Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Int Urol Nephrol ; 50(12): 2153-2165, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30324580

ABSTRACT

PURPOSE: The purpose of the study was to assess safety and efficacy of autologous muscle derived cells for urinary sphincter repair (AMDC-USR) in female subjects with predominant stress urinary incontinence. METHODS: A randomized, double-blind, multicenter trial examined intra-sphincteric injection of 150 × 106 AMDC-USR versus placebo in female subjects with stress or stress predominant, mixed urinary incontinence. AMDC-USR products were generated from vastus lateralis needle biopsies. Subjects were randomized 2:1 to receive AMDC-USR or placebo and 1:1 to receive 1 or 2 treatments (6 months after the first). Primary outcome was composite of ≥ 50% reduction in stress incontinence episode frequency (IEF), 24-h or in-office pad weight tests at 12 months. Other outcome data included validated subject-recorded questionnaires. Subjects randomized to placebo could elect to receive open-label AMDC-USR treatment after 12 months. Subject follow-up was up to 2 years. RESULTS: AMDC-USR was safe and well-tolerated with no product-related serious adverse events or discontinuations due to adverse events. Interim analysis revealed an unexpectedly high placebo response rate (90%) using the composite primary outcome which prevented assessment of treatment effect as designed and thus enrollment was halted at 61% of planned subjects. Post hoc analyses suggested that more stringent endpoints lowered placebo response rates and revealed a possible treatment effect. CONCLUSIONS: Although the primary efficacy finding was inconclusive, these results inform future trial design of AMDC-USR to identify clinically meaningful efficacy endpoints based on IEF reduction, understanding of placebo response rate, and refinement of subject selection criteria to more appropriately align with AMDC-USR's proposed mechanism of action.


Subject(s)
Muscle Cells/transplantation , Urethra/surgery , Urinary Incontinence, Stress/therapy , Adult , Aged , Aged, 80 and over , Double-Blind Method , Dysuria/etiology , Female , Humans , Middle Aged , Nausea/etiology , Pain/etiology , Quadriceps Muscle/cytology , Severity of Illness Index , Surveys and Questionnaires , Transplantation, Autologous/adverse effects , Treatment Outcome , Urinary Tract Infections/etiology , Young Adult
2.
Neurourol Urodyn ; 37(8): 2932-2937, 2018 11.
Article in English | MEDLINE | ID: mdl-29953660

ABSTRACT

BACKGROUND AND AIMS: Stakeholders from around the world came together to address the unmet needs of underactive bladder (UAB) at the 3rd International Congress for Underactive Bladder. METHODS: The main recommendation from the regulatory working group is a need for a meeting of UAB stakeholders and regulatory agencies including the FDA to discuss guidance for regulatory trial design for devices, drugs, and/or biologics for UAB. RESULTS: The following issues to be discussed and agreed upon for UAB trials: 1) Appropriate inclusion and exclusion criteria. 2) Should residual urine volume be the primary outcome parameter and how often should it be measured? 3) Are there secondary measures that should have a place in UAB trials, such as change in the number of catheterizations, quality of life measures, etc.? 4) Use and format of bladder voiding and catheterization diary for trials. 5) Define role and technique of urodynamics in UAB trials. Are urodynamics required to monitor, and possibly exclude, individuals with high pressure voiding induced by bladder prokinetic therapies? 6) Development and use of UAB questionnaires. DISCUSSION AND CONCLUSION: The UAB regulatory working group recognizes the path forward should include engaging the FDA and other regulatory organizations that may harmonize and formalize guidance for regulatory trial designs for therapeutics for UAB.


Subject(s)
Technology Assessment, Biomedical/methods , Urinary Bladder, Underactive/therapy , Bethanechol/therapeutic use , Clinical Trials as Topic , Electric Stimulation Therapy , Humans , Muscarinic Agonists/therapeutic use , Quality of Life , Research Design , Surveys and Questionnaires , Treatment Outcome , United States , United States Food and Drug Administration , Urinary Bladder, Underactive/psychology , Urinary Catheterization/statistics & numerical data , Urodynamics
3.
Int Urol Nephrol ; 47(3): 465-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25663053

ABSTRACT

We conducted the first-regenerative medicine cellular therapