bba - molecular basis of disease · 2018. 11. 15. · delivery of zf-atfs, we chose recombinant...

11
Contents lists available at ScienceDirect BBA - Molecular Basis of Disease journal homepage: www.elsevier.com/locate/bbadis Utrophin up-regulation by articial transcription factors induces muscle rescue and impacts the neuromuscular junction in mdx mice Cinzia Pisani a, , Georgios Strimpakos b,c , Francesca Gabanella b , Maria Grazia Di Certo b , Annalisa Onori a , Cinzia Severini b , Siro Luvisetto b,c , Stefano Farioli-Vecchioli b,c , Irene Carrozzo a , Antonio Esposito d , Tamara Canu d , Elisabetta Mattei b,c , Nicoletta Corbi a , Claudio Passananti a, a CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italy b CNR-Cell Biology and Neurobiology Institute, Rome, Italy c IRCCS Santa Lucia Foundation, Rome, Italy d Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raaele Scientic Institute, Vita-Salute San Raaele University, Italy ARTICLE INFO Keywords: ZF-ATF Utrophin DMD NMJ AAV Gene therapy 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 articial zinc nger transcription factors (ZF-ATFs) that target the utrophin ʻAʼ promoter. We have previously shown that the ZF-ATF Jazz, either by transgenic manipulation or by systemic adeno- associated viral delivery, induces signicant rescue of muscle function in dystrophic mdxmice. We present the full characterization of an upgraded version of Jazz gene named JZif1designed 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 Apromoter. Here, we show that JZif1 induces remarkable amelioration of the pathological phenotype in mdx mice. To investigate the molecular mechanisms underlying Jazz and JZif1 in- duced 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 eects 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. 1. Introduction Duchenne Muscular Dystrophy (DMD) is a severe X-linked muscle degenerative disease caused by the absence of the cytoskeletal protein dystrophin [1]. Despite the considerable progress in understanding the pathogenesis and advancement of therapeutic approaches, there is no eective cure for DMD [24]. A promising strategy to cure DMD aims to up-regulate utrophin gene expression, a ubiquitously expressed auto- somal paralogue of dystrophin gene [5,6]. The strategy is based on the ability of utrophin protein to partially replace defective dystrophin. Utrophin displays about 80% homology with dystrophin and it is able to perform similar functions. Notably, utrophin up-regulation does not raise any immune response. Many research groups and companies are studying the possibility of modulating utrophin gene expression using pharmacological approaches [3,4,79]. Several small molecules, in- cluding heregulin, nabumetone, L-arginine and SMT C1100 are able to up-regulate the utrophin gene by activating the utrophin Apromoter [3]. In particular, the drug SMT C1100/Ezutromid is currently in clinical trials by Summit Therapeutics [4,10]. In addition, a post-tran- scriptional modulation of utrophin is achieved by a recombinant bi- glycan (rhBGN), which can recruit utrophin to the sarcolemma [11,12]. Our strategy is based on the use of zinc nger articial transcription factors (ZF-ATFs) designed ad hoc to re-program utrophin gene ex- pression. ZF-ATFs are small transcriptional regulators, active at very low concentration and mimic the body's natural regulatory mechanism, resulting in the production of all the dierent isoforms of the targeted protein [13]. Moreover, ZF-ATFs are well tolerated and do not aect global transcription when expressed in animal models, as demonstrated with the ZF-ATF prototype gene Jazz [13,14]. In addition, our last generation of ZF-ATFs has been designed to minimize immunogenicity. Using selective amino acid substitution, the natural Zif268/Egr1 tran- scription factor was specically re-directed to the utrophin Apro- moter, giving rise to the novel ZF-ATF named JZif1. Importantly JZif1 and Jazz genes share the same 9 base pair DNA target sequence. For https://doi.org/10.1016/j.bbadis.2018.01.030 Received 26 October 2017; Received in revised form 16 January 2018; Accepted 25 January 2018 Corresponding authors at: CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy. E-mail addresses: [email protected] (C. Pisani), [email protected] (C. Passananti). BBA - Molecular Basis of Disease 1864 (2018) 1172–1182 Available online 31 January 2018 0925-4439/ © 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/). T

Upload: others

Post on 05-Dec-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

Contents lists available at ScienceDirect

BBA - Molecular Basis of Disease

journal homepage: www.elsevier.com/locate/bbadis

Utrophin up-regulation by artificial transcription factors induces musclerescue and impacts the neuromuscular junction in mdx mice

Cinzia Pisania,⁎, Georgios Strimpakosb,c, Francesca Gabanellab, Maria Grazia Di Certob,Annalisa Onoria, Cinzia Severinib, Siro Luvisettob,c, Stefano Farioli-Vecchiolib,c, Irene Carrozzoa,Antonio Espositod, Tamara Canud, Elisabetta Matteib,c, Nicoletta Corbia, Claudio Passanantia,⁎

a CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Rome, Italyb CNR-Cell Biology and Neurobiology Institute, Rome, Italyc IRCCS Santa Lucia Foundation, Rome, Italyd Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Italy

A R T I C L E I N F O

Keywords:ZF-ATFUtrophinDMDNMJAAVGene therapy

A B S T R A C T

Up-regulation of the dystrophin-related gene utrophin represents a promising therapeutic strategy for thetreatment of Duchenne Muscular Dystrophy (DMD). In order to re-program the utrophin expression level inmuscle, we engineered artificial zinc finger transcription factors (ZF-ATFs) that target the utrophin ʻAʼ 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 thefull characterization of an upgraded version of Jazz gene named “JZif1” designed to minimize any possible hostimmune response. JZif1 was engineered on the Zif268 gene-backbone using selective amino acid substitutions toaddress JZif1 to the utrophin ‘A’ promoter. Here, we show that JZif1 induces remarkable amelioration of thepathological phenotype in mdx mice. To investigate the molecular mechanisms underlying Jazz and JZif1 in-duced muscle functional rescue, we focused on utrophin related pathways. Coherently with utrophin subcellularlocalization and role in neuromuscular junction (NMJ) plasticity, we found that our ZF-ATFs positively impactthe NMJ. We report on ZF-ATF effects on post-synaptic membranes in myogenic cell line, as well as in wild typeand mdx mice. These results candidate our ZF-ATFs as novel therapeutic molecules for DMD treatment.

1. Introduction

Duchenne Muscular Dystrophy (DMD) is a severe X-linked muscledegenerative disease caused by the absence of the cytoskeletal proteindystrophin [1]. Despite the considerable progress in understanding thepathogenesis and advancement of therapeutic approaches, there is noeffective cure for DMD [2–4]. A promising strategy to cure DMD aims toup-regulate utrophin gene expression, a ubiquitously expressed auto-somal paralogue of dystrophin gene [5,6]. The strategy is based on theability of utrophin protein to partially replace defective dystrophin.Utrophin displays about 80% homology with dystrophin and it is ableto perform similar functions. Notably, utrophin up-regulation does notraise any immune response. Many research groups and companies arestudying the possibility of modulating utrophin gene expression usingpharmacological approaches [3,4,7–9]. Several small molecules, in-cluding heregulin, nabumetone, L-arginine and SMT C1100 are able toup-regulate the utrophin gene by activating the utrophin ‘A’ promoter

[3]. In particular, the drug SMT C1100/Ezutromid is currently inclinical trials by Summit Therapeutics [4,10]. In addition, a post-tran-scriptional modulation of utrophin is achieved by a recombinant bi-glycan (rhBGN), which can recruit utrophin to the sarcolemma [11,12].Our strategy is based on the use of zinc finger artificial transcriptionfactors (ZF-ATFs) designed ad hoc to re-program utrophin gene ex-pression. ZF-ATFs are small transcriptional regulators, active at verylow concentration and mimic the body's natural regulatory mechanism,resulting in the production of all the different isoforms of the targetedprotein [13]. Moreover, ZF-ATFs are well tolerated and do not affectglobal transcription when expressed in animal models, as demonstratedwith the ZF-ATF prototype gene Jazz [13,14]. In addition, our lastgeneration of ZF-ATFs has been designed to minimize immunogenicity.Using selective amino acid substitution, the natural Zif268/Egr1 tran-scription factor was specifically re-directed to the utrophin ‘A’ pro-moter, giving rise to the novel ZF-ATF named ‘JZif1’. Importantly JZif1and Jazz genes share the same 9 base pair DNA target sequence. For

https://doi.org/10.1016/j.bbadis.2018.01.030Received 26 October 2017; Received in revised form 16 January 2018; Accepted 25 January 2018

⁎ Corresponding authors at: CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy.E-mail addresses: [email protected] (C. Pisani), [email protected] (C. Passananti).

BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

Available online 31 January 20180925-4439/ © 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).

T

Page 2: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

delivery of ZF-ATFs, we chose recombinant Adeno-associated viral(AAV) vectors which have been described as powerful and safe “genetransfer” vehicles for the treatment of neuromuscular disorders[15–18]. The AAV vector was engineered to express therapeutic genesunder the control of the human alpha-actin muscle specific promoter:“muscle-AAV” (mAAV). The mAAV has been combined with the use ofthe muscle-targeting AAV-serotype “8”. Intraperitoneal injection ofmAAV8-JZif1 or mAAV8-Jazz into dystrophin-deficient mdx mice(5 days after birth) resulted in therapeutic re-programming of themuscle utrophin gene, with a consequent amelioration of morpholo-gical as well as functional parameters of disease progression. Here wedemonstrate the beneficial effects of our ZF-ATFs in mdx mice anddescribe the molecular mechanisms through which ZF-ATFs up-regulateutrophin, induce rescue of muscle function and positively impact theneuromuscular junction (NMJ). NMJ are structurally impaired in dys-trophin-deficient mice, displaying an aberrant fragmented distributionof Acetylcholine receptors (AChRs), resulting in discontinuous anddispersed motor end-plate morphology [19–22]. AChR spatial dis-tribution at the post-synaptic membrane is crucial for synaptic function[23,24]. Recent studies indicate that an impaired NMJ morphologyleads to NMJ dysfunction that can play a crucial role in DMD patho-genesis [25,26]. The beneficial effect of our ZF-ATFs on post-synapticclustering of AChRs and on NMJ morphology/plasticity could thereforecontribute to improve various aspects of muscle physiology and ame-liorate DMD-like symptoms in the mdx mice.

2. Material and methods

2.1. Constructs

DNA fragments containing ZF-ATF zinc finger domains were syn-thetized by GenScript (New Jersey, USA). JZif1 gene was cloned intomAAV vector [15] and into the pAAV vector (Stratagene, La Jolla, CA,USA). The pXP luciferase reporter construct contains a portion of theutrophin ‘A’ promoter [27].

2.2. Cell cultures, transfections and reporter gene assay

Human HeLa and AAV-293 cell lines were grown in DMEM (Gibco-BRL, Grand Island, NY, USA) supplemented with 10% foetal bovineserum (Gibco-BRL), L-glutamine and penicillin/streptomycin. MouseC2C12 myoblasts cell line [28] was grown in DMEM supplemented with20% foetal bovine serum. Cell cultures were maintained at 37 °C in ahumidified atmosphere of 5% CO2. Transient transfections were per-formed using Lipofectin® Transfection Reagent and PLUS™ Reagent (forHeLa cells) (Invitrogen, Carlsbad, CA, USA), or TransIT®-LT1 (forC2C12 cells) Transfection Reagent (Mirus Bio, Madison, WI, USA). Cellextracts were prepared and assayed for luciferase (LUC) activity aspreviously described [29].

2.3. Immunofluorescence

Cells were fixed in 4% formaldehyde for 10min, permeabilized with0.1% Triton X-100 for 5min and incubated with mouse monoclonalanti-myc 9E10 antibody (9E10 clone; 1:20, DSHB, Iowa City, IO).Immunofluorescence on tissue was performed as previously described[13]. The following antibodies were used: anti-utrophin monoclonalantibody (610896, 1:100, BD Transduction Laboratories, FranklinLakes, NJ, USA) and anti-laminin polyclonal antibody (L9393, 1:500,Sigma Aldrich Corporation, St. Louis, Missouri, USA). Samples wereanalyzed by conventional epifluorescence microscope (Olympus BX51;Tokyo, Japan). Images were captured using a digital camera andmerged using the IAS2000 software.

2.4. Western blot analysis

Western blotting was performed as previously described [29].Quadriceps muscle samples were homogenized in lysis buffer (1% SDS,1% NP40, 5% glycerol, 5 mM EDTA) supplemented with proteinaseinhibitors using a rotary-blade homogenizer (OMNI International GLH,Kennesaw, Georgia, USA). Anti-myc monoclonal antibody (9E10 clone;1:100, DSHB, Iowa City, IO); anti β-actin monoclonal antibody (A5441,1:10,000, Sigma-Aldrich Corporation, St. Louis, Missouri, USA); anti-utrophin monoclonal antibody (Mancho3, 8A4 clone; 1:100 DSHB); antiEgr-1 rabbit polyclonal antibody (C-19 clone, 1:200, Santa Cruz, SantaCruz, CA, USA); anti-laminin polyclonal antibody (L9393, 1:2,000,Sigma-Aldrich Corporation) were used. ImageJ software (National In-stitutes of Health, USA) was used to quantify the densitometry of theimmunoblot bands.

2.5. Chromatin immunoprecipitation (ChIP) assay

Chromatin immunoprecipitation assays were performed as pre-viously described [29]. Equal amounts of chromatin from each samplewere immunoprecipitated overnight with anti-myc 9E10 monoclonalantibody (9E10 clone; 5 μg, DSHB, Iowa City, IO). The im-munoprecipitated sonicated chromatin was amplified using humanutrophin promoter-specific primers forward 5′-CGGCACGCACGGTTCACTCTGGAGCGC-3′ and reverse 5′-CAGCAACTTTGTTCCGGAAGATCAGCC-3′; human thymidine kinase (TK)-specific primers forward5′-GCCCCTTTAAACTTGGTGGGCGG-3′ and reverse 5′-TTGCGCCTCCGGGAAGTTCACG-3′; human dystrophin promoter-specific primers for-ward 5′-GTGTTTTAAGAATTGGCACCAG-3′ and reverse 5′-AGTCTGAATAAGAGAAGCAGCA-3′; human TopBP1 promoter-specific primersforward 5′-TGCTCACCTCCACGTTTGACA-3′ and reverse 5′-GCCTCACTTACCTCGTTGGAG-3′ [30]; human chromosome 16 region–specificprimers forward 5′-AGGACCACTCGCTGGGTAAGCA-3′ and reverse5′-CGCGGAGGGTGACATGGGGT-3′; human chromosome 17 re-gion–specific primers forward 5′-ACCTGTGTGTGGGTGGTGAGA-3′ andreverse 5′-CTGTAGGGCCCCAGGCACCAT-3′; human chromosome 19region–specific primers forward 5′-AGTGCCTGGCATCAGCTT-3′ andreverse 5′-CGCCATTGCACTCCAGCAT-3′. The DNA sequence of thepotential off target site present in chromosome 16 is: 5′-GCTGCTGgG-3′, with 8 out of 9 base pair matches with JZif1 target sequence.The DNA sequence of the potential off target site present in chromo-some 17 is: 5′-GCTGCTGCG-3′, 100% match with JZif1 target sequence.The DNA sequence of the potential off target site present in chromo-some 19 is: 5′-GCTGCTGCG-3′, with 100% match with JZif1 target se-quence.

2.6. Quantitative real-time PCR (qPCR)

RNA was isolated using TRIzol® reagent (Gibco-BRL) and was thenreverse transcribed using a High Capacity cDNA Reverse Transcriptionkit (Applied Biosystems, Foster City, CA, USA). Quantitative real-timePCR assays were performed using the TaqMan Universal PCR MasterMix (Applied Biosystems) in an ABI Prism 7000 Sequence DetectionSystem (Applied Biosystems) according to the manufacturer's protocol.β2M was used for the normalization of mRNA and relative expressionwas calculated using the comparative Ct methods (2−ΔΔCt). PCR pri-mers and probes for the target gene (UTRN) and for housekeeping gene(β2M) were purchased as TaqMan Gene Expression Assays (AppliedBiosystems) [15].

