Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Blood ; 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861668

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapies have demonstrated transformative efficacy in treating B-cell malignancies. However, high cost and manufacturing complexities hinder their widespread use. To overcome these hurdles, we have developed the VivoVecTM platform, a lentiviral vector capable of generating CAR T-cells in vivo. Here we describe the incorporation of T cell activation and costimulatory signals onto the surface of VivoVecTM particles (VVPs) in the form of a multi-domain fusion protein and show enhanced in vivo transduction and improved CAR-T cell antitumor functionality. Furthermore, in the absence of lymphodepleting chemotherapy, administration of VVPs into non-human primates resulted in the robust generation of anti-CD20 CAR T-cells and the complete depletion of B cells for more than 10 weeks. These data validate the VivoVecTM platform in a translationally relevant model and support its transition into human clinical testing, offering a paradigm shift in the field of CAR T-cell therapies.

2.
Blood Adv ; 6(12): 3557-3568, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35427415

ABSTRACT

A safe, effective, and inclusive gene therapy will significantly benefit a large population of patients with hemophilia. We used a minimally invasive transcutaneous ultrasound-mediated gene delivery (UMGD) strategy combined with microbubbles (MBs) to enhance gene transfer into 4 canine livers. A mixture of high-expressing, liver-specific human factor VIII (hFVIII) plasmid and MBs was injected into the hepatic vein via balloon catheter under fluoroscopy guidance with simultaneous transcutaneous UMGD treatment targeting a specific liver lobe. Therapeutic levels of hFVIII expression were achieved in all 4 dogs, and hFVIII levels were maintained at a detectable level in 3 dogs throughout the 60-day experimental period. Plasmid copy numbers correlated with hFVIII antigen levels, and plasmid-derived messenger RNA (mRNA) was detected in treated livers. Liver transaminase levels and histology analysis indicated minimal liver damage and a rapid recovery after treatment. These results indicate that liver-targeted transcutaneous UMGD is promising as a clinically feasible therapy for hemophilia A and other diseases.


Subject(s)
Hemophilia A , Hemostatics , Animals , Dogs , Factor VIII/genetics , Factor VIII/therapeutic use , Gene Transfer Techniques , Genetic Therapy/methods , Hemophilia A/genetics , Hemophilia A/therapy , Hemophilia A/veterinary , Humans , Liver/metabolism
4.
Transplantation ; 105(5): 1008-1016, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33065723

ABSTRACT

BACKGROUND: Chronic graft-versus-host disease (GVHD) is a significant cause of morbidity and mortality in transplant patients. We have previously shown that 3 doses of an anti-inducible costimulator (ICOS) mAb transiently ameliorated symptoms and extended survival of dogs affected by chronic GVHD over that of control dogs. The purpose of this study was to specifically correlate changes in T-cell populations in the peripheral blood with anti-ICOS treatment and chronic GVHD progression and regression to reach a better understanding of the mechanism of the disease and prioritize future studies. METHODS: Peripheral blood cells from canines transplanted with DLA-mismatched bone marrow and peripheral blood mononuclear cells to generate chronic GVHD were analyzed by flow cytometry using a panel of antibodies specific to helper and cytolytic T cells. RESULTS: Chronic GVHD was specifically associated with an increase in CD4+ICOS+ cells, ICOS+ cells expressing IL-17A, and CD8+ cells generating granzyme B. Treatment with anti-ICOS mAb at onset of chronic GVHD symptoms specifically targeted IL-17A+-expressing cells, transiently relieved symptoms, and lengthened survival but was unable to reduce the percentage of CD8+ T-cells expressing granzyme B. CONCLUSIONS: These studies suggested a role for both CD4+ and CD8+ T cells in pathogenesis of chronic GVHD in the canine model. We propose that future studies should focus on further extending survival by developing a treatment that would control both CD4+ and CD8+ T cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Graft vs Host Disease/drug therapy , Immunosuppressive Agents/pharmacology , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , Interleukin-17/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chronic Disease , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Dogs , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Granzymes/metabolism , Inducible T-Cell Co-Stimulator Protein/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism
5.
Transplant Direct ; 6(12): e632, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33225057

