101
|
Bott LC, Salomons FA, Maric D, Liu Y, Merry D, Fischbeck KH, Dantuma NP. The polyglutamine-expanded androgen receptor responsible for spinal and bulbar muscular atrophy inhibits the APC/C(Cdh1) ubiquitin ligase complex. Sci Rep 2016; 6:27703. [PMID: 27312068 PMCID: PMC4911547 DOI: 10.1038/srep27703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/24/2016] [Indexed: 01/05/2023] Open
Abstract
Polyglutamine expansion in the androgen receptor (AR) causes spinal and bulbar muscular atrophy (SBMA), an X-linked neuromuscular disease that is fully manifest only in males. It has been suggested that proteins with expanded polyglutamine tracts impair ubiquitin-dependent proteolysis due to their propensity to aggregate, but recent studies indicate that the overall activity of the ubiquitin-proteasome system is preserved in SBMA models. Here we report that AR selectively interferes with the function of the ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C), which, together with its substrate adaptor Cdh1, is critical for cell cycle arrest and neuronal architecture. We show that both wild-type and mutant AR physically interact with the APC/CCdh1 complex in a ligand-dependent fashion without being targeted for proteasomal degradation. Inhibition of APC/CCdh1 by mutant but not wild-type AR in PC12 cells results in enhanced neurite outgrowth which is typically followed by rapid neurite retraction and mitotic entry. Our data indicate a role of AR in neuronal differentiation through regulation of APC/CCdh1 and suggest abnormal cell cycle reactivation as a pathogenic mechanism in SBMA.
Collapse
Affiliation(s)
- Laura C Bott
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 17177, Sweden.,National Institute of Neurological Disorders and Stroke, Neurogenetics Branch, Bethesda, MD 20892, USA
| | - Florian A Salomons
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Dragan Maric
- Flow Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Yuhong Liu
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Diane Merry
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Kenneth H Fischbeck
- National Institute of Neurological Disorders and Stroke, Neurogenetics Branch, Bethesda, MD 20892, USA
| | - Nico P Dantuma
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 17177, Sweden
| |
Collapse
|
102
|
Transcriptional induction of the heat shock protein B8 mediates the clearance of misfolded proteins responsible for motor neuron diseases. Sci Rep 2016; 6:22827. [PMID: 26961006 PMCID: PMC4785366 DOI: 10.1038/srep22827] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/22/2016] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases (NDs) are often associated with the presence of misfolded protein inclusions. The chaperone HSPB8 is upregulated in mice, the human brain and muscle structures affected during NDs progression. HSPB8 exerts a potent pro-degradative activity on several misfolded proteins responsible for familial NDs forms. Here, we demonstrated that HSPB8 also counteracts accumulation of aberrantly localized misfolded forms of TDP-43 and its 25 KDa fragment involved in most sporadic cases of Amyotrophic Lateral Sclerosis (sALS) and of Fronto Lateral Temporal Dementia (FLTD). HSPB8 acts with BAG3 and the HSP70/HSC70-CHIP complex enhancing the autophagic removal of misfolded proteins. We performed a high-through put screening (HTS) to find small molecules capable of inducing HSPB8 in neurons for therapeutic purposes. We identified two compounds, colchicine and doxorubicin, that robustly up-regulated HSPB8 expression. Both colchicine and doxorubicin increased the expression of the master regulator of autophagy TFEB, the autophagy linker p62/SQSTM1 and the autophagosome component LC3. In line, both drugs counteracted the accumulation of TDP-43 and TDP-25 misfolded species responsible for motoneuronal death in sALS. Thus, analogs of colchicine and doxorubicin able to induce HSPB8 and with better safety and tolerability may result beneficial in NDs models.
Collapse
|
103
|
Davey RA, Grossmann M. Androgen Receptor Structure, Function and Biology: From Bench to Bedside. Clin Biochem Rev 2016; 37:3-15. [PMID: 27057074 PMCID: PMC4810760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The actions of androgens such as testosterone and dihydrotestosterone are mediated via the androgen receptor (AR), a ligand-dependent nuclear transcription factor and member of the steroid hormone nuclear receptor family. Given its widespread expression in many cells and tissues, the AR has a diverse range of biological actions including important roles in the development and maintenance of the reproductive, musculoskeletal, cardiovascular, immune, neural and haemopoietic systems. AR signalling may also be involved in the development of tumours in the prostate, bladder, liver, kidney and lung. Androgens can exert their actions via the AR in a DNA binding-dependent manner to regulate target gene transcription, or in a non-DNA binding-dependent manner to initiate rapid, cellular events such as the phosphorylation of 2(nd) messenger signalling cascades. More recently, ligand-independent actions of the AR have also been identified. Given the large volume of studies relating to androgens and the AR, this review is not intended as an extensive review of all studies investigating the AR, but rather as an overview of the structure, function, signalling pathways and biology of the AR as well as its important role in clinical medicine, with emphasis on recent developments in this field.
Collapse
Affiliation(s)
- Rachel A Davey
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, 3084, Australia
| | - Mathis Grossmann
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, 3084, Australia
| |
Collapse
|
104
|
MiR-298 Counteracts Mutant Androgen Receptor Toxicity in Spinal and Bulbar Muscular Atrophy. Mol Ther 2016; 24:937-45. [PMID: 26755334 PMCID: PMC4881766 DOI: 10.1038/mt.2016.13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/04/2016] [Indexed: 01/18/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a currently untreatable adult-onset neuromuscular disease caused by expansion of a polyglutamine repeat in the androgen receptor (AR). In SBMA, as in other polyglutamine diseases, a toxic gain of function in the mutant protein is an important factor in the disease mechanism; therefore, reducing the mutant protein holds promise as an effective treatment strategy. In this work, we evaluated a microRNA (miRNA) to reduce AR expression. From a list of predicted miRNAs that target human AR, we selected microRNA-298 (miR-298) for its ability to downregulate AR mRNA and protein levels when transfected in cells overexpressing wild-type and mutant AR and in SBMA patient-derived fibroblasts. We showed that miR-298 directly binds to the 3'-untranslated region of the human AR transcript, and counteracts AR toxicity in vitro. Intravenous delivery of miR-298 with adeno-associated virus serotype 9 vector resulted in efficient transduction of muscle and spinal cord and amelioration of the disease phenotype in SBMA mice. Our findings support the development of miRNAs as a therapeutic strategy for SBMA and other neurodegenerative disorders caused by toxic proteins.
Collapse
|
105
|
Rinaldi C, Malik B, Greensmith L. Targeted Molecular Therapies for SBMA. J Mol Neurosci 2015; 58:335-42. [PMID: 26576772 DOI: 10.1007/s12031-015-0676-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/01/2015] [Indexed: 12/11/2022]
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a late-onset neuromuscular disease caused by a polyglutamine expansion in the androgen receptor gene which results in progressive spinal and bulbar motor neuron degeneration, and muscle atrophy. Although the causative genetic defect is known, until recently, the molecular pathogenesis of the disease was unclear, resulting in few, if any, targets for therapy development. However, over the past decade, our understanding of the pathomechanisms that play a role in SBMA has increased dramatically, and several of these pathways and mechanisms have now been investigated as possible therapeutic targets. In this review, we discuss some of the key pathomechanisms implicated in SBMA and describe some of the therapeutic strategies that have been tested in SBMA to date, which fall into four main categories: (i) gene silencing; (ii) protein quality control and/or increased protein degradation; (iii) androgen deprivation; and (iv) modulation of AR function. Finally, it is also now clear that in addition to a greater understanding of the molecular mechanisms that underlie disease, the development of an effective disease modifying therapy for SBMA will require the coordinated, collaborative effort of research teams with diverse areas of expertise, clinicians, pharmaceutical companies as well as patient groups.
