multipotent stromal cells alleviate inflammation, neuropathology, and symptoms associated with...

21
Author contributions: B.A.S.: Conception and design, collection and/or assembly of data, data analysis and interpretation, manuscript writing; X.Z.: Conception and design, collection and/or assembly of data, data analysis and interpretation; A.C.B.: Collection and/or assembly of data, data analysis and interpretation; P.A.G.: Collection and/or assembly of data, data analysis and interpretation; J.A.S.: Conception and design, collection and/or assembly of data, data analysis and interpretation; J M.F-P.: Collection and/or assembly of data; S.Z.: Collection and/or assembly of data, data analysis and interpretation; R.W.B.: Data analysis and interpretation; L.M.: Data analysis and interpretation; S.C.L.: Collection and/or assembly of data, data analysis and interpretation; A.V.K.: Conception and design, data analysis and interpretation; B.A.B.: Conception and design, financial support, administrative support, data analysis and interpretation, final approval of manuscript *Corresponding author: Bruce A. Bunnell, PhD, Tulane University School of Medicine, Center for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA, USA 70112, Phone: 1-(504) 988-7711, Fax: 1-(504) 988-7710, [email protected]; Grants and Funding: R21-NS059665 from the National Institutes of Neurological Disorders and Stroke (NIH) and Tulane University.; Received January 20, 2013; accepted for publication March 18, 2013; access option. 1066-5099/2013/$30.00/0 doi: 10.1002/stem.1397 This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1002/stem.1397 STEM CELLS ® REGENERATIVE MEDICINE Multipotent Stromal Cells Alleviate Inflammation, Neuropathology, and Symptoms Associated with Globoid Cell Leukodystrophy in the Twitcher Mouse Brittni A. Scruggs 1,2 , Xiujuan Zhang 1 , Annie C. Bowles 1,3 , Peter A. Gold 1 , Julie A. Semon 1 , Jeanne M. Fisher-Perkins 4 , Shijia Zhang 1,2 , Ryan W. Bonvillain 1 , Leann Myers 5 , Su Chen Li 6 , Allan V. Kalueff 2 , Bruce A. Bunnell 1,2,4* 1 Center for Stem Cell Research and Regenerative Medicine, SL-99, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA, 2 Department of Pharmacology, SL-83, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA, 3 Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, 2000 Percival Stern Hall, 6400 Freret Street, New Orleans, LA 70118, USA, 4 Division of Regenerative Medicine, Tulane University National Primate Center, 18703 Three Rivers Road, Covington, LA 70433, USA, 5 Department of Biostatistics & Bioinformatics, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Suite 2001, New Orleans, LA 70112, USA, 6 Department of Biochemistry and Molecular Biology, SL- 43, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA Key words. Globoid cell leukodystrophy mesenchymal stem cells/multipotent stromal cells/MSCs twitcher mouse bone marrow-derived stem cells/BMSCs stem cell transplantation ABSTRACT Globoid cell leukodystrophy (GLD) is a common neurodegenerative lysosomal storage disorder caused by a deficiency in galactocerebrosidase (GALC), an enzyme that cleaves galactocerebroside during myelination. Bone marrow transplantation has shown promise when administered to late-onset GLD patients. However, the side effects (e.g., graft versus host disease), harsh conditioning regimens (e.g., myelosuppression), and variable therapeutic effects make this an unsuitable option for infantile GLD patients. We previously reported modest improvements in the twitcher mouse model of GLD after intracerebroventricular (ICV) injections of a low dose of multipotent stromal cells (MSCs). Goals of this study were to improve bone marrow-derived MSC (BMSC) therapy for GLD by increasing the cell dosage and comparing cell type (e.g., transduced v. native), treatment timing (e.g., single v. weekly), and administration route (e.g., ICV v. intraperitoneal). Neonatal twitcher mice received (1) 2x10 5 BMSCs by ICV injection, (2) 1x10 6 BMSCs by intraperitoneal (IP) injection, (3) weekly IP injections of 1x10 6 BMSCs, or (4) 1x10 6 lentiviral-transduced BMSCs overexpressing GALC (GALC-BMSC) by IP injection. All treated mice lived longer than untreated mice. However, the mice receiving peripheral MSC therapy had improved motor function (e.g., hind limb strength and rearing ability), twitching symptoms, and weight compared to both the untreated and ICV-treated mice. Inflammatory cell, globoid cell, and apoptotic cell levels in the sciatic nerves were significantly decreased as a result of the GALC-BMSC or weekly IP injections. The

Upload: independent

Post on 28-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Author contributions: B.A.S.: Conception and design, collection and/or assembly of data, data analysis and interpretation, manuscript writing; X.Z.: Conception and design, collection and/or assembly of data, data analysis and interpretation; A.C.B.: Collection and/or assembly of data, data analysis and interpretation; P.A.G.: Collection and/or assembly of data, data analysis and interpretation; J.A.S.: Conception and design, collection and/or assembly of data, data analysis and interpretation; J M.F-P.: Collection and/or assembly of data; S.Z.: Collection and/or assembly of data, data analysis and interpretation; R.W.B.: Data analysis and interpretation; L.M.: Data analysis and interpretation; S.C.L.: Collection and/or assembly of data, data analysis and interpretation; A.V.K.: Conception and design, data analysis and interpretation; B.A.B.: Conception and design, financial support, administrative support, data analysis and interpretation, final approval of manuscript

*Corresponding author: Bruce A. Bunnell, PhD, Tulane University School of Medicine, Center for Stem Cell Research and Regenerative Medicine, 1430 Tulane Avenue, SL-99, New Orleans, LA, USA 70112, Phone: 1-(504) 988-7711, Fax: 1-(504) 988-7710, [email protected]; Grants and Funding: R21-NS059665 from the National Institutes of Neurological Disorders and Stroke (NIH) and Tulane University.; Received January 20, 2013; accepted for publication March 18, 2013; access option. 1066-5099/2013/$30.00/0 doi: 10.1002/stem.1397

This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1002/stem.1397

STEM CELLS®

REGENERATIVE MEDICINE

Multipotent Stromal Cells Alleviate Inflammation, Neuropathology, and Symptoms Associated with Globoid Cell Leukodystrophy in the Twitcher Mouse

Brittni A. Scruggs1,2, Xiujuan Zhang1, Annie C. Bowles1,3, Peter A. Gold1, Julie A. Semon1, Jeanne M. Fisher-Perkins4, Shijia Zhang1,2, Ryan W. Bonvillain1, Leann Myers5, Su Chen Li6, Allan V. Kalueff2,Bruce A. Bunnell1,2,4*

1Center for Stem Cell Research and Regenerative Medicine, SL-99, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA, 2Department of Pharmacology, SL-83, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA, 3Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, 2000 Percival Stern Hall, 6400 Freret Street, New Orleans, LA 70118, USA, 4Division of Regenerative Medicine, Tulane University National Primate Center, 18703 Three Rivers Road, Covington, LA 70433, USA, 5Department of Biostatistics & Bioinformatics, Tulane University School of Public Health and Tropical Medicine,1440 Canal Street, Suite 2001, New Orleans, LA 70112, USA, 6Department of Biochemistry and Molecular Biology, SL-43, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA

Key words. Globoid cell leukodystrophy mesenchymal stem cells/multipotent stromal cells/MSCs twitcher mouse

bone marrow-derived stem cells/BMSCs stem cell transplantation

ABSTRACTGloboid cell leukodystrophy (GLD) is a common neurodegenerative lysosomal storage disorder caused by a deficiency in galactocerebrosidase (GALC), an enzyme that cleaves galactocerebroside during myelination. Bone marrow transplantation has shown promise when administered to late-onset GLD patients. However, the side effects (e.g., graft versus host disease), harsh conditioning regimens (e.g., myelosuppression), and variable therapeutic effects make this an unsuitable option for infantile GLD patients. We previously reported modest improvements in the twitcher mouse model of GLD after intracerebroventricular (ICV) injections of a low dose of multipotent stromal cells (MSCs). Goals of this study were to improve bone marrow-derived MSC (BMSC) therapy for GLD by increasing the cell dosage and

comparing cell type (e.g., transduced v. native), treatment timing (e.g., single v. weekly), and administration route (e.g., ICV v. intraperitoneal). Neonatal twitcher mice received (1) 2x105 BMSCs by ICV injection, (2) 1x106 BMSCs by intraperitoneal (IP) injection, (3) weekly IP injections of 1x106 BMSCs, or (4) 1x106 lentiviral-transduced BMSCs overexpressing GALC (GALC-BMSC) by IP injection. All treated mice lived longer than untreated mice. However, the mice receiving peripheral MSC therapy had improved motor function (e.g., hind limb strength and rearing ability), twitching symptoms, and weight compared to both the untreated and ICV-treated mice. Inflammatory cell, globoid cell, and apoptotic cell levels in the sciatic nerves were significantly decreased as a result of the GALC-BMSC or weekly IP injections. The

Peripheral Stem Cell Therapy in GLD Mice

2

results of this study indicate a promising future for peripheral MSC therapy as a non-invasive, adjunct

therapy for patients affected with GLD.

