specifically, we show that FUS association with polyribosomes is increased by Torin1 treatment or when cells are cultured in nutrient-deficient media, but not when cells are treated with rapamycin, the allosteric inhibitor of mTORC1. Moreover, we report that FUS is necessary for efficient stalling of translation because deficient cells are refractory to the inhibition of mTOR-dependent signaling by Torin1. We also show that ALS-linked FUS mutants R521G and P525L associate abundantly with polyribosomes and decrease global protein synthesis. Importantly, the inhibitory effect on translation by FUS is impaired by mutations that reduce its RNA-binding affinity. These findings demonstrate that FUS is an important RNA-binding protein that mediates translational repression through mTOR-dependent signaling and that ALS-linked FUS mutants can cause a toxic gain of function in the cytoplasm by repressing the translation of mRNA at polyribosomes.
Collapse
Affiliation(s)
- Myriam Sévigny
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Isabelle Bourdeau Julien
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Janani Priya Venkatasubramani
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Jeremy B Hui
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Paul A Dutchak
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Chantelle F Sephton
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada.
| |
Collapse
|
34
|
Chen L, Wang Y, Xie J. A Human iPSC Line Carrying a de novo Pathogenic FUS Mutation Identified in a Patient With Juvenile ALS Differentiated Into Motor Neurons With Pathological Characteristics. Front Cell Neurosci 2020; 14:273. [PMID: 33093822 PMCID: PMC7507938 DOI: 10.3389/fncel.2020.00273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/31/2020] [Indexed: 11/14/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) are used to establish patient-specific cell lines and are ideal models to mirror the pathological features of diseases and investigate their underlying mechanisms in vitro, especially for rare genic diseases. Here, a de novo mutation c.1509dupA (p.R503fs) in fused in sarcoma (FUS) was detected in a patient with sporadic juvenile amyotrophic lateral sclerosis (JALS). JALS is a rare and severe form of ALS with unclear pathogenesis and no effective cure. An induced pluripotent stem cell (iPSC) line carrying the de novo mutation was established, and it represents a good tool to study JALS pathogenesis and gene therapy strategies for the treatment of this condition. The established human iPSC line carrying the de novoFUS mutation strongly expressed pluripotency markers and could be differentiated into three embryonic germ layers with no gross chromosomal aberrations. Furthermore, the iPSCs could be successfully differentiated into motor neurons exhibiting the pathological characteristics of ALS. Our results indicate that this line may be useful for uncovering the pathogenesis of sporadic JALS and screen for drugs to treat this disorder.
Collapse
Affiliation(s)
- Li Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yali Wang
- Department of Neurology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jie Xie
- Help Stem Cell Innovations, Nanjing Life Science and Technology Innovation Park, Nanjing, China
| |
Collapse
|
35
|
Chen L. FUS mutation is probably the most common pathogenic gene for JALS, especially sporadic JALS. Rev Neurol (Paris) 2020; 177:333-340. [PMID: 33036763 DOI: 10.1016/j.neurol.2020.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/15/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
Juvenile amyotrophic lateral sclerosis (JALS) is a rare and severe form of ALS. The development of gene sequencing methods has resulted in increased reports of JALS cases in recent years, and additional gene mutations in FUS have been identified. Fused in sarcoma (FUS) mutations, appeared rarely in classical ALS but indeed were the most frequent pathogenic mutations in JALS, especially in sporadic JALS. After studied the reports in the last 10 years about JALS cases, the case characteristics caused by FUS mutations and the commonality of the mutation sites were summarized in this review. FUS mutation associated with more than half of JALS and the very majority of sporadic JALS. It's worth noting that almost all of the mutations occur in nuclear localization signal (NLS) of FUS in sporadic JALS. This discovery emphasized a new perspective focus on NLS for the diagnosis and etiology of sporadic JALS as well as for further study about new treatment.
Collapse
Affiliation(s)
- L Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Janshe East Road, Zhengzhou, 450000 Henan Province, China.
| |
Collapse
|
36
|
Sukhanova MV, Singatulina AS, Pastré D, Lavrik OI. Fused in Sarcoma (FUS) in DNA Repair: Tango with Poly(ADP-ribose) Polymerase 1 and Compartmentalisation of Damaged DNA. Int J Mol Sci 2020; 21:E7020. [PMID: 32987654 PMCID: PMC7582374 DOI: 10.3390/ijms21197020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022] Open
Abstract
The fused in sarcoma (FUS) protein combines prion-like properties with a multifunctional DNA/RNA-binding domain and has functions spanning the regulation of RNA metabolism, including transcription, pre-mRNA splicing, mRNA transport and translation. In addition to its roles in RNA metabolism, FUS is implicated in the maintenance of DNA integrity. In this review, we examine the participation of FUS in major DNA repair pathways, focusing on DNA repair associated with poly(ADP-ribosyl)ation events and on how the interaction of FUS with poly(ADP-ribose) may orchestrate transient compartmentalisation of DNA strand breaks. Unravelling how prion-like RNA-binding proteins control DNA repair pathways will deepen our understanding of the pathogenesis of some neurological diseases and cancer as well as provide the basis for the development of relevant innovative therapeutic technologies. This knowledge may also extend the range of applications of poly(ADP-ribose) polymerase inhibitors to the treatment of neurodegenerative diseases related to RNA-binding proteins in the cell, e.g., amyotrophic lateral sclerosis and frontotemporal lobar degeneration.
Collapse
Affiliation(s)
- Maria V. Sukhanova
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia; (A.S.S.); (O.I.L.)
| | - Anastasia S. Singatulina
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia; (A.S.S.); (O.I.L.)
| | - David Pastré
- Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, INSERM U1204, Université Paris-Saclay, 91025 Evry, France;
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 630090 Novosibirsk, Russia; (A.S.S.); (O.I.L.)
| |
Collapse
|
37
|
Riancho J, Delgado-Alvarado M, Andreu MD, Paz-Fajardo L, Arozamena S, Gil-Bea FJ, López de Munaín A. Amyotrophic lateral sclerosis (ALS), cancer, autoimmunity and metabolic disorders: An unsolved tantalizing challenge. Br J Pharmacol 2020; 178:1269-1278. [PMID: 32497246 DOI: 10.1111/bph.15151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/21/2020] [Accepted: 05/28/2020] [Indexed: 12/27/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) commonly referred to as motor neurone disease, is a neurodegenerative disease of unknown pathogenesis that progresses rapidly and has attracted an increased amount of scholarly interest in recent years. The current conception of amyotrophic lateral sclerosis has transitioned into a more complex theory in which individual genetic risk, ageing and environmental factors interact, leading to disease onset in subjects in whom the sum of these factors reach a determined threshold. Based on this conceptualization, the environmental conditions, particularly those that are potentially modifiable, are becoming increasingly relevant. In this review, the current integrative model of the disease is discussed. In addition, we explore the role of cancer, autoimmunity and metabolic diseases as examples of novel, non-genetic and environmental factors. Together with the potential triggers or perpetuating pathogenic mechanisms along with new insights into potential lines of future research are provided. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Javier Riancho
- Service of Neurology, Hospital Sierrallana-IDIVAL, Torrelavega, Spain.,Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas, CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Manuel Delgado-Alvarado
- Service of Neurology, Hospital Sierrallana-IDIVAL, Torrelavega, Spain.,Biomedical Research Networking Center for Mental Health (CIBERSAM), ISC III, Madrid, Spain
| | | | - Lucía Paz-Fajardo
- Service of Internal Medicina, Hospital Sierrallana-IDIVAL, Torrelavega, Spain
| | - Sara Arozamena
- Service of Neurology, Hospital Sierrallana-IDIVAL, Torrelavega, Spain
| | - Francisco Javier Gil-Bea
- Centro de Investigación en Red de Enfermedades Neurodegenerativas, CIBERNED, Instituto Carlos III, Madrid, Spain.,Neurosciences Area, Biodonostia Research Institute, San Sebastián, Spain
| | - Adolfo López de Munaín
- Centro de Investigación en Red de Enfermedades Neurodegenerativas, CIBERNED, Instituto Carlos III, Madrid, Spain.,Neurosciences Area, Biodonostia Research Institute, San Sebastián, Spain.,Neurology Department, Donostia University Hospital, OSAKIDETZA, San Sebastián, Spain.,Neurosciences Department, Basque Country University, San Sebastián, Spain
| |
Collapse
|
38
|
Chen L, Li J, Lu H, Liu Y. A de novo c.1509dupA:p.R503fs mutation of FUS: report of a girl with sporadic juvenile amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:635-637. [PMID: 32501131 DOI: 10.1080/21678421.2020.1775256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Li Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Janshe East Road, Zhengzhou, Henan Province, 450000, China
| | - Junmin Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Janshe East Road, Zhengzhou, Henan Province, 450000, China
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Janshe East Road, Zhengzhou, Henan Province, 450000, China
| | - Yanru Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Janshe East Road, Zhengzhou, Henan Province, 450000, China
| |
Collapse
|
39
|
Mestre-Fos S, Penev PI, Richards JC, Dean WL, Gray RD, Chaires JB, Williams LD. Profusion of G-quadruplexes on both subunits of metazoan ribosomes. PLoS One 2019; 14:e0226177. [PMID: 31834895 PMCID: PMC6910669 DOI: 10.1371/journal.pone.0226177] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/03/2019] [Indexed: 01/17/2023] Open
Abstract
Mammalian and bird ribosomes are nearly twice the mass of prokaryotic ribosomes in part because of their extraordinarily long rRNA tentacles. Human rRNA tentacles are not fully observable in current three-dimensional structures and their conformations remain to be fully resolved. In previous work we identified sequences that favor G-quadruplexes in silico and in vitro in rRNA tentacles of the human large ribosomal subunit. We demonstrated by experiment that these sequences form G-quadruplexes in vitro. Here, using a more recent motif definition, we report additional G-quadruplex sequences on surfaces of both subunits of the human ribosome. The revised sequence definition reveals expansive arrays of potential G-quadruplex sequences on LSU tentacles. In addition, we demonstrate by a variety of experimental methods that fragments of the small subunit rRNA form G-quadruplexes in vitro. Prior to this report rRNA sequences that form G-quadruplexes were confined to the large ribosomal subunit. Our combined results indicate that the surface of the assembled human ribosome contains numerous sequences capable of forming G-quadruplexes on both ribosomal subunits. The data suggest conversion between duplexes and G-quadruplexes in response to association with proteins, ions, or other RNAs. In some systems it seems likely that the integrated population of RNA G-quadruplexes may be dominated by rRNA, which is the most abundant cellular RNA.
Collapse
Affiliation(s)
- Santi Mestre-Fos
- Center for the Origin of Life, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Petar I. Penev
- Center for the Origin of Life, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - John Colin Richards
- Center for the Origin of Life, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - William L. Dean
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Robert D. Gray
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Jonathan B. Chaires
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Loren Dean Williams
- Center for the Origin of Life, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
40
|
Fernandopulle M, Wang G, Nixon-Abell J, Qamar S, Balaji V, Morihara R, St George-Hyslop PH. Inherited and Sporadic Amyotrophic Lateral Sclerosis and Fronto-Temporal Lobar Degenerations arising from Pathological Condensates of Phase Separating Proteins. Hum Mol Genet 2019; 28:R187-R196. [PMID: 31595953 PMCID: PMC6872449 DOI: 10.1093/hmg/ddz162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
Recent work on the biophysics of proteins with low complexity, intrinsically disordered domains that have the capacity to form biological condensates has profoundly altered the concepts about the pathogenesis of inherited and sporadic neurodegenerative disorders associated with pathological accumulation of these proteins. In the present review, we use the FUS, TDP-43 and A11 proteins as examples to illustrate how missense mutations and aberrant post-translational modifications of these proteins cause amyotrophic lateral sclerosis (ALS) and fronto-temporal lobar degeneration (FTLD).
Collapse
Affiliation(s)
- Michael Fernandopulle
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - GuoZhen Wang
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Jonathon Nixon-Abell
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Seema Qamar
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Varun Balaji
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| | - Ryuta Morihara
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| | - Peter H St George-Hyslop
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| |
Collapse
|
41
|
Wu Q, Ma J, Meng W, Hui P. DLX6-AS1 promotes cell proliferation, migration and EMT of gastric cancer through FUS-regulated MAP4K1. Cancer Biol Ther 2019; 21:17-25. [PMID: 31591939 DOI: 10.1080/15384047.2019.1647050] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is the second most prevalent carcinoma resulting in cancer-related deaths in the world, with differences among geographic areas. Although the incidence and mortality rates of GC in Asia are decreasing, the search for diverse and effective therapies of GC is still needed to be fully inquired. The present research explored the expression pattern, functional role and underlying mechanism of DLX6-AS1 in GC. Firstly, we measured DLX6-AS1 expression in GC and then found the elevated level of DLX6-AS1. To further inspect the function role of DLX6-AS1 involved in GC, we performed lost-of-function assays. The silencing of DLX6-AS1 suppressed cell proliferation, migration and EMT process of GC cells. Subsequently, we uncovered that MAP4K1 was also up-regulated in GC and could be positively regulated by DLX6-AS1. Moreover, MAP4K1 down-regulation similarly inhibited GC progression. In addition, DLX6-AS1 stabilized MAP4K1 via modulating FUS. In summary, DLX6-AS1 modulated GC progression through FUS-regulated MAP4K1. Our paper exposed the role and regulatory mechanism of DLX6-AS1 in GC, which suggested a novel and valid therapy for GC patients.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiali Ma
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenying Meng
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pingping Hui
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
Lysikova EA, Kukharsky MS, Chaprov KD, Vasilieva NA, Roman AY, Ovchinnikov RK, Deykin AV, Ninkina N, Buchman VL. Behavioural impairments in mice of a novel FUS transgenic line recapitulate features of frontotemporal lobar degeneration. GENES BRAIN AND BEHAVIOR 2019; 18:e12607. [DOI: 10.1111/gbb.12607] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Ekaterina A. Lysikova
- School of BiosciencesCardiff University Cardiff UK
- Institute of Physiologically Active Compounds, Russian Academy of Sciences Chernogolovka Russian Federation
| | - Michail S. Kukharsky
- Institute of Physiologically Active Compounds, Russian Academy of Sciences Chernogolovka Russian Federation
| | - Kirill D. Chaprov
- School of BiosciencesCardiff University Cardiff UK
- Institute of Physiologically Active Compounds, Russian Academy of Sciences Chernogolovka Russian Federation
| | - Nataliia A. Vasilieva
- Institute of Physiologically Active Compounds, Russian Academy of Sciences Chernogolovka Russian Federation
| | - Andrei Y. Roman
- Institute of Physiologically Active Compounds, Russian Academy of Sciences Chernogolovka Russian Federation
| | - Ruslan K. Ovchinnikov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences Chernogolovka Russian Federation
| | - Alexey V. Deykin
- Institute of Gene Biology, Russian Academy of Sciences Moscow Russian Federation
| | - Natalia Ninkina
- School of BiosciencesCardiff University Cardiff UK
- Institute of Physiologically Active Compounds, Russian Academy of Sciences Chernogolovka Russian Federation
| | - Vladimir L. Buchman
- School of BiosciencesCardiff University Cardiff UK
- Institute of Physiologically Active Compounds, Russian Academy of Sciences Chernogolovka Russian Federation
| |
Collapse
|
43
|