Collapse
Affiliation(s)
- Carlo Rinaldi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX, UK.
| | - Bilal Malik
- Sobell Department of Motor Neuroscience and Movement Disorders, Queen Square, London, WC1N 3BG, UK
| | - Linda Greensmith
- Sobell Department of Motor Neuroscience and Movement Disorders, Queen Square, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
106
|
The Role of the Protein Quality Control System in SBMA. J Mol Neurosci 2015; 58:348-64. [PMID: 26572535 DOI: 10.1007/s12031-015-0675-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/01/2015] [Indexed: 12/13/2022]
Abstract
Spinal and bulbar muscular atrophy (SBMA) or Kennedy's disease is an X-linked disease associated with the expansion of the CAG triplet repeat present in exon 1 of the androgen receptor (AR) gene. This results in the production of a mutant AR containing an elongated polyglutamine tract (polyQ) in its N-terminus. Interestingly, the ARpolyQ becomes toxic only after its activation by the natural androgenic ligands, possibly because of aberrant androgen-induced conformational changes of the ARpolyQ, which generate misfolded species. These misfolded ARpolyQ species must be cleared from motoneurons and muscle cells, and this process is mediated by the protein quality control (PQC) system. Experimental evidence suggested that failure of the PQC pathways occurs in disease, leading to ARpolyQ accumulation and toxicity in the target cells. In this review, we summarized the overall impact of mutant and misfolded ARpolyQ on the PQC system and described how molecular chaperones and the degradative pathways (ubiquitin-proteasome system (UPS), the autophagy-lysosome pathway (ALP), and the unfolded protein response (UPR), which activates the endoplasmic reticulum-associated degradation (ERAD)) are differentially affected in SBMA. We also extensively and critically reviewed several molecular and pharmacological approaches proposed to restore a global intracellular activity of the PQC system. Collectively, these data suggest that the fine and delicate equilibrium existing among the different players of the PQC system could be restored in a therapeutic perspective by the synergic/additive activities of compounds designed to tackle sequential or alternative steps of the intracellular defense mechanisms triggered against proteotoxic misfolded species.
Collapse
|
107
|
Abstract
Spinal and bulbar muscular atrophy is an X-linked neuromuscular disease caused by an expanded repeat in the androgen receptor gene. The mutant protein is toxic to motor neurons and muscle. The toxicity is ligand-dependent and likely involves aberrant interaction of the mutant androgen receptor with other nuclear factors leading to transcriptional dysregulation. Various therapeutic strategies have been effective in transgenic animal models, and the challenge now is to translate these strategies into safe and effective treatment in patients.
Collapse
|
108
|
Pandey SK, Wheeler TM, Justice SL, Kim A, Younis HS, Gattis D, Jauvin D, Puymirat J, Swayze EE, Freier SM, Bennett CF, Thornton CA, MacLeod AR. Identification and characterization of modified antisense oligonucleotides targeting DMPK in mice and nonhuman primates for the treatment of myotonic dystrophy type 1. J Pharmacol Exp Ther 2015; 355:329-40. [PMID: 26330536 PMCID: PMC4613955 DOI: 10.1124/jpet.115.226969] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/31/2015] [Indexed: 01/07/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common form of muscular dystrophy in adults. DM1 is caused by an expanded CTG repeat in the 3'-untranslated region of DMPK, the gene encoding dystrophia myotonica protein kinase (DMPK). Antisense oligonucleotides (ASOs) containing 2',4'-constrained ethyl-modified (cEt) residues exhibit a significantly increased RNA binding affinity and in vivo potency relative to those modified with other 2'-chemistries, which we speculated could translate to enhanced activity in extrahepatic tissues, such as muscle. Here, we describe the design and characterization of a cEt gapmer DMPK ASO (ISIS 486178), with potent activity in vitro and in vivo against mouse, monkey, and human DMPK. Systemic delivery of unformulated ISIS 486718 to wild-type mice decreased DMPK mRNA levels by up to 90% in liver and skeletal muscle. Similarly, treatment of either human DMPK transgenic mice or cynomolgus monkeys with ISIS 486178 led to up to 70% inhibition of DMPK in multiple skeletal muscles and ∼50% in cardiac muscle in both species. Importantly, inhibition of DMPK was well tolerated and was not associated with any skeletal muscle or cardiac toxicity. Also interesting was the demonstration that the inhibition of DMPK mRNA levels in muscle was maintained for up to 16 and 13 weeks post-treatment in mice and monkeys, respectively. These results demonstrate that cEt-modified ASOs show potent activity in skeletal muscle, and that this attractive therapeutic approach warrants further clinical investigation to inhibit the gain-of-function toxic RNA underlying the pathogenesis of DM1.
Collapse
Affiliation(s)
- Sanjay K Pandey
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Thurman M Wheeler
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Samantha L Justice
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Aneeza Kim
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Husam S Younis
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Danielle Gattis
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Dominic Jauvin
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Jack Puymirat
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Eric E Swayze
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Susan M Freier
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - C Frank Bennett
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Charles A Thornton
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - A Robert MacLeod
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| |
Collapse
|
109
|
Araki K, Nakanishi H, Nakamura T, Atsuta N, Yamada S, Hijikata Y, Hashizume A, Suzuki K, Katsuno M, Sobue G. Myotonia-like symptoms in a patient with spinal and bulbar muscular atrophy. Neuromuscul Disord 2015; 25:913-5. [DOI: 10.1016/j.nmd.2015.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 08/11/2015] [Accepted: 08/14/2015] [Indexed: 10/23/2022]
|
110
|
Aberrant Autophagic Response in The Muscle of A Knock-in Mouse Model of Spinal and Bulbar Muscular Atrophy. Sci Rep 2015; 5:15174. [PMID: 26490709 PMCID: PMC4614888 DOI: 10.1038/srep15174] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/18/2015] [Indexed: 12/12/2022] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is characterized by loss of motoneurons and sensory neurons, accompanied by atrophy of muscle cells. SBMA is due to an androgen receptor containing a polyglutamine tract (ARpolyQ) that misfolds and aggregates, thereby perturbing the protein quality control (PQC) system. Using SBMA AR113Q mice we analyzed proteotoxic stress-induced alterations of HSPB8-mediated PQC machinery promoting clearance of misfolded proteins by autophagy. In muscle of symptomatic AR113Q male mice, we found expression upregulation of Pax-7, myogenin, E2-ubiquitin ligase UBE2Q1 and acetylcholine receptor (AchR), but not of MyoD, and of two E3-ligases (MuRF-1 and Cullin3). TGFβ1 and PGC-1α were also robustly upregulated. We also found a dramatic perturbation of the autophagic response, with upregulation of most autophagic markers (Beclin-1, ATG10, p62/SQSTM1, LC3) and of the HSPB8-mediated PQC response. Both HSPB8 and its co-chaperone BAG3 were robustly upregulated together with other specific HSPB8 interactors (HSPB2 and HSPB3). Notably, the BAG3:BAG1 ratio increased in muscle suggesting preferential misfolded proteins routing to autophagy rather than to proteasome. Thus, mutant ARpolyQ induces a potent autophagic response in muscle cells. Alteration in HSPB8-based PQC machinery may represent muscle-specific biomarkers useful to assess SBMA progression in mice and patients in response to pharmacological treatments.
Collapse
|
111
|
Lieberman AP, Iniguez-Lluhi JA. Undoing SUMO aids polyQ AR. Oncotarget 2015; 6:32283-4. [PMID: 26431497 PMCID: PMC4741679 DOI: 10.18632/oncotarget.5862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Andrew P Lieberman
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
112
|
Huang S, Yang S, Guo J, Yan S, Gaertig MA, Li S, Li XJ. Large Polyglutamine Repeats Cause Muscle Degeneration in SCA17 Mice. Cell Rep 2015; 13:196-208. [PMID: 26387956 PMCID: PMC4598297 DOI: 10.1016/j.celrep.2015.08.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 07/23/2015] [Accepted: 08/21/2015] [Indexed: 01/17/2023] Open
Abstract
In polyglutamine (polyQ) diseases, large polyQ repeats cause juvenile cases with different symptoms than those of adult-onset patients, who carry smaller expanded polyQ repeats. The mechanisms behind the differential pathology mediated by different polyQ repeat lengths remain unknown. By studying knockin mouse models of spinal cerebellar ataxia-17 (SCA17), we found that a large polyQ (105 glutamines) in the TATA-box-binding protein (TBP) preferentially causes muscle degeneration and reduces the expression of muscle-specific genes. Direct expression of TBP with different polyQ repeats in mouse muscle revealed that muscle degeneration is mediated only by the large polyQ repeats. Different polyQ repeats differentially alter TBP's interaction with neuronal and muscle-specific transcription factors. As a result, the large polyQ repeat decreases the association of MyoD with TBP and DNA promoters. Our findings suggest that specific alterations in protein interactions by large polyQ repeats may account for the unique pathology in juvenile polyQ diseases.
