14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Mechanism of STMN2 cryptic splice-polyadenylation and its correction for TDP-43 proteinopathies

      Read this article at

      ScienceOpenPublisherPubMed
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Loss of nuclear TDP-43 is a hallmark of neurodegeneration in TDP-43 proteinopathies, including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). TDP-43 mislocalization results in cryptic splicing and polyadenylation of pre–messenger RNAs (pre-mRNAs) encoding stathmin-2 (also known as SCG10), a protein that is required for axonal regeneration. We found that TDP-43 binding to a GU-rich region sterically blocked recognition of the cryptic 3′ splice site in STMN2 pre-mRNA. Targeting dCasRx or antisense oligonucleotides (ASOs) suppressed cryptic splicing, which restored axonal regeneration and stathmin-2–dependent lysosome trafficking in TDP-43–deficient human motor neurons. In mice that were gene-edited to contain human STMN2 cryptic splice-polyadenylation sequences, ASO injection into cerebral spinal fluid successfully corrected Stmn2 pre-mRNA misprocessing and restored stathmin-2 expression levels independently of TDP-43 binding.

          Abstract

          Rescue from TDP-43 proteinopathies

          Loss of the RNA-binding protein TDP-43 from the nuclei of affected neurons is a hallmark of neurodegeneration in TDP-43 proteinopathies, including amyotrophic lateral sclerosis and frontotemporal dementia. The RNA most affected by reduced TDP-43, STMN2 , encodes stathmin-2, a protein required for axonal regeneration after injury. Baughn et al . found that TDP-43 sterically blocks recognition of a cryptic splice site in STMN2 pre-mRNA (see the Perspective by O’Brien and Mizielinska). The CRISPR effector dCasRx or antisense oligonucleotides could block STMN2 pre-mRNA cryptic splicing. This approach was able to rescue stathmin-2 levels in TDP-43–deficient human motor neurons and mouse genes edited to contain human STMN2 cryptic splice/polyadenylation sequences. —SMH

          Abstract

          An antisense oligonucleotide approach shows potential for therapeutic restoration of stathmin-2 in TDP-43 proteinopathies.

          Abstract

          INTRODUCTION

          Nuclear clearance and cytoplasmic aggregation of the RNA-binding protein TDP-43 is the hallmark of neurodegenerative diseases called TDP-43 proteinopathies. This includes almost all instances of amyotrophic lateral sclerosis (ALS) and about half of frontotemporal dementia. In ALS, the motor neurons that innervate and trigger contraction of skeletal muscles degenerate, resulting in paralysis. One of the most highly abundant motor neuron mRNAs encodes stathmin-2, a protein necessary for axonal regeneration and maintenance of neuromuscular junctions (NMJs). Loss of functional TDP-43 is accompanied by misprocessing of the STMN2 RNA precursor, which is driven by use of cryptic splicing and polyadenylation sites, and producing a truncated RNA that encodes a nonfunctional stathmin-2 fragment.

          RATIONALE

          Recognizing that stathmin-2 is essential for axonal recovery after injury and NMJ maintenance, a central interest in TDP-43 proteinopathies is to determine the mechanism through which TDP-43 enables correct processing of STMN2 mRNAs and to develop methods to restore stathmin-2 synthesis in neurons with TDP-43 dysfunction.

          RESULTS

          We found that TDP-43 binding to a 24-base, GU-rich motif within the first intron of the STMN2 pre-mRNA was required to suppress cryptic splicing and polyadenylation. Conversion of this GU-rich binding motif into a 19-base sequence bound by the MS2 bacteriophage coat protein (MCP) ablated TDP-43 binding and produced constitutive misprocessing of STMN2 . Correct processing of this modified STMN2 pre-mRNA was restored by binding of MCP, suggesting that TDP-43 normally functions by sterically blocking access to the cryptic sites of RNA-processing factors. Further genome editing revealed that the cryptic 3′ splice acceptor, not the cryptic polyadenylation site, was essential for initiating STMN2 pre-mRNA misprocessing.