Crivello M, Hogg MC, Jirström E, Halang L, Woods I, Rayner M, Coughlan KS, Lewandowski SA, Prehn JHM. Vascular regression precedes motor neuron loss in the FUS (1-359) ALS mouse model. Dis Model Mech 2019; 12:dmm.040238. [PMID: 31383794 PMCID: PMC6737946 DOI: 10.1242/dmm.040238] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/12/2019] [Indexed: 11/28/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) presents a poorly understood pathogenesis. Evidence from patients and mutant SOD1 mouse models suggests vascular damage may precede or aggravate motor dysfunction in ALS. We have previously shown angiogenin (ANG) treatment enhances motor neuron survival, delays motor dysfunction and prevents vascular regression in the SOD1G93A ALS model. However, the existence of vascular defects at different stages of disease progression remains to be established in other ALS models. Here, we assessed vascular integrity in vivo throughout different disease stages, and investigated whether ANG treatment reverses vascular regression and prolongs motor neuron survival in the FUS (1-359) mouse model of ALS. Lumbar spinal cord tissue was collected from FUS (1-359) and non-transgenic control mice at postnatal day (P)50, P90 and P120. We found a significant decrease in vascular network density in lumbar spinal cords from FUS (1-359) mice by day 90, at which point motor neuron numbers were unaffected. ANG treatment did not affect survival or counter vascular regression. Endogenous Ang1 and Vegf expression were unchanged at P50 and P90; however, we found a significant decrease in miRNA 126 at P50, indicating vascular integrity in FUS mice may be compromised via an alternative pathway. Our study demonstrates that vascular regression occurs before motor neuron degeneration in FUS (1-359) mice, and highlights that heterogeneity in responses to novel ALS therapeutics can already be detected in preclinical mouse models of ALS. This article has an associated First Person interview with the joint first authors of the paper. Summary: Vascular regression is observed prior to motor neuron loss in the FUS (1-359) mouse model of ALS, yet is not rescued by angiogenin treatment.
Collapse
Affiliation(s)
- Martin Crivello
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Marion C Hogg
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| | - Elisabeth Jirström
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| | - Luise Halang
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Ina Woods
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Megan Rayner
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Karen S Coughlan
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Sebastian A Lewandowski
- Tissue Biology Laboratory, Department of Medical Biochemistry and Biophysics, Center for Molecular Medicine, Karolinska Institute, Scheeles v. 2, 17177 Stockholm, Sweden.,Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,FutureNeuro Research Centre, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
44
|
Rossaert E, Pollari E, Jaspers T, Van Helleputte L, Jarpe M, Van Damme P, De Bock K, Moisse M, Van Den Bosch L. Restoration of histone acetylation ameliorates disease and metabolic abnormalities in a FUS mouse model. Acta Neuropathol Commun 2019; 7:107. [PMID: 31277703 PMCID: PMC6612190 DOI: 10.1186/s40478-019-0750-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022] Open
Abstract
Dysregulation of epigenetic mechanisms is emerging as a central event in neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). In many models of neurodegeneration, global histone acetylation is decreased in the affected neuronal tissues. Histone acetylation is controlled by the antagonistic actions of two protein families -the histone acetyltransferases (HATs) and the histone deacetylases (HDACs). Drugs inhibiting HDAC activity are already used in the clinic as anti-cancer agents. The aim of this study was to explore the therapeutic potential of HDAC inhibition in the context of ALS. We discovered that transgenic mice overexpressing wild-type FUS ("Tg FUS+/+"), which recapitulate many aspects of human ALS, showed reduced global histone acetylation and alterations in metabolic gene expression, resulting in a dysregulated metabolic homeostasis. Chronic treatment of Tg FUS+/+ mice with ACY-738, a potent HDAC inhibitor that can cross the blood-brain barrier, ameliorated the motor phenotype and substantially extended the life span of the Tg FUS+/+ mice. At the molecular level, ACY-738 restored global histone acetylation and metabolic gene expression, thereby re-establishing metabolite levels in the spinal cord. Taken together, our findings link epigenetic alterations to metabolic dysregulation in ALS pathology, and highlight ACY-738 as a potential therapeutic strategy to treat this devastating disease.