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA; Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, China
| | - Su Yang
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA
| | - Jifeng Guo
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA
| | - Sen Yan
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 10010, China
| | - Marta A Gaertig
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA
| | - Shihua Li
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA.
| | - Xiao-Jiang Li
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 10010, China.
| |
Collapse
|
113
|
Abstract
Spinal and bulbar muscular atrophy, or Kennedy disease, is a slowly progressive X-linked neuromuscular disease caused by a trinucleotide (CAG) repeat expansion in the androgen receptor gene. Affected males typically develop weakness in their mid-40s as well as evidence of androgen insensitivity with reduced fertility and gynecomastia. Diagnosis is often delayed because of decreased awareness of the disease, although genetic testing allows for direct diagnosis. Therapeutic strategies to block the toxicity of the mutant androgen receptor have been unsuccessful thus far, and evaluation of additional candidate therapies is underway.
Collapse
Affiliation(s)
- Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, 35 Convent Drive, Bethesda, MD 20892, USA.
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, 35 Convent Drive, Bethesda, MD 20892, USA
| |
Collapse
|
114
|
Wächter N, Storch A, Hermann A. Human TDP-43 and FUS selectively affect motor neuron maturation and survival in a murine cell model of ALS by non-cell-autonomous mechanisms. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16:431-41. [PMID: 26174443 DOI: 10.3109/21678421.2015.1055275] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
TAR DNA-binding protein 43 (TDP-43) and fused in sarcoma (FUS) were recently found to cause familial and sporadic amyotrophic lateral sclerosis (ALS). The mechanisms by which mutations within these genes cause ALS are not understood. We established murine embryonic stem cell (ESC)-based cell models that stably express the human wild-type (WT) and various ALS causing mutations of TDP-43 (A315T) and FUS (R514S, R521C and P525L). We investigated their effect on pan-neuron as well as motor neuron degeneration. Finally, non-cell-autonomous mediated neurodegeneration by muscle cells was investigated. Expression of mutant hTDP-43, but not wild-type TDP-43, as well as wild-type and mutant hFUS proteins induced neuronal degeneration with partial selectivity for motor neurons. Motor neuron loss was accompanied by abnormal neurite morphology and length. In chimeric coculture experiments with control motor neurons and mutant muscle cells (as their major target cells), we detected that mutant hTDP-43 A315T as well as wild-type and hFUS P525L expression only in muscle cells is sufficient to exert degenerative effects on control motor neurons. In conclusion, our data indicate that a selective vulnerability of motor neurons expressing the pathogenic ALS-causing genes TDP-43 and FUS, is, at least in part, mediated through non-cell-autonomous mechanisms.
Collapse
Affiliation(s)
- Nicole Wächter
- a Division for Neurodegenerative Diseases, Department of Neurology , Technische Universität Dresden , Dresden , Germany.,b German Centre for Neurodegenerative Diseases (DZNE) , Dresden , Germany
| | - Alexander Storch
- a Division for Neurodegenerative Diseases, Department of Neurology , Technische Universität Dresden , Dresden , Germany.,b German Centre for Neurodegenerative Diseases (DZNE) , Dresden , Germany.,c DFG-Centre for Regenerative Therapies Dresden Cluster of Excellence / Technische Universität Dresden , Dresden , Germany
| | - Andreas Hermann
- a Division for Neurodegenerative Diseases, Department of Neurology , Technische Universität Dresden , Dresden , Germany.,b German Centre for Neurodegenerative Diseases (DZNE) , Dresden , Germany
| |
Collapse
|
115
|
Todd TW, Kokubu H, Miranda HC, Cortes CJ, La Spada AR, Lim J. Nemo-like kinase is a novel regulator of spinal and bulbar muscular atrophy. eLife 2015; 4:e08493. [PMID: 26308581 PMCID: PMC4577982 DOI: 10.7554/elife.08493] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 08/24/2015] [Indexed: 01/03/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a progressive neuromuscular disease caused by polyglutamine expansion in the androgen receptor (AR) protein. Despite extensive research, the exact pathogenic mechanisms underlying SBMA remain elusive. In this study, we present evidence that Nemo-like kinase (NLK) promotes disease pathogenesis across multiple SBMA model systems. Most remarkably, loss of one copy of Nlk rescues SBMA phenotypes in mice, including extending lifespan. We also investigated the molecular mechanisms by which NLK exerts its effects in SBMA. Specifically, we have found that NLK can phosphorylate the mutant polyglutamine-expanded AR, enhance its aggregation, and promote AR-dependent gene transcription by regulating AR-cofactor interactions. Furthermore, NLK modulates the toxicity of a mutant AR fragment via a mechanism that is independent of AR-mediated gene transcription. Our findings uncover a crucial role for NLK in controlling SBMA toxicity and reveal a novel avenue for therapy development in SBMA. DOI:http://dx.doi.org/10.7554/eLife.08493.001 Spinal and bulbar muscular atrophy (SBMA) is an inherited disease that eventually leads to degeneration in motor neurons and weakness in muscles. It is caused by a specific genetic mutation in the gene that encodes the androgen receptor protein, which leads to the production of a mutant protein that is larger than normal. Similar mutations in other genes can lead to the development of other so-called ‘polyglutamine’ diseases such as Huntington's disease and spinocerebellar ataxia. However, the precise details of how these mutations lead to disease symptoms are not known, and there are currently no effective ways of treating these conditions. Previous research has shown that an enzyme called Nemo-like kinase (or NLK for short) regulates the normal androgen receptor in cancer cells. NLK has kinase activity, that is, it adds phosphate molecules to other proteins to regulate their activity. Todd et al. used human cells, fruit flies, and mice as model systems to investigate whether NLK is involved in the development of SBMA. The experiments show that NLK promotes the development of features associated with SBMA in all three models. The kinase activity of NLK is required for these features to develop. Todd et al. also found that NLK can bind to and add phosphate molecules to the mutant version of the androgen receptor protein. This causes the mutant androgen receptor proteins to accumulate and increases the ability of the mutant proteins to activate particular genes. Todd et al.'s findings suggest that NLK promotes the development of SBMA by interacting with the mutant androgen receptor. Previous studies have shown that NLK is able to modulate the development of spinocerebellar ataxia type 1, which suggests that NLK may also play an important role in other polyglutamine diseases. The next challenge will be to fully understand the role of NLK in these diseases, which may aid future efforts to develop new treatments. DOI:http://dx.doi.org/10.7554/eLife.08493.002
Collapse
Affiliation(s)
- Tiffany W Todd
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Genetics, Yale School of Medicine, New Haven, United States
| | - Hiroshi Kokubu
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Genetics, Yale School of Medicine, New Haven, United States
| | - Helen C Miranda
- Departments of Cellular and Molecular Medicine, Neurosciences, and Pediatrics, Division of Biological Sciences, Institute for Genomic Medicine, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, United States
| | - Constanza J Cortes
- Departments of Cellular and Molecular Medicine, Neurosciences, and Pediatrics, Division of Biological Sciences, Institute for Genomic Medicine, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, United States
| | - Albert R La Spada
- Departments of Cellular and Molecular Medicine, Neurosciences, and Pediatrics, Division of Biological Sciences, Institute for Genomic Medicine, Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, United States
| | - Janghoo Lim
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Genetics, Yale School of Medicine, New Haven, United States
| |
Collapse
|
116
|
Agarwala A, Jones P, Nambi V. The role of antisense oligonucleotide therapy in patients with familial hypercholesterolemia: risks, benefits, and management recommendations. Curr Atheroscler Rep 2015; 17:467. [PMID: 25398643 DOI: 10.1007/s11883-014-0467-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Antisense oligonucleotide therapy is a promising approach for the treatment of a broad variety of medical conditions. It functions at the cellular level by interfering with RNA function, often leading to degradation of specifically targeted abnormal gene products implicated in the disease process. Mipomersen is a novel antisense oligonucleotide directed at apolipoprotein (apoB)-100, the primary apolipoprotein associated with low-density lipoprotein cholesterol (LDL-C), which has recently been approved for the treatment of familial hypercholesterolemia. A number of clinical studies have demonstrated its efficacy in lowering LDL-C and apoB levels in patients with elevated LDL-C despite maximal medical therapy using conventional lipid-lowering agents. This review outlines the risks and benefits of therapy and provides recommendations on the use of mipomersen.
