1
|
Happi Mbakam C, Lamothe G, Tremblay JP. Therapeutic Strategies for Dystrophin Replacement in Duchenne Muscular Dystrophy. Front Med (Lausanne) 2022; 9:859930. [PMID: 35419381 PMCID: PMC8995704 DOI: 10.3389/fmed.2022.859930] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked hereditary disease characterized by progressive muscle wasting due to modifications in the DMD gene (exon deletions, nonsense mutations, intra-exonic insertions or deletions, exon duplications, splice site defects, and deep intronic mutations) that result in a lack of functional dystrophin expression. Many therapeutic approaches have so far been attempted to induce dystrophin expression and improve the patient phenotype. In this manuscript, we describe the relevant updates for some therapeutic strategies for DMD aiming to restore dystrophin expression. We also present and analyze in vitro and in vivo ongoing experimental approaches to treat the disease.
Collapse
Affiliation(s)
- Cedric Happi Mbakam
- Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Gabriel Lamothe
- Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Jacques P Tremblay
- Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
2
|
Therapeutic potential of highly functional codon-optimized microutrophin for muscle-specific expression. Sci Rep 2022; 12:848. [PMID: 35039573 PMCID: PMC8764061 DOI: 10.1038/s41598-022-04892-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/03/2022] [Indexed: 12/26/2022] Open
Abstract
High expectations have been set on gene therapy with an AAV-delivered shortened version of dystrophin (µDys) for Duchenne muscular dystrophy (DMD), with several drug candidates currently undergoing clinical trials. Safety concerns with this therapeutic approach include the immune response to introduced dystrophin antigens observed in some DMD patients. Recent reports highlighted microutrophin (µUtrn) as a less immunogenic functional dystrophin substitute for gene therapy. In the current study, we created a human codon-optimized µUtrn which was subjected to side-by-side characterization with previously reported mouse and human µUtrn sequences after rAAV9 intramuscular injections in mdx mice. Long-term studies with systemic delivery of rAAV9-µUtrn demonstrated robust transgene expression in muscles, with localization to the sarcolemma, functional improvement of muscle performance, decreased creatine kinase levels, and lower immunogenicity as compared to µDys. An extensive toxicity study in wild-type rats did not reveal adverse changes associated with high-dose rAAV9 administration and human codon-optimized µUtrn overexpression. Furthermore, we verified that muscle-specific promoters MHCK7 and SPc5-12 drive a sufficient level of rAAV9-µUtrn expression to ameliorate the dystrophic phenotype in mdx mice. Our results provide ground for taking human codon-optimized µUtrn combined with muscle-specific promoters into clinical development as safe and efficient gene therapy for DMD.
Collapse
|
3
|
PMO-based let-7c site blocking oligonucleotide (SBO) mediated utrophin upregulation in mdx mice, a therapeutic approach for Duchenne muscular dystrophy (DMD). Sci Rep 2020; 10:21492. [PMID: 33298994 PMCID: PMC7726560 DOI: 10.1038/s41598-020-76338-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Upregulation of utrophin, a dystrophin related protein, is considered a promising therapeutic approach for Duchenne muscular dystrophy (DMD). Utrophin expression is repressed at the post-transcriptional level by a set of miRNAs, among which let-7c is evolutionarily highly conserved. We designed PMO-based SBOs complementary to the let-7c binding site in UTRN 3′UTR, with the goal of inhibiting let-7c interaction with UTRN mRNA and thus upregulating utrophin. We used the C2C12UTRN5′luc3′ reporter cell line in which the 5′- and 3′-UTRs of human UTRN sequences flank luciferase, for reporter assays and the C2C12 cell line for utrophin western blots, to independently evaluate the site blocking efficiency of a series of let-7c PMOs in vitro. Treatment of one-month old mdx mice with the most effective let-7c PMO (i.e. S56) resulted in ca. two-fold higher utrophin protein expression in skeletal muscles and the improvement in dystrophic pathophysiology in mdx mice, in vivo. In summary, we show that PMO-based let-7c SBO has potential applicability for upregulating utrophin expression as a therapeutic approach for DMD.
Collapse
|
4
|
Sengupta K, Mishra MK, Loro E, Spencer MJ, Pyle AD, Khurana TS. Genome Editing-Mediated Utrophin Upregulation in Duchenne Muscular Dystrophy Stem Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:500-509. [PMID: 33230452 PMCID: PMC7554652 DOI: 10.1016/j.omtn.2020.08.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/25/2020] [Indexed: 12/26/2022]
Abstract
Utrophin upregulation is considered a promising therapeutic strategy for Duchenne muscular dystrophy (DMD). A number of microRNAs (miRNAs) post-transcriptionally regulate utrophin expression by binding their cognate sites in the 3′ UTR. Previously we have shown that miRNA: UTRN repression can be alleviated using miRNA let-7c site blocking oligonucleotides (SBOs) to achieve utrophin upregulation and functional improvement in mdx mice. Here, we used CRISPR/Cas9-mediated genome editing to delete five miRNA binding sites (miR-150, miR-296-5p, miR-133b, let-7c, miR-196b) clustered in a 500 bp inhibitory miRNA target region (IMTR) within the UTRN 3′ UTR, for achieving higher expression of endogenous utrophin. Deleting the UTRN IMTR in DMD patient-derived human induced pluripotent stem cells (DMD-hiPSCs) resulted in ca. 2-fold higher levels of utrophin protein. Differentiation of the UTRN edited DMD-hiPSCs (UTRNΔIMTR) by MyoD overexpression resulted in increased sarcolemmal α-sarcoglycan staining consistent with improved dystrophin glycoprotein complex (DGC) restoration. These results demonstrate that CRISPR/Cas9-based UTRN genome editing offers a novel utrophin upregulation therapeutic strategy applicable to all DMD patients, irrespective of the dystrophin mutation status.