          Rescue of stathmin-2 expression and axonal regeneration after injury in human motor neurons depleted of TDP-43 was achieved with steric binding antisense oligonucleotides (ASOs). Humanization (by insertion of the human STMN2 cryptic exon) sensitized mouse Stmn2 to TDP-43 expression level. Mice alternately humanized with the cryptic exon containing a disrupted TDP-43 binding site produced chronic Stmn2 pre-mRNA misprocessing independent of TDP-43 level. ASOs were identified that when injected into cerebral spinal fluid of mice with constitutive humanized Stmn2 RNA misprocessing, restored stathmin-2 mRNA and protein levels.

          CONCLUSION

          We determined that TDP-43 binding in the first intron of the STMN2 pre-mRNA sterically blocked access of RNA processing factors that would otherwise recognize and use a cryptic 3′ splice site. We identified RNA-targeted CRISPR effectors and ASOs that restored STMN2 levels despite reduced TDP-43. ASO injection into cerebral spinal fluid, an approach feasible for human therapy, rescued stathmin-2 protein levels in the central nervous system of mice with chronically misprocessed Stmn2 pre-mRNAs.

          Related collections

          Most cited references73

          • Record: found
          • Abstract: not found
          • Article: not found

          Enzymatic assembly of DNA molecules up to several hundred kilobases.

          We describe an isothermal, single-reaction method for assembling multiple overlapping DNA molecules by the concerted action of a 5' exonuclease, a DNA polymerase and a DNA ligase. First we recessed DNA fragments, yielding single-stranded DNA overhangs that specifically annealed, and then covalently joined them. This assembly method can be used to seamlessly construct synthetic and natural genes, genetic pathways and entire genomes, and could be a useful molecular engineering tool.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis.

            Ubiquitin-positive, tau- and alpha-synuclein-negative inclusions are hallmarks of frontotemporal lobar degeneration with ubiquitin-positive inclusions and amyotrophic lateral sclerosis. Although the identity of the ubiquitinated protein specific to either disorder was unknown, we showed that TDP-43 is the major disease protein in both disorders. Pathologic TDP-43 was hyper-phosphorylated, ubiquitinated, and cleaved to generate C-terminal fragments and was recovered only from affected central nervous system regions, including hippocampus, neocortex, and spinal cord. TDP-43 represents the common pathologic substrate linking these neurodegenerative disorders.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Decoding ALS: from genes to mechanism.

              Amyotrophic lateral sclerosis (ALS) is a progressive and uniformly fatal neurodegenerative disease. A plethora of genetic factors have been identified that drive the degeneration of motor neurons in ALS, increase susceptibility to the disease or influence the rate of its progression. Emerging themes include dysfunction in RNA metabolism and protein homeostasis, with specific defects in nucleocytoplasmic trafficking, the induction of stress at the endoplasmic reticulum and impaired dynamics of ribonucleoprotein bodies such as RNA granules that assemble through liquid-liquid phase separation. Extraordinary progress in understanding the biology of ALS provides new reasons for optimism that meaningful therapies will be identified.
                Bookmark

                Author and article information

                Contributors
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                (View ORCID Profile)
                Journal
                Science
                Science
                American Association for the Advancement of Science (AAAS)
                0036-8075
                1095-9203
                March 17 2023
                March 17 2023
                : 379
                : 6637
                : 1140-1149
                Affiliations
                [1 ]Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, CA 92093, USA.
                [2 ]Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
                [3 ]Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Israel.
                [4 ]Ionis Pharmaceuticals, Carlsbad, CA 92010, USA.
                [5 ]Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME 04609, USA.
                [6 ]Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
                [7 ]Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA.
                Article
                10.1126/science.abq5622
                36927019
                12700c14-1605-459f-89cd-92da1167309a
                © 2023
                History

                Comments

                Comment on this article