Collapse
|
45
|
Yu JSE, Colborne S, Hughes CS, Morin GB, Nielsen TO. The FUS-DDIT3 Interactome in Myxoid Liposarcoma. Neoplasia 2019; 21:740-751. [PMID: 31220736 PMCID: PMC6584455 DOI: 10.1016/j.neo.2019.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022] Open
Abstract
Myxoid liposarcoma is a malignant lipogenic tumor that develops in deep soft tissues. While local control rates are good, current chemotherapy options remain ineffective against metastatic disease. Myxoid liposarcoma is characterized by the FUS-DDIT3 fusion oncoprotein that is proposed to function as an aberrant transcription factor, but its exact mechanism of action has remained unclear. To identify the key functional interacting partners of FUS-DDIT3, this study utilized immunoprecipitation-mass spectrometry (IP-MS) to identify the FUS-DDIT3 interactome in whole cell lysates of myxoid liposarcoma cells, and results showed an enrichment of RNA processing proteins. Further quantitative MS analyses of FUS-DDIT3 complexes isolated from nuclear lysates showed that members of several chromatin regulatory complexes were present in the FUS-DDIT3 interactome, including NuRD, SWI/SNF, PRC1, PRC2, and MLL1 COMPASS-like complexes. Co-immunoprecipitation validated the associations of FUS-DDIT3 with BRG1/SMARCA4, BAF155/SMARCC1, BAF57/SMARCE1, and KDM1A. Data from this study provides candidates for functional validation as potential therapeutic targets, particularly for emerging epigenetic drugs.
Collapse
Affiliation(s)
- Jamie S E Yu
- Department of Pathology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| | - Shane Colborne
- British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada.
| | | | - Gregg B Morin
- British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada.
| | - Torsten O Nielsen
- Department of Pathology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
46
|
Wang H, Hegde ML. New Mechanisms of DNA Repair Defects in Fused in Sarcoma-Associated Neurodegeneration: Stage Set for DNA Repair-Based Therapeutics? J Exp Neurosci 2019; 13:1179069519856358. [PMID: 31217692 PMCID: PMC6558540 DOI: 10.1177/1179069519856358] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/21/2019] [Indexed: 11/25/2022] Open
Abstract
Genome damage and defective DNA repair are etiologically linked to several
neurodegenerative disorders, including fused in sarcoma (FUS)–associated
amyotrophic lateral sclerosis (ALS). However, the underlying mechanisms remain
enigmatic, which is a roadblock for exploiting genome repair-targeted therapies.
Our recent studies identified defects in DNA nick ligation and oxidative damage
repair caused by mutations in the RNA/DNA-binding protein FUS in familial ALS
patients. In healthy neurons, FUS protects the genome by facilitating
PARP1-dependent recruitment of XRCC1/DNA Ligase IIIα (LigIII) to oxidized genome
sites and activating LigIII via direct interaction. This is a critical step in
the repair of oxidative genome damage, a foremost challenge for postmitotic
neurons due to their high oxygen consumption. We discovered that mutant FUS
significantly inhibited the recruitment of XRCC1/LigIII to DNA strand breaks,
causing defects in DNA ligation during the repair of oxidative DNA damage, which
contributed to neurodegeneration. While the FUS loss of function was responsible
for the repair defects, increased oxidative genome damage due to mutant FUS
aggregation could exacerbate the phenomenon. We highlight how these new
molecular insights into previously undescribed DNA repair defect linked to
FUS-associated neurodegeneration could provide an important opportunity for
exploring DNA repair-based therapeutic avenues.