Collapse
Affiliation(s)
- Kasturi Sengupta
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Manoj K Mishra
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emanuele Loro
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Melissa J Spencer
- Molecular Biology Institute, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Neurology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - April D Pyle
- Molecular Biology Institute, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tejvir S Khurana
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Abstract
KIBRA has been suggested as a key regulator of the hippo pathway, regulating organ size, cell contact inhibition as well as tissue regeneration and tumorigenesis. Recently, alterations of KIBRA expression caused by promotor methylation have been reported for several types of cancer. Our current study aimed to design an artificial transcription factor capable of re-activating expression of the tumor suppressor KIBRA and the hippo pathway. We engineered a new gene named 'ZFP226' encoding for a ~23 kDa fusion protein. ZFP226 belongs to the Cys2-His2 zinc finger type and recognizes a nine base-pair DNA sequence 5'-GGC-GGC-GGC-3' in the KIBRA core promoter P1a. ZFP226 showed nuclear localization in human immortalized kidney epithelial cells and activated the KIBRA core promoter (p < 0.001) resulting in significantly increased KIBRA mRNA and protein levels (p < 0.001). Furthermore, ZFP226 led to activation of hippo signaling marked by elevated YAP and LATS phosphorylation. In Annexin V flow cytometry assays ZFP226 overexpression showed strong pro-apoptotic capacity on MCF-7 breast cancer cells (p < 0.01 early-, p < 0.001 late-apoptotic cells). We conclude that the artificial transcription factor ZFP226 can be used for target KIBRA and hippo pathway activation. This novel molecule may represent a molecular tool for the development of future applications in cancer treatment.
Collapse
|
6
|
Utrophin up-regulation by artificial transcription factors induces muscle rescue and impacts the neuromuscular junction in mdx mice. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1172-1182. [PMID: 29408646 PMCID: PMC5851675 DOI: 10.1016/j.bbadis.2018.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/16/2018] [Accepted: 01/25/2018] [Indexed: 01/31/2023]
Abstract
Up-regulation of the dystrophin-related gene utrophin represents a promising therapeutic strategy for the treatment of Duchenne Muscular Dystrophy (DMD). In order to re-program the utrophin expression level in muscle, we engineered artificial zinc finger transcription factors (ZF-ATFs) that target the utrophin 'A' promoter. We have previously shown that the ZF-ATF "Jazz", either by transgenic manipulation or by systemic adeno-associated viral delivery, induces significant rescue of muscle function in dystrophic "mdx" mice. We present the full characterization of an upgraded version of Jazz gene named "JZif1" designed to minimize any possible host immune response. JZif1 was engineered on the Zif268 gene-backbone using selective amino acid substitutions to address JZif1 to the utrophin 'A' promoter. Here, we show that JZif1 induces remarkable amelioration of the pathological phenotype in mdx mice. To investigate the molecular mechanisms underlying Jazz and JZif1 induced muscle functional rescue, we focused on utrophin related pathways. Coherently with utrophin subcellular localization and role in neuromuscular junction (NMJ) plasticity, we found that our ZF-ATFs positively impact the NMJ. We report on ZF-ATF effects on post-synaptic membranes in myogenic cell line, as well as in wild type and mdx mice. These results candidate our ZF-ATFs as novel therapeutic molecules for DMD treatment.
Collapse
|
7
|
Meng Z, Gao P, Chen L, Peng J, Huang J, Wu M, Chen K, Zhou Z. Artificial Zinc-Finger Transcription Factor of A20 Suppresses Restenosis in Sprague Dawley Rats after Carotid Injury via the PPARα Pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:123-131. [PMID: 28918014 PMCID: PMC5493820 DOI: 10.1016/j.omtn.2017.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 06/13/2017] [Accepted: 06/14/2017] [Indexed: 01/25/2023]
Abstract
The inhibition of inflammation and vascular smooth muscle cell (VSMC) proliferation is an ideal strategy to suppress intimal hyperplasia after percutaneous transluminal angioplasty (PTA). Evidence has indicated that overexpression of A20 suppresses neointima formation, but its low transfection efficiency limits its application. Hence, we upregulated A20 expression via transfection of rAd.ATF (recombinant adenovirus vector of artificial transcription factor) and rAd.A20 in rat carotid arteries after balloon dilatation (in vivo) and isolated VSMCs (in vitro). In vivo, we found that after rAd.ATF and rAd.A20 transfection, A20 expression was markedly increased, whereas proliferating cell nuclear antigen (PCNA) and nuclear factor κB p65 (NF-κBp65) protein levels were significantly decreased, and intimal hyperplasia and secretion of proinflammatory factors were significantly reduced when compared with empty vector and saline control groups. Most importantly, the rAd.ATF-treated group showed more significant inhibition on intimal hyperplasia and expression of PCNA than the rAd.A20-treated group. In vitro, compared with the control group, transfection of rAd.ATF and rAd.A20 significantly increased A20 expression, which upregulated the proliferator-activated receptor (PPAR) level for both mRNA and protein, and reduced migration and proliferation of VSMCs and lipopolysaccharide (LPS)-induced inflammation. Furthermore, the PPARα agonist GW6471 could partially restore the effect of A20 on VSMCs. Our findings indicate that the ATF of A20 inhibits neointimal hyperplasia and, therefore, constitutes a novel potential alternative to prevent restenosis.
Collapse
Affiliation(s)
- Zhaoyou Meng
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Pan Gao
- Department of Geratology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lin Chen
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jing Peng
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jialu Huang
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Min Wu
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Kangning Chen
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Zhenhua Zhou
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
8
|
RNA activation of haploinsufficient Foxg1 gene in murine neocortex. Sci Rep 2016; 6:39311. [PMID: 27995975 PMCID: PMC5172352 DOI: 10.1038/srep39311] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/22/2016] [Indexed: 11/18/2022] Open
Abstract
More than one hundred distinct gene hemizygosities are specifically linked to epilepsy, mental retardation, autism, schizophrenia and neuro-degeneration. Radical repair of these gene deficits via genome engineering is hardly feasible. The same applies to therapeutic stimulation of the spared allele by artificial transactivators. Small activating RNAs (saRNAs) offer an alternative, appealing approach. As a proof-of-principle, here we tested this approach on the Rett syndrome-linked, haploinsufficient, Foxg1 brain patterning gene. We selected a set of artificial small activating RNAs (saRNAs) upregulating it in neocortical precursors and their derivatives. Expression of these effectors achieved a robust biological outcome. saRNA-driven activation (RNAa) was limited to neural cells which normally express Foxg1 and did not hide endogenous gene tuning. saRNAs recognized target chromatin through a ncRNA stemming from it. Gene upregulation required Ago1 and was associated to RNApolII enrichment throughout the Foxg1 locus. Finally, saRNA delivery to murine neonatal brain replicated Foxg1-RNAa in vivo.