INTRODUCTION

Lysosomal storage diseases (LSDs) affect 1 out of every 5,000-7,000 live births and, collectively, represent some of the most common inherited diseases in pediatric populations [1-3]. Globoid cell leukodystrophy (GLD; Krabbe’s disease) is an autosomal recessive neurodegenerative LSD caused by a deficiency of galactocerebrosidase (GALC), an enzyme responsible for the catabolism of myelin sphingolipids, such as galactocerebroside (GalCer). Although asymptomatic at birth, infantile GLD patients present with symptoms before six months of age, deteriorating rapidly due to toxic substrate accumulation (e.g.,psychosine) and extensive demyelination of the central and peripheral nervous systems [4-6]. The symptoms resulting from such pathology include febrile episodes, inconsolable fits, arrest of mental and motor development, hypertonicity, seizures, and paralysis followed by premature death within two years [5, 7-9].

The twitcher mouse, a model of GLD, has striking pathological similarities to the human patient, including axonopathy, severe gliosis, globoid cell formation, widespread apoptosis and inflammation, and a diffuse demyelinating peripheral neuropathy [10-12]. Between postnatal days (PND) 15-20, the GALC deficient mouse begins to show symptoms of motor function deterioration, weight reduction, decreased locomotion, and behavioral abnormalities [13-15]. If left untreated, twitcher mice experience rapid motor and neurological deterioration and death by PND30-40. The severity of the twitcher phenotype makes these mice a useful genetic model for high-throughput development and screening of therapeutic approaches for GLD and other LSDs.

Hematopoietic stem cell transplantation (HSCT) has recently shown promising results (e.g.,

delayed onset of symptoms and increased lifespan) when administered to asymptomatic late-onset GLD patients [16, 17]. This therapy has also been supportive in several infantile GLD patients and twitcher mice [16, 18-20]; these patients and mice, however, have all succumbed to a progressive disease [9, 21, 22]. Furthermore, there are serious risks associated with HSCT due to complications of the procedure (e.g., graft versus host disease, GVHD) or the aggressive conditioning regimens [23, 24]. Studies using the twitcher mouse have shown that HSCT, specifically bone marrow transplantation (BMT), exacerbates the tremor, fails to provide substantial peripheral nerve improvement, and has a 20% mortality rate due to severe hemorrhage [19, 25]. The lack of effective treatment options, in addition to the severity of GLD, highlights the need for development of innovative therapeutic approaches.

Recent studies have demonstrated that enzyme replacement therapy (ERT) [26, 27], substrate reduction therapy (SRT) [28], vector therapy [29-31], and anti-inflammatory agents [32] improve the twitcher mouse pathology and modestly prolong lifespan. However, these therapeutic approaches are only partially successful even when combined with BMT [22, 29]. An alternative approach - transplantation of multipotent mesenchymal stromal cells (MSCs)- has been studied for several LSDs due to the potential of these cells to migrate to sites of tissue injury, deliver therapeutic gene products and trophic factors, replace affected cells through engraftment and differentiation, and decrease inflammation in neurodegenerative disease states [33-37]. MSCs, which can undergo both self-renewal and multi-lineage differentiation, are easily harvested and expanded ex vivo and are capable of providing neuroprotection and immunomodulation while evading host immune surveillance [35, 38, 39]. Interestingly, MSCs are currently being used in

Peripheral Stem Cell Therapy in GLD Mice

3

conjunction with BMT to decrease the risk of GVHD and improve engraftment in the clinical setting [40-42]. Thus, alleviation of symptoms in the twitcher mouse by using MSC therapy would likely lead to development of a potentially safer and more effective HSCT treatment for GLD patients.

Our group recently reported that administration of a single, low dose of MSCs through intraperitoneal (IP) or intracerebroventricular (ICV) injections improved the phenotype of the twitcher mouse by reducing the levels of inflammation [13, 43]. The current study aims to enhance MSC therapy for GLD by increasing the functional GALC levels and anti-inflammatory effects in the twitcher mouse. To accomplish these goals, twitcher mice received peripheral or central-directed MSC therapy in higher cell numbers or increased injection frequency (i.e.,weekly IP injections) compared to our previous studies. All MSC treatments were evaluated for effectiveness by assessing improvements in apoptosis and inflammation in peripheral nerves, twitching symptoms, body weight, and hind limb strength. The goal of the novel stem cell therapy in this study is to improve pathology or phenotype in the twitcher mouse in order to develop a pre-clinical model for a non-invasive adjunct therapy for patients affected with LSDs.

MATERIALS AND METHODS

AnimalsAdult (3 month-old) heterozygote (Het; Galctwi/+) C57Bl/6J (B6.CE-Galctwi/J) mice were originally obtained from The Jackson Laboratory (Bangor, ME) and used as breeder pairs to generate homozygous (Twi; Galctwi/twi)twitcher mice and wild type (WT) C57Bl/6J mice. All animal procedures were approved by the Institutional Animal Care and Use Committee (IACUC) at Tulane University and conformed to the requirements of the Animal Welfare Act. In total, there were six mouse groups studied: 15 ICV BMSC-treated wild type (WT, Galc+/+), 26 untreated twitcher (Twi, Galc-

/-), 18 single IP BMSC-treated twitcher (IP, Galc-

/-), 8 weekly IP BMSC-treated twitcher (Weekly, Galc-/-), 9 IP GALC-transduced BMSC-treated twitcher (GALC, Galc-/-), and 7 ICV BMSC-treated twitcher (ICV, Galc-/-) male and female mice were used in this study (Figure 1). The specific Galc mutation was confirmed as previously described [44].

Harvesting, Culture, and Characterization of Murine eGFPTgBMSCs BMSCs were obtained from male eGFP transgenic mice (C57Bl/6-Tg(UBC-GFP)30Scha/J strain; Jackson Laboratory) between 4 and 6 months of age. BMSCs were isolated, characterized, and cultured from the femurs and tibiae of each mouse as previously described [45]. Briefly, the ends of each tibia and femur were removed to expose the marrow. The marrow was pushed out of the bone using a syringe with complete expansion media (CEM), re-suspended in CEM, and filtered through a 70

m nylon mesh filter. The mixture was then centrifuged at 400 x g for 10 minutes at 4°C, and the pellet was re-suspended in 3 mL CEM. CEM consists of Iscove's Modified Dulbecco's Medium (IMDM, Invitrogen, Carlsbad, CA) supplemented with 9% fetal bovine serum (FBS; Atlanta Biologicals, Lawrenceville, GA), 9% horse serum (HS; Hyclone Laboratories, Logan UT), 100 U/mL penicillin (Invitrogen), 100

g/mL streptomycin (Invitrogen), 0.25 g/mL amphotericin B (Invitrogen), and 12 M L-glutamine (Invitrogen). The cells were then plated, washed with media, and stored in liquid nitrogen or expanded further exactly as described in Ripoll, et al. [45]. The cells were characterized as MSCs using osteogenesis, adipogenesis, and chondrogenic differentiation assays, FACS analysis for detection of cell surface markers, growth rate examination, and a colony forming unit (CFU) assay as previously described [45].

Intraperitoneal Injections of Murine eGFPTgBMSCsPassage-eight (P8) murine eGFPTgBMSCs were recovered from cryopreservation in CEM, cultured to 70% confluence, and harvested for

Peripheral Stem Cell Therapy in GLD Mice

4

infusion as previously described [13]. A 50 µL Hamilton syringe with a 30-gauge stainless steel needle was used to administer 50 µL of the cell suspension (1 x 106 total eGFPTgBMSCs) or HBSS into the left side of the peritoneal cavity on PND5-6. The pups were then returned to their mothers.

Lentiviral Transduction of eGFPTgBMSCs The GALC viral particles were produced by calcium phosphate transient transfection of 293T cells [46] with the GALC transgene-containing plasmid (pNL-EF1 -GALC-TMH/WPRE 03, a gift from Dr. Jakob Reiser, FDA/CBER) and two helper plasmids (pMD.G, pHR- 8.9). Briefly, the 293T cells were seeded in 15-cm cell culture plates with a concentration of 8×106 cells per plate. The transfection by calcium phosphate with the 3 plasmids in the presence of 25 µM chloroquine (Sigma) was carried out 24 h later. The transfectate was replaced by fresh ultraculture serum-free medium (Lonza, Basel, Switzerland) after 16-18 h, and the unconcentrated GALC viral supernatant was collected 48 h later. The titer of the viral particles was determined by the lentiviral titer kit from MellGen Laboratories Inc (Surrey, BC, Canada).

For cell transduction, the GFP+ BMSCs were seeded on 6-well cell culture plates with 8×104

cells per well. After a 24-hour incubation period, the cells in each well were treated with 1 mL GALC lentivirus in the presence of 0.5 mL IMDM with 10% heat-inactivated FBS and 8 µg/mL polybrene. After 24 h, the medium containing the GALC lentivirus was replaced with new medium and another 1 mL of GALC lentivirus. The same procedure was repeated again 24 h later, and then new medium without GALC lentivirus was added and the cells were incubated for another 72 h. The GALC transduced GFP+ BMSCs were isolated by single cell colony selection. The cells were seeded on 10-cm cell culture plate with 100 cells per plate, and cultured for 14 days. The Nikon Object Marker of the microscope was used to mark the desired colonies, and a cloning ring (Fisher) with

vacuum grease was put around each desired colony. Isolated colonies were lifted by trypsinization and seeded onto 48-well cell culture plates for expansion. After expansion, each selected colony was stained by a modified

-galactosidase assay [47] to determine the GALC transduction efficiency, and eGFPTgBMSCs with over 90% GALC transduction efficiency were used for IP injections as described.