2.7. Production, purification and titration of recombinant mAAV8 stocks

Recombinant mAAV8 vector stocks were packaged by triple trans-fection of AAV-293 cells using the plasmid method “AAV Helper-FreeSystem” (Stratagene). mAAV8 viral particles were purified from DMEMgrowth medium 72 h after transfection. The cell growth medium was

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1173

Page 3: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

extensively centrifuged, purified and concentrated 20:1. The viral titerwas quantified by real-time PCR (v.p./mL) [15].

2.8. Animal care

Dystrophin-deficient C57BL/10ScSn-DMDmdx/J mice (mdx) andC57BL/6 wt mice were maintained under conventional housing con-ditions. All procedures on mice were carried out in accordance with theDirective 2010/63/EU of the European Community for the care and useof laboratory animals and our experimental protocol was approved bythe Ethical Committee Italian Health Ministry (Permit numbers 498/2015 PR). Housing of the animals meets the behavioral needing of thespecies and was supervised by the responsible veterinarian.

2.9. Mice treatments

Mice were injected with viruses at 5 days of age. Systemic infectionwas achieved by intraperitoneal (i.p.) injection of 150 μl of viral sus-pension (5×1012 v.p./ml) (75 μl on each side of the lower quadrant ofthe abdomen) using a 0.3ml Accu-Fine syringe (Roche Diagnostics).Control mice were injected with the same volume of saline solution.Mice were analyzed at two/three months of age.

2.10. Muscle tissue preparation

Mice were sacrificed 8–12weeks after injections. Muscles weredissected and embedded in Tissue-Tek O.C.T. medium (Sakura Finetechnical, NL) or directly snap frozen in liquid nitrogen for RNA andprotein extraction. For histological and immunofluorescence analysis,transversal sections were obtained by cryostate CM1850UV at 2208C(Leica Microsystem GmbH, Wetzlar, Germany), placed onto polysine-coated microscope slides (Menzel Gmbh & Co, Iserlohn, Germany) andfixed with formaldehyde 4% in PBS for 10min.

2.11. Histological analysis

Transversal sections (9 μm thick) of muscles were stained with he-matoxylin and eosin (H&E; Roth, Germany) following the manufac-turer's instructions. The entire cross-section, taken at mid-belly, wasanalyzed by microscope Olympus BX51 (Olympus Corporation)[13,15]. Images were captured using a digital camera at 10 x magni-fication and analyzed by ImageJ software. Five mice per group wereanalyzed and at least 200 myofibers were counted in each section.

2.12. Serum creatine kinase (CK) assay

The serum CK assay was performed as described by Radley-Crabbet al. [31]. CK activity was quantified in duplicate using the CK NAC Kit(Greiner Diagnostic GmbH, Bahlingen, Germany), and analyzed kine-tically using the multilabel counter Victor 3 (PerkinElmer, Waltham,MA, USA). Data are expressed as units per liter (U/L).

2.13. Assays of mice performances by treadmill running

Exercise studies were performed as previously described and afterthe treadmill performance some mice were injected with Evans Bluedye [15].

2.14. Mechanical response of isolated muscles

As previously described [15], muscle excitability was examined ex-vivo by physiological assessment of the muscle force on isolated ex-tensor digitorum longus (EDL) preparations of both hind limbs and ofabdominal longitudinal strips (ABD), following direct electrical stimu-lation with rectilinear pulses of 0.5ms duration at 0.05–0.2 Hz usingvariable voltage, until reaching the supramaximal voltage. At the end of

the tension recordings, EDL and ABD muscles were subjected to aperiod of repetitive stimulations by trains of stimuli at a frequency of40 Hz for 250ms every second for 3min. Following this procedure, ableto obtain the muscle fatigue, tissues were removed from the chamberand placed in a Procion Orange solution (0.2%), in order to identify thepercentage of damaged muscular fibers [13].

2.15. AChR clustering C2C12 myotubes

C2C12 myoblasts were seeded at a density of 1.7× 104 cells/cm2 inLab-Tek® Chamber Slides (Sigma-Aldrich Co.) previously coated withlaminin‑1 (L2020; Sigma-Aldrich Co.) at 1 μg/cm2. 24 h after thetransfection, the growth media was replaced with differentiation mediacontaining 2% horse serum (Gibco-BRL). After 48 h Rat agrin (R&DSystems, Inc., Minneapolis, MN, USA) was included in the media at10 ng/ml. Then, 18 h later, cultures were fixed in 4% formaldehyde for30min a room temperature, permeabilized for 10min with 0.2%Nonidet P40, incubated with α‑bungarotoxin Alexa Fluor® 594 con-jugate (B13423, 1:1000, Thermo Fisher Scientific, Waltham, MA, USA)at 1 μg/ml for 1 h at room temperature and mounted with ProLong Goldwith Dapi (Thermo Fisher Scientific). Samples were examined by con-ventional epifluorescence microscope (Olympus BX51; OlympusCorporation). Images were captured using a digital camera at 20×magnification by a SPOT RT3 camera and elaborated by IAS2000software. About twenty fields for each category were randomly cap-tured and three independent experiments were averaged. Image ana-lysis was performed with ImageJ software.

2.16. Morphological analysis of neuromuscular junctions (NMJ)

Mice were anesthetized with 400mg/kg choral hydrate (SigmaAldrich Co.) and transcardially perfused with phosphate buffered saline(PBS) followed by 4% paraformaldehyde (PFA) in PBS. The entirediaphragm was dissected and completely immersed in PBS. All sub-sequent incubation steps were performed on a rotating platform, atroom temperature. Diaphragm was immersed in blocking buffer (3%BSA+0.5% Triton-X in PBS) for at least 3 h, incubated in quenchingsolution (2mg/ml sodium borohydride, in PBS) for three times, 10mineach time, and then washed three times with PBS. To visualize post-synaptic acetylcholine receptors (AChRs), diaphragms were incubatedwith Alexa Fluor 594-conjugated or Alexa Fluor 488 α-bungarotoxin (α-BTX) (B13423, B13422, 1:1,000, Thermo Fisher Scientific, Waltham,MA, USA), diluted in BSA 1%+Triton X-100 0.1%, for at least 2 h,washed 3 times with PBS, and then placed on a slide and mounted withProLong Gold with Dapi (Termo Fisher Scientific). In order to identifyeach individual NMJ, the diaphragm was further flattened applying apressure. Stained diaphragm was examined by confocal microscope(TCS-SP5, Leica Microsystem) acquiring a z-series of 20–25 opticalsections at 4–4.5 μm intervals, by a ×20 magnification lens.Morphological analysis of neuromuscular junctions was carried out ontwo-dimensional projections at maximum intensity of each z-seriesgenerated with the LAF AF software platform (Leyca Microsystem) inthe Tif format. For each hemi-diaphragm muscle, 3 separate regionswere imaged following the trajectory of the post-synaptic receptorclusters. Approximately 200–250 junctions for the whole diaphragmmuscle were analyzed in at least 3 animals per experimental group.AChR clusters were classified as continuous if they presented 3 or lesscontinuous regions of receptor clustering and discontinuous if theypresented>3 regions of receptor clustering.

2.17. Magnetic resonance imaging

MRI studies were performed on a 7T preclinical magnetic resonancescanner (Bruker, BioSpec 70/30 USR; Karlsruhe, Germany), equippedwith 450/675mT/m gradients (slew-rate: 3,400-4,500 T/m/s; rise-time: 140 μs). A phased-array rat-heart coil with four internal

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1174

Page 4: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

preamplifiers was used as receiver, coupled with a 72mm linear-vo-lume coil as transmitter. Mice were analyzed under general anesthesiaobtained by 1.5–2% isoflurane (Forane®; Abbott, North Chicago, Il,USA) vaporized in 100% oxygen, in prone position, with the right legfixed in the center of the coil. Breathing and body temperature wasmonitored during MRI (SA Instruments Inc., Stony Brook, NY, USA).MRI protocol included T2-mapping (inflammation and necrosis detec-tion), diffusion-mapping (fibers disruption and tissue reorganization),and Proton spectrometry (presence of fat and connective tissue) [32].MRI post-processing and image analysis were performed with Paravi-sion-5.0 software (Bruker).

2.18. Statistical analysis

Data are presented as either the mean ± S.D. or± SEM. The dif-ferences between means were analyzed with a t-test. Statistical sig-nificance was defined at P < 0.05. Analysis of variance (ANOVA) forrepeated measures was used to analyze the effect of treadmill running.