Collapse
|
9
|
Fimiani C, Goina E, Mallamaci A. Upregulating endogenous genes by an RNA-programmable artificial transactivator. Nucleic Acids Res 2015; 43:7850-64. [PMID: 26152305 PMCID: PMC4652751 DOI: 10.1093/nar/gkv682] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/22/2015] [Indexed: 11/12/2022] Open
Abstract
To promote expression of endogenous genes ad libitum, we developed a novel, programmable transcription factor prototype. Kept together via an MS2 coat protein/RNA interface, it includes a fixed, polypeptidic transactivating domain and a variable RNA domain that recognizes the desired gene. Thanks to this device, we specifically upregulated five genes, in cell lines and primary cultures of murine pallial precursors. Gene upregulation was small, however sufficient to robustly inhibit neuronal differentiation. The transactivator interacted with target gene chromatin via its RNA cofactor. Its activity was restricted to cells in which the target gene is normally transcribed. Our device might be useful for specific applications. However for this purpose, it will require an improvement of its transactivation power as well as a better characterization of its target specificity and mechanism of action.
Collapse
Affiliation(s)
- Cristina Fimiani
- Laboratory of Cerebral Cortex Development, SISSA, Trieste, 34136, Italy
| | - Elisa Goina
- Laboratory of Cerebral Cortex Development, SISSA, Trieste, 34136, Italy
| | | |
Collapse
|
10
|
Betts CA, Saleh AF, Carr CA, Hammond SM, Coenen-Stass AML, Godfrey C, McClorey G, Varela MA, Roberts TC, Clarke K, Gait MJ, Wood MJA. Prevention of exercised induced cardiomyopathy following Pip-PMO treatment in dystrophic mdx mice. Sci Rep 2015; 5:8986. [PMID: 25758104 PMCID: PMC4355666 DOI: 10.1038/srep08986] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/03/2015] [Indexed: 01/16/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disorder caused by mutations in the Dmd gene. In addition to skeletal muscle wasting, DMD patients develop cardiomyopathy, which significantly contributes to mortality. Antisense oligonucleotides (AOs) are a promising DMD therapy, restoring functional dystrophin protein by exon skipping. However, a major limitation with current AOs is the absence of dystrophin correction in heart. Pip peptide-AOs demonstrate high activity in cardiac muscle. To determine their therapeutic value, dystrophic mdx mice were subject to forced exercise to model the DMD cardiac phenotype. Repeated peptide-AO treatments resulted in high levels of cardiac dystrophin protein, which prevented the exercised induced progression of cardiomyopathy, normalising heart size as well as stabilising other cardiac parameters. Treated mice also exhibited significantly reduced cardiac fibrosis and improved sarcolemmal integrity. This work demonstrates that high levels of cardiac dystrophin restored by Pip peptide-AOs prevents further deterioration of cardiomyopathy and pathology following exercise in dystrophic DMD mice.
Collapse
Affiliation(s)
- Corinne A Betts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Amer F Saleh
- 1] Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK [2] AstraZeneca R&D, Discovery Safety, Drug safety and Metabolism, Alderley Park, Macclesfield, SK10 4TG, UK
| | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Suzan M Hammond
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Caroline Godfrey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Graham McClorey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Miguel A Varela
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Thomas C Roberts
- 1] Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX [2] Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 NTorrey Pines Road, La Jolla, CA 92037, USA
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Michael J Gait
- Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| |
Collapse
|
11
|
Gintjee TJJ, Magh ASH, Bertoni C. High throughput screening in duchenne muscular dystrophy: from drug discovery to functional genomics. BIOLOGY 2014; 3:752-80. [PMID: 25405319 PMCID: PMC4280510 DOI: 10.3390/biology3040752] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 01/16/2023]
Abstract
Centers for the screening of biologically active compounds and genomic libraries are becoming common in the academic setting and have enabled researchers devoted to developing strategies for the treatment of diseases or interested in studying a biological phenomenon to have unprecedented access to libraries that, until few years ago, were accessible only by pharmaceutical companies. As a result, new drugs and genetic targets have now been identified for the treatment of Duchenne muscular dystrophy (DMD), the most prominent of the neuromuscular disorders affecting children. Although the work is still at an early stage, the results obtained to date are encouraging and demonstrate the importance that these centers may have in advancing therapeutic strategies for DMD as well as other diseases. This review will provide a summary of the status and progress made toward the development of a cure for this disorder and implementing high-throughput screening (HTS) technologies as the main source of discovery. As more academic institutions are gaining access to HTS as a valuable discovery tool, the identification of new biologically active molecules is likely to grow larger. In addition, the presence in the academic setting of experts in different aspects of the disease will offer the opportunity to develop novel assays capable of identifying new targets to be pursued as potential therapeutic options. These assays will represent an excellent source to be used by pharmaceutical companies for the screening of larger libraries providing the opportunity to establish strong collaborations between the private and academic sectors and maximizing the chances of bringing into the clinic new drugs for the treatment of DMD.
Collapse
Affiliation(s)
- Thomas J J Gintjee
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90095, USA.
| | - Alvin S H Magh
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90095, USA.
| | - Carmen Bertoni
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90095, USA.