Collapse
Affiliation(s)
- Anandita Agarwala
- Department of Medicine, Baylor College of Medicine, Michael E. DeBakey Veterans Affairs Medical Center, One Baylor Plaza, Houston, TX, 77030, USA,
| | | | | |
Collapse
|
117
|
Sahashi K, Katsuno M, Hung G, Adachi H, Kondo N, Nakatsuji H, Tohnai G, Iida M, Bennett CF, Sobue G. Silencing neuronal mutant androgen receptor in a mouse model of spinal and bulbar muscular atrophy. Hum Mol Genet 2015; 24:5985-94. [PMID: 26231218 DOI: 10.1093/hmg/ddv300] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/21/2015] [Indexed: 12/25/2022] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA), an adult-onset neurodegenerative disease that affects males, results from a CAG triplet repeat/polyglutamine expansions in the androgen receptor (AR) gene. Patients develop progressive muscular weakness and atrophy, and no effective therapy is currently available. The tissue-specific pathogenesis, especially relative pathological contributions between degenerative motor neurons and muscles, remains inconclusive. Though peripheral pathology in skeletal muscle caused by toxic AR protein has been recently reported to play a pivotal role in the pathogenesis of SBMA using mouse models, the role of motor neuron degeneration in SBMA has not been rigorously investigated. Here, we exploited synthetic antisense oligonucleotides to inhibit the RNA levels of mutant AR in the central nervous system (CNS) and explore its therapeutic effects in our SBMA mouse model that harbors a mutant AR gene with 97 CAG expansions and characteristic SBMA-like neurogenic phenotypes. A single intracerebroventricular administration of the antisense oligonucleotides in the presymptomatic phase efficiently suppressed the mutant gene expression in the CNS, and delayed the onset and progression of motor dysfunction, improved body weight gain and survival with the amelioration of neuronal histopathology in motor units such as spinal motor neurons, neuromuscular junctions and skeletal muscle. These findings highlight the importance of the neurotoxicity of mutant AR protein in motor neurons as a therapeutic target.
Collapse
Affiliation(s)
- Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan,
| | - Gene Hung
- Isis Pharmaceuticals, Carlsbad, CA 92008, USA and
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Naohide Kondo
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hideaki Nakatsuji
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Genki Tohnai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Madoka Iida
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan,
| |
Collapse
|
118
|
Distinct Etiological Roles for Myocytes and Motor Neurons in a Mouse Model of Kennedy's Disease/Spinobulbar Muscular Atrophy. J Neurosci 2015; 35:6444-51. [PMID: 25904795 DOI: 10.1523/jneurosci.3599-14.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Polyglutamine (polyQ) expansion of the androgen receptor (AR) causes Kennedy's disease/spinobulbar muscular atrophy (KD/SBMA) through poorly defined cellular mechanisms. Although KD/SBMA has been thought of as a motor neuron disease, recent evidence indicates a key role for skeletal muscle. To resolve which early aspects of the disease can be caused by neurogenic or myogenic mechanisms, we made use of the tet-On and Cre-loxP genetic systems to selectively and acutely express polyQ AR in either motor neurons (NeuroAR) or myocytes (MyoAR) of transgenic mice. After 4 weeks of transgene induction in adulthood, deficits in gross motor function were seen in NeuroAR mice, but not MyoAR mice. Conversely, reduced size of fast glycolytic fibers and alterations in expression of candidate genes were observed only in MyoAR mice. Both NeuroAR and MyoAR mice exhibited reduced oxidative capacity in skeletal muscles, as well as a shift in fast fibers from oxidative to glycolytic. Markers of oxidative stress were increased in the muscle of NeuroAR mice and were reduced in motor neurons of both NeuroAR and MyoAR mice. Despite secondary pathology in skeletal muscle and behavioral deficits, no pathological signs were observed in motor neurons of NeuroAR mice, possibly due to relatively low levels of polyQ AR expression. These results indicate that polyQ AR in motor neurons can produce secondary pathology in muscle. Results also support both neurogenic and myogenic contributions of polyQ AR to several acute aspects of pathology and provide further evidence for disordered cellular respiration in KD/SBMA skeletal muscle.
Collapse
|
119
|
Geary RS, Norris D, Yu R, Bennett CF. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev 2015; 87:46-51. [PMID: 25666165 DOI: 10.1016/j.addr.2015.01.008] [Citation(s) in RCA: 618] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/23/2015] [Accepted: 01/29/2015] [Indexed: 01/16/2023]
Abstract
Pharmacokinetic properties of oligonucleotides are largely driven by chemistry of the backbone and thus are sequence independent within a chemical class. Tissue bioavailability (% of administered dose) is assisted by plasma protein binding that limits glomerular filtration and ultimate urinary excretion of oligonucleotides. The substitution of one non-bridging oxygen with the more hydrophobic sulfur atom (phosphorothioate) increases both plasma stability and plasma protein binding and thus, ultimately, tissue bioavailability. Additional modifications of the sugar at the 2' position, increase RNA binding affinity and significantly increase potency, tissue half-life and prolong RNA inhibitory activity. Oligonucleotides modified in this manner consistently exhibit the highest tissue bioavailability (>90%). Systemic biodistribution is broad, and organs typically with highest concentrations are liver and kidney followed by bone marrow, adipocytes, and lymph nodes. Cell uptake is predominantly mediated by endocytosis. Both size and charge for most oligonucleotides prevents distribution across the blood brain barrier. However, modified single-strand oligonucleotides administered by intrathecal injection into the CSF distribute broadly in the CNS. The majority of intracellular oligonucleotide distribution following systemic or local administration occurs rapidly in just a few hours following administration and is facilitated by rapid endocytotic uptake mechanisms. Further understanding of the intracellular trafficking of oligonucleotides may provide further enhancements in design and ultimate potency of antisense oligonucleotides in the future.
Collapse
Affiliation(s)
- Richard S Geary
- Isis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA.
| | - Daniel Norris
- Isis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Rosie Yu
- Isis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - C Frank Bennett
- Isis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA
| |
Collapse
|
120
|
Shrader JA, Kats I, Kokkinis A, Zampieri C, Levy E, Joe GO, Woolstenhulme JG, Drinkard BE, Smith MR, Ching W, Ghosh L, Fox D, Auh S, Schindler AB, Fischbeck KH, Grunseich C. A randomized controlled trial of exercise in spinal and bulbar muscular atrophy. Ann Clin Transl Neurol 2015; 2:739-47. [PMID: 26273686 PMCID: PMC4531056 DOI: 10.1002/acn3.208] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/30/2015] [Accepted: 04/01/2015] [Indexed: 01/22/2023] Open
Abstract
Objective To determine the safety and efficacy of a home-based functional exercise program in spinal and bulbar muscular atrophy (SBMA). Methods Subjects were randomly assigned to participate in 12 weeks of either functional exercises (intervention) or a stretching program (control) at the National Institutes of Health in Bethesda, MD. A total of 54 subjects enrolled, and 50 completed the study with 24 in the functional exercise group and 26 in the stretching control group. The primary outcome measure was the Adult Myopathy Assessment Tool (AMAT) total score, and secondary measures included total activity by accelerometry, muscle strength, balance, timed up and go, sit-to-stand test, health-related quality of life, creatine kinase, and insulin-like growth factor-1. Results Functional exercise was well tolerated but did not lead to significant group differences in the primary outcome measure or any of the secondary measures. The functional exercise did not produce significantly more adverse events than stretching, and was not perceived to be difficult. To determine whether a subset of the subjects may have benefited, we divided them into high and low functioning based on baseline AMAT scores and performed a post hoc subgroup analysis. Low-functioning individuals receiving the intervention increased AMAT functional subscale scores compared to the control group. Interpretation Although these trial results indicate that functional exercise had no significant effect on total AMAT scores or on mobility, strength, balance, and quality of life, post hoc findings indicate that low-functioning men with SBMA may respond better to functional exercises, and this warrants further investigation with appropriate exercise intensity.