3. Results

3.1. The novel artificial transcription factor JZif1

We previously demonstrated that the ZF-ATF artificial gene named‘Jazz’ (Fig. 1A, top) (GenBank accession number AJ243577.1.), eitherby transgenic manipulation or by systemic adeno-associated viralvector delivery, induces significant rescue of muscle function in dys-trophic mdx mice [13–15]. With the intent to move toward clinicalapplication and to minimize potential deleterious immune responses,we developed a novel “almost natural” ZF-ATF gene using the backboneof the well-characterized Zif268/EGR1 human transcription factor(NCBI accession number NM_001964). As summarized in Fig. 1A, bysubstituting key amino acids in Zif268 alpha-helix zinc finger motifs,we re-directed the modified Zif268, re-named ‘JZif1’ (GenBank acces-sion number MF377379) to utrophin ‘A’ promoter. The JZif1 DNAbinding domain was designed following the same zinc finger signaturepreviously used to engineer Jazz prototype gene [33–35]. JZif1 andJazz bind the same nine base pair target sequence (5′-GCTCGTGCG-3′)presents in both the mouse and human utrophin gene ‘A’ promoters(Fig. 1A) (EMBL accession No. X95523). To verify whether JZif1overcomes any impediments derived from its own primary structureand/or chromatin infrastructure in the utrophin promoter locus, weinvestigated its nuclear localization and performed chromatin im-munoprecipitation experiments (ChIPs) (Fig. 1B and C). As shown inFig. 1C, ChIP experiments in HeLa cells demonstrated that JZif1, as wellas Jazz prototype gene, displayed binding specificity for the humangenomic utrophin ‘A’ promoter. As expected, JZif1 did not bind theunrelated promoters belonging to thymidine kinase (TK) gene anddystrophin gene. Importantly, again by ChIP experiments, we showedthat JZif1 did not bind the TopBP1 gene promoter that has been re-ported to be targeted and regulated by Zif268/Egr1 [30]. In addition,we verified that JZif1 did not bind the potential chromosomal “off-target” sites belonging respectively to Chromosome 16 (carrying eightbase pair matches out of nine), to Chromosome 17 and to Chromosome19 (both 100% nine base pair DNA target match).

The relative transcriptional activity of the JZif1artificial transcrip-tion factor was evaluated using luciferase activity assay; as shownFig. 1D, expression of JZif1 revealed an approximately 20-fold induc-tion of pXP-Luc vector [27], containing a portion of the utrophin ‘A’promoter, while co-transfection of the control pGL2-promoter luciferasevector with JZif1 did not yield any luciferase activity. Next, we eval-uated the capacity of JZif1 to up-regulate the endogenous utrophingene expression by quantitative real-time PCR (qPCR) analysis of theutrophin mRNA levels in differentiated C2C12 myogenic cells. Asshown in Fig. 1E, we observed an approximate 1.8-fold increase inutrophin transcript level in the presence of mAAV-JZif1 compared to

the mAAV empty vector control. Western blot analysis using anti-utrophin antibody verified a direct correlation between utrophin mRNAexpression and utrophin protein level (Fig. 1F).

3.2. Expression of JZif1 and utrophin up-regulation in mdx mice

To characterize the biological activity of the JZif1 protein in vivo,we delivered JZif1 in mdx mice by infection with “muscle-AAV serotype8” (mAAV8-JZif1) [15]. As shown in Fig. 2A, real-time PCR experi-ments revealed about 2-fold increase of utrophin mRNA expression inquadriceps and diaphragm isolated from mdx mice mAAV8-JZif1 in-traperitoneally injected (i.p.) at 5 days and analyzed at two months ofage. To determine whether increase of utrophin gene transcription,upon JZif1 treatment, was correlated with utrophin protein levels,Western blot analysis was performed (Fig. 1SA). Moreover, the histo-logical analysis of the diaphragm stained with the utrophin antibodyrevealed a significant increase and consequent re-localization along thesarcolemma of utrophin protein in mAAV8-JZif1 mdx mice compared tocontrol animals (Fig. 2B). These data demonstrate that the delivery ofJZif1 by mAAV8 in mdx mice is able to re-program utrophin expressionin muscle both at transcription and translation levels. To verify thepossible therapeutic effects of utrophin up-regulation in mdx micetreated with mAAV8-JZif1, several DMD diagnostic parameters wereevaluated. We quantified the extent of mononuclear inflammatory cellinfiltration, the frequency of centrally nucleated myofibers (CNFs) andthe fiber CSA (Cross section area), in muscle slices stained with he-matoxylin and eosin (H&E) (Fig. 2C and Fig. 1SB). The results clearlyshowed a dramatic reduction in muscle pathology, as displayed bybetter tissue architecture as well as less pronounced degenerative/re-generative processes in mAAV8-JZif1 treated muscles. Moreover, in asubset of mice, we checked serum creatine kinase (CK) levels, an in-dicator of muscle damage in mdx mice. As shown in Fig. 2D, serum CKlevels measured at 3months of age, were elevated, as expected, inuntreated mdx mice compared to wild type C57BL/6 mice and sig-nificantly reduced in JZif1-treated mdx mice.

3.3. JZif1 delivery induces muscle functional recovery in mdx mice

Utrophin up-regulation achieved by JZif1 delivery effectively pre-vented muscle damage in mdx mice. In addition to histochemicalanalyses, we assessed the effects of JZif1 on dystrophic muscle functionin vivo. Single session performance (first and second panel) and totalrunning time performance (third panel) relative to four weekly tread-mill trials with exhaustive exercise protocol were determined in un-treated mdx mice, mAAV8-EGFP and mAAV8-JZif1 treated mdx mice(Fig. 3A). The mAAV8-JZif1 treated mdx mice showed significant im-provement in exercise performance as compared to mAAV8-EGFPtreated or untreated mdx mice. Notably, mAAV8-JZif1 treated mdxmice displayed running times close to those of wt mice (Fig. 1SC). JZif1and Jazz prototype genes delivered in mdx mice induced a similarperformance in treadmill trials (Fig. 1SC). Furthermore, JZif1 expres-sion improved mdx sarcolemma integrity post exercise as shown byreduced in vivo Evan's blue dye uptake (Fig. 2SA). To verify whetherJZif1 expression improved the mechanical response in dystrophicmuscle, we also measured the contractile activity ex vivo. Isolated ab-dominal muscle strips and extensor digitorium longus (EDL) were ex-amined by physiological assessment of muscle excitability using vari-able voltages until reaching the supramaximal value. As shown inFig. 3B, both the abdomen and EDL muscles from mAAV8-JZif1 treatedmdx mice showed a significant decrease in stimulation threshold for themuscle fibers, compared with control mice. Similarly, to the treadmilltest, mAAV8-JZif1 and mAAV8-Jazz treated mdx mice displayed me-chanical force response close to the wild type mice category (Fig. 1SD).At the end of the force test, we also assessed contraction-induced injuryof the sarcolemma by staining each muscle with Procion Orange dye.Quantification of the dye-positive area in each section confirmed the

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1175

Page 5: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

increased ability of JZif1 to maintain the membrane integrity ofstressed fibers (Fig. 2SB). Our results demonstrate the remarkablefunctional improvement of muscle performance in mdx mice expressingeither JZif1or its prototype Jazz [15].

3.4. JZif1 and Jazz enhance the formation of post-synaptic acetylcholinereceptor (AChR) clusters

To investigate the molecular mechanisms underlying JZif1 and Jazzrescue of muscular function, we focused on utrophin related pathways,at the neuromuscular junction (NMJ) where utrophin co-localizes withacetylcoline receptors (AChRs) at the crests of post-junctional folds. Asfirst step, we investigated a possible effect of JZif1 and Jazz on post-synaptic AChR clustering in C2C12 myotubes expressing control vector,mAAV-JZif1 or mAAV-Jazz. Myotubes were subsequently treated withrecombinant neural agrin (10 ng/ml for 18 h) to induce the basal

formation of AChR post-synaptic clustering and visualized by alpha-bungarotoxin (BTX) staining. A 2–3 fold increase of AChR clusters wasobserved in the presence of either Jazz or JZif1 gene expression com-pared to control cells (Fig. 4A). Next, the effects of ZF-ATFs on max-imum z-stack projections of whole diaphragm were evaluated in wildtype mice treated with either Jazz or JZif1. With both ZF-ATFs, weobserved an increase in the number of AChR clusters/motor endplatescompared to control wt mice (Fig. 4B).