ABSTRACT

BACKGROUND: Complementary, marrow donor-derived peripheral blood T-lymphocyte infusions enable consistent hematopoietic engraftment in lethally irradiated dog leukocyte antigen (DLA)-haploidentical littermate recipients, but at the cost of severe graft versus host disease (GVHD). Here, we explored whether CD94-selected and in vitro-expanded natural killer (NK) cells could be substituted for T-lymphocytes for enhancing marrow engraftment without causing severe GVHD. METHODS: Five dogs were conditioned with 700 cGy total body irradiation followed by infusion of DLA-haploidentical donor marrow and CD94-selected, in vitro-expanded NK cells. NK cells were infused at a median of 140 000 (range 78 000-317 000) cells/kg. RESULTS: Four dogs rejected their marrow grafts, whereas 1 dog fully engrafted and developed GVHD. We observed an increase in peripheral blood NK cells after infusion of CD94-selected, ex vivo-expanded NK in 2 dogs. Peripheral blood lymphocyte counts peaked at day 7 or 8 posttransplant in the 4 rejecting dogs, whereas in the fully engrafted dog, lymphocyte counts remained stable at suboptimal levels. CONCLUSIONS: Our study indicates NK cells can be expanded in vitro and safely infused into DLA-haploidentical recipients. Within the range of CD94-selected and expanded cells infused we concluded that they failed to both uniformly promote engraftment and avert GVHD.

6.
Vet Immunol Immunopathol ; 211: 10-18, 2019 May.
Article in English | MEDLINE | ID: mdl-31084888

ABSTRACT

Natural killer (NK) cells are non-T, non-B lymphocytes are part of the innate immune system and function without prior activation. The human NK cell surface determinant, CD94, plays a critical role in regulation of NK cell activity as a heterodimer with NKG2 subclasses. Canine NK cells are not as well defined as the human and murine equivalents, due in part to the paucity of reagents specific to cell surface markers. Canines possess NK/NKT cells that have similar morphological characteristics to those found in humans, yet little is known about their functional characteristics nor of cell surface expression of CD94. Here, we describe the development and function of a monoclonal antibody (mAb) to canine (ca) CD94. Freshly isolated canine CD94+ cells were CD3+/-, CD8+/-, CD4-, CD21-, CD5low, NKp46+, and were cytotoxic against a canine target cell line. Anti-caCD94 mAb proved useful in enriching NK/NKT cells from PBMC for expansion on CTAC feeder cells in the presence of IL-2 and IL-15. The cultured cells were highly cytolytic with co-expression of NKp46 and reduced expression of CD3. Transmission electron microscopy revealed expanded CD94+ lymphocytes were morphologically large granular lymphocytes with large electron dense granules. Anti-caCD94 (mAb) can serve to enrich NK/NKT cells from dog peripheral blood for ex vivo expansion for HCT and is a potentially valuable reagent for studying NK/NKT regulation in the dog.


Subject(s)
Antibodies, Monoclonal/immunology , Dogs/immunology , NK Cell Lectin-Like Receptor Subfamily D/immunology , Animals , Antibody Specificity/immunology , Cloning, Molecular , Female , Flow Cytometry/veterinary , Killer Cells, Natural/immunology , Male , Mice/immunology , Natural Killer T-Cells/immunology , Polymerase Chain Reaction/veterinary
7.
ILAR J ; 59(3): 263-275, 2018 12 31.
Article in English | MEDLINE | ID: mdl-30010833