Intracerebroventricular Injection of Murine eGFPTgBMSCsGFP+ BMSCs were harvested, cultured, and lifted as previously described for the IP injections. The cell suspension was taken up via a sterile 30G needle that was attached by plastic tubing to a 10 µL Hamilton syringe. A stereotaxic instrument was used to immobilize the heads of the cryoanesthesized twitcher and unaffected littermate pups, while 2 µL of the BMSC suspension (5 x 104 BMSCs/µL in HBBS) or HBSS was injected into each lateral ventricle on PND3-4 using previously defined coordinates [43]. The needle was kept immobilized for 2 min before withdrawing. The pups were then resuscitated on a heating pad before being returned to their mothers.

Motor Function Testing Beginning on PND14, motor function tests were performed three times per week to monitor the twitching frequency and severity, wire hang, and hind stride length. Mice were scored based on the scoring systems explained in our previous studies [43]. Video and behavior recording of each mouse was conducted weekly using a LifeCam Cinema webcam with True 720p HD video and Debut Video Capture Software (Microsoft Corp., Redmond, WA) as described in our previous study [13, 43]. Briefly, all video files were uploaded for analysis using the EthoVision XT7 software for quantification of various spatial endpoints [13]. Twitcher mice were euthanized once they lost 20% of their maximum body weight or became moribund.

Peripheral Stem Cell Therapy in GLD Mice

5

Tissue Processing and Histological Staining Animals were euthanized by CO2 asphyxiation and perfused with sterile PBS through the left ventricle after rupture of the right atrium. After intracardial perfusion, the brain and sciatic nerves were removed and stored for up to three months at 4°C in Allprotect tissue reagent solution (Qiagen, Valencia, CA) or stored at room temperature in 10% neutral buffered formalin. Paraffin-embedded sections were cut at 5 microns, mounted, and subsequently stained with hematoxylin and eosin (H&E). Slides were scanned using an Aperio ScanScope® CS instrument (Aperio Technologies, Inc., Vista, CA), and the images were analyzed with Fiji/Image J software (National Institutes of Health, Bethesda, Maryland, http://imagej.nih.gov/ij/) [48]. For each treatment group, H&E stains of three mice (10 sections/mouse) were analyzed.

TUNEL Assay to Detect DNA Fragmentation Unstained slides of three mice per treatment group (3 sections/mouse) for terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and immunohistochemistry (IHC) were prepared by sectioning paraffin-embedded samples at a thickness of 5 µm. All slides were warmed on a heating platform at 57°C for 30 minutes prior to deparaffinization. For deparaffinization, all slides were submerged in HistoChoice (Amresco, Solon, OH) twice for 5 min, 100% ethanol twice for 2 min, 95% ethanol twice for 2 min, 70% ethanol for 2 min, 50% ethanol for 2 min, distilled (DI) water twice for 2 min, and 1X tris-buffered saline (TBS, pH 10.0) twice for 5 min. The slides were rinsed with DI water, air dried, outlined using an ImmEdge Pen (Vector Laboratories, Burlingame, CA), washed with 20mM Tris-HCl (pH 8.0), and then incubated with a 1:1000 proteinase K solution in Tris-HCl pH 10.0 (20 µg/ml) for 15 min at 37°C in a humidified chamber.

For any given slide, one section was designated as the positive control and incubated at room temperature (RT) with a DNase I (Invitrogen)

solution in PBS (15U/mL) for 10 min. Using the In Situ Cell Death/Fluorescein Detection Kit (Roche Diagnostics, Indianapolis, IN), all slides were incubated with 50 µL of TUNEL solution for 1 h at 37°C in a humidified chamber. The slides were washed three times in 1X PBS for 5 min before incubation with a 0.4 mM DAPI/TBS solution. ProLong Gold Antifade Reagent (Invitrogen) was then used to mount coverslips. Fluorescent images were acquired at 5X and 10X using a Leica DMRXA2 deconvolution microscope (Leica Microsystems, Buffalo Grove, IL).

ImmunohistochemistryThe deparaffinized slides were submerged in 700mL of citrate buffer pH 6.0 (10mM) and heated for 20 min in a microwave using a low heat setting. After cooling, the slides were washed for 5 min in 1X PBS and subsequently washed with PBS-FSG-Tx-100 (10% v/v 10X PBS, 0.2% v/v fish skin gelatin, and 0.1% v/v Triton x-100) for 5 min before incubation for 1 h in a humidified chamber at RT with blocking solution, which consisted of 10% normal goat serum (NGS) in PBS-FSG (10% v/v 10X PBS and 0.2% v/v fish skin gelatin). The primary antibody to EGFP (anti-GFP; 1:100, Invitrogen: A-11121 or 11122), mature macrophages (F4/80; 1:10, Santa Cruz: SC-59171 Rat IgG2b), neuronal nuclei (NeuN; 1:50, Chemicon: MAB377 Ms IgG1), neural crest cells (S-100; 1:1000, Sigma: S-2644 Rb), or astrocytes (GFAP; 1:200, Sigma: C9205 Ms IgG1) was diluted in 10% NGS solution and applied to appropriate experimental sections for 1 hour incubation in a humidified chamber at RT. Control slides were treated with secondary antibody-only (2° only). Following incubation, the slides were washed in PBS-FSG-Tx-100 and PBS-FSG, each for 10 min. The sections were then incubated in a humidified chamber at RT for 1 hour with the secondary antibody (e.g.,anti-rat, anti-rabbit, or anti-mouse IgG antibody conjugated with Alexa 488 or 594) in 10% NGS solution. Slides were then washed twice in PBS-FSG-Tx-100 and once in PBS-FSG. The slides were incubated with a DAPI (50 µM) or TO-Pro-

Peripheral Stem Cell Therapy in GLD Mice

6

3 (1:1000)/10% NGS solution, mounted with ProLong medium, and imaged using either a deconvolution or a confocal microscope.

Protein Isolation For protein isolation, Allprotect-preserved brain, kidney, and sciatic nerve samples were homogenized in 0.2% NP-40 in PBS using a plastic motorized pestle. Approximately 10 mL of fluid were used per g of sample. All samples were exposed to 10 rounds of sonication, where each round consisted of 5 s of sonication followed by 20 s on ice without sonication. The solutions were cleared twice by centrifugation at 12,000Xg; the supernatants were removed and saved in aliquots at -20°C. Protein concentration was determined using the bicinchoninic acid (BCA) assay kit (Pierce, Rockford, IL), and all protein lysates were centrifuged using centrifugal filter units with microporous membranes (Millipore, Billerica, MA).

Western Blotting Protein lysate (30-100µg), NuPage Reducing Sample Agent (10X, Invitrogen), NuPage LDS Sample buffer (4X, Invitrogen), and DI water were mixed, centrifuged at 1000 g for 1 min, then boiled for 5 min in a PCR PTC-200 thermal cycler (MJ Research, Waltham, MA). The samples (32µL total) and 2-3µL of a MagicMark XP ladder (Invitrogen) were run through a NuPage 4-12% Bis-Tris 1.5 mm gel in 1X MOPS (Invitrogen) running buffer for 1 h at 200V; NuPage antioxidant (0.5mL, Invitrogen) was added to the MOPS in the inner chamber. An iBlot system (Invitrogen) was used with the iBlot transfer stack (Invitrogen) using program 3 for 7 minutes to transfer the protein to a nitrocellulose membrane. The membrane was then incubated with 10 mL of premade blocking solution (Bløk noise canceling reagent for chemiluminescent detection, Millipore) at RT, rinsed, and then incubated in PBST (1X PBS with 0.1% Tween-20) for 1 h at RT followed by overnight at 4°C with the primary antibody to GALC (anti-GALC; 1:2000, ProteinTech: 11991-1-AP), GFP (anti-GFP; 1:1000, Invitrogen: A-11122), or Actin (anti-actin;

1:3000, Sigma-Aldrich: A2066). The membranes were washed three times in 10 mL of PBST with shaking before being gently rocked at RT for 1 h with an HRP (horseradish peroxidase)-conjugated anti-rabbit IgG secondary antibody (1:1000, Cell Signaling) in blocking solution. After three washes with PBST, the membrane was incubated for 2 min with the Novex ECL HRP chemiluminescent substrate reagents (Invitrogen) and then developed using an ImageQuant LAS 4000 imager (GE Healthcare Biosciences, Pittsburgh, PA).