3.5. Correlation among MRI, treadmill performance and quantity/qualityof AChR clusters in mdx mice

To further examine the range of responses to ZF-ATFs expression invivo, the same sub-set of mdx mice were comparatively analyzed bymagnetic resonance imaging (MRI), treadmill performance and AChRclusters analysis. MRI was used to monitor changes in the transverse

Fig. 1. The novel artificial transcription factor JZif1.a. Schematic view of the novel artificial/natural transcription factor JZif1 and its prototype ‘Jazz’. Varied aminoacids in the Zif268 zinc finger region and transcription factors bindingsites are indicated. b. Immunofluorescence images showing the nuclear localization of over-expressed myc-JZif1 in HeLa cells (top panel). The fluorescence intensity was normalizedagainst the background signal. Western blot analysis of cell lysates from HeLa cells transiently transfected with myc-JZif1 (bottom panel). c. Chromatin immuno-precipitation (ChIP) wasperformed in HeLa cells (untransfected or transfected with the myc-JZif1 expression vector) using myc-tag monoclonal antibody or with no antibodies (no-Ab). Immuno-precipitates fromeach sample were analyzed by PCR performed with primers specific for the human utrophin promoter ‘A’. The PCR of unrelated promoters of thymidine kinase and dystrophin genes wereincluded as controls. The PCR of TopBP1 gene promoter containing Zif268/Egr1 binding sites and the PCR of specific ‘off-target’ regions from chromosome 16, chromosome 17 andchromosome 19 were also included. A sample representing the linear amplification of the total input chromatin (input) was included (lane 1). d. Luciferase activity obtained upon co-transfection among myc-JZif1 and pXP-Luc utrophin promoter ‘A’ construct or the control reporter pGL2-Promoter (Promega) in HeLa cells. The data are presented as the means ± S.D.of three independent experiments that were performed in triplicate. e. Quantitative real time RT-PCR (qPCR) analysis of the utrophin gene mRNA expression in differentiated C2C12 cellstransfected with the mAAV-JZif1 or mAAV control vectors. The gene expression ratio of utrophin, normalized as indicated, is shown as the mean ± S.D. from an experiment performed intriplicate. f. Representative Western blot analysis of utrophin protein levels in differentiated C2C12 cells transfected with the mAAV-JZif1 or mAAV control vectors. The antibodies usedare indicated. Densitometric analysis represents the mean ± S.D. of four independent experiments (right panel).

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1176

Page 6: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

relaxation time constant (T2) muscle. Because alterations in muscle T2correlate with muscle damage, edema, inflammatory infiltration andfibrosis [36], T2 MRI was used to monitor the pathological changes inlower hindlimb muscles in untreated wt (C57BL/6) untreated or treatedmdx mice at different times before and after exhaustive treadmill ex-ercise (Fig. 5A). mAAV8-JZif1 and mAAV8-Jazz treated mdx mice de-monstrated a better recovery from muscle damage five days aftertreadmill challenge, as compared with untreated littermates. Fig. 5A(top) shows representative MRI images of the fourth of 16 consecutiveNMR sections of the lower hindlimbs taken for all experimental groups;the lighter areas indicate the presence of inflammatory infiltration ormuscle damage. Part of the cohort of mice analyzed by MRI andtreadmill test was also examined by NMJ evaluation. Maximum z-stackprojections of the whole diaphragm in mdx mice showed a significantincrease in the total number of the AChR clusters at post-synaptic

membranes (Fig. 5B and C). Moreover, the AChR clusters analysis re-vealed that JZif1 and Jazz also improve the morphology of post-sy-naptic membrane, as indicated by an increase number of continuous(not fragmented) AChR clusters/motor endplates (Fig. 5D). Sig-nificantly, in the same groups of mdx mice, we found a positive cor-relation among: 1) profile of “T2 relaxation time parameter” (MRI); 2)muscle functionality (treadmill-running times); and 3) quantity/qualityof NMJ post-synaptic membranes (BTX staining). Overall, these ob-servations are consistent with an improvement in muscle functionmediated by ectopic expression of ZF-ATFs.

4. Discussion

ZF-ATFs designed ad hoc to target disease-related genes offer ther-apeutic promise in gene therapy [37–41,44]. We engineered several ZF-

Fig. 2. Expression of JZif1 and utrophin up-regulation in mdx mice.a. Quantification by real-time PCR (qPCR) of utrophin transcripts from quadriceps and diaphragm muscles isolated from 2-month old mdx male mice either untreated or intraperitonealinjected at 5 days with mAAV8-JZif1. The gene expression ratio between utrophin and β2-microglobulin (β2M) is shown. Values are expressed as means ± S.D. of at least five differentmice. (*P < 0.05, **P < 0.002). b. Immunohistochemistry of the diaphragm muscle isolated from 2-month old untreated, mAAV8-JZif1 or mAAV8-EGFP-treated mdx mice (four micewere analyzed in each group) and stained with the anti-utrophin antibody (green). The extracellular matrix is counterstained with the anti-laminin polyclonal antibody (red) and nucleiwith Dapi (blue). The fluorescence intensity was normalized against the background signal. c. Representative images (H&E staining) and quantification graphs of inflammatory infiltrate(a), Centrally Nucleated Fibers (CNF) (b), Fiber Cross-sectional Area (CSA) (c) of quadriceps muscle from 2-month old mdx mice treated at 5 days of age with mAAV8-JZif1 as comparedwith mAAV8-EGFP treated or untreated mdx mice (five mice were analyzed in each group). Images quality: 640×512 pixels. All values are expressed as the mean ± SEM thestatistically significant differences were calculated by Student t-test (*P < 0.05 JZif1 vs mdx). d. Quantification of serum CK levels in wt C57BL/6 mice (n=5), untreated mdx (n= 5)and mAAV8-JZif1 treated mdx mice (n=5) at 3 months of age. Data represent means (± S.D.) and statistically significant differences were calculated by Student t-test. (**P < 0.002JZif1 vs mdx).

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1177

Page 7: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

ATFs that up-regulated utrophin gene expression in muscle, and tar-geted both the human and mouse utrophin ‘A’ promoters[13–15,33,35,45,46]. The prototype ZF-ATF named ‘Jazz’ was able toinduce muscle functional recovery and to correct dystrophic pathologyin mdx mice, making ZF-ATFs strong candidate for DMD treatment[13–15]. In this context, moving toward a possible clinical application,we focused on different crucial aspects: i) safe and tissue-restricted AAVdelivery; ii) ZF-ATF tissue-restricted and persistent expression; iii) ab-sence/reduction of immunogenicity induced by both ZF-ATFs and theirdelivery vehicles; and iv) characterization of molecular mechanismsunderlying the therapeutic effects of ZF-ATFs. Our previous work facedsome of these aspects, in particular our engineered AAV-vectors ex-pressing therapeutic molecules are muscle restricted thanks to the useof both muscle specific promoter (mAAV) and AAV “serotype-8” [15].Importantly, the muscle specific combination of AAV serotype ‘8’ withour mAAV vector, per se contributed to lowering host immune response[18]. Here, we report on the characterization of an upgraded version ofthe Jazz gene, that we named ‘JZif1’. Jazz prototype gene was en-gineered assembling the selected zinc finger domains, the viral Vp16activation domain, the viral SV40 nuclear localization signal and themyc-tag [45]. JZif1 was designed on the natural human Zif268 genebackbone in order to minimize any possible host immune response.Notably, only thirteen amino acid substitutions were properly

distributed in the alpha helix region of Zif268 three zinc finger domainsto direct the novel ‘JZif1’ to both human and mouse utrophin ‘A’ pro-moters. It is important to underline that Zif268/Egr1 gene is highlyconserved in mammalian, in particular between human and mouse andwe have chosen to use human Zif268 as backbone for JZif1 gene in avision of a possible gene therapy approach. We speculate that fewamino acid changes in Zif268 alpha helix do not activate host immuneresponse, since the alpha helix of thousands of natural zinc finger do-mains (> 3,000 in the human genome) vary in these key positions inorder to bind different DNA targets [38]. JZif1, as well as Jazz proto-type gene, achieves muscle utrophin up-regulation inducing remarkableamelioration of the pathological phenotype in mdx mice. Morpholo-gical analysis of JZif1-treated mdx mice displayed better tissue archi-tecture as well as less pronounced degenerative/regenerative processes.Consistently with data obtained with Jazz, JZif1 treated mdx micedisplayed running times very close to wild type mice in treadmill test.The mdx muscle functional recovery was confirmed by ex vivo test onisolated muscles, where JZif1 expression clearly improves the me-chanical response. To investigate the molecular mechanisms underlyingJZif1 and Jazz mdx muscle rescue, we focused on the neuromuscularjunction (NMJ), since utrophin localizes to and plays a role at the NMJ[23,24,47–50]. Utrophin in partnership with the scaffold protein rapsynmay serve as backbone for consolidating post-synaptic domains,