ABSTRACT

Since its inception in the 1950s, hematopoietic cell transplantation (HCT) has become a highly effective clinical treatment for malignant and nonmalignant hematological disorders. This milestone in cancer therapy was only possible through decades of intensive research using murine and canine animal models that overcame what appeared in the early days to be insurmountable obstacles. Conditioning protocols for tumor ablation and immunosuppression of the recipient using irradiation and chemotherapeutic drugs were developed in mouse and dog models as well as postgrafting immunosuppression methods essential for dependable donor cell engraftment. The random-bred canine was particularly important in defining the role of histocompatibility barriers and the development of the nonmyeloablative transplantation procedure, making HCT available to elderly patients with comorbidities. Two complications limit the success of HCT: disease relapse and graft versus host disease. Studies in both mice and dogs have made significant progress toward reducing and to some degree eliminating patient morbidity and mortality associated with both disease relapse and graft versus host disease. However, more investigation is needed to make HCT more effective, safer, and available as a treatment modality for other non-life-threatening diseases such as autoimmune disorders. Here, we focus our review on the contributions made by both the murine and canine models for the successful past and future development of HCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Models, Animal , Animals , Dogs , Graft vs Host Disease/therapy , Mice
8.
Biol Blood Marrow Transplant ; 24(1): 50-54, 2018 01.
Article in English | MEDLINE | ID: mdl-28958896

ABSTRACT

In murine model systems inducible costimulator (ICOS) signaling has been implicated in the formation of chronic graft-versus-host disease (GVHD). Previously, we showed that chronic GVHD can be reproducibly produced in the dog hematopoietic cell transplantation (HCT) model and that ICOS expression is upregulated on T cells in dogs with chronic GVHD. The goal of the present study was to determine whether administration of a short course of anti-canine ICOS mAb could alter the rapid and progressive course of chronic GVHD. Five dogs underwent HCT from dog leukocyte antigen mismatched unrelated donors after total body irradiation. Postgrafting immunosuppression consisted of methotrexate (days 1, 3, 6, and 11) and cyclosporine (days -1 through 78). Anti-ICOS mAb (3 injections, 72 hours apart) was administered upon diagnosis of GVHD. One dog failed to respond to anti-ICOS mAb therapy and succumbed to chronic GVHD in a time course similar to control untreated dogs. Overall, anti-ICOS-treated dogs experienced a significant prolongation in survival from the time of diagnosis of chronic GVHD compared with control dogs. Within the limitations of the number of study dogs we suggest that a short course of anti-ICOS mAb may be useful in the treatment of chronic canine GVHD.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Graft vs Host Disease/therapy , Inducible T-Cell Co-Stimulator Protein/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Surface , Disease Models, Animal , Dogs , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation , Immunosuppression Therapy/methods , Survival Rate , Treatment Outcome
9.
Biol Blood Marrow Transplant ; 23(3): 420-427, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28013013

ABSTRACT

In long-term survivors of allogeneic hematopoietic cell transplantation (HCT), chronic graft-versus-host disease (GVHD) is the major cause of morbidity and mortality and a major determinant of quality of life. Chronic GVHD responds poorly to current immunosuppressive drugs, and while T cell depletion may be preventive, this gain is offset by increased relapse rates. A significant impediment to progress in treating chronic GVHD has been the limitations of existing animal models. The goal of this study was to develop a reproducible comprehensive model of chronic GVHD in the dog. Ten recipient dogs received 920 cGy total body irradiation, infusion of marrow, and an infusion of buffy coat cells from a dog leukocyte antigen (DLA)-mismatched unrelated donor. Postgrafting immunosuppression consisted of methotrexate (days 1, 3, 6, 11) and cyclosporine. The duration of cyclosporine administration was limited to 80 days instead of the clinically used 180 days. This was done to contain costs, as chronic GVHD was expected to develop at earlier time points. All recipients were given ursodiol for liver protection. One dog had graft failure and 9 dogs showed stable engraftment. Eight of the 9 developed de novo chronic GVHD. Dogs progressed with clinical signs of chronic GVHD over a period of 43 to 164 (median, 88) days after discontinuation of cyclosporine. Target organs showed the spectrum of chronic GVHD manifestations that are typically seen clinically. These included lichenoid changes of the skin, fasciitis, ocular involvement (xerophthalmia), conjunctivitis, bronchiolitis obliterans, salivary gland involvement, gingivitis, esophageal involvement, and hepatic involvement. Peripheral blood lymphocyte surface antigen expression of CD28 and inducible costimulator was elevated in dogs with GHVD compared with those in normal dogs, but not significantly so. Serum levels of IL-8 and monocyte chemotactic protein-1 in GVHD-affected dogs at time of euthanasia were elevated, whereas levels of IL-15 were depressed compared with those in normal dogs. Results indicate that the canine model is well suited for future studies aimed at preventing or treating chronic GVHD.