For detection of GFP specific antibodies in the sera of cell-transplanted mice, purified GFP (BioVision, San Francisco, CA) was run in a 1-well NuPage 10% Bis-Tris 1.0 mm gel. After transfer of the protein to a nitrocellulose membrane, the membrane was cut into strips, which were individually blocked and incubated for one hour at RT followed by overnight at 4°C with mouse serum (1:20 in PBST) of a single treatment group. After washing, the membranes were incubated with HRP-conjugated anti-mouse IgG secondary antibody (1:1000, Cell Signaling: 7076) in blocking solution and then imaged with an ImageQuant LAS 4000 imager (GE Healthcare Biosciences) as described above.

GALC Activity Assay For preparation of the BMSCs or GALC-BMSCs, the cells were trypsinized and pelleted as performed for injection purposes and then lysed by sonication in 200 µL of 20mM acetate buffer (pH4.5) while on ice. A total of 150 µL lysed cell suspension or 500 µL brain protein lysates in 25 µL of 200 mM acetate buffer (pH 4.5) was incubated with 50 µL of a 3H-GalCer(Moravek Biochemicals, Brea, CA) substrate mixture (approximately 31,000 cpm) that was prepared as previously described [49]. All enzymatic reactions were stopped, vortexed, centrifuged, extracted, and measured for radioactivity using a scintillation counter (TRI-CARB 1600CA, PACARD) as previously described without modifications [43]. GALC activity is expressed as nanomoles of GalCer hydrolyzed per h per milligram of protein

Peripheral Stem Cell Therapy in GLD Mice

7

(nmol/h/mg protein) or per 1X106 cells (nmol/h/1x106cells).

Fluorescent Substrate Cleavage Assay In order to calculate the activity of three

endogenous enzymes, beta-galactosidase ( -gal),

alpha-galactosidase ( -gal), and beta-

hexosaminidase ( -hex), a total of 5 µL of the lysed BMSC or GALC-BMSC cell suspension was incubated with 10 µL (20 nmoles) of a 4-methylumbelliferyl-beta-D-galactopyranoside (4MU-beta-gal), 4MU-alpha-D-galactopyranoside, or 4MU-beta-N-acetyl-D-glucosaminide, 40 µL of 50mM acetate buffer at pH 4.0 or 7.0, and 50 µL DI water for 0.5-3 h. At the preset time, the enzymatic reactions were quenched by adding 0.7 mL of 20 mM borate buffer pH 9.8. A Turner Quantech fluorometer (Barnstead/Thermolyne Corporation, Dubuque, IA) was used to measure the fluorescence of the liberated 4MU in each sample [50]. The nanomoles of substrate cleaved per h per 10,000 cells (nmol/h/1X104 cells) were calculated based on a standard curve.

Statistical Analysis For comparisons of body weight, wire hang, twitching, and hind stride length, mixed models regression methods were implemented via PROC MIXED in Statistical Analysis Software (SAS, version 9.2). For tests with a significant (p<0.05) time X group interaction, pairwise comparisons of least square means were made to further investigate the interaction. When applicable, the WT group was compared to all twitcher groups. The untreated twitcher group was compared to the treated twitcher groups. Lastly, the treated groups were compared to each other. To correct for multiple pairwise comparisons, the p-values were adjusted using a Bonferroni correction, and significance was determined if p<0.01 for the weight, hind stride, wire hang, and twitching studies. For the wire hang and twitching data, exact chi-square tests were also performed at each time point to test for differences between the treatment groups.

For Kaplan-Meier survival curves, analyses were performed using the log rank test, and specific lifespan differences between the Twi group and the multiple treatment groups were determined using the Sidak-adjusted log rank test. For the behavioral, neurophenotyping, and histological studies, statistical analysis of three or more groups was performed using one-way analysis of variance (ANOVA) followed by pairwise comparisons of the mouse groups using post-hoctesting with Bonferroni correction. For any study where the mouse groups were monitored for three consecutive weeks, two-way ANOVA (factors: time and mouse group) with repeated measures and subsequent Bonferroni post hoc testing was performed. Significance for the overall group effect and individual pairwise comparisons was defined as p<0.05. One- and two-way ANOVA tests were performed using GraphPad Prism 4.0b for Macintosh (GraphPad Software, San Diego, CA), and all values were reported as mean ± SEM.

RESULTS

Weight and longevity The Twi mice had a median lifespan of 36 days, whereas the ICV, IP, Weekly, and GALC groups had median survival times of 43, 42, 41.5, and 44 days, respectively (Fig. 1A), compared to a 44-day median lifespan of mice receiving BMT with low radiation conditioning in a previous study [22]. Using Sidak-adjusted differences, we found that the Twi group had a significantly lower median survival time when compared to any of the four treated groups (p<0.0001). There were no significant differences between the survival curves of the treatment groups. At PND42, only 3.6% of Twi mice were alive compared to 37.5, 38.9, 66.7, and 77.8% for the Weekly, IP, ICV, and GALC mice, respectively. Weight was an objective measurement used to compare all mouse groups between PND16 and PND40, and there was a significant (p<0.0001) group x time interaction (Fig. 1B). By PND28, the Twi group was significantly lower in weight than the Weekly and GALC groups, and the Twi mice had lower weights than every treatment

Peripheral Stem Cell Therapy in GLD Mice

8

group between PND30-40. The Weekly and GALC groups had increased weight after PND30 and PND36, respectively, when compared to the IP group. The treatment also had significant effects on the maximum body weight achieved by each mouse group (F(5, 72)=126.90, p<0.0001); there was a significant increase in maximum weight for all treatment groups compared to Twi mice (p<0.05-0.001; Fig. 1B).

Assessment of motor function using observational and automated phenotyping methodsTwitching is an early presenting symptom in twitcher mice, and comparison of twitching severity across mouse groups showed a significant group x time interaction (p=0.0003; Fig. 1C). Pairwise comparisons of twitching severity indicated that the Twi mice had more severe twitching compared to the IP, Weekly, and GALC groups starting at PND18 and the ICV group after PND32. Although the Weekly and GALC groups achieved lower (i.e., less severe) scores for every time point compared to the IP group, only after PND38 did these groups differ significantly. Furthermore, the treatment type significantly affected the day of twitching onset (F(4, 59)=61.80; p<0.0001); the Twi group presented with twitching symptoms at PND18.70+/-0.50, which was significantly lower than the onset of all treatment groups (p<0.001). The IP, Weekly, and GALC groups had twitching onsets at PND29.88+/-0.92, 34.25+/-0.45, and 30.20+/-0.96; the ICV group exhibited this symptom at PND24.57+/-1.56, which was significantly earlier than all IP treated groups (p<0.01-0.001). Additionally, the twitching onset of the Weekly group was delayed compared to the mice receiving only a single IP injection (p<0.05; Fig. 1C).

The wire hang (Fig. 1D), hind stride length (Fig. 1E), and rearing abilities (Fig. 1F) were tested for each mouse group. For the wire hang study, there was a significant group x time interaction (p=0.0004), and no differences were found between any mouse group until PND24, at which time the Twi mice had significantly less ability

to hang on the wire compared to all treated mice (p<0.01). The Weekly and IP groups had improved wire hang ability compared to the Twi mice at all time points between PND26 and PND38. Compared to the Twi mice, the GALC group also had improved hanging ability for all time points, except PND28 and PND38, In contrast, the ICV mice showed no wire hang improvement after PND26. Additionally, the ICV group had decreased hanging ability compared to the IP, Weekly, and GALC groups at PND34-38 (Fig. 1D). The group x time interaction was also significant for the hind stride study (p<0.0001). The hind stride of the WT was significantly greater than the Twi and IP-treated twitcher mice starting at PND28, and it was not until PND32 that the ICV group differed from the WT mice. Interestingly, the Weekly and GALC groups had similar hind stride lengths compared to the WT group until PND38. The Twi mice had shorter hind stride lengths compared to the GALC group at PND24-38 and the Weekly group at PND34. Furthermore, the GALC and Weekly groups were shown to have longer hind stride lengths compared to the IP group at PND24 and PND34, respectively.

Twi animals demonstrated deficient locomotor activity during the test period, and this deficit was rescued by IP, Weekly, and GALC treatments. Specifically, the Twi mice had significantly fewer rearing (protected and unprotected) v. WT (p<0.001) at PND23-29. The Weekly and GALC treated mice showed improved vertical rearing compared to Twi mice (p<0.05; Fig. 1F). As shown by the representative track visualizations (Fig. 2A), the Twi mice showed marked thigmotaxis for all weeks and minimal movement in the arena after PND30. In contrast, all IP-treated mice showed striking differences from both the ICV and Twi mouse groups. The improvements seen in the track densities were further supported by improvements in the total distance traveled (F(5, 60)=2.76, p<0.026), average velocity (F(5, 60)=2.76, p<0.02), and time spent in the inner zone (F(5, 60)=6.21, p<0.0001) at PND23-29 in

Peripheral Stem Cell Therapy in GLD Mice

9

the Weekly and GALC mouse groups (Fig. 2B-C). Comparing angular velocity over three weeks for all mouse groups, significant differences in group X time interaction (F(10, 144)=2.41, p=0.01) were found with all groups compared to WT mice at PND30-35 (Fig. 2D; p<0.01-0.001). The Twi group was the only twitcher cohort that had increased angular velocity compared to the WT mice at PND23-29 (p<0.001), and both the GALC and IP treatments significantly corrected this behavior at PND30-35 (p<0.01), although not to the baseline level.