Fig. 3. JZif1 delivery induces muscle functional recovery in mdx mice.a. Single session performance (left) and total running time (right) relative to four weekly treadmill trials with exhaustive exercise protocol. Untreated mdx mice, mAAV8-EGFP (controls)or mAAV8-JZif1 mdx mice injected at day 5 after birth were tested at 3 months of age. For each group, lines indicate the mean duration of running time during each trial (left), andcolumns indicate the cumulative running time over the four consecutive trials (right). Number of animals is n=11 for each treated group and n=7 for untreated group. Data representmeans (± SEM) and statistically significant differences were calculated by Student t-test. (*P < 0.05 vs mdx, **P < 0.002 vs mdx, *** P < 0.001 vs mdx). b. Mechanical response ofisolated muscles of resting female and male mdx mice treated at 5 days and examined at 2 months. Effects of electrical stimulation on isotonic contractions in abdominal muscles and EDLfrom mAAV8-JZif1 and mAAV8-EGFP treated or untreated control mice. Muscles contractile force for each voltage was determined and considered as % of maximal contraction. Datarepresent means (± SEM) from n muscle samples and statistically significant differences were calculated by Student t-test. (*P < 0.05 JZif1vs mdx).

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1178

Page 8: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

influencing different aspects, including AChR geometry/distribution,anchorage (to dystrophin glycoprotein complex DGC) and stability[26,51]. Importantly, several studies highlight that an impaired NMJmorphology/activity could contribute to DMD pathogenesis [25,26,52].JZif1 and Jazz expression positively affects NMJ morphology/plasticity,improving the post-synaptic clustering of AChRs. In C2C12 myotubes,ZF-ATF utrophin up-regulation correlated with increased number ofAChR clusters. ZF-ATF utrophin up-regulation, in both healthy wildtype and mdx mice exerted a positive effect on NMJ at post-synapticlevel, increasing the number of AChR clusters. These results are in goodagreement with the improvement of muscular force previously ob-served in our wild type-Jazz transgenic mice [45]. Significantly, ZF-ATFexpression also improved the morphology of post-synaptic membranesin mdx mice, as indicated by the percentage of continuous (not frag-mented) AChR clusters versus discontinuous endplates. In ZF-ATFtreated mdx mice, MRI analysis showed a better recovery after tread-mill exercise. Significantly, in a sub-group of ZF-ATF treated andtreadmill exercised mdx mice we were able to detect a positive corre-lation, highly consistent with a muscle functional rescue, among: 1)treadmill-running times, 2) quantity/quality of NMJ post-synapticmembranes, 3) profile of MRI-T2 relaxation time parameter. Re-markably, utrophin quality/quantity obtained by ZF-ATFs treatment in

muscle is able to provide therapeutic effects. Significantly, our ZF-ATFtherapeutic strategy is feasible independently from the type of deletion/mutation occurring in DMD patients and it can be combined and sy-nergized with appropriate pharmacological treatments [53].

We can speculate that, inside the fiber syncytium, ZF-ATF expres-sion increases the number of nuclei that maintain active utrophin genetranscription. This speculation could partially explain the results ob-tained on NMJ both in C2C12 myotubes and in wild type ZF-ATFtreated mice. On the other hand, muscle up-regulation of all utrophinisoforms, obtained with ZF-ATF treatment in mdx mice, triggeredfunctional and morphological muscle rescue differently from normallyoccurring utrophin up-regulation described in mdx untreated mice [9].Here, we demonstrated that JZif1, as well as its prototype Jazz gene,delivered/expressed in muscle tissues by muscle AAV vector induce aremarkable amelioration of the pathological phenotype in mdx mice.We described a positive impact of our ZF-ATF expression on NMJ, bothin physiological and pathological conditions. As depicted in the model(Fig. 6) we propose that the expression our ZF-ATFs improve themorphology/plasticity and augment the number of NMJ dictating aproper structural order in NMJ and sub-membrane domains. Moreover,ZF-ATF mediated utrophin up-regulation could exert a positive effectnot only in receiving the signal-stimulus coming from the nerve, but

Fig. 4. JZif1 and Jazz positively impact the formation of post-synaptic Acetylcholine Receptor clusters (AChR).a. Myogenic cell line C2C12 was transiently transfected with either mAAV-Jazz and mAAV-JZif1 or the empty vector as control and the differentiated myotubes were treated with agrin(10 ng/ml for 18 h) to induce a basal formation of AChR clusters. The clusters were visualized with alpha-bungarotoxin (BTX) staining. The fields (about twenty for each category) wererandomly captured in three independent experiments and data represent means (± S.D.). ***P < 0.001 indicate statistical significance by Student t-test (Jazz vs control, JZif1vscontrol). b. Maximum z-stack projections of whole diaphragm in wt C57BL/6mice, untreated and treated with mAAV8-Jazz or mAAV8-JZif1. Muscles were stained with BTX to visualizeand count AChR clusters (right panel). Approximately, a total of 250 AChR clusters were examined for each diaphragm. Data represent the average ± SEM of four mice per group andstatistically significant differences were calculated by Student t-test. (*P < 0.05 JZif1 vs wt).

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1179

Page 9: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

also in promoting a retrograde stimulus toward the nerve [25,54,55].These results candidate our ZF-ATFs for the treatment of DMD andpotentially other diseases related to NMJ deficits. Our ongoing workpoints to further investigate utrophin related DMD pathways positivelyaffected by our ZF-ATF treatments with the aim to deeply characterizethe molecular mechanisms underlying JZif1 and Jazz induced func-tional rescue. The fine identification of ZF-ATF affected pathways re-lated to DMD pathogenesis can suggest novel targets, as key factorswithin the NMJ network, for either gene therapy or novel pharmaco-logical treatments.

Supplementary data to this article can be found online at https://doi.org/10.1016/j.bbadis.2018.01.030.

Transparency document

The Transparency document associated with this article can be

found in online version.

Acknowledgments

We thank Dr. John Hiscott and Dr. Libera Berghella for editing andcritical reading of the manuscript. We thank Dr. R. G. Ruscitti for herprecious constant assistance. This work was supported by Telethon-Italy (Grant no. GGP14073), Zingenix Ltd. Company, CNR-InterOmicsFlagship Projects 2017 (Grant no. 19699) and FARMM-Onlus.