| |
Collapse
|
12
|
Strimpakos G, Corbi N, Pisani C, Di Certo MG, Onori A, Luvisetto S, Severini C, Gabanella F, Monaco L, Mattei E, Passananti C. Novel adeno-associated viral vector delivering the utrophin gene regulator jazz counteracts dystrophic pathology in mdx mice. J Cell Physiol 2014; 229:1283-91. [PMID: 24469912 PMCID: PMC4303978 DOI: 10.1002/jcp.24567] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 01/24/2014] [Indexed: 01/28/2023]
Abstract
Over-expression of the dystrophin-related gene utrophin represents a promising therapeutic strategy for Duchenne muscular dystrophy (DMD). The strategy is based on the ability of utrophin to functionally replace defective dystrophin. We developed the artificial zinc finger transcription factor “Jazz” that up-regulates both the human and mouse utrophin promoter. We observed a significant recovery of muscle strength in dystrophic Jazz-transgenic mdx mice. Here we demonstrate the efficacy of an experimental gene therapy based on the systemic delivery of Jazz gene in mdx mice by adeno-associated virus (AAV). AAV serotype 8 was chosen on the basis of its high affinity for skeletal muscle. Muscle-specific expression of the therapeutic Jazz gene was enhanced by adding the muscle α-actin promoter to the AAV vector (mAAV). Injection of mAAV8-Jazz viral preparations into mdx mice resulted in muscle-specific Jazz expression coupled with up-regulation of the utrophin gene. We show a significant recovery from the dystrophic phenotype in mAAV8-Jazz-treated mdx mice. Histological and physiological analysis revealed a reduction of fiber necrosis and inflammatory cell infiltration associated with functional recovery in muscle contractile force. The combination of ZF-ATF technology with the AAV delivery can open a new avenue to obtain a therapeutic strategy for treatment of DMD. J. Cell. Physiol. 229: 1283–1291, 2014. © 2014 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Georgios Strimpakos
- Institute of Cell Biology and Neurobiology CNR, IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Wilson KA, Chateau ML, Porteus MH. Design and Development of Artificial Zinc Finger Transcription Factors and Zinc Finger Nucleases to the hTERT Locus. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e87. [PMID: 23612114 PMCID: PMC3650244 DOI: 10.1038/mtna.2013.12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 02/05/2013] [Indexed: 01/07/2023]
Abstract
The ability to direct human telomerase reverse transcriptase (hTERT) expression through either genetic control or tunable regulatory factors would advance not only our understanding of the transcriptional regulation of this gene, but also potentially produce new strategies for addressing telomerase-associated disease. In this work, we describe the engineering of artificial zinc finger transcription factors (ZFTFs) and ZF nucleases (ZFNs) to target sequences within the hTERT promoter and exon-1. We were able to identify several active ZFTFs that demonstrate a broadly tunable response when screened by a cell-based transcriptional reporter assay. Using the same DNA-binding domains, we generated ZFNs that were screened in combinatorial pairs in cell-based extrachromosomal single-strand annealing (SSA) assays and in gene-targeting assays using stably integrated constructs. Selected ZFN pairs were tested for the ability to induce sequence changes in a Cel1 assay and we observed frequencies of genomic modification up to 18.7% at the endogenous hTERT locus. These screening strategies have pinpointed several ZFN pairs that may be useful in gene editing of the hTERT locus. Our work provides a foundation for using engineered ZF proteins (ZFPs) for modulation of the hTERT locus.Molecular Therapy - Nucleic Acids (2013) 2, e87; doi:10.1038/mtna.2013.12; published online 23 April 2013.
Collapse
Affiliation(s)
- Kimberly A Wilson
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Morgan L Chateau
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Matthew H Porteus
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Pediatrics, Stanford University, Stanford, California, USA
| |
Collapse
|
14
|
Moorwood C, Khurana TS. Duchenne muscular dystrophy drug discovery - the application of utrophin promoter activation screening. Expert Opin Drug Discov 2013; 8:569-81. [PMID: 23473647 DOI: 10.1517/17460441.2013.777040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a devastating genetic muscle wasting disease caused by mutations in the DMD gene that in turn lead to an absence of dystrophin. Currently, there is no definitive therapy for DMD. Gene- and cell-based therapies designed to replace dystrophin have met some degree of success, as have strategies that seek to improve the dystrophic pathology independent of dystrophin. AREAS COVERED In this review the authors focus on utrophin promoter activation-based strategies and their implications on potential therapeutics for DMD. These strategies in common are designed to identify drugs/small molecules that can activate the utrophin promoter and would allow the functional substitution of dystrophin by upregulating utrophin expression in dystrophic muscle. The authors provide an overview of utrophin biology with a focus on regulation of the utrophin promoter and discuss current attempts in identifying utrophin promoter-activating molecules using high-throughput screening (HTS). EXPERT OPINION The characterisation of utrophin promoter regulatory mechanisms coupled with advances in HTS have allowed researchers to undertake screens and identify a number of promising lead compounds that may prove useful for DMD. In principle, these pharmacological compounds offer significant advantages from a translational viewpoint for developing DMD therapeutics.
Collapse
Affiliation(s)
- Catherine Moorwood
- University of Pennsylvania School of Dental Medicine, Department of Anatomy & Cell Biology, 438 Levy Research Building, 240 S. 40th Street, Philadelphia, PA 19104, USA
| | | |
Collapse
|
15
|
Onori A, Pisani C, Strimpakos G, Monaco L, Mattei E, Passananti C, Corbi N. UtroUp is a novel six zinc finger artificial transcription factor that recognises 18 base pairs of the utrophin promoter and efficiently drives utrophin upregulation. BMC Mol Biol 2013; 14:3. [PMID: 23363418 PMCID: PMC3576267 DOI: 10.1186/1471-2199-14-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 12/12/2012] [Indexed: 02/08/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is the most common X-linked muscle degenerative disease and it is due to the absence of the cytoskeletal protein dystrophin. Currently there is no effective treatment for DMD. Among the different strategies for achieving a functional recovery of the dystrophic muscle, the upregulation of the dystrophin-related gene utrophin is becoming more and more feasible. Results We have previously shown that the zinc finger-based artificial transcriptional factor “Jazz” corrects the dystrophic pathology in mdx mice by upregulating utrophin gene expression. Here we describe a novel artificial transcription factor, named “UtroUp”, engineered to further improve the DNA-binding specificity. UtroUp has been designed to recognise an extended DNA target sequence on both the human and mouse utrophin gene promoters. The UtroUp DNA-binding domain contains six zinc finger motifs in tandem, which is able to recognise an 18-base-pair DNA target sequence that statistically is present only once in the human genome. To achieve a higher transcriptional activation, we coupled the UtroUp DNA-binding domain with the innovative transcriptional activation domain, which was derived from the multivalent adaptor protein Che-1/AATF. We show that the artificial transcription factor UtroUp, due to its six zinc finger tandem motif, possesses a low dissociation constant that is consistent with a strong affinity/specificity toward its DNA-binding site. When expressed in mammalian cell lines, UtroUp promotes utrophin transcription and efficiently accesses active chromatin promoting accumulation of the acetylated form of histone H3 in the utrophin promoter locus. Conclusions This novel artificial molecule may represent an improved platform for the development of future applications in DMD treatment.