Histological analysis of the sciatic nerve The H&E sciatic nerve images were analyzed with Fiji/Image J software to quantify the

number of total cells (i.e., cells of area 5 µm2)

and the number of large cells (i.e., cells of area 40 µm2) per field at 400X magnification [22].

Treatment had a significant effect on both the total number of cells (F(5, 152)=63.57, p<0.0001) and large cells (F(5, 152)=44.26, p<0.0001) in the sciatic nerve. These large cells (Fig. 3A, white arrow) were positive for F4/80, a macrophage surface marker (data not shown). Compared to the Twi group, the IP, Weekly, and GALC treatments led to a significant (p<0.001) decrease in total cell number (Fig. 3B). Additionally, all IP-treated groups had a significant (p<0.001) decrease in peripheral globoid cell formation (Fig. 3C). The sciatic nerves from the ICV mice had globoid cell formation and inflammatory infiltration levels that were not different from the Twi group. Interestingly, only the GALC mice had infiltrating and globoid cell numbers that were corrected to WT levels. Taken together, these data showed significant improvements in sciatic nerve pathophysiology for all three IP-treated groups compared to the ICV treated and control Twi mice (Fig. 3A-C).

TUNEL staining of the brain and sciatic nerve for detection of apoptosis The presence of darkly stained nuclei in the Twi and ICV H&E stained sciatic nerves indicated that apoptotic or necrotic cells were likely present in these samples. To investigate this

further, unstained sciatic nerves were tested for apoptosis or necrosis activity using the TUNEL assay. All sciatic nerves, regardless of mouse group, displayed similar positive staining when exposed to DNase treatment as a positive control (Fig. 4A). The percentage of apoptotic cells based on the total number of DAPI+ cells was quantified using Image J for all experimental samples. There was no treatment effect on the percentage of TUNEL+ cells (F(5, 29)=2.19, p=0.07). However, by visual inspection, the experimental WT and IP-treated samples showed minimal TUNEL activity in any section (Fig. 4A-B). Conversely, the Twi showed DNA fragmentation in every section tested, as evidenced by the presence of 17.4%+8.4%apoptotic or necrotic cells detected by the TUNEL assay per field (Fig.4B).

GALC protein levels and enzymatic activity in the brain The presence of GALC protein was determined for all treatment groups using Western blotting (Fig. 5A). Each lane in Fig. 5A represents the brain lysates of three mice pooled together, and the results showed GALC specific bands at 50 and 30 KDa for the WT, GALC, and Weekly mouse groups. Comparisons of the actin-normalized 30 KDa band intensities for GALC and Weekly groups showed 1.8- and 1.3-fold increases in GALC protein levels, compared to WT levels (Fig. 5B). The normalized bands for the ICV and IP groups were 21% and 10% the intensity of the 30 KDa WT band.

A radioactive GalCer assay was performed to assess the activity of the GALC enzyme (Fig. 5C). This assay involved incubating a 3H-GalCersubstrate in the presence of brain lysate, and any detected radioactivity represented the presence of cleaved substrate and, thus, active GALC enzyme. For each treatment group, brain lysates of 3-5 mice were tested, and it was found that the GALC and Weekly groups had GALC activity that was 4.4% and 2.4% of the WT activity. The ICV, IP, and Twi groups had negligible enzymatic activities, with 0.5%, 0.0%, and 0.0% of WT activity, respectively.

Peripheral Stem Cell Therapy in GLD Mice

10

To confirm that the transduced and untransduced eGFPTgBMSCs produce functional GALC enzyme, three experiments were performed. First, the cells were expanded in culture and

stained with a modified -galactosidase assay

[48]. The GALC-BMSCs had functional -galactosidase enzyme capable of cleaving the X-gal substrate in over 90% of the cells (Fig. 5D); as expected, the staining of the untransduced BMSCs was of a lesser intensity and in fewer cells. Second, the cells were lysed and immediately incubated with 4Mu-glycosides.

After normalization of the -gal levels to both -

hex and -gal, it was found that the GALC-

BMSCs had -gal activity that was 2.4 times greater than that of the untransduced cells (Fig. 5E). Furthermore, the enzymatic activities were calculated at either pH 4.0 or pH 7.0 (Fig. 5F), and the ratio of activities of the GALC-BMSCs at 4.0 vs. 7.0 was 1.8-fold greater than the BMSCs. Lastly, the radioactive 3H-GalCer assay was performed on eGFPTgBMSCs and GALC-transduced eGFPTgBMSCs to determine the activity of the GALC enzyme present in these cell types (Fig. 5G). The GALC-BMSCs had a GALC activity of 2.9 nmol of cleaved substrate per hr per 1,000,000 cells, whereas the untransduced BMSCs cleaved the substrate at 1.05 nmol/h/1,000,000 cells.

Detection of green fluorescence protein in stem cell injected mice by Western blotting and immunofluorescence To determine if eGFP was present in brain lysates at the time of euthanasia (PND33-48), Western blotting was performed using a GFP specific antibody for all treatment groups (Fig. 6A). A specific GFP band at 27 KDa was detected for the ICV and WT mice that received eGFPTgBMSCs administered directly to the brain. None of the IP treatment groups showed evidence of GFP signal in their respective lanes, indicating an absence of the cells at this terminal time point. Liver and kidney lysates were also tested using Western blotting with the same GFP specific antibody. No treatment group had detectable GFP protein in the kidney lysates, and the Weekly group was the only treatment group

to have a GFP specific band in the liver samples (data not shown).

GFP+ BMSCs were identified in frozen sections of the ICV mouse brains by IHC using a GFP antibody. These cells were found in the hindbrain, peri-ventricular region, and forebrain; however, there was no evidence of co-localization when stained for NeuN, GFAP, or S-100 (Fig, 6B-D).

To determine if an antibody response formed in the mice against the GFP+ BMSCs after multiple injections, the sera of the Twi, IP, and Weekly mice were incubated as the primary antibody solution for Western blotting of purified GFP (Fig. 6E). When compared to a control lane, which had been incubated with a known antibody against GFP, the Weekly treatment group had a similar band (white arrows), although of a lesser intensity.

DISCUSSION

To develop a safer, more effective therapy for GLD patients, this study investigates the therapeutic effects of central and peripheral MSC injections in the twitcher mouse. Affected mice were grouped according to the injection conditions (e.g., administration route, injection frequency, and cell transduction status) used to administer the BMSCs, and all treated cohorts were compared to Twi mice for assessment of therapeutic effectiveness. The effects of 1) increased injection frequency or 2) transduced BMSCs overexpressing the GALC enzyme have not, to our knowledge, been assessed as potential treatments. For this, stem cell persistence, brain and sciatic nerve pathology, levels of GALC, and GLD-related symptoms were assessed for all mice to compare various transplantation conditions. This study demonstrates that twitcher mice receiving weekly IP injections of BMSCs or a single IP injection of GALC-transduced BMSCs have significant improvements in lifespan, twitching onset and severity, body weight, motor function (e.g., wire hang ability, hind stride length), general motor activity (e.g.,

Peripheral Stem Cell Therapy in GLD Mice

11

velocity, distance traveled), and sciatic nerve histopathology (e.g., decreased globoid cell formation and apoptotic cell death) vs. Twi mice and/or ICV mice. Such results corroborate recent studies demonstrating the therapeutic potential of peripherally injected MSCs in LSDs [41, 51-53] and indicate a promising future for MSC therapy in the GLD patient.

Previous studies in our laboratory by Ripoll, etal. [43], Wicks, et al. [54], and Scruggs, et al.[13] have shown modest improvements in the twitcher phenotype and/or lifespan after suboptimal doses of BMSCs were injected ICV, intrastriatally, or IP, respectively. In an effort to further improve the outcomes of MSC therapy, the number of cells administered in this study was increased 5-fold, yet all treatment groups

received doses (e.g., 1x107 cells/kg) that are comparable to those currently used in MSC-related clinical trials. Increasing the ICV administered cell number from 40,000 [43] to 200,000 total BMSCs achieved greater persistence and distribution of the stem cells in the twitcher brain, as was evident by the presence of GFP+ cells in all ICV injected mice even at the time of euthanasia. Furthermore, the ICV mice had a significant increase in median lifespan compared to mice receiving fewer cells [43]. However, the increased cell persistence in the brain did not result in increased GALC activity or transdifferentiation of the BMSCs to neurons, astrocytes, or glial cells; such results are consistent with recent findings that BMSCs provide neuroprotection through anti-inflammatory and immunomodulatory actions, not cell or enzyme replacement [38, 43].