Subject(s)
Bone Marrow Transplantation/adverse effects , Disease Models, Animal , Graft vs Host Disease , Transplantation Immunology , Animals , Blood Buffy Coat/transplantation , Bone Marrow Transplantation/methods , Chronic Disease , Dogs , Graft Survival , Histocompatibility , Immunosuppressive Agents/therapeutic use , Lymphocyte Depletion , Transplantation Conditioning/methods , Transplantation, Homologous , Unrelated Donors , Whole-Body Irradiation
10.
Methods Mol Biol ; 1460: 209-21, 2016.
Article in English | MEDLINE | ID: mdl-27492175

ABSTRACT

Comparing the functional myogenic potential of various human cell populations is an important step in the preclinical evaluation of cell transplantation as a means to treat human muscle disease and degeneration. Culture systems allow one to gage the potential of cell populations to proliferate and undergo myogenic differentiation under specific conditions. An in vivo assay evaluates the ability of cells to differentiate and generate muscle fibers within a natural environment, and importantly, evaluates the potential of donor cells to reconstitute the satellite cell niche. In this chapter, we describe a technique for isolating mononuclear cells from human muscle samples, and a method of xenotransplantation for assessing functional myogenic potential in vivo. Briefly, cell populations are injected into the pre-irradiated and regenerating muscle of immunodeficient mice. The injected muscle is frozen at specific time points after injection and cryosections analyzed by immunostaining. The number of human dystrophin-expressing fibers and the number of Pax7(+) human lamin A/C(+) nuclei are determined, which provides a quantitative method of comparing the in vivo functional potential of cell populations.


Subject(s)
Cell Differentiation , Muscle Development , Muscle Fibers, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Stem Cells/cytology , Animals , Cell Separation/methods , Graft Survival , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Regeneration , Stem Cell Niche , Stem Cell Transplantation , Stem Cells/metabolism
11.
Front Genet ; 6: 59, 2015.
Article in English | MEDLINE | ID: mdl-25750654

ABSTRACT

Skeletal muscle is a striated tissue composed of multinucleated fibers that contract under the control of the somatic nervous system to direct movement. The stem cells of skeletal muscle, known as satellite cells, are responsible for muscle fiber growth, turnover, and regeneration. Satellite cells are activated and proliferate in response to stimuli, and simplistically, have two main fates-to repopulate the satellite cell niche, or differentiate to regenerate or repair muscle fibers. However, the ability to regenerate muscle and replace lost myofibers declines with age. This loss of function may be a result of extrinsic changes in the niche, such as alterations in signaling or modifications to the extracellular matrix. However, intrinsic epigenetic changes within satellite cells may also affect cell fate and cause a decline in regenerative capacity. This review will describe the mechanisms that regulate cell fate decisions in adult skeletal muscle, and how changes during aging affect muscle fiber turnover and regeneration.