Collapse
Affiliation(s)
- Annalisa Onori
- Institute of Molecular Biology and Pathology CNR, Department of Molecular Medicine, University Sapienza, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
16
|
Langlois C, Del Gatto A, Arseneault G, Lafrance-Vanasse J, De Simone M, Morse T, de Paola I, Lussier-Price M, Legault P, Pedone C, Zaccaro L, Omichinski JG. Structure-based design of a potent artificial transactivation domain based on p53. J Am Chem Soc 2012; 134:1715-23. [PMID: 22191432 DOI: 10.1021/ja208999e] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Malfunctions in transcriptional regulation are associated with a number of critical human diseases. As a result, there is considerable interest in designing artificial transcription activators (ATAs) that specifically control genes linked to human diseases. Like native transcriptional activator proteins, an ATA must minimally contain a DNA-binding domain (DBD) and a transactivation domain (TAD) and, although there are several reliable methods for designing artificial DBDs, designing artificial TADs has proven difficult. In this manuscript, we present a structure-based strategy for designing short peptides containing natural amino acids that function as artificial TADs. Using a segment of the TAD of p53 as the scaffolding, modifications are introduced to increase the helical propensity of the peptides. The most active artificial TAD, termed E-Cap-(LL), is a 13-mer peptide that contains four key residues from p53, an N-capping motif and a dileucine hydrophobic bridge. In vitro analysis demonstrates that E-Cap-(LL) interacts with several known p53 target proteins, while in vivo studies in a yeast model system show that it is a 20-fold more potent transcriptional activator than the native p53-13 peptide. These results demonstrate that structure-based design represents a promising approach for developing artificial TADs that can be combined with artificial DBDs to create potent and specific ATAs.
Collapse
Affiliation(s)
- Chantal Langlois
- Département de Biochimie, Université de Montréal, C.P. 6128 Succursale, Centre-Ville, Montréal, Quebec H3C 3J7, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
|
18
|
Tinsley JM, Fairclough RJ, Storer R, Wilkes FJ, Potter AC, Squire SE, Powell DS, Cozzoli A, Capogrosso RF, Lambert A, Wilson FX, Wren SP, De Luca A, Davies KE. Daily treatment with SMTC1100, a novel small molecule utrophin upregulator, dramatically reduces the dystrophic symptoms in the mdx mouse. PLoS One 2011; 6:e19189. [PMID: 21573153 PMCID: PMC3089598 DOI: 10.1371/journal.pone.0019189] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 03/22/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a lethal, progressive muscle wasting disease caused by a loss of sarcolemmal bound dystrophin, which results in the death of the muscle fibers leading to the gradual depletion of skeletal muscle. There is significant evidence demonstrating that increasing levels of the dystrophin-related protein, utrophin, in mouse models results in sarcolemmal bound utrophin and prevents the muscular dystrophy pathology. The aim of this work was to develop a small molecule which increases the levels of utrophin in muscle and thus has therapeutic potential. METHODOLOGY AND PRINCIPAL FINDINGS We describe the in vivo activity of SMT C1100; the first orally bioavailable small molecule utrophin upregulator. Once-a-day daily-dosing with SMT C1100 reduces a number of the pathological effects of dystrophin deficiency. Treatment results in reduced pathology, better muscle physiology leading to an increase in overall strength, and an ability to resist fatigue after forced exercise; a surrogate for the six minute walk test currently recommended as the pivotal outcome measure in human trials for DMD. CONCLUSIONS AND SIGNIFICANCE This study demonstrates proof-of-principle for the use of in vitro screening methods in allowing identification of pharmacological agents for utrophin transcriptional upregulation. The best compound identified, SMT C1100, demonstrated significant disease modifying effects in DMD models. Our data warrant the full evaluation of this compound in clinical trials in DMD patients.
Collapse
Affiliation(s)
| | - Rebecca J. Fairclough
- MRC Functional Genomics Unit, Department of Physiology Anatomy and
Genetics, University of Oxford, Oxford, United Kingdom
| | | | | | - Allyson C. Potter
- MRC Functional Genomics Unit, Department of Physiology Anatomy and
Genetics, University of Oxford, Oxford, United Kingdom
| | - Sarah E. Squire
- MRC Functional Genomics Unit, Department of Physiology Anatomy and
Genetics, University of Oxford, Oxford, United Kingdom
| | - Dave S. Powell
- MRC Functional Genomics Unit, Department of Physiology Anatomy and
Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Cozzoli
- Unit of Pharmacology, Department of Pharmaco-biology, University of Bari
“A. Moro”, Bari, Italy
| | - Roberta F. Capogrosso
- Unit of Pharmacology, Department of Pharmaco-biology, University of Bari
“A. Moro”, Bari, Italy
| | | | | | | | - Annamaria De Luca
- Unit of Pharmacology, Department of Pharmaco-biology, University of Bari
“A. Moro”, Bari, Italy
| | - Kay E. Davies
- MRC Functional Genomics Unit, Department of Physiology Anatomy and
Genetics, University of Oxford, Oxford, United Kingdom
- * E-mail: (JMT); (KED)
| |
Collapse
|
19
|
Wilton SD, Fletcher S. Novel compounds for the treatment of Duchenne muscular dystrophy: emerging therapeutic agents. APPLICATION OF CLINICAL GENETICS 2011; 4:29-44. [PMID: 23776365 PMCID: PMC3681176 DOI: 10.2147/tacg.s8762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The identification of dystrophin and the causative role of mutations in this gene in Duchenne and Becker muscular dystrophies (D/BMD) was expected to lead to timely development of effective therapies. Despite over 20 years of research, corticosteroids remain the only available pharmacological treatment for DMD, although significant benefits and extended life have resulted from advances in the clinical care and management of DMD individuals. Effective treatment of DMD will require dystrophin restitution in skeletal, cardiac, and smooth muscles and nonmuscle tissues; however, modulation of muscle loss and regeneration has the potential to play an important role in altering the natural history of DMD, particularly in combination with other treatments. Emerging biological, molecular, and small molecule therapeutics are showing promise in ameliorating this devastating disease, and it is anticipated that regulatory environments will need to display some flexibility in order to accommodate the new treatment paradigms.