The majority of GLD symptoms in twitcher mice result from severe PNS inflammation and demyelination [12, 55, 56]. Twitcher mice display symptoms related to widespread peripheral neuropathy, including severe muscle wasting, hind limb weakness, gait abnormalities, spatial abnormalities, and decreased rearing [13, 14]. Similarly, GLD patients often show decreased reflexes and limb strength, muscle atrophy, early coordination disturbance, poor

head control, spasticity, and slowed motor nerve conduction velocity [7, 8, 57, 58]. Such symptoms result from the extensive myelin sheath degeneration, axonal death, and inflammatory infiltration (e.g., globoid cells, eosinophils, mast cells) present in peripheral nerves. Striking differences were detected between the sciatic nerves of the ICV and IP-treated mice. These results suggest that MSC do not exert an anti-inflammatory on peripheral nerves when administered directly into the brain. Globoid cell and infiltrating cells in the PNS may be a main contributor to the poor motor function of the ICV mice. Conversely, peripheral MSC injections have been shown to be both anti-inflammatory and anti-apoptotic in the PNS, which is supported by the motor function results. In support of this conclusion, the GALC sciatic nerves showed no histological evidence of inflammation. The most significant motor function improvements were seen after IP administration of BMSCs or GALC-BMSCs, yet the results showed that the IP injected cells did not home to the CNS. Recent studies have tracked MSCs after systemic administration in neurodegenerative and autoimmune mouse models, and all have demonstrated similar beneficial effects after MSC injection due to the cells locating to secondary lymphatic organs and mediating immune responses [59-63]. Therefore, it is likely that the BMSCs and GALC-BMSCs of this study were capable of exerting potent systemic effects after IP administration by homing to sites, such as lymph nodes, and acting as immune effectors.

Many LSDs with peripheral manifestations have been successfully treated with ERT [33, 64], and recent studies using the twitcher mouse show a beneficial, albeit transient, effect on the presentation and neuropathology of the affected mice [26]. In an effort to treat twitcher mice by providing a more stable supply of GALC enzyme, BMSCs that were engineered to overexpress GALC were delivered or the frequency of delivery of naïve BMSC was increased. Despite increased GALC protein in the brain and modest improvement in lifespan,

Peripheral Stem Cell Therapy in GLD Mice

12

enzyme activity assays showed no detectable GALC activity in the brains of the treated mice. These findings suggest that the GALC enzyme, over time, is unable to cleave its substrates that have accumulated in the brain at the time of euthanasia; this result was consistent with the observation that all treated mice died before PND50. The combination of increased GALC protein, active GALC enzyme in the BMSCs, and absent GALC activity in the transplanted brain may indicate that the detected enzyme, although initially active, is not transported by cross-correction mechanisms to inside the affected brain cells [65]. Such faulty localization has been documented previously [30], and this may cause the enzyme to become partially degraded, incompletely processed, incorrectly folded, and/or irreversibly bound to an inhibitor.Our laboratory has ongoing experiments aimed to understand this phenomenon for improving cross-correction. Based on the improved motor function and pathology in the GALC and Weekly groups, it is expected that weekly administration of GALC-transduced BMSCs would increase the amount of GALC protein in the PNS and CNS more than either group alone. This novel treatment group will be tested in our laboratory upon production of adequate numbers of GALC-transduced BMSCs, and based on our current results we expect that this treatment will improve the cross-correction of GALC, lifespan, and quality of life for twitcher mice.

BMSCs are exceptional candidates for restoration of damaged tissue [35, 66] and have the potential, if injected under optimized conditions, to significantly alleviate peripheral inflammation and apoptotic cell death in the twitcher mouse. Specifically, BMSCs secrete a variety of cytokines and factors that have paracrine activities [38], promote differentiation of the host’s stem cells, and modulate immune reactions [35, 37, 66-69]. Although these beneficial effects are clearly demonstrated in the present study using peripheral MSC transplantation, all treated mice eventually died in a moribund state after a rapid decline in health. Based on the GALC activity results, it is

likely that the psychosine levels in the brain were not decreased, leading to increased oxidative stress and apoptotic death of myelin producing cells and other brain cells [6, 65]. Taken together, these results suggest a promising future for peripherally administered MSCs if combined with a therapy that increases GALC levels in the brain.

CONCLUSION

Transplantation of BMSCs requires extensive optimization in animal models before being implemented in clinical trials as a novel therapy for neurodegenerative diseases. In order to understand the mechanism of action of these cells and to improve their therapeutic efficacy in the twitcher mouse model of GLD, this study aimed to increase MSC distribution, persistence, and anti-inflammatory effects through optimization of the cell type, injection frequency, and administration route. Significant improvements in lifespan, motor function, and GLD-related symptoms after IP transplantation of MSCs were associated with a significant reduction in inflammation in the PNS. The results of this study indicate that repeated injections of BMSCs or GALC-BMSCs in the periphery could be an effective adjunct therapy for HSCT to decrease risks GVHD and improve PNS outcomes of patients affected with GLD.

ACKNOWLEDGMENTS

The authors thank Siddharth Gaikwad, Jeremy Green, Evan Kyzar, and the Neurophenotyping Core at Tulane University School of Medicine for their instruction, advice, and suggestions regarding the video recording studies. We also thank the veterinary and vivarial staff of the TUHSC Animal Facility for the daily care of the mice. The authors also thank Michelle E. Scarritt for help with the immunofluorescence experiments, Kathleen MP Imhof for assistance with the motor function tests, Krystal Brown for help with the deconvolution microscope, Dr. Xavier Alvarez of the Tulane National Primate Research Center (TNPRC) for assistance in

Peripheral Stem Cell Therapy in GLD Mice

13

analyzing the histological samples using the confocal microscope, Alan Tucker for performing flow cytometry for characterization of the MSC populations, and Dina Gaupp and Claire Llamas in the Tulane Histology Core for performance of the histological staining for this study. The multiplex plates were run by the staff at the Pathogen Detection and Quantification Core (PDQC) at the TNPRC under grant number RR00164. The GALC activity assay was

performed with the assistance of Dr. Yu-Teh Li of the Tulane University Biochemistry Department. The research was supported by grant R21-NS059665 from the National Institutes of Neurological Disorders and Stroke (NIH) and by Tulane University.

Disclosure of potential conflicts of interest No potential conflicts of interest were disclosed.

REFERENCES

1. Hawkins-Salsbury JA, Reddy AS, Sands MS: Combination therapies for lysosomal storage disease: is the whole greater than the sum of its parts? Hum Mol Genet 2011, 20:R54-60.

2. Heese BA: Current strategies in the management of lysosomal storage diseases. Semin Pediatr Neurol 2008, 15:119-126.

3. Hodges BL, Cheng SH: Cell and gene-based therapies for the lysosomal storage diseases. Curr Gene Ther 2006, 6:227-241.

4. Suzuki K, Suzuki Y: Globoid cell leucodystrophy (Krabbe's disease): deficiency of galactocerebroside beta-galactosidase. Proc Natl Acad Sci U S A 1970, 66:302-309.

5. Kolodny EH: Globoid Leukodystrophy. Elsevier Science; 1996.

6. Sakai N: Pathogenesis of leukodystrophy for Krabbe disease: molecular mechanism and clinical treatment. Brain Dev 2009, 31:485-487.

7. Duffner PK, Barczykowski A, Jalal K, Yan L, Kay DM, Carter RL: Early infantile Krabbe disease: results of the World-Wide Krabbe Registry. Pediatr Neurol 2011, 45:141-148.

8. Barone R, Brühl K, Stoeter P, Fiumara A, Pavone L, Beck M: Clinical and neuroradiological findings in classic infantile and late-onset globoid-cell leukodystrophy (Krabbe disease). Am J Med Genet 1996, 63:209-217.

9. Costello DJ, Eichler AF, Eichler FS: Leukodystrophies: classification, diagnosis, and treatment. Neurologist 2009, 15:319-328.

10. Castelvetri LC, Givogri MI, Zhu H, Smith B, Lopez-Rosas A, Qiu X, van Breemen R, Bongarzone ER: Axonopathy is a compounding factor in the pathogenesis of Krabbe disease. Acta Neuropathol 2011, 122:35-48.

11. Duchen LW, Eicher EM, Jacobs JM, Scaravilli F, Teixeira F: Hereditary leucodystrophy in the mouse: the new mutant twitcher. Brain 1980, 103:695-710.

12. Smith B, Galbiati F, Cantuti-Castelvetri L, Givogri MI, Lopez-Rosas A, Bongarzone ER: Peripheral

neuropathy in the Twitcher mouse involves the activation of axonal caspase 3. ASN Neuro 2011.

13. Scruggs BA, Bowles AC, Zhang X, Semon JA, Kyzar EJ, Myers L, Kalueff AV, Bunnell BA: High-throughput screening of stem cell therapy for globoid cell leukodystrophy using automated neurophenotyping of twitcher mice. Behav Brain Res 2012, 236C:35-47.

14. Olmstead CE: Neurological and neurobehavioral development of the mutant 'twitcher' mouse. Behav Brain Res 1987, 25:143-153.

15. Kobayashi T, Yamanaka T, Jacobs JM, Teixeira F, Suzuki K: The Twitcher mouse: an enzymatically authentic model of human globoid cell leukodystrophy (Krabbe disease). Brain Res 1980, 202:479-483.

16. McGraw P, Liang L, Escolar M, Mukundan S, Kurtzberg J, Provenzale JM: Krabbe disease treated with hematopoietic stem cell transplantation: serial assessment of anisotropy measurements--initial experience. Radiology 2005, 236:221-230.