13.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2C.4, 2013.
Article in English | MEDLINE | ID: mdl-23661246

ABSTRACT

Studies in mice showed tremendous promise for the eventual clinical utility of myoblast transplantation to treat human muscular dystrophies. Initial attempts to translate the murine studies to humans, however, were not successful, due in part to limited engraftability of expanded donor myoblasts. Conventionally, muscle cells have been cultured on collagen-coated tissue culture-treated polystyrene. However, this promotes lineage progression and differentiation of cells, which limits engraftment potential. This unit describes the isolation of canine muscle-derived cells, ex vivo expansion of cells on plates coated with a modified Notch ligand, and the xenotransplant method used to evaluate engraftment potential. Activation of Notch signaling in freshly isolated canine muscle-derived cells with Delta-1(ext)-IgG inhibits myogenic differentiation, and maintains cells earlier in myogenic lineage progression. Delta-1(ext)-IgG-expanded cells engraft into the regenerating muscle of NOD/SCID mice more effectively than control cells expanded on human IgG, as evidenced by a significant increase in the number of muscle fibers expressing canine dystrophin in recipient murine muscle. Therefore, this protocol provides the basis for further developing culture conditions for ex vivo expansion of donor muscle cells for transplant.


Subject(s)
Cell Culture Techniques/methods , Muscle Cells/cytology , Muscle Cells/transplantation , Animals , Cell Count , Cell Proliferation , Cell Separation , Cells, Cultured , Cryoultramicrotomy , Dogs , Humans , Immunoglobulin G/metabolism , Injections , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/transplantation , Mice , Mice, SCID
14.
Stem Cells ; 30(10): 2212-20, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22865615

ABSTRACT

Transplantation of myogenic stem cells possesses great potential for long-term repair of dystrophic muscle. However, a single donor muscle biopsy is unlikely to provide enough cells to effectively transplant the muscle mass of a patient affected by muscular dystrophy. Expansion of cells ex vivo using traditional culture techniques significantly reduces engraftment potential. We hypothesized that activation of Notch signaling during ex vivo expansion would maintain donor cell engraftment potential. In this study, we expanded freshly isolated canine muscle-derived cells on tissue culture plates coated with Delta-1(ext) -IgG to activate Notch signaling or with human IgG as a control. A model of canine-to-murine xenotransplantation was used to quantitatively compare canine muscle cell engraftment and determine whether engrafted donor cells could function as satellite cells in vivo. We show that Delta-1(ext) -IgG inhibited differentiation of canine muscle-derived cells and increased the level of genes normally expressed in myogenic precursors. Moreover, cells expanded on Delta-1(ext) -IgG resulted in a significant increase in the number of donor-derived fibers, as compared to cells expanded on human IgG, reaching engraftment levels similar to freshly isolated cells. Importantly, cells expanded on Delta-1(ext) -IgG engrafted to the recipient satellite cell niche and contributed to further regeneration. A similar strategy of expanding human muscle-derived cells on Notch ligand might facilitate engraftment and muscle regeneration for patients affected with muscular dystrophy.


Subject(s)
Graft Survival , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism , Animals , Cell Communication , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dogs , Humans , Immunoglobulin G/pharmacology , Mice , Mice, SCID , Muscle Cells/cytology , Muscle Cells/transplantation , Muscle, Skeletal/cytology , Regeneration , Signal Transduction , Species Specificity , Stem Cell Transplantation , Stem Cells/cytology , Transplantation, Heterologous
15.
Skelet Muscle ; 2(1): 6, 2012 05 19.
Article in English | MEDLINE | ID: mdl-22541644

ABSTRACT

BACKGROUND: Mice lacking MyoD exhibit delayed skeletal muscle regeneration and markedly enhanced numbers of satellite cells. Myoblasts isolated from MyoD-/- myoblasts proliferate more rapidly than wild type myoblasts, display a dramatic delay in differentiation, and continue to incorporate BrdU after serum withdrawal. METHODS: Primary myoblasts isolated from wild type and MyoD-/- mutant mice were examined by microarray analysis and further characterized by cell and molecular experiments in cell culture. RESULTS: We found that NF-κB, a key regulator of cell-cycle withdrawal and differentiation, aberrantly maintains nuclear localization and transcriptional activity in MyoD-/- myoblasts. As a result, expression of cyclin D is maintained during serum withdrawal, inhibiting expression of muscle-specific genes and progression through the differentiation program. Sustained nuclear localization of cyclin E, and a concomitant increase in cdk2 activity maintains S-phase entry in MyoD-/- myoblasts even in the absence of mitogens. Importantly, this deficit was rescued by forced expression of IκBαSR, a non-degradable mutant of IκBα, indicating that inhibition of NF-κB is sufficient to induce terminal myogenic differentiation in the absence of MyoD. CONCLUSION: MyoD-induced cytoplasmic relocalization of NF-κB is an essential step in linking cell-cycle withdrawal to the terminal differentiation of skeletal myoblasts. These results provide important insight into the unique functions of MyoD in regulating the switch from progenitor proliferation to terminal differentiation.