Collapse
Affiliation(s)
- Steve D Wilton
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Crawley, Perth, WA, Australia
| | | |
Collapse
|
20
|
Mussolino C, Sanges D, Marrocco E, Bonetti C, Di Vicino U, Marigo V, Auricchio A, Meroni G, Surace EM. Zinc-finger-based transcriptional repression of rhodopsin in a model of dominant retinitis pigmentosa. EMBO Mol Med 2011; 3:118-28. [PMID: 21268285 PMCID: PMC3085076 DOI: 10.1002/emmm.201000119] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 12/19/2010] [Accepted: 12/27/2010] [Indexed: 01/09/2023] Open
Abstract
Despite the recent success of gene-based complementation approaches for genetic recessive traits, the development of therapeutic strategies for gain-of-function mutations poses great challenges. General therapeutic principles to correct these genetic defects mostly rely on post-transcriptional gene regulation (RNA silencing). Engineered zinc-finger (ZF) protein-based repression of transcription may represent a novel approach for treating gain-of-function mutations, although proof-of-concept of this use is still lacking. Here, we generated a series of transcriptional repressors to silence human rhodopsin (hRHO), the gene most abundantly expressed in retinal photoreceptors. The strategy was designed to suppress both the mutated and the wild-type hRHO allele in a mutational-independent fashion, to overcome mutational heterogeneity of autosomal dominant retinitis pigmentosa due to hRHO mutations. Here we demonstrate that ZF proteins promote a robust transcriptional repression of hRHO in a transgenic mouse model of autosomal dominant retinitis pigmentosa. Furthermore, we show that specifically decreasing the mutated human RHO transcript in conjunction with unaltered expression of the endogenous murine Rho gene results in amelioration of disease progression, as demonstrated by significant improvements in retinal morphology and function. This zinc-finger-based mutation-independent approach paves the way towards a 'repression-replacement' strategy, which is expected to facilitate widespread applications in the development of novel therapeutics for a variety of disorders that are due to gain-of-function mutations.
Collapse
|
21
|
Gonçalves MAFV, Janssen JM, Nguyen QG, Athanasopoulos T, Hauschka SD, Dickson G, de Vries AAF. Transcription factor rational design improves directed differentiation of human mesenchymal stem cells into skeletal myocytes. Mol Ther 2011; 19:1331-41. [PMID: 21266958 DOI: 10.1038/mt.2010.308] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
There is great interest in transdifferentiating cells from one lineage into those of another and in dedifferentiating mature cells back into a stem/progenitor cell state by deploying naturally occurring transcription factors (TFs). Often, however, steering cellular differentiation pathways in a predictable and efficient manner remains challenging. Here, we investigated the principle of combining domains from different lineage-specific TFs to improve directed cellular differentiation. As proof-of-concept, we engineered the whole-human TF MyoDCD, which has the NH(2)-terminal transcription activation domain (TAD) and adjacent DNA-binding motif of MyoD COOH-terminally fused to the TAD of myocardin (MyoCD). We found via reporter gene and marker protein assays as well as by a cell fusion readout system that, targeting the TAD of MyoCD to genes normally responsive to the skeletal muscle-specific TF MyoD enforces more robust myogenic reprogramming of nonmuscle cells than that achieved by the parental, prototypic master TF, MyoD. Human mesenchymal stem cells (hMSCs) transduced with a codon-optimized microdystrophin gene linked to a synthetic striated muscle-specific promoter and/or with MyoD or MyoDCD were evaluated for complementing the genetic defect in Duchenne muscular dystrophy (DMD) myocytes through heterotypic cell fusion. Cotransduction of hMSCs with MyoDCD and microdystrophin led to chimeric myotubes containing the highest dystrophin levels.
Collapse
Affiliation(s)
- Manuel A F V Gonçalves
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
22
|
Crisp A, Yin H, Goyenvalle A, Betts C, Moulton HM, Seow Y, Babbs A, Merritt T, Saleh AF, Gait MJ, Stuckey DJ, Clarke K, Davies KE, Wood MJA. Diaphragm rescue alone prevents heart dysfunction in dystrophic mice. Hum Mol Genet 2010; 20:413-21. [PMID: 21062902 DOI: 10.1093/hmg/ddq477] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused, in most cases, by the complete absence of the 427 kDa cytoskeletal protein, dystrophin. There is no effective treatment, and affected individuals die from respiratory failure and cardiomyopathy by age 30. Here, we investigated whether cardiomyopathy could be prevented in animal models of DMD by increasing diaphragm utrophin or dystrophin expression and thereby restoring diaphragm function. In a transgenic mdx mouse, where utrophin was over expressed in the skeletal muscle and the diaphragm, but not in the heart, we found cardiac function, specifically right and left ventricular ejection fraction as measured using in vivo magnetic resonance imaging, was restored to wild-type levels. In mdx mice treated with a peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) that resulted in high levels of dystrophin restoration in the skeletal muscle and the diaphragm only, cardiac function was also restored to wild-type levels. In dystrophin/utrophin-deficient double-knockout (dKO) mice, a more severely affected animal model of DMD, treatment with a PPMO again produced high levels of dystrophin only in the skeletal muscle and the diaphragm, and once more restored cardiac function to wild-type levels. In the dKO mouse, there was no difference in heart function between treatment of the diaphragm plus the heart and treatment of the diaphragm alone. Restoration of diaphragm and other respiratory muscle function, irrespective of the method used, was sufficient to prevent cardiomyopathy in dystrophic mice. This novel mechanism of treating respiratory muscles to prevent cardiomyopathy in dystrophic mice warrants further investigation for its implications on the need to directly treat the heart in DMD.
Collapse
Affiliation(s)
- Alastair Crisp
- MRC Annex Building, MRC Clinical Sciences Centre, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
De Arcangelis V, Serra F, Cogoni C, Vivarelli E, Monaco L, Naro F. β1-syntrophin modulation by miR-222 in mdx mice. PLoS One 2010; 5:e12098. [PMID: 20856896 PMCID: PMC2938373 DOI: 10.1371/journal.pone.0012098] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Accepted: 07/18/2010] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND In mdx mice, the absence of dystrophin leads to the deficiency of other components of the dystrophin-glycoprotein complex (DAPC), making skeletal muscle fibers more susceptible to necrosis. The mechanisms involved in the disappearance of the DAPC are not completely understood. The muscles of mdx mice express normal amounts of mRNA for the DAPC components, thus suggesting post-transcriptional regulation. METHODOLOGY/PRINCIPAL FINDINGS We investigated the hypothesis that DAPC reduction could be associated with the microRNA system. Among the possible microRNAs (miRs) found to be upregulated in the skeletal muscle tissue of mdx compared to wt mice, we demonstrated that miR-222 specifically binds to the 3'-UTR of β1-syntrophin and participates in the downregulation of β1-syntrophin. In addition, we documented an altered regulation of the 3'-UTR of β1-syntrophin in muscle tissue from dystrophic mice. CONCLUSION/SIGNIFICANCE These results show the importance of the microRNA system in the regulation of DAPC components in dystrophic muscle, and suggest a potential role of miRs in the pathophysiology of dystrophy.