17. Prasad VK, Kurtzberg J: Cord blood and bone marrow transplantation in inherited metabolic diseases: scientific basis, current status and future directions. Br J Haematol 2010, 148:356-372.

18. Escolar ML, Poe MD, Provenzale JM, Richards KC, Allison J, Wood S, Wenger DA, Pietryga D, Wall D, Champagne M, et al: Transplantation of umbilical-cord blood in babies with infantile Krabbe's disease. N Engl J Med 2005, 352:2069-2081.

19. Hoogerbrugge PM, Poorthuis BJ, Romme AE, van de Kamp JJ, Wagemaker G, van Bekkum DW: Effect of bone marrow transplantation on enzyme levels and clinical course in the neurologically affected twitcher mouse. J Clin Invest 1988, 81:1790-1794.

20. Luzi P, Rafi MA, Zaka M, Rao HZ, Curtis M, Vanier MT, Wenger DA: Biochemical and pathological evaluation of long-lived mice with globoid cell leukodystrophy after bone marrow transplantation. Mol Genet Metab 2005, 86:150-159.

21. Wenger DA, Rafi MA, Luzi P, Datto J, Costantino-Ceccarini E: Krabbe disease: genetic aspects and progress toward therapy. Mol Genet Metab 2000, 70:1-9.

22. Lin D, Donsante A, Macauley S, Levy B, Vogler C, Sands MS: Central nervous system-directed AAV2/5-

Peripheral Stem Cell Therapy in GLD Mice

14

mediated gene therapy synergizes with bone marrow transplantation in the murine model of globoid-cell leukodystrophy. Mol Ther 2007, 15:44-52.

23. Qin EY, Hawkins-Salsbury JA, Jiang X, Reddy AS, Farber NB, Ory DS, Sands MS: Bone marrow transplantation increases efficacy of central nervous system-directed enzyme replacement therapy in the murine model of globoid cell leukodystrophy. Mol Genet Metab 2012, 107:186-196.

24. Martin PL, Carter SL, Kernan NA, Sahdev I, Wall D, Pietryga D, Wagner JE, Kurtzberg J: Results of the cord blood transplantation study (COBLT): outcomes of unrelated donor umbilical cord blood transplantation in pediatric patients with lysosomal and peroxisomal storage diseases. Biol Blood Marrow Transplant 2006, 12:184-194.

25. Reddy A, Wozniak D, Farber N, Dearborn J, Fowler S, Sands M: Bone Marrow Transplantation Alters the Tremor Phenotype in the Murine Model of Globoid-Cell Leukodystrophy. Journal of Clinical Medicine 2012, 1:1-14.

26. Lee WC, Courtenay A, Troendle FJ, Stallings-Mann ML, Dickey CA, DeLucia MW, Dickson DW, Eckman CB: Enzyme replacement therapy results in substantial improvements in early clinical phenotype in a mouse model of globoid cell leukodystrophy. Faseb J 2005, 19:1549-1551.

27. Lee WC, Tsoi YK, Troendle FJ, DeLucia MW, Ahmed Z, Dicky CA, Dickson DW, Eckman CB: Single-dose intracerebroventricular administration of galactocerebrosidase improves survival in a mouse model of globoid cell leukodystrophy. Faseb J 2007, 21:2520-2527.

28. Biswas S, LeVine SM: Substrate-reduction therapy enhances the benefits of bone marrow transplantation in young mice with globoid cell leukodystrophy. Pediatr Res 2002, 51:40-47.

29. Galbiati F, Givogri MI, Cantuti L, Rosas AL, Cao H, van Breemen R, Bongarzone ER: Combined hematopoietic and lentiviral gene-transfer therapies in newborn Twitcher mice reveal contemporaneous neurodegeneration and demyelination in Krabbe disease. J Neurosci Res 2009, 87:1748-1759.

30. Lin D, Fantz CR, Levy B, Rafi MA, Vogler C, Wenger DA, Sands MS: AAV2/5 vector expressing galactocerebrosidase ameliorates CNS disease in the murine model of globoid-cell leukodystrophy more efficiently than AAV2. Mol Ther 2005, 12:422-430.

31. Dolcetta D, Perani L, Givogri MI, Galbiati F, Amadio S, Del Carro U, Finocchiaro G, Fanzani A, Marchesini S, Naldini L, et al: Design and optimization of lentiviral vectors for transfer of GALC expression in Twitcher brain. J Gene Med 2006, 8:962-971.

32. Kagitani-Shimono K, Mohri I, Fujitani Y, Suzuki K, Ozono K, Urade Y, Taniike M: Anti-inflammatory therapy by ibudilast, a phosphodiesterase inhibitor, in demyelination of twitcher, a genetic demyelination model. J Neuroinflammation 2005, 2:10.

33. Beck M: Therapy for lysosomal storage disorders. IUBMB Life 2010, 62:33-40.

34. Le Blanc K, Ringden O: Mesenchymal stem cells: properties and role in clinical bone marrow transplantation. Curr Opin Immunol 2006, 18:586-591.

35. Hardy SA, Maltman DJ, Przyborski SA: Mesenchymal stem cells as mediators of neural differentiation. Curr Stem Cell Res Ther 2008, 3:43-52.

36. Kassis I, Grigoriadis N, Gowda-Kurkalli B, Mizrachi-Kol R, Ben-Hur T, Slavin S, Abramsky O, Karussis D: Neuroprotection and immunomodulation with mesenchymal stem cells in chronic experimental autoimmune encephalomyelitis. Arch Neurol 2008, 65:753-761.

37. Uccelli A, Moretta L, Pistoia V: Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008, 8:726-736.

38. Bunnell BA, Betancourt AM, Sullivan DE: New concepts on the immune modulation mediated by mesenchymal stem cells. Stem Cell Res Ther 2010, 1:34.

39. Uccelli A, Benvenuto F, Laroni A, Giunti D: Neuroprotective features of mesenchymal stem cells. Best Pract Res Clin Haematol 2011, 24:59-64.

40. Ringden O, Le Blanc K: Mesenchymal stem cells for treatment of acute and chronic graft-versus-host disease, tissue toxicity and hemorrhages. Best Pract Res Clin Haematol 2011, 24:65-72.

41. Giordano A, Galderisi U, Marino IR: From the laboratory bench to the patient's bedside: an update on clinical trials with mesenchymal stem cells. J Cell Physiol 2007, 211:27-35.

42. Ringden O, Uzunel M, Rasmusson I, Remberger M, Sundberg B, Lonnies H, Marschall HU, Dlugosz A, Szakos A, Hassan Z, et al: Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation 2006, 81:1390-1397.

43. Ripoll CB, Flaat M, Klopf-Eiermann J, Fisher-Perkins JM, Trygg CB, Scruggs BA, McCants ML, Leonard HP, Lin AF, Zhang S, et al: Mesenchymal-Lineage Stem Cells Have Pronounced Anti-Inflammatory Effects in the Twitcher Mouse Model of Krabbe's Disease. Stem Cells 2011, 29:67-77.

44. Terrell KA, Rasmussen TA, Trygg C, Bunnell BA, Buck WR: Molecular beacon genotyping for globoid cell leukodystrophy from hair roots in the twitcher mouse and rhesus macaque. J Neurosci Methods 2007, 163:60-66.

45. Ripoll CB, Bunnell BA: Comparative characterization of mesenchymal stem cells from eGFP transgenic and non-transgenic mice. BMC Cell Biol 2009, 10:3.

46. Kutner RH, Zhang XY, Reiser J: Production, concentration and titration of pseudotyped HIV-1-based lentiviral vectors. Nat Protoc 2009, 4:495-505.

47. Dolcetta D, Perani L, Givogri MI, Galbiati F, Orlacchio A, Martino S, Roncarolo MG, Bongarzone E: Analysis of galactocerebrosidase activity in the

Peripheral Stem Cell Therapy in GLD Mice

15

mouse brain by a new histological staining method. JNeurosci Res 2004, 77:462-464.

48. Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, Preibisch S, Rueden C, Saalfeld S, Schmid B, et al: Fiji: an open-source platform for biological-image analysis. Nat Methods 2012, 9:676-682.

49. Raghavan S, Krusell A: Optimal assay conditions for enzymatic characterization of homozygous and heterozygous twitcher mouse. Biochim Biophys Acta 1986, 877:1-8.

50. Potier M, Mameli L, Bélisle M, Dallaire L, Melançon SB: Fluorometric assay of neuraminidase with a sodium (4-methylumbelliferyl-alpha-D-N-acetylneuraminate) substrate. Anal Biochem 1979, 94:287-296.

51. Miranda CO, Teixeira CA, Liz MA, Sousa VF, Franquinho F, Forte G, Di Nardo P, Pinto-Do OP, Sousa MM: Systemic Delivery of Bone Marrow-Derived Mesenchymal Stromal Cells Diminishes Neuropathology in a Mouse Model of Krabbe's Disease. Stem Cells 2011, 29:1738-1751.

52. Jin HK, Carter JE, Huntley GW, Schuchman EH: Intracerebral transplantation of mesenchymal stem cells into acid sphingomyelinase-deficient mice delays the onset of neurological abnormalities and extends their life span. J Clin Invest 2002, 109:1183-1191.