Author contributions

Conceived and designed the experiments: C.P., N.C., M.G.DC., E.M.,Cl.P. Performed the experiments: C.P., G.S., F.G., I.C., M.G.DC., N.C.,A.O., S.L., C.S. Analyzed the data: C.P., N.C., Cl.P., A.O., M.G.DC., G.S.,E.M. Contributed reagents/materials/tools/services: A.E., T.C. Wrote

Fig. 5. Correlation among MRI, treadmill performance and quantity/quality of AChR clusters in mdx mice.The same set of mdx mice were included in a cohort analyzed by: magnetic resonance imaging (MRI); treadmill performance and AChR clusters analysis. a. MRI was used to monitorchanges in the transverse relaxation time constant (T2) muscle. On the top representative MRI images of the fourth of 16 consecutive NMR sections of the lower hind limbs for allexperimental groups are shown. Alterations in muscle T2 are correlated to muscle damage, edema, inflammatory infiltration and fibrosis. T2 MRI changes in mdx mice lower hind limbmuscles in untreated wt and untreated or treated mdx mice with mAAV8-Jazz or mAAV8-JZif1 at 5 days. All experimental groups were examined at the times indicated before and after anexhaustive treadmill exercise until the age of about 3months. Statistical analysis with ‘unpaired t-test’ showed a significant difference in T2 relaxation time between untreated and treatedmdx mice indicated by (*P < 0.05). The lighter areas indicate the presence of inflammatory infiltration or muscle damage. b. The table shows the correlation between treadmill runningtimes and counts of AChR clusters. c. Maximum z-stack projections of whole diaphragm in mdx mice, untreated and treated with mAAV8-JZif1 ormAAV8-Jazz, analyzed post-treadmill.Muscles were stained with alpha-bungarotoxin to visualize and count AChR clusters at the postsynaptic membrane (right panel). Data represent the average ± SEM of 3 mice/group andstatistically significant differences were calculated by Student t-test. (*P < 0.05 JZif1 vs mdx, Jazz vs mdx). d. The AChR clusters analysis, performed in mdx diaphragm reveals thatJZif1 and Jazz improve also the morphology of postsynaptic membrane, as indicated by an increase number of continuous (not fragmented) AChR clusters. Approximately, a total of 200AChR clusters were examined for each diaphragm. Data represent the average ± SEM of three mice per group and statistically significant differences were calculated by Student t-test.(*P < 0.05 JZif1 vs mdx, Jazz vs mdx).

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1180

Page 10: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

the paper: C.P., N.C., Cl.P. All authors read and approved the finalmanuscript.

Conflicts of interest

C.P., G.S., M.G.DC., S.L., E.M., N.C., and Cl.P. are named inventorsof the following National Research Council patent applications:

1) U.S. Patent Application n. 15/328,833; European Patent Applicationn. 14002611.3

2) U.S. Patent Application n. 14/909,854; European Patent Applicationn. 14746954.8

All other authors declare no competing interests.

References

[1] E.M. McNally, P. Pytel, Muscle diseases: the muscular dystrophies, Annu. Rev.Pathol. 2 (2007) 87–109.

[2] P. Konieczny, K. Swiderski, J.S. Chamberlain, Gene and cell-mediated therapies formuscular dystrophy, Muscle Nerve 47 (2013) 649–663.

[3] S. Guiraud, A. Aartsma-Rus, N.M. Vieira, K.E. Davies, G.J. Van Ommen,L.M. Kunkel, The pathogenesis and therapy of muscular dystrophies, Annu. Rev.Genomics Hum. Genet. 16 (2015) 281–308.

[4] S. Guiraud, K.E. Davies, Pharmacological advances for treatment in Duchennemuscular dystrophy, Curr. Opin. Pharmacol. 34 (2017) 36–48.

[5] P. Miura, B.J. Jasmin, Utrophin upregulation for treating Duchenne or Beckermuscular dystrophy: how close are we? Trends Mol. Med. 12 (2006) 122–129.

[6] R.J. Fairclough, M.J. Wood, K.E. Davies, Therapy for Duchenne muscular dys-trophy: renewed optimism from genetic approaches, Nat. Rev. Genet. 14 (2013)373–378.

[7] S. Guiraud, H. Chen, D.T. Burns, K.E. Davies, Advances in genetic therapeuticstrategies for Duchenne muscular dystrophy, Exp. Physiol. 100 (2015) 1458–1467.

[8] V. Malik, L.R. Rodino-Klapac, J.R. Mendell, Emerging drugs for Duchenne musculardystrophy, Expert Opin. Emerg. Drugs 17 (2012) 261–277.

[9] C. Moorwood, T.S. Khurana, Duchenne muscular dystrophy drug discovery - theapplication of utrophin promoter activation screening, Expert Opin. Drug Discovery8 (2013) 569–581.

[10] V. Ricotti, S. Spinty, H. Roper, I. Hughes, B. Tejura, N. Robinson, G. Layton,K. Davies, F. Muntoni, J. Tinsley, Safety, tolerability, and pharmacokinetics of SMTC1100, a 2‑arylbenzoxazole utrophin modulator, following single- and multiple-dose administration to pediatric patients with Duchenne muscular dystrophy, PLoSOne 11 (2016) e0152840.

[11] A.R. Amenta, A. Yilmaz, S. Bogdanovich, B.A. McKechnie, M. Abedi, T.S. Khurana,J.R. Fallon, Biglycan recruits utrophin to the sarcolemma and counters dystrophicpathology in mdx mice, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 762–767.

[12] M. Ito, Y. Ehara, J. Li, K. Inada, K. Ohno, Protein-anchoring therapy of biglycan for

Mdx Mouse Model of Duchenne muscular dystrophy, Hum. Gene Ther. 28 (2017)428–436.

[13] M.G. Di Certo, N. Corbi, G. Strimpakos, A. Onori, S. Luvisetto, C. Severini,A. Guglielmotti, E.M. Batassa, C. Pisani, A. Floridi, B. Benassi, M. Fanciulli,A. Magrelli, E. Mattei, C. Passananti, The artificial gene Jazz, a transcriptionalregulator of utrophin, corrects the dystrophic pathology in mdx mice, Hum. Mol.Genet. 19 (2010) 752–760.

[14] C. Passananti, N. Corbi, A. Onori, M.G. Di Certo, E. Mattei, Transgenic mice ex-pressing an artificial zinc finger regulator targeting an endogenous gene, MethodsMol. Biol. 649 (2010) 183–206.

[15] G. Strimpakos, N. Corbi, C. Pisani, M.G. Di Certo, A. Onori, S. Luvisetto, C. Severini,F. Gabanella, L. Monaco, E. Mattei, C. Passananti, Novel adeno-associated viralvector delivering the utrophin gene regulator jazz counteracts dystrophic pathologyin mdx mice, J. Cell. Physiol. 229 (2014) 1283–1291.

[16] J. Ramos, J.S. Chamberlain, Gene therapy for Duchenne muscular dystrophy,Expert Opin. Orphan Drugs 3 (2015) 1255–1266.

[17] K. Hollinger, J.S. Chamberlain, Viral vector-mediated gene therapies, Curr. Opin.Neurol. 28 (2015) 522–527.

[18] J.R. Chamberlain, J.S. Chamberlain, Progress toward gene therapy for Duchennemuscular dystrophy, Mol. Ther. 25 (2017) 1125–1131.

[19] P.R. Lyons, C.R. Slater, Structure and function of the neuromuscular junction inyoung adult mdx mice, J. Neurocytol. 20 (1991) 969–981.

[20] M. van Putten, D. Kumar, M. Hulsker, W.M. Hoogaars, J.J. Plomp, A. van Opstal,M. van Iterson, P. Admiraal, G.J. van Ommen, P.A. 't Hoen, A. Aartsma-Rus,Comparison of skeletal muscle pathology and motor function of dystrophin andutrophin deficient mouse strains, Neuromuscul. Disord. 22 (2012) 406–411.

[21] S.J. Pratt, S.B. Shah, C.W. Ward, M.P. Inacio, J.P. Stains, R.M. Lovering, Effects of invivo injury on the neuromuscular junction in healthy and dystrophic muscles, J.Physiol. 591 (2013) 559–570.

[22] S.J. Pratt, S.B. Shah, C.W. Ward, J.P. Kerr, J.P. Stains, R.M. Lovering, Recovery ofaltered neuromuscular junction morphology and muscle function in mdx mice afterinjury, Cell. Mol. Life Sci. 72 (2015) 153–164.

[23] J.R. Sanes, J.W. Lichtman, Development of the vertebrate neuromuscular junction,Annu. Rev. Neurosci. 22 (1999) 389–442.

[24] E. Ferraro, F. Molinari, L. Berghella, Molecular control of neuromuscular junctiondevelopment, J. Cachexia. Sarcopenia Muscle 3 (2012) 13–23.

[25] S.J. Pratt, A.P. Valencia, G.K. Le, S.B. Shah, R.M. Lovering, Pre- and postsynapticchanges in the neuromuscular junction in dystrophic mice, Front. Physiol. 9 (2015)252.

[26] E.M. Van der Pijl, M. Van Putten, E.H. Niks, J.J. Verschuuren, A. Aartsma-Rus,J.J. Plomp, Characterization of neuromuscular synapse function abnormalities inmultiple Duchenne muscular dystrophy mouse models, Eur. J. Neurosci. 43 (2016)1623–1635.