16.
Skelet Muscle ; 2(1): 4, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-22340947

ABSTRACT

BACKGROUND: Transplantation of myogenic stem cells possesses great potential for long-term repair of dystrophic muscle. In murine-to-murine transplantation experiments, CXCR4 expression marks a population of adult murine satellite cells with robust engraftment potential in mdx mice, and CXCR4-positive murine muscle-derived SP cells home more effectively to dystrophic muscle after intra-arterial delivery in mdx5cv mice. Together, these data suggest that CXCR4 plays an important role in donor cell engraftment. Therefore, we sought to translate these results to a clinically relevant canine-to-canine allogeneic transplant model for Duchenne muscular dystrophy (DMD) and determine if CXCR4 is important for donor cell engraftment. METHODS: In this study, we used a canine-to-murine xenotransplantation model to quantitatively compare canine muscle cell engraftment, and test the most effective cell population and modulating factor in a canine model of DMD using allogeneic transplantation experiments. RESULTS: We show that CXCR4 expressing cells are important for donor muscle cell engraftment, yet FACS sorted CXCR4-positive cells display decreased engraftment efficiency. However, diprotin A, a positive modulator of CXCR4-SDF-1 binding, significantly enhanced engraftment and stimulated sustained proliferation of donor cells in vivo. Furthermore, the canine-to-murine xenotransplantation model accurately predicted results in canine-to-canine muscle cell transplantation. CONCLUSIONS: Therefore, these results establish the efficacy of diprotin A in stimulating muscle cell engraftment, and highlight the pre-clinical utility of a xenotransplantation model in assessing the relative efficacy of muscle stem cell populations.

17.
Dev Cell ; 18(4): 662-74, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20412780

ABSTRACT

Recent studies have demonstrated that MyoD initiates a feed-forward regulation of skeletal muscle gene expression, predicting that MyoD binds directly to many genes expressed during differentiation. We have used chromatin immunoprecipitation and high-throughput sequencing to identify genome-wide binding of MyoD in several skeletal muscle cell types. As anticipated, MyoD preferentially binds to a VCASCTG sequence that resembles the in vitro-selected site for a MyoD:E-protein heterodimer, and MyoD binding increases during differentiation at many of the regulatory regions of genes expressed in skeletal muscle. Unanticipated findings were that MyoD was constitutively bound to thousands of additional sites in both myoblasts and myotubes, and that the genome-wide binding of MyoD was associated with regional histone acetylation. Therefore, in addition to regulating muscle gene expression, MyoD binds genome wide and has the ability to broadly alter the epigenome in myoblasts and myotubes.


Subject(s)
Gene Expression Regulation , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Amino Acid Motifs , Animals , Binding Sites , Cell Differentiation , Cell Line , Fibroblasts/metabolism , Genome , Histones/chemistry , Mice , Models, Biological , Muscle Fibers, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis
18.
Mol Ther ; 16(7): 1340-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18500253