53. Lee H, Lee JK, Min WK, Bae JH, He X, Schuchman EH, Bae JS, Jin HK: Bone marrow-derived mesenchymal stem cells prevent the loss of Niemann-Pick type C mouse Purkinje neurons by correcting sphingolipid metabolism and increasing sphingosine-1-phosphate. Stem Cells 2010, 28:821-831.

54. Wicks SE, Londot H, Zhang B, Dowden J, Klopf-Eiermann J, Fisher-Perkins JM, Trygg CB, Scruggs BA, Zhang X, Gimble JM, et al: Effect of intrastriatal mesenchymal stromal cell injection on progression of a murine model of Krabbe disease. Behav Brain Res 2011, 225:415-425.

55. Kagitani-Shimono K, Mohri I, Yagi T, Taniike M, Suzuki K: Peripheral neuropathy in the twitcher mouse: accumulation of extracellular matrix in the endoneurium and aberrant expression of ion channels. Acta Neuropathol 2008, 115:577-587.

56. Suzuki K, Taniike M: Murine model of genetic demyelinating disease: the twitcher mouse. Microsc Res Tech 1995, 32:204-214.

57. Zafeiriou DI, Anastasiou AL, Michelakaki EM, Augoustidou-Savvopoulou PA, Katzos GS, Kontopoulos EE: Early infantile Krabbe disease: deceptively normal magnetic resonance imaging and serial neurophysiological studies. Brain Dev 1997, 19:488-491.

58. Sasaki M, Sakuragawa N, Takashima S, Hanaoka S, Arima M: MRI and CT findings in Krabbe disease. Pediatr Neurol 1991, 7:283-288.

59. Madec AM, Mallone R, Afonso G, Abou Mrad E, Mesnier A, Eljaafari A, Thivolet C: Mesenchymal stem cells protect NOD mice from diabetes by inducing regulatory T cells. Diabetologia 2009, 52:1391-1399.

60. Chiesa S, Morbelli S, Morando S, Massollo M, Marini C, Bertoni A, Frassoni F, Bartolomé ST, Sambuceti G, Traggiai E, Uccelli A: Mesenchymal stem cells impair in vivo T-cell priming by dendritic cells. Proc Natl Acad Sci U S A 2011, 108:17384-17389.

61. Chamberlain G, Fox J, Ashton B, Middleton J: Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells 2007, 25:2739-2749.

62. Zappia E, Casazza S, Pedemonte E, Benvenuto F, Bonanni I, Gerdoni E, Giunti D, Ceravolo A, Cazzanti F, Frassoni F, et al: Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood 2005, 106:1755-1761.

63. Von Lüttichau I, Notohamiprodjo M, Wechselberger A, Peters C, Henger A, Seliger C, Djafarzadeh R, Huss R, Nelson PJ: Human adult CD34- progenitor cells functionally express the chemokine receptors CCR1, CCR4, CCR7, CXCR5, and CCR10 but not CXCR4. Stem Cells Dev 2005, 14:329-336.

64. Cox TM: Gaucher disease: clinical profile and therapeutic developments. Biologics 2010, 4:299-313.

65. White AB, Galbiati F, Givogri MI, Lopez Rosas A, Qiu X, van Breemen R, Bongarzone ER: Persistence of psychosine in brain lipid rafts is a limiting factor in the therapeutic recovery of a mouse model for Krabbe disease. J Neurosci Res 2011, 89:352-364.

66. Chen X, Armstrong MA, Li G: Mesenchymal stem cells in immunoregulation. Immunol Cell Biol 2006, 84:413-421.

67. Croitoru-Lamoury J, Williams KR, Lamoury FM, Veas LA, Ajami B, Taylor RM, Brew BJ: Neural transplantation of human MSC and NT2 cells in the twitcher mouse model. Cytotherapy 2006, 8:445-458.

68. Gimble JM, Katz AJ, Bunnell BA: Adipose-derived stem cells for regenerative medicine. Circ Res 2007, 100:1249-1260.

69. Koc ON, Peters C, Aubourg P, Raghavan S, Dyhouse S, DeGasperi R, Kolodny EH, Yoseph YB, Gerson SL, Lazarus HM, et al: Bone marrow-derived mesenchymal stem cells remain host-derived despite successful hematopoietic engraftment after allogeneic transplantation in patients with lysosomal and peroxisomal storage diseases. Exp Hematol 1999, 27:1675-1681.

Peripheral Stem Cell Therapy in GLD Mice

16

Figure 1. Weight, lifespan and motor function. A. A Kaplan-Meier survival curve for the untreated and BMSC treated twitcher groups. B. Body weight was measured starting at PND 16. C. Twitching severity was assessed using the conventional twitching clinical scoring systems. D. Hind leg strength was assessed using the wire hang test. E. Hind stride length was measured for assessment of gait. F. Comparative analysis of the total number of rears performed during PND23-29. G. Table of different mouse groups tested in this study. The genotype, wild-type (GALC+/+) or twitcher (GALC-/-), of each mouse group is listed. The number of animals per group and the details of each treatment are provided. Significant differences are denoted by ***P<0.001 vs. WT and #P<0.05 vs. Twi mice. All tests were performed for all mouse groups three times per week. ICV, intracerebroventricular; IP, intraperitoneal.

Peripheral Stem Cell Therapy in GLD Mice

17

Figure 2. Monitoring disease progression using automated phenotyping. A. Representative images of track visualizations of the six mouse groups over 4 consecutive weeks, PND16-22, 23-29, 30-35, and 36-45. For any given time period, a mouse was recorded once for 5 min, using Ethovision XT7 to generate a track. B. The total distance traveled in 5 min was compared across all mouse groups at PND23-29. C. Time spent in the inner zone over 5 min was compared for all mouse groups at PND23-29. D. The average angular velocities at 3 different time points were compared across mouse groups. Significant differences are denoted as *P<0.05; **P<0.01; ***P<0.001 vs. WT mice and ##P<0.01 vs. Twi mice. Comparisons between mouse groups were made using one-way ANOVA or two-way ANOVA with repeated measures and Bonferroni’s post-hoc testing.

Peripheral Stem Cell Therapy in GLD Mice

18

Figure 3. Histological staining of sciatic nerves. A. Sciatic nerves from WT, treated Twi, and untreated Twi mice (N=3 mice/group; 10 sections/mouse) were processed using Hematoxylin and Eosin (H&E). The scale bar represents 50 µm for all larger images (400X), and each smaller image (1600X) is a four-fold magnification of the larger image (upper panel). B. The total number of cells in the binary images (lower panel) was calculated per field at 400X using ImageJ software. (C). The total number of globoid cells was calculated per field at 400X using ImageJ software. Comparisons across all mouse groups were made using one-way ANOVA and Bonferroni’s post-hoc testing.***P<0.001vs. WT, ##P<0.01; ###P<0.0001 vs. Twi mice, and +++P<0.0001 vs. ICV mice.

Peripheral Stem Cell Therapy in GLD Mice

19

Figure 4. Detection of TUNEL+ cells in the sciatic nerve. A. Sciatic nerves (N=3 mice/group, 3 sections/mouse) were deparaffinized and tested for apoptotic events (green) using an in situ cell death/fluorescein detection kit. For each nerve tested, a positive control was performed using DNase to test efficiency of the TUNEL assay (upper panel). B. The percentage of TUNEL+ events per total DAPI+ cells was calculated using ImageJ software. Comparisons across all mouse groups using one-way ANOVA and Bonferroni’s post-hoc testing revealed no significant differences between any treatment group.

Peripheral Stem Cell Therapy in GLD Mice

20

Figure 5. GALC protein levels and enzymatic activity assays. A. Brain protein samples were tested by Western blot, and each lane represents the brain lysates of three mice pooled for protein analysis. Bands present at 30 and 50 KDa denoted GALC enzyme, and actin protein was detected at 42 KDa for each sample on a simultaneously run blot. B. Comparisons of the actin-normalized 30 KDa band intensities for GALC and Weekly groups. C. GALC activity was determined in the brain lysates of all mouse groups (N=3-5 mice/group) using the radioactive 3H-GalCer assay. D. X-gal staining was

performed using a modified -Galactosidase assay for BMSC and GALC-transduced cells. E-F. Celllysates were incubated with 4MU-glycosides to determine the activity of three enzymes: beta-galactosidase, alpha-galactosidase, and beta-hexosaminidase at pH 4.0 or 7.0. G. The radioactive 3H-GalCer assay was performed on lysates of GALC-transduced and untransduced BMSCs

Peripheral Stem Cell Therapy in GLD Mice

21

Figure 6. GFP protein and antibody detection in MSC-injected mice. A. eGFP was detected in brain lysates by Western blotting as a band at 27 KDa. B. eGFP+ cells (green) were located in deparaffinized sections of injected brains at euthanasia and immunostained with NeuN (red), C. GFAP (red), or D.S100 (red) to investigate cellular differentiation along neural, astrocyte, or glial lineages, respectively. Nuclei were stained with To-Pro-3 (blue). E. Sera of the Twi, IP, and Weekly mice were incubated as the primary antibody solution for Western blotting of purified GFP.