Collapse
Affiliation(s)
- Haibo Wang
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, USA.,Neurological Institute, Institute of Academic Medicine, Houston Methodist, Houston, TX, USA
| | - Muralidhar L Hegde
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, USA.,Neurological Institute, Institute of Academic Medicine, Houston Methodist, Houston, TX, USA.,Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
47
|
Burk K, Pasterkamp RJ. Disrupted neuronal trafficking in amyotrophic lateral sclerosis. Acta Neuropathol 2019; 137:859-877. [PMID: 30721407 PMCID: PMC6531423 DOI: 10.1007/s00401-019-01964-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/19/2019] [Accepted: 01/19/2019] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, adult-onset neurodegenerative disease caused by degeneration of motor neurons in the brain and spinal cord leading to muscle weakness. Median survival after symptom onset in patients is 3-5 years and no effective therapies are available to treat or cure ALS. Therefore, further insight is needed into the molecular and cellular mechanisms that cause motor neuron degeneration and ALS. Different ALS disease mechanisms have been identified and recent evidence supports a prominent role for defects in intracellular transport. Several different ALS-causing gene mutations (e.g., in FUS, TDP-43, or C9ORF72) have been linked to defects in neuronal trafficking and a picture is emerging on how these defects may trigger disease. This review summarizes and discusses these recent findings. An overview of how endosomal and receptor trafficking are affected in ALS is followed by a description on dysregulated autophagy and ER/Golgi trafficking. Finally, changes in axonal transport and nucleocytoplasmic transport are discussed. Further insight into intracellular trafficking defects in ALS will deepen our understanding of ALS pathogenesis and will provide novel avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Katja Burk
- Department of Neurologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, Von-Siebold-Str. 3A, 37075, Göttingen, Germany.
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
48
|
Barton SK, Gregory JM, Chandran S, Turner BJ. Could an Impairment in Local Translation of mRNAs in Glia be Contributing to Pathogenesis in ALS? Front Mol Neurosci 2019; 12:124. [PMID: 31164803 PMCID: PMC6536688 DOI: 10.3389/fnmol.2019.00124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/26/2019] [Indexed: 12/13/2022] Open
Abstract
One of the key pathways implicated in amyotrophic lateral sclerosis (ALS) pathogenesis is abnormal RNA processing. Studies to date have focussed on defects in RNA stability, splicing, and translation, but this review article will focus on the largely overlooked RNA processing mechanism of RNA trafficking, with particular emphasis on the importance of glia. In the central nervous system (CNS), oligodendrocytes can extend processes to myelinate and metabolically support up to 50 axons and astrocytes can extend processes to cover up to 100,000 synapses, all with differing local functional requirements. Furthermore, many of the proteins required in these processes are large, aggregation-prone proteins which would be difficult to transport in their fully translated, terminally-folded state. This, therefore, highlights a critical requirement in these cells for local control of protein translation, which is achieved through specific trafficking of mRNAs to each process and local translation therein. Given that a large number of RNA-binding proteins have been implicated in ALS, and RNA-binding proteins are essential for trafficking mRNAs from the nucleus to glial processes for local translation, RNA misprocessing in glial cells is a likely source of cellular dysfunction in ALS. To date, neurons have been the focus of ALS research, but an intrinsic deficit in glia, namely astrocytes and oligodendrocytes, could have an additive effect on declining neuronal function in ALS. This review article aims to highlight the key evidence that supports the contention that RNA trafficking deficits in astrocytes and oligodendrocytes may contribute to in ALS.