Collapse
Affiliation(s)
| | - Filippo Serra
- Department of Histology and Medical Embryology, University Sapienza, Rome, Italy
| | - Carlo Cogoni
- Department of Cellular Biotechnology and Ematology, University Sapienza, Rome, Italy
| | - Elisabetta Vivarelli
- Department of Histology and Medical Embryology, University Sapienza, Rome, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology, University Sapienza, Rome, Italy
| | - Fabio Naro
- Department of Histology and Medical Embryology, University Sapienza, Rome, Italy
- IIM, Pavia, Italy
| |
Collapse
|
24
|
Passananti C, Corbi N, Onori A, Di Certo MG, Mattei E. Transgenic mice expressing an artificial zinc finger regulator targeting an endogenous gene. Methods Mol Biol 2010; 649:183-206. [PMID: 20680835 DOI: 10.1007/978-1-60761-753-2_11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Zinc finger (ZF) proteins belonging to the Cys2-His2 class provide a simple and versatile framework to design novel artificial transcription factors (ATFs) targeted to the desired genes. Our work is based on ZF ATFs engineered to up-regulate the expression level of the dystrophin-related gene utrophin in Duchenne muscular dystrophy (DMD). In particular, on the basis of the "recognition code" that defines specific rules between zinc finger primary structure and potential DNA-binding sites we engineered and selected a new family of artificial transcription factors, whose DNA-binding domain consists in a three zinc finger peptide called "Jazz." Jazz protein binds specifically the 9 bp DNA sequence (5(')-GCT-GCT-GCG-3(')) present in the promoter region of both the human and mouse utrophin gene. We generated a transgenic mouse expressing Jazz protein fused to the strong transcriptional activation domain VP16 and under the control of the muscle specific promoter of the myosin light chain gene. Vp16-Jazz mice display a strong up-regulation of the utrophin at both mRNA and protein levels. To our knowledge, this represents the first example of a transgenic mouse expressing an artificial gene coding for a zinc finger-based transcription factor.
Collapse
|
25
|
Kawai M. [Establishment of an evaluation method for muscular dystrophy and a patient registration system for clinical trials]. Rinsho Shinkeigaku 2009; 49:863-6. [PMID: 20030232 DOI: 10.5692/clinicalneurol.49.863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
About 20 years have passed since the discovery of the causative protein of Duchenne muscular dystrophy, in 1987, and treatments targeting causative factors such as exon skipping, read-through of stop codons, and the upregulation of utrophin are approaching practical levels. In Japan, also, clinical trials are planned as the final stage of treatment development. In this field, an appropriate outcome measure has not been established due to the lack of experience in clinical trials. Treatments for muscular dystrophy are deemed effective only when increases in the muscle mass and muscle strength and improvements in the ADL and QOL as well as biological marker levels at target points have been demonstrated. The Muscular Dystrophy Clinical Study Group has addressed the development of these evaluation methods since 2002. Also, as treatments for muscular dystrophy being developed today are so-called tailor-made treatments aimed at specific mutations, a system that facilitates identification of the type and site of mutation in each individual must be prepared. The Gene Analysis Center was only just established in the National Center of Neurology and Psychiatry in 2009. Also, it is expected to be difficult to secure a sufficient number of subjects to start a clinical trial in a short period. Therefore, the Registry of Muscular Dystrophy (REMUDY), a system for the registration of patients with muscular dystrophy including their clinical and genetic information was implemented. This system, which provides information concerning the number of patients required by the protocol to researchers and pharmaceutical companies and the latest information regarding the development of treatments to patients, is expected to serve as a prototype for the establishment of the basis of clinical trials against rare diseases.
Collapse
Affiliation(s)
- Mitsuru Kawai
- National Hospital Organization, Higashisaitama National Hospital
| |
Collapse
|
26
|
Di Certo MG, Corbi N, Strimpakos G, Onori A, Luvisetto S, Severini C, Guglielmotti A, Batassa EM, Pisani C, Floridi A, Benassi B, Fanciulli M, Magrelli A, Mattei E, Passananti C. The artificial gene Jazz, a transcriptional regulator of utrophin, corrects the dystrophic pathology in mdx mice. Hum Mol Genet 2009; 19:752-60. [PMID: 19965907 DOI: 10.1093/hmg/ddp539] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The absence of the cytoskeletal protein dystrophin results in Duchenne muscular dystrophy (DMD). The utrophin protein is the best candidate for dystrophin replacement in DMD patients. To obtain therapeutic levels of utrophin expression in dystrophic muscle, we developed an alternative strategy based on the use of artificial zinc finger transcription factors (ZF ATFs). The ZF ATF 'Jazz' was recently engineered and tested in vivo by generating a transgenic mouse specifically expressing Jazz at the muscular level. To validate the ZF ATF technology for DMD treatment we generated a second mouse model by crossing Jazz-transgenic mice with dystrophin-deficient mdx mice. Here, we show that the artificial Jazz protein restores sarcolemmal integrity and prevents the development of the dystrophic disease in mdx mice. This exclusive animal model establishes the notion that utrophin-based therapy for DMD can be efficiently developed using ZF ATF technology and candidates Jazz as a novel therapeutic molecule for DMD therapy.