Collapse
Affiliation(s)
- Joseph A Shrader
- Rehabilitation Medicine Department, Clinical Center, National Institutes of HealthBethesda, Maryland
| | - Ilona Kats
- Neurogenetics Branch, National Institute of Neurological Disorders and StrokeBethesda, Maryland
| | - Angela Kokkinis
- Neurogenetics Branch, National Institute of Neurological Disorders and StrokeBethesda, Maryland
| | - Cris Zampieri
- Rehabilitation Medicine Department, Clinical Center, National Institutes of HealthBethesda, Maryland
| | - Ellen Levy
- Rehabilitation Medicine Department, Clinical Center, National Institutes of HealthBethesda, Maryland
| | - Galen O Joe
- Rehabilitation Medicine Department, Clinical Center, National Institutes of HealthBethesda, Maryland
| | - Joshua G Woolstenhulme
- Rehabilitation Medicine Department, Clinical Center, National Institutes of HealthBethesda, Maryland
| | - Bart E Drinkard
- Rehabilitation Medicine Department, Clinical Center, National Institutes of HealthBethesda, Maryland
| | - Michaele R Smith
- Rehabilitation Medicine Department, Clinical Center, National Institutes of HealthBethesda, Maryland
| | - Willie Ching
- Rehabilitation Medicine Department, Clinical Center, National Institutes of HealthBethesda, Maryland
| | - Laboni Ghosh
- Neurogenetics Branch, National Institute of Neurological Disorders and StrokeBethesda, Maryland
| | - Derrick Fox
- Neurogenetics Branch, National Institute of Neurological Disorders and StrokeBethesda, Maryland
| | - Sungyoung Auh
- Clinical Neuroscience Program, National Institute of Neurological Disorders and StrokeBethesda, Maryland
| | - Alice B Schindler
- Neurogenetics Branch, National Institute of Neurological Disorders and StrokeBethesda, Maryland
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute of Neurological Disorders and StrokeBethesda, Maryland
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and StrokeBethesda, Maryland
- Correspondence Christopher Grunseich, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892. Tel: 301-402-5423; Fax: 301-480-3365; E-mail:
| |
Collapse
|
121
|
Halievski K, Henley CL, Domino L, Poort JE, Fu M, Katsuno M, Adachi H, Sobue G, Breedlove SM, Jordan CL. Androgen-dependent loss of muscle BDNF mRNA in two mouse models of SBMA. Exp Neurol 2015; 269:224-32. [PMID: 25929689 DOI: 10.1016/j.expneurol.2015.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/03/2015] [Accepted: 04/21/2015] [Indexed: 10/23/2022]
Abstract
Transgenic expression of neurotrophic factors in skeletal muscle has been found to protect mice from neuromuscular disease, including spinal bulbar muscular atrophy (SBMA), triggering renewed interest in neurotrophic factors as therapeutic agents for treating neuromuscular disease. Because SBMA is an androgen-dependent disease, and brain-derived neurotrophic factor (BDNF) mediates effects of androgens on neuromuscular systems, we asked whether BDNF expression is impaired in two different transgenic (Tg) mouse models of SBMA, the so called "97Q" and "myogenic" SBMA models. The 97Q model globally overexpresses a full length human AR with 97 glutamine repeats whereas the myogenic model of SBMA overexpresses a wild-type rat androgen receptor (AR) only in skeletal muscle fibers. Using quantitative PCR, we find that muscle BDNF mRNA declines in an androgen-dependent manner in both models, paralleling changes in motor function, with robust deficits (6-8 fold) in both fast and slow twitch muscles of impaired Tg males. Castration rescues or reverses disease-related deficits in muscle BDNF mRNA in both models, paralleling its effect on motor function. Moreover, when disease is acutely induced in Tg females, both motor function and muscle BDNF mRNA expression plummet, with the deficit in muscle BDNF emerging before overt motor dysfunction. That androgen-dependent motor dysfunction is tightly associated with a robust and early down-regulation of muscle BDNF mRNA suggests that BDNF delivered to skeletal muscle may have therapeutic value for SBMA.
Collapse
Affiliation(s)
- Katherine Halievski
- Neuroscience Program, Michigan State University, 293 Farm Lane, 108 Giltner, East Lansing, MI 48824-1101, USA.
| | - Casey L Henley
- Neuroscience Program, Michigan State University, 293 Farm Lane, 108 Giltner, East Lansing, MI 48824-1101, USA
| | - Laurel Domino
- Neuroscience Program, Michigan State University, 293 Farm Lane, 108 Giltner, East Lansing, MI 48824-1101, USA
| | - Jessica E Poort
- Neuroscience Program, Michigan State University, 293 Farm Lane, 108 Giltner, East Lansing, MI 48824-1101, USA
| | - Martina Fu
- Neuroscience Program, Michigan State University, 293 Farm Lane, 108 Giltner, East Lansing, MI 48824-1101, USA
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, Fukuoka 807-8555, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - S Marc Breedlove
- Neuroscience Program, Michigan State University, 293 Farm Lane, 108 Giltner, East Lansing, MI 48824-1101, USA
| | - Cynthia L Jordan
- Neuroscience Program, Michigan State University, 293 Farm Lane, 108 Giltner, East Lansing, MI 48824-1101, USA
| |
Collapse
|
122
|
Cortes CJ, La Spada AR. Autophagy in polyglutamine disease: Imposing order on disorder or contributing to the chaos? Mol Cell Neurosci 2015; 66:53-61. [PMID: 25771431 DOI: 10.1016/j.mcn.2015.03.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/07/2015] [Accepted: 03/09/2015] [Indexed: 12/13/2022] Open
Abstract
Autophagy is an essential, fundamentally important catabolic pathway in which double membrane-bound vesicles form in the cytosol and encircle macromolecules and organelles to permit their degradation after fusion with lysosomes. More than a decade of research has revealed that autophagy is required for normal central nervous system (CNS) function and plays a central role in maintaining protein and organelle quality controls in neurons. Neurodegenerative diseases occur when misfolded proteins accumulate and disrupt normal cellular processes, and autophagy has emerged as a key arbiter of the cell's homeostatic response to this threat. One class of inherited neurodegenerative disease is known as the CAG/polyglutamine repeat disorders, and these diseases all result from the expansion of a CAG repeat tract in the coding regions of distinct genes. Polyglutamine (polyQ) repeat diseases result in the production polyQ-expanded proteins that misfold to form inclusions or aggregates that challenge the main cellular proteostasis system of the cell, the ubiquitin proteasome system (UPS). The UPS cannot efficiently degrade polyQ-expanded disease proteins, and components of the UPS are enriched in polyQ disease aggregate bodies found in degenerating neurons. In addition to components of the UPS, polyQ protein cytosolic aggregates co-localize with key autophagy proteins, even in autophagy deficient cells, suggesting that they probably do not reflect the formation of autophagosomes but rather the sequestration of key autophagy components. Furthermore, recent evidence now implicates polyQ proteins in the regulation of the autophagy pathway itself. Thus, a complex model emerges where polyQ proteins play a dual role as both autophagy substrates and autophagy offenders. In this review, we consider the role of autophagy in polyQ disorders and the therapeutic potential for autophagy modulation in these diseases. This article is part of a Special Issue entitled "Neuronal Protein".