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is the most common and severe form of muscular dystrophy in humans. The goal of myogenic stem cell transplant therapy for DMD is to increase dystrophin expression in existing muscle fibers and to provide a source of stem cells for future muscle generation. Although syngeneic myogenic stem cell transplants have been successful in mice, allogeneic transplants of myogenic stem cells were ineffective in several human trials. To determine whether allogeneic muscle progenitor cells can be successfully transplanted in an immune-tolerant recipient, we induced immune tolerance in two DMD-affected (cxmd) dogs through hematopoietic cell transplantation (HCT). Injection of freshly isolated muscle-derived cells from the HCT donor into either fully or partially chimeric xmd recipients restored dystrophin expression up to 6.48% of wild-type levels, reduced the number of centrally located nuclei, and improved muscle structure. Dystrophin expression was maintained for at least 24 weeks. Taken together, these data indicate that immune tolerance to donor myoblasts provides an important platform from which to further improve myoblast transplantation, with the goal of restoring dystrophin expression to patients with DMD.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunosuppression Therapy/methods , Muscular Dystrophy, Duchenne/surgery , Myoblasts/transplantation , Animals , Dogs , Immune Tolerance , Myoblasts/immunology
19.
Mol Cell Biol ; 26(15): 5771-83, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16847330

ABSTRACT

The MyoD family of basic helix-loop-helix transcription factors function as heterodimers with members of the E-protein family to induce myogenic gene activation. The E-protein HEB is alternatively spliced to generate alpha and beta isoforms. While the function of these molecules has been studied in other cell types, questions persist regarding the molecular functions of HEB proteins in skeletal muscle. Our data demonstrate that HEB alpha expression remains unchanged in both myoblasts and myotubes, whereas HEB beta is upregulated during the early phases of terminal differentiation. Upon induction of differentiation, a MyoD-HEB beta complex bound the E1 E-box of the myogenin promoter leading to transcriptional activation. Importantly, forced expression of HEB beta with MyoD synergistically lead to precocious myogenin expression in proliferating myoblasts. However, after differentiation, HEB alpha and HEB beta synergized with myogenin, but not MyoD, to activate the myogenin promoter. Specific knockdown of HEB beta by small interfering RNA in myoblasts blocked differentiation and inhibited induction of myogenin transcription. Therefore, HEB alpha and HEB beta play novel and central roles in orchestrating the regulation of myogenic factor activity through myogenic differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , MyoD Protein/metabolism , Myoblasts/physiology , Protein Isoforms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cell Proliferation , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/physiology , MyoD Protein/genetics , Myoblasts/cytology , Myogenin/genetics , Myogenin/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic
20.
J Cell Biol ; 166(6): 865-76, 2004 Sep 13.
Article in English | MEDLINE | ID: mdl-15364961

ABSTRACT

To investigate the requirement for pRb in myogenic differentiation, a floxed Rb allele was deleted either in proliferating myoblasts or after differentiation. Myf5-Cre mice, lacking pRb in myoblasts, died immediately at birth and exhibited high numbers of apoptotic nuclei and an almost complete absence of myofibers. In contrast, MCK-Cre mice, lacking pRb in differentiated fibers, were viable and exhibited a normal muscle phenotype and ability to regenerate. Induction of differentiation of Rb-deficient primary myoblasts resulted in high rates of apoptosis and a total inability to form multinucleated myotubes. Upon induction of differentiation, Rb-deficient myoblasts up-regulated myogenin, an immediate early marker of differentiation, but failed to down-regulate Pax7 and exhibited growth in low serum conditions. Primary myoblasts in which Rb was deleted after expression of differentiated MCK-Cre formed normal multinucleated myotubes that did not enter S-phase in response to serum stimulation. Therefore, Rb plays a crucial role in the switch from proliferation to differentiation rather than maintenance of the terminally differentiated state.


Subject(s)
Cell Differentiation , Muscle Fibers, Skeletal/physiology , Myoblasts/physiology , Retinoblastoma Protein/physiology , Adenoviridae/genetics , Alleles , Animals , Apoptosis/genetics , Cell Division , Cells, Cultured , Culture Media, Serum-Free , Gene Deletion , Gene Expression Regulation, Developmental , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Mice, Transgenic , Muscle, Skeletal/cytology , Myoblasts/cytology , Myogenin/physiology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...