[27] C.L. Dennis, J.M. Tinsley, A.E. Deconinck, K.E. Davies, Molecular and functionalanalysis of the utrophin promoter, Nucleic Acids Res. 24 (1996) 1646–1652.

[28] D. Yaffe, O. Saxel, Serial passaging and differentiation of myogenic cells isolatedfrom dystrophic mouse muscle, Nature 270 (1977) 725–727.

[29] A. Onori, C. Pisani, G. Strimpakos, L. Monaco, E. Mattei, C. Passananti, N. Corbi,UtroUp is a novel six zinc finger artificial transcription factor that recognises 18base pairs of the utrophin promoter and efficiently drives utrophin upregulation,BMC Mol. Biol. 14 (2013) 3.

[30] C. Usskilat, C. Skerka, H.P. Saluz, F. Hänel, The transcription factor Egr-1 is a

Fig. 6. The artificial transcription factors JZif1 and its prototype Jazz impact the neuromuscular junction (NMJ) by utrophin up-regulation.The panels illustrate a schematic representation of the utrophin-associated protein complex at mammalian neuromuscular junction (NMJ). Dystroglycan (DG) associates with rapsyn andrapsyn links Acetylcholine Receptors (AChRs) to the utrophin associated protein complex. Utrophin links the entire complex to the F-actin cytoskeleton. The artificial transcription factorsJZif1 and Jazz bind the utrophin promoter ‘A’ at the 9-base pair long target sequence, thereby enhancing transcription of utrophin gene. The expression of JZif1 (and Jazz) gene improvesthe morphology/plasticity of neuromuscular junction.

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1181

Page 11: BBA - Molecular Basis of Disease · 2018. 11. 15. · delivery of ZF-ATFs, we chose recombinant Adeno-associated viral (AAV) vectors which have been described as powerful and safe

regulator of the human TopBP1 gene, Gene 380 (2006) 144–1450.[31] H.G. Radley-Crabb, M.L. Fiorotto, M.D. Grounds, The different impact of a high fat

diet on dystrophic mdx and control C57Bl/10 mice, PLoS Curr. 3 (2011) RRN1276.[32] U. Walter, K. Krolikowski, B. Tarnacka, R. Benecke, A. Czlonkowska, D. Dressler,

Sonographic detection of basal ganglia lesions in asymptomatic and symptomaticWilson disease, Neurology 64 (2005) 1726–1732.

[33] N. Corbi, V. Libri, M. Fanciulli, J.M. Tinsley, K.E. Davies, C. Passananti, The arti-ficial zinc finger coding gene ‘Jazz’ binds the utrophin promoter and activatestranscription, Gene Ther. 7 (2000) 1076–1083.

[34] N. Corbi, V. Libri, A. Onori, C. Passananti, Synthetic zinc finger peptides: old andnovel applications, Biochem. Cell Biol. 82 (2004) 428−436.

[35] Y. Choo, A. Klug, Physical basis of a protein-DNA recognition code, Curr. Opin.Struct. Biol. 7 (1997) 117–125.

[36] J. Park, J. Wicki, S.E. Knoblaugh, J.S. Chamberlain, D. Lee, Multi-parametric MRI at14T for muscular dystrophy mice treated with AAV vector-mediated gene therapy,PLoS One 10 (2015) e0124914.

[37] A. Klug, The discovery of zinc fingers and their development for practical appli-cations in gene regulation and genome manipulation, Q. Rev. Biophys. 43 (2010)1–21.

[38] C.A. Gersbach, T. Gaj, C.F. Barbas, Synthetic zinc finger proteins: the advent oftargeted gene regulation and genome modification technologies, Acc. Chem. Res.47 (2014) 2309–2318.

[39] A. Eguchi, G.O. Lee, F. Wan, G.S. Erwin, A.Z. Ansari, Controlling gene networks andcell fate with precision-targeted DNA-binding proteins and small-molecule-basedgenome readers, Biochem. J. 462 (2014) 397–413.

[40] T. Gaj, S.J. Sirk, S.L. Shui, J. Liu, Genome-editing technologies: principles andapplications, Cold Spring Harb. Perspect. Biol. 8 (2016) (pii: a023754).

[41] Sangamo Therapeutic Inc, http://www.sangamo.com, (2017) , Accessed date: 13December 2017.

[44] C.E. Nelson, J.N. Robinson-Hamm, C.A. Gersbach, Genome engineering: a newapproach to gene therapy for neuromuscular disorders, Nat. Rev. Neurol. 13 (2017)647–661.

[45] E. Mattei, N. Corbi, M.G. Di Certo, G. Strimpakos, C. Severini, A. Onori, A. Desantis,V. Libri, S. Buontempo, A. Floridi, M. Fanciulli, D. Baban, K.E. Davies,C. Passananti, Utrophin up-regulation by an artificial transcription factor in

transgenic mice, PLoS One 2 (2007) e774.[46] A. Desantis, A. Onori, M.G. Di Certo, E. Mattei, M. Fanciulli, C. Passananti, N. Corbi,

Novel activation domain derived from Che−1 cofactor coupled with the artificialprotein Jazz drives utrophin upregulation, Neuromuscul. Disord. 19 (2009)158–162.

[47] A.E. Deconinck, A.C. Potter, J.M. Tinsley, S.J. Wood, R. Vater, C. Young,L. Metzinger, A. Vincent, C.R. Slater, K.E. Davies, Postsynaptic abnormalities at theneuromuscular junctions of utrophin-deficient mice, J. Cell Biol. 136 (1997)883–894.

[48] J.A. Rafael, E.R. Townsend, S.E. Squire, A.C. Potter, J.S. Chamberlain, K.E. Davies,Dystrophin and utrophin influence fiber type composition and post-synapticmembrane structure, Hum. Mol. Genet. 9 (2000) 1357–1367.

[49] G.B. Banks, C. Fuhrer, M.E. Adams, S.C. Froehner, The postsynaptic submembranemachinery at the neuromuscular junction: requirement for rapsyn and the utro-phin/dystrophin-associated complex, J. Neurocytol. 32 (2003) 709–726.

[50] R.M. Grady, H. Zhou, J.M. Cunningham, M.D. Henry, K.P. Campbell, J.R. Sanes,Maturation and maintenance of the neuromuscular synapse: genetic evidence forroles of the dystrophin—glycoprotein complex, Neuron 25 (2000) 279–293.

[51] M. Aittaleb, Y. Martinez-Pena, I. Valenzuela, M. Akaaboune, Spatial distributionand molecular dynamics of dystrophin glycoprotein components at the neuromus-cular junction in vivo, J. Cell Sci. 130 (2017) 1752–1759.

[52] R. Rudolf, M.M. Khan, S. Labeit, M.R. Deschenes, Degeneration of neuromuscularjunction in age and dystrophy, Front. Aging Neurosci. 6 (2014) 99.

[53] V. De Arcangelis, G. Strimpakos, F. Gabanella, N. Corbi, S. Luvisetto, A. Magrelli,A. Onori, C. Passananti, C. Pisani, S. Rome, C. Severini, F. Naro, E. Mattei, M.G. DiCerto, L. Monaco, Pathways implicated in Tadalafil amelioration of Duchennemuscular dystrophy, J. Cell. Physiol. (2016) 224–233.

[54] G. Dobrowolny, M. Aucello, E. Rizzuto, S. Beccafico, C. Mammucari,S. Boncompagni, S. Belia, F. Wannenes, C. Nicoletti, Z. Del Prete, N. Rosenthal,M. Molinaro, F. Protasi, G. Fanò, M. Sandri, A. Musarò, Skeletal muscle is a primarytarget of SOD1G93A-mediated toxicity, Cell Metab. 8 (2008) 425–436.

[55] J. Penney, K. Tsurudome, E.H. Liao, F. Elazzouzi, M. Livingstone, M. Gonzalez,N. Sonenberg, A.P. Haghighi, TOR is required for the retrograde regulation of sy-naptic homeostasis at the Drosophila neuromuscular junction, Neuron 12 (2012)166–178.

C. Pisani et al. BBA - Molecular Basis of Disease 1864 (2018) 1172–1182

1182