Collapse
Affiliation(s)
- Constanza J Cortes
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92037, USA
| | - Albert R La Spada
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92037, USA; Department of Cellular & Molecular Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA 92037, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Rady Children's Hospital, San Diego, CA 92193, USA.
| |
Collapse
|
123
|
Oki K, Halievski K, Vicente L, Xu Y, Zeolla D, Poort J, Katsuno M, Adachi H, Sobue G, Wiseman RW, Breedlove SM, Jordan CL. Contractile dysfunction in muscle may underlie androgen-dependent motor dysfunction in spinal bulbar muscular atrophy. J Appl Physiol (1985) 2015; 118:941-52. [PMID: 25663674 DOI: 10.1152/japplphysiol.00886.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 02/03/2015] [Indexed: 01/11/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is characterized by progressive muscle weakness linked to a polyglutamine expansion in the androgen receptor (AR). Current evidence indicates that mutant AR causes SBMA by acting in muscle to perturb its function. However, information about how muscle function is impaired is scant. One fundamental question is whether the intrinsic strength of muscles, an attribute of muscle independent of its mass, is affected. In the current study, we assess the contractile properties of hindlimb muscles in vitro from chronically diseased males of three different SBMA mouse models: a transgenic (Tg) model that broadly expresses a full-length human AR with 97 CAGs (97Q), a knock-in (KI) model that expresses a humanized AR containing a CAG expansion in the first exon, and a Tg myogenic model that overexpresses wild-type AR only in skeletal muscle fibers. We found that hindlimb muscles in the two Tg models (97Q and myogenic) showed marked losses in their intrinsic strength and resistance to fatigue, but were minimally affected in KI males. However, diseased muscles of all three models showed symptoms consistent with myotonic dystrophy type 1, namely, reduced resting membrane potential and deficits in chloride channel mRNA. These data indicate that muscle dysfunction is a core feature of SBMA caused by at least some of the same pathogenic mechanisms as myotonic dystrophy. Thus mechanisms controlling muscle function per se independent of mass are prime targets for SBMA therapeutics.
Collapse
Affiliation(s)
- Kentaro Oki
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | | | - Laura Vicente
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Youfen Xu
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Donald Zeolla
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Jessica Poort
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Fukuoka, Japan; and
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Robert W Wiseman
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - S Marc Breedlove
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Cynthia L Jordan
- Neuroscience Program, Michigan State University, East Lansing, Michigan;
| |
Collapse
|
124
|
Chua JP, Reddy SL, Yu Z, Giorgetti E, Montie HL, Mukherjee S, Higgins J, McEachin RC, Robins DM, Merry DE, Iñiguez-Lluhí JA, Lieberman AP. Disrupting SUMOylation enhances transcriptional function and ameliorates polyglutamine androgen receptor-mediated disease. J Clin Invest 2015; 125:831-45. [PMID: 25607844 DOI: 10.1172/jci73214] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 11/25/2014] [Indexed: 12/20/2022] Open
Abstract
Expansion of the polyglutamine (polyQ) tract within the androgen receptor (AR) causes neuromuscular degeneration in individuals with spinobulbar muscular atrophy (SBMA). PolyQ AR has diminished transcriptional function and exhibits ligand-dependent proteotoxicity, features that have both been implicated in SBMA; however, the extent to which altered AR transcriptional function contributes to pathogenesis remains controversial. Here, we sought to dissociate effects of diminished AR function from polyQ-mediated proteotoxicity by enhancing the transcriptional activity of polyQ AR. To accomplish this, we bypassed the inhibitory effect of AR SUMOylation (where SUMO indicates small ubiquitin-like modifier) by mutating conserved lysines in the polyQ AR that are sites of SUMOylation. We determined that replacement of these residues by arginine enhances polyQ AR activity as a hormone-dependent transcriptional regulator. In a murine model, disruption of polyQ AR SUMOylation rescued exercise endurance and type I muscle fiber atrophy; it also prolonged survival. These changes occurred without overt alterations in polyQ AR expression or aggregation, revealing the favorable trophic support exerted by the ligand-activated receptor. Our findings demonstrate beneficial effects of enhancing the transcriptional function of the ligand-activated polyQ AR and indicate that the SUMOylation pathway may be a potential target for therapeutic intervention in SBMA.
Collapse
|
125
|
Wan WB, Migawa MT, Vasquez G, Murray HM, Nichols JG, Gaus H, Berdeja A, Lee S, Hart CE, Lima WF, Swayze EE, Seth PP. Synthesis, biophysical properties and biological activity of second generation antisense oligonucleotides containing chiral phosphorothioate linkages. Nucleic Acids Res 2014; 42:13456-68. [PMID: 25398895 PMCID: PMC4267618 DOI: 10.1093/nar/gku1115] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 10/23/2014] [Accepted: 10/24/2014] [Indexed: 12/19/2022] Open
Abstract
Bicyclic oxazaphospholidine monomers were used to prepare a series of phosphorothioate (PS)-modified gapmer antisense oligonucleotides (ASOs) with control of the chirality of each of the PS linkages within the 10-base gap. The stereoselectivity was determined to be 98% for each coupling. The objective of this work was to study how PS chirality influences biophysical and biological properties of the ASO including binding affinity (Tm), nuclease stability, activity in vitro and in vivo, RNase H activation and cleavage patterns (both human and E. coli) in a gapmer context. Compounds that had nine or more Sp-linkages in the gap were found to be poorly active in vitro, while compounds with uniform Rp-gaps exhibited activity very similar to that of the stereo-random parent ASOs. Conversely, when tested in vivo, the full Rp-gap compound was found to be quickly metabolized resulting in low activity. A total of 31 ASOs were prepared with control of the PS chirally of each linkage within the gap in an attempt to identify favorable Rp/Sp positions. We conclude that a mix of Rp and Sp is required to achieve a balance between good activity and nuclease stability.
Collapse
Affiliation(s)
- W Brad Wan
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Michael T Migawa
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Guillermo Vasquez
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Heather M Murray
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Josh G Nichols
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Hans Gaus
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Andres Berdeja
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Sam Lee
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | | | - Walt F Lima
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Eric E Swayze
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| | - Punit P Seth
- Isis Pharmaceuticals, Inc., 2855 Gazelle Ct, Carlsbad, CA 92010, USA
| |
Collapse
|
126
|
Dossena M, Bedini G, Rusmini P, Giorgetti E, Canazza A, Tosetti V, Salsano E, Sagnelli A, Mariotti C, Gellera C, Navone SE, Marfia G, Alessandri G, Corsi F, Parati EA, Pareyson D, Poletti A. Human adipose-derived mesenchymal stem cells as a new model of spinal and bulbar muscular atrophy. PLoS One 2014; 9:e112746. [PMID: 25392924 PMCID: PMC4231043 DOI: 10.1371/journal.pone.0112746] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 10/13/2014] [Indexed: 01/08/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) or Kennedy's disease is an X-linked CAG/polyglutamine expansion motoneuron disease, in which an elongated polyglutamine tract (polyQ) in the N-terminal androgen receptor (ARpolyQ) confers toxicity to this protein. Typical markers of SBMA disease are ARpolyQ intranuclear inclusions. These are generated after the ARpolyQ binds to its endogenous ligands, which promotes AR release from chaperones, activation and nuclear translocation, but also cell toxicity. The SBMA mouse models developed so far, and used in preclinical studies, all contain an expanded CAG repeat significantly longer than that of SBMA patients. Here, we propose the use of SBMA patients adipose-derived mesenchymal stem cells (MSCs) as a new human in vitro model to study ARpolyQ toxicity. These cells have the advantage to express only ARpolyQ, and not the wild type AR allele. Therefore, we isolated and characterized adipose-derived MSCs from three SBMA patients (ADSC from Kennedy's patients, ADSCK) and three control volunteers (ADSCs). We found that both ADSCs and ADSCKs express mesenchymal antigens, even if only ADSCs can differentiate into the three typical cell lineages (adipocytes, chondrocytes and osteocytes), whereas ADSCKs, from SBMA patients, showed a lower growth potential and differentiated only into adipocyte. Moreover, analysing AR expression on our mesenchymal cultures we found lower levels in all ADSCKs than ADSCs, possibly related to negative pressures exerted by toxic ARpolyQ in ADSCKs. In addition, with proteasome inhibition the ARpolyQ levels increased specifically in ADSCKs, inducing the formation of HSP70 and ubiquitin positive nuclear ARpolyQ inclusions. Considering all of this evidence, SBMA patients adipose-derived MSCs cultures should be considered an innovative in vitro human model to understand the molecular mechanisms of ARpolyQ toxicity and to test novel therapeutic approaches in SBMA.