Collapse
Affiliation(s)
- Maria Grazia Di Certo
- Istituto di Neurobiologia e Medicina Molecolare, CNR, IRCCS Fondazione S. Lucia, 00143 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Markert CD, Atala A, Cann JK, Christ G, Furth M, Ambrosio F, Childers MK. Mesenchymal stem cells: emerging therapy for Duchenne muscular dystrophy. PM R 2009; 1:547-59. [PMID: 19627945 DOI: 10.1016/j.pmrj.2009.02.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 02/20/2009] [Accepted: 02/25/2009] [Indexed: 12/31/2022]
Abstract
Multipotent cells that can give rise to bone, cartilage, fat, connective tissue, and skeletal and cardiac muscle are termed mesenchymal stem cells. These cells were first identified in the bone marrow, distinct from blood-forming stem cells. Based on the embryologic derivation, availability, and various pro-regenerative characteristics, research exploring their use in cell therapy shows great promise for patients with degenerative muscle diseases and a number of other conditions. In this review, the authors explore the potential for mesenchymal stem cell therapy in the emerging field of regenerative medicine with a focus on treatment for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Chad D Markert
- Department of Neurology, School of Medicine, and Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Zinc-finger nucleases: a powerful tool for genetic engineering of animals. Transgenic Res 2009; 19:363-71. [DOI: 10.1007/s11248-009-9323-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Accepted: 09/10/2009] [Indexed: 10/20/2022]
|
29
|
Sera T. Zinc-finger-based artificial transcription factors and their applications. Adv Drug Deliv Rev 2009; 61:513-26. [PMID: 19394375 DOI: 10.1016/j.addr.2009.03.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 03/10/2009] [Indexed: 11/28/2022]
Abstract
Artificial transcription factors (ATFs) are potentially a powerful molecular tool to modulate endogenous target gene expression in living cells and organisms. To date, many DNA-binding molecules have been developed as the DNA-binding domains for ATFs. Among them, ATFs comprising Cys(2)His(2)-type zinc-finger proteins (ZFPs) as the DNA-binding domain have been extensively explored. The zinc-finger-based ATFs specifically recognize targeting sites in chromosomes and effectively up- and downregulate expression of their target genes not only in vitro, but also in vivo. In this review, after briefly introducing Cys(2)His(2)-type ZFPs, I will review the studies of endogenous human gene regulation by zinc-finger-based ATFs and other applications as well.
Collapse
Affiliation(s)
- Takashi Sera
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyotodaigaku-Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| |
Collapse
|
30
|
Arnett ALH, Chamberlain JR, Chamberlain JS. Therapy for neuromuscular disorders. Curr Opin Genet Dev 2009; 19:290-7. [PMID: 19411172 DOI: 10.1016/j.gde.2009.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 03/17/2009] [Accepted: 03/20/2009] [Indexed: 12/21/2022]
Abstract
Research into therapeutic approaches for both recessive and dominant neuromuscular disorders has made great progress over the past few years. In the field of gene therapy, antisense-mediated exon skipping is being applied to bypass deleterious mutations in the dystrophin gene and restore dystrophin expression in animal models of muscular dystrophy. Approaches for the dominant genetic muscle diseases have turned toward elimination of the mutant gene product with anti-sense oligonucleotide therapy and RNA interference techniques. Refinements of adeno-associated viral vectors and strategies for their delivery are also leading towards future clinical trials. The discovery of new, multipotent cell lineages, some of which possess the ability to successfully engraft muscle following vascular delivery, presents exciting prospects for the field of stem cell therapy. These discoveries represent steady progress towards the development of effective therapies for a wide range of neuromuscular disorders.
Collapse
Affiliation(s)
- Andrea L H Arnett
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195-7720, United States.
| | | | | |
Collapse
|
31
|
Desantis A, Onori A, Di Certo MG, Mattei E, Fanciulli M, Passananti C, Corbi N. Novel activation domain derived from Che-1 cofactor coupled with the artificial protein Jazz drives utrophin upregulation. Neuromuscul Disord 2009; 19:158-62. [PMID: 19162479 DOI: 10.1016/j.nmd.2008.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 10/31/2008] [Accepted: 11/10/2008] [Indexed: 10/21/2022]
Abstract
Our aim is to upregulate the expression level of the dystrophin related gene utrophin in Duchenne muscular dystrophy, thus complementing the lack of dystrophin functions. To this end, we have engineered synthetic zinc finger based transcription factors. We have previously shown that the artificial three-zinc finger protein named Jazz fused with the Vp16 activation domain, is able to bind utrophin promoter A and to increase the endogenous level of utrophin in transgenic mice. Here, we report on an innovative artificial protein, named CJ7, that consists of Jazz DNA binding domain fused to a novel activation domain derived from the regulatory multivalent adaptor protein Che-1/AATF. This transcriptional activation domain is 100 amino acids in size and it is very powerful as compared to the Vp16 activation domain. We show that CJ7 protein efficiently promotes transcription and accumulation of the acetylated form of histone H3 on the genomic utrophin promoter locus.
Collapse
Affiliation(s)
- Agata Desantis
- Istituto di Biologia e Patologia Molecolari, CNR, c/o Regina Elena Cancer Institute, Via delle Messi d'Oro 156, 00158 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
32
|
Lu Y, Tian C, Danialou G, Gilbert R, Petrof BJ, Karpati G, Nalbantoglu J. Targeting artificial transcription factors to the utrophin A promoter: effects on dystrophic pathology and muscle function. J Biol Chem 2008; 283:34720-7. [PMID: 18945675 PMCID: PMC3259868 DOI: 10.1074/jbc.m804518200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Revised: 10/15/2008] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy is caused by a genetic defect in the dystrophin gene. The absence of dystrophin results in muscle fiber necrosis and regeneration, leading to progressive muscle fiber loss. Utrophin is a close analogue of dystrophin. A substantial, ectopic expression of utrophin in the extrasynaptic sarcolemma of dystrophin-deficient muscle fibers can prevent deleterious effects of dystrophin deficiency. An alternative approach for the extrasynaptic up-regulation of utrophin involves the augmentation of utrophin transcription via the endogenous utrophin A promoter using custom-designed transcriptional activator proteins with zinc finger (ZFP) motifs. We tested a panel of custom-designed ZFP for their ability to activate the utrophin A promoter. Expression of one such ZFP efficiently increased, in a time-dependent manner, utrophin transcript and protein levels both in vitro and in vivo. In dystrophic mouse (mdx) muscles, administration of adenoviral vectors expressing this ZFP led to significant enhancement of muscle function with decreased necrosis, restoration of the dystrophin-associated proteins, and improved resistance to eccentric contractions. These studies provide evidence that specifically designed ZFPs can act as strong transcriptional activators of the utrophin A promoter. These may thus serve as attractive therapeutic agents for dystrophin deficiency states such as Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Yifan Lu
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Chai Tian
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Gawiyou Danialou
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Rénald Gilbert
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Basil J. Petrof
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - George Karpati
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Josephine Nalbantoglu
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| |
Collapse
|