Collapse
Affiliation(s)
- Samantha K Barton
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jenna M Gregory
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.,UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
| | - Siddharthan Chandran
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.,UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
| | - Bradley J Turner
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
49
|
TDP-43 induces mitochondrial damage and activates the mitochondrial unfolded protein response. PLoS Genet 2019; 15:e1007947. [PMID: 31100073 PMCID: PMC6524796 DOI: 10.1371/journal.pgen.1007947] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 01/08/2019] [Indexed: 02/07/2023] Open
Abstract
Mutations in or dys-regulation of the TDP-43 gene have been associated with TDP-43 proteinopathy, a spectrum of neurodegenerative diseases including Frontotemporal Lobar Degeneration (FTLD) and Amyotrophic Lateral Sclerosis (ALS). The underlying molecular and cellular defects, however, remain unclear. Here, we report a systematic study combining analyses of patient brain samples with cellular and animal models for TDP-43 proteinopathy. Electron microscopy (EM) analyses of patient samples revealed prominent mitochondrial impairment, including abnormal cristae and a loss of cristae; these ultrastructural changes were consistently observed in both cellular and animal models of TDP-43 proteinopathy. In these models, increased TDP-43 expression induced mitochondrial dysfunction, including decreased mitochondrial membrane potential and elevated production of reactive oxygen species (ROS). TDP-43 expression suppressed mitochondrial complex I activity and reduced mitochondrial ATP synthesis. Importantly, TDP-43 activated the mitochondrial unfolded protein response (UPRmt) in both cellular and animal models. Down-regulating mitochondrial protease LonP1 increased mitochondrial TDP-43 levels and exacerbated TDP-43-induced mitochondrial damage as well as neurodegeneration. Together, our results demonstrate that TDP-43 induced mitochondrial impairment is a critical aspect in TDP-43 proteinopathy. Our work has not only uncovered a previously unknown role of LonP1 in regulating mitochondrial TDP-43 levels, but also advanced our understanding of the pathogenic mechanisms for TDP-43 proteinopathy. Our study suggests that blocking or reversing mitochondrial damage may provide a potential therapeutic approach to these devastating diseases. TDP-43 proteinopathy is a group of fatal neurological diseases. Here, we report a systematic examination of the role of mitochondrial damage in TDP-43 proteinopathy using patient brain tissues, as well as cellular and animal models. Our data show that TDP-43 induces severe mitochondrial damage, accompanied by activation of UPRmt in both cellular and animal models of TDP-43 proteinopathy. LonP1, one of the key mitochondrial proteases in UPRmt, protects against TDP-43 induced cytotoxicity and neurodegeneration. Our study uncovers LonP1 as a modifier gene for TDP-43 proteinopathy and suggests protecting against or reversing mitochondrial damage as a potential therapeutic approach to these neurodegenerative disorders.
Collapse
|
50
|
Wang B, Maxwell BA, Joo JH, Gwon Y, Messing J, Mishra A, Shaw TI, Ward AL, Quan H, Sakurada SM, Pruett-Miller SM, Bertorini T, Vogel P, Kim HJ, Peng J, Taylor JP, Kundu M. ULK1 and ULK2 Regulate Stress Granule Disassembly Through Phosphorylation and Activation of VCP/p97. Mol Cell 2019; 74:742-757.e8. [PMID: 30979586 PMCID: PMC6859904 DOI: 10.1016/j.molcel.2019.03.027] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/08/2019] [Accepted: 03/22/2019] [Indexed: 12/22/2022]
Abstract
Disturbances in autophagy and stress granule dynamics have been implicated as potential mechanisms underlying inclusion body myopathy (IBM) and related disorders. Yet the roles of core autophagy proteins in IBM and stress granule dynamics remain poorly characterized. Here, we demonstrate that disrupted expression of the core autophagy proteins ULK1 and ULK2 in mice causes a vacuolar myopathy with ubiquitin and TDP-43-positive inclusions; this myopathy is similar to that caused by VCP/p97 mutations, the most common cause of familial IBM. Mechanistically, we show that ULK1/2 localize to stress granules and phosphorylate VCP, thereby increasing VCP's activity and ability to disassemble stress granules. These data suggest that VCP dysregulation and defective stress granule disassembly contribute to IBM-like disease in Ulk1/2-deficient mice. In addition, stress granule disassembly is accelerated by an ULK1/2 agonist, suggesting ULK1/2 as targets for exploiting the higher-order regulation of stress granules for therapeutic intervention of IBM and related disorders.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brian A Maxwell
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Joung Hyuck Joo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Youngdae Gwon
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - James Messing
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Ashutosh Mishra
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Timothy I Shaw
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amber L Ward
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Honghu Quan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sadie Miki Sakurada
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Tulio Bertorini
- Department of Neurology, University of Tennessee Heath Science Center, Memphis, TN 38163, USA
| | - Peter Vogel
- Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hong Joo Kim
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - J Paul Taylor
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Mondira Kundu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|