Collapse
Affiliation(s)
- Marta Dossena
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gloria Bedini
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro Interdipartimentale sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milan, Italy
| | - Elisa Giorgetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro Interdipartimentale sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milan, Italy
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, United States of America
| | - Alessandra Canazza
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Valentina Tosetti
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ettore Salsano
- Clinic of Central and Peripheral Degenerative Neuropathies Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Anna Sagnelli
- Clinic of Central and Peripheral Degenerative Neuropathies Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Caterina Mariotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Department of Diagnostic and Applied Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cinzia Gellera
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Department of Diagnostic and Applied Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefania Elena Navone
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanni Marfia
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fabio Corsi
- Surgery Division, Department of Clinical Sciences, University of Milan, “Luigi Sacco” Hospital, Milan, Italy
| | - Eugenio Agostino Parati
- Cellular Neurobiology Laboratory, Unit of Cerebrovascular Disease, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Davide Pareyson
- Clinic of Central and Peripheral Degenerative Neuropathies Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- * E-mail: (DP); (AP)
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro Interdipartimentale sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milan, Italy
- * E-mail: (DP); (AP)
| |
Collapse
|
127
|
Galbiati M, Crippa V, Rusmini P, Cristofani R, Cicardi ME, Giorgetti E, Onesto E, Messi E, Poletti A. ALS-related misfolded protein management in motor neurons and muscle cells. Neurochem Int 2014; 79:70-8. [PMID: 25451799 DOI: 10.1016/j.neuint.2014.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/11/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is the most common form of adult-onset motor neuron disease. It is now considered a multi-factorial and multi-systemic disorder in which alterations of the crosstalk between neuronal and non-neuronal cell types might influence the course of the disease. In this review, we will provide evidence that dysfunctions of affected muscle cells are not only a marginal consequence of denervation associated to motor neurons loss, but a direct consequence of cell muscle toxicity of mutant SOD1. In muscle, the misfolded state of mutant SOD1 protein, unlike in motor neurons, does not appear to have direct effects on protein aggregation and mitochondrial functionality. Muscle cells are, in fact, more capable than motor neurons to handle misfolded proteins, suggesting that mutant SOD1 toxicity in muscle is not mediated by classical mechanisms of intracellular misfolded proteins accumulation. Several recent works indicate that a higher activation of molecular chaperones and degradative systems is present in muscle cells, which for this reason are possibly able to better manage misfolded mutant SOD1. However, several alterations in gene expression and regenerative potential of skeletal muscles have also been reported as a consequence of the expression of mutant SOD1 in muscle. Whether these changes in muscle cells are causative of ALS or a consequence of motor neuron alterations is not yet clear, but their elucidation is very important, since the understanding of the mechanisms involved in mutant SOD1 toxicity in muscle may facilitate the design of treatments directed toward this specific tissue to treat ALS or at least to delay disease progression.
Collapse
Affiliation(s)
- Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy; InterUniversity Center on Neurodegenerative Diseases (CIMN), Università degli Studi di Firenze, Roma "Tor Vergata", Genova and Milano, Italy
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy; InterUniversity Center on Neurodegenerative Diseases (CIMN), Università degli Studi di Firenze, Roma "Tor Vergata", Genova and Milano, Italy
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy; InterUniversity Center on Neurodegenerative Diseases (CIMN), Università degli Studi di Firenze, Roma "Tor Vergata", Genova and Milano, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy; InterUniversity Center on Neurodegenerative Diseases (CIMN), Università degli Studi di Firenze, Roma "Tor Vergata", Genova and Milano, Italy
| | - Maria Elena Cicardi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy
| | - Elisa Giorgetti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Elisa Onesto
- Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Elio Messi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy; InterUniversity Center on Neurodegenerative Diseases (CIMN), Università degli Studi di Firenze, Roma "Tor Vergata", Genova and Milano, Italy.
| |
Collapse
|
128
|
Maguire CA, Ramirez SH, Merkel SF, Sena-Esteves M, Breakefield XO. Gene therapy for the nervous system: challenges and new strategies. Neurotherapeutics 2014; 11:817-39. [PMID: 25159276 PMCID: PMC4391389 DOI: 10.1007/s13311-014-0299-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Current clinical treatments for central nervous system (CNS) diseases, such as Parkinson's disease and glioblastoma do not halt disease progression and have significant treatment morbidities. Gene therapy has the potential to "permanently" correct disease by bringing in a normal gene to correct a mutant gene deficiency, knocking down mRNA of mutant alleles, and inducing cell-death in cancer cells using transgenes encoding apoptosis-inducing proteins. Promising results in clinical trials of eye disease (Leber's congenital aumorosis) and Parkinson's disease have shown that gene-based neurotherapeutics have great potential. The recent development of genome editing technology, such as zinc finger nucleases, TALENS, and CRISPR, has made the ultimate goal of gene correction a step closer. This review summarizes the challenges faced by gene-based neurotherapeutics and the current and recent strategies designed to overcome these barriers. We have chosen the following challenges to focus on in this review: (1) delivery vehicles (both virus and nonviral), (2) use of promoters for vector-mediated gene expression in CNS, and (3) delivery across the blood-brain barrier. The final section (4) focuses on promising pre-clinical/clinical studies of neurotherapeutics.
Collapse
Affiliation(s)
- Casey A Maguire
- Department of Neurology, Massachusetts General Hospital, and Neuroscience Program, Harvard Medical School, Molecular Neurogenetics Unit, 13th Street, Building 149, Charlestown, MA, 02129, USA,
| | | | | | | | | |
Collapse
|
129
|
Katsuno M, Watanabe H, Yamamoto M, Sobue G. Potential therapeutic targets in polyglutamine-mediated diseases. Expert Rev Neurother 2014; 14:1215-28. [PMID: 25190502 DOI: 10.1586/14737175.2014.956727] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Polyglutamine diseases are a group of inherited neurodegenerative disorders that are caused by an abnormal expansion of a trinucleotide CAG repeat, which encodes a polyglutamine tract in the protein-coding region of the respective disease genes. To date, nine polyglutamine diseases are known, including Huntington's disease, spinal and bulbar muscular atrophy, dentatorubral-pallidoluysian atrophy and six forms of spinocerebellar ataxia. These diseases share a salient molecular pathophysiology including the aggregation of the mutant protein followed by the disruption of cellular functions such as transcriptional regulation and axonal transport. The intraneuronal accumulation of mutant protein and resulting cellular dysfunction are the essential targets for the development of disease-modifying therapies, some of which have shown beneficial effects in animal models. In this review, the current status of and perspectives on therapy development for polyglutamine diseases will be discussed.
Collapse
Affiliation(s)
- Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | |
Collapse
|
130
|
Iida M, Katsuno M, Nakatsuji H, Adachi H, Kondo N, Miyazaki Y, Tohnai G, Ikenaka K, Watanabe H, Yamamoto M, Kishida K, Sobue G. Pioglitazone suppresses neuronal and muscular degeneration caused by polyglutamine-expanded androgen receptors. Hum Mol Genet 2014; 24:314-29. [DOI: 10.1093/hmg/ddu445] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
131
|
|
132
|
Giorgetti E, Rusmini P, Crippa V, Cristofani R, Boncoraglio A, Cicardi ME, Galbiati M, Poletti A. Synergic prodegradative activity of Bicalutamide and trehalose on the mutant androgen receptor responsible for spinal and bulbar muscular atrophy. Hum Mol Genet 2014; 24:64-75. [PMID: 25122660 PMCID: PMC4262493 DOI: 10.1093/hmg/ddu419] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is an X-linked motoneuron disease due to a CAG triplet-repeat expansion in the androgen receptor (AR) gene, which is translated into an elongated polyglutamine (polyQ) tract in AR protein (ARpolyQ). ARpolyQ toxicity is activated by the AR ligand testosterone (or dihydrotestosterone), and the polyQ triggers ARpolyQ misfolding and aggregation in spinal cord motoneurons and muscle cells. In motoneurons, testosterone triggers nuclear toxicity by inducing AR nuclear translocation. Thus, (i) prevention of ARpolyQ nuclear localization, combined with (ii) an increased ARpolyQ cytoplasmic clearance, should reduce its detrimental activity. Using the antiandrogen Bicalutamide (Casodex®), which slows down AR activation and nuclear translocation, and the disaccharide trehalose, an autophagy activator, we found that, in motoneurons, the two compounds together reduced ARpolyQ insoluble forms with higher efficiency than that obtained with single treatments. The ARpolyQ clearance was mediated by trehalose-induced autophagy combined with the longer cytoplasmic retention of ARpolyQ bound to Bicalutamide. This allows an increased recognition of misfolded species by the autophagic system prior to their migration into the nucleus. Interestingly, the combinatory use of trehalose and Bicalutamide was also efficient in the removal of insoluble species of AR with a very long polyQ (Q112) tract, which typically aggregates into the cell nuclei. Collectively, these data suggest that the combinatory use of Bicalutamide and trehalose is a novel approach to facilitate ARpolyQ clearance that has to be tested in other cell types target of SBMA (i.e. muscle cells) and in vivo in animal models of SBMA.
Collapse
Affiliation(s)
- Elisa Giorgetti
- Sezione di Biomedicina ed Endocrinologia, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milano 20133, Italy Centro InterUniversitario sulle Malattie Neurodegenerative, Università degli Studi di Firenze, Genova e Roma Tor Vergata, Milano 20133, Italy Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA and
| | - Paola Rusmini
- Sezione di Biomedicina ed Endocrinologia, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milano 20133, Italy Centro InterUniversitario sulle Malattie Neurodegenerative, Università degli Studi di Firenze, Genova e Roma Tor Vergata, Milano 20133, Italy
| | - Valeria Crippa
- Sezione di Biomedicina ed Endocrinologia, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milano 20133, Italy Centro InterUniversitario sulle Malattie Neurodegenerative, Università degli Studi di Firenze, Genova e Roma Tor Vergata, Milano 20133, Italy
| | - Riccardo Cristofani
- Sezione di Biomedicina ed Endocrinologia, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milano 20133, Italy Centro InterUniversitario sulle Malattie Neurodegenerative, Università degli Studi di Firenze, Genova e Roma Tor Vergata, Milano 20133, Italy
| | - Alessandra Boncoraglio
- Sezione di Biomedicina ed Endocrinologia, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milano 20133, Italy Centro InterUniversitario sulle Malattie Neurodegenerative, Università degli Studi di Firenze, Genova e Roma Tor Vergata, Milano 20133, Italy Department of Cell Biology, University Medical Center of Groningen, RB 9700 Groningen, The Netherlands
| | - Maria E Cicardi
- Sezione di Biomedicina ed Endocrinologia, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milano 20133, Italy Centro InterUniversitario sulle Malattie Neurodegenerative, Università degli Studi di Firenze, Genova e Roma Tor Vergata, Milano 20133, Italy
| | - Mariarita Galbiati
- Sezione di Biomedicina ed Endocrinologia, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milano 20133, Italy Centro InterUniversitario sulle Malattie Neurodegenerative, Università degli Studi di Firenze, Genova e Roma Tor Vergata, Milano 20133, Italy
| | - Angelo Poletti
- Sezione di Biomedicina ed Endocrinologia, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milano 20133, Italy Centro InterUniversitario sulle Malattie Neurodegenerative, Università degli Studi di Firenze, Genova e Roma Tor Vergata, Milano 20133, Italy
| |
Collapse
|
133
|
Cortes CJ, Ling SC, Guo LT, Hung G, Tsunemi T, Ly L, Tokunaga S, Lopez E, Sopher BL, Bennett CF, Shelton GD, Cleveland DW, La Spada AR. Muscle expression of mutant androgen receptor accounts for systemic and motor neuron disease phenotypes in spinal and bulbar muscular atrophy. Neuron 2014; 82:295-307. [PMID: 24742458 DOI: 10.1016/j.neuron.2014.03.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2014] [Indexed: 02/07/2023]
Abstract
X-linked spinal and bulbar muscular atrophy (SBMA) is characterized by adult-onset muscle weakness and lower motor neuron degeneration. SBMA is caused by CAG-polyglutamine (polyQ) repeat expansions in the androgen receptor (AR) gene. Pathological findings include motor neuron loss, with polyQ-AR accumulation in intranuclear inclusions. SBMA patients exhibit myopathic features, suggesting a role for muscle in disease pathogenesis. To determine the contribution of muscle, we developed a BAC mouse model featuring a floxed first exon to permit cell-type-specific excision of human AR121Q. BAC fxAR121 mice develop systemic and neuromuscular phenotypes, including shortened survival. After validating termination of AR121 expression and full rescue with ubiquitous Cre, we crossed BAC fxAR121 mice with Human Skeletal Actin-Cre mice. Muscle-specific excision prevented weight loss, motor phenotypes, muscle pathology, and motor neuronopathy and dramatically extended survival. Our results reveal a crucial role for muscle expression of polyQ-AR in SBMA and suggest muscle-directed therapies as effective treatments.
Collapse
Affiliation(s)
- Constanza J Cortes
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shuo-Chien Ling
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ling T Guo
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gene Hung
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Taiji Tsunemi
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Linda Ly
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seiya Tokunaga
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Edith Lopez
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bryce L Sopher
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - C Frank Bennett
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - G Diane Shelton
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Don W Cleveland
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Albert R La Spada
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Rady Children's Hospital, San Diego, CA 92123, USA.
| |
Collapse
|
134
|
Abstract
Polyglutamine expansion in the androgen receptor causes Kennedy's disease. Two recent reports, Cortes et al. (2014) in this issue of Neuron and Lieberman et al. (2014) in Cell Reports, raise the possibility that targeting expression of the mutant protein in skeletal muscle, instead of the nervous system, may mitigate manifestations of this disorder.
Collapse
Affiliation(s)
- Carlo Rinaldi
- Neurogenetics Branch, National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura C Bott
- Neurogenetics Branch, National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; Department of Cell and Molecular Biology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
135
|
Cortes CJ, La Spada AR. Motor neuron degeneration in spinal and Bulbar Muscular Atrophy is a skeletal muscle-driven process: Relevance to therapy development and implications for related motor neuron diseases. Rare Dis 2014; 2:e962402. [PMID: 26942099 PMCID: PMC4755247 DOI: 10.4161/2167549x.2014.962402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/20/2014] [Accepted: 08/29/2014] [Indexed: 11/19/2022] Open
Abstract
Non-cell autonomous degeneration has arisen as an important mechanism in neurodegenerative disorders. Using a novel line of BAC androgen receptor (AR) transgenic mice with a floxed transgene (BAC fxAR121), we uncovered a key role for skeletal muscle in X-linked Spinal and Bulbar Muscular Atrophy (SBMA), a motor neuronopathy caused by a polyglutamine expansion in exon 1 of the AR gene. By excising the mutant AR transgene from muscle only, we achieved complete rescue of neuromuscular phenotypes in these mice, despite retaining strong CNS expression. Furthermore, we delivered an antisense oligonucleotide (ASO) directed against the human AR transgene by peripheral injection, and documented that peripheral ASO delivery could rescue muscle weakness and premature death in BAC fxAR121 mice. Our results reveal a crucial role for skeletal muscle in SBMA disease pathogenesis, and offer an appealing avenue for therapy development for SBMA and perhaps also for related motor neuron diseases.
Collapse
Affiliation(s)
- Constanza J Cortes
- Department of Pediatrics; Cellular & Molecular Medicine and Neurosciences; Division of Biological Sciences; Institute for Genomic Medicine; Sanford Consortium for Regenerative Medicine; University of California ; San Diego, CA USA
| | - Albert R La Spada
- Department of Pediatrics; Cellular & Molecular Medicine and Neurosciences; Division of Biological Sciences; Institute for Genomic Medicine; Sanford Consortium for Regenerative Medicine; University of California; San Diego, CA USA; Rady Children's Hospital; San Diego, CA USA
| |
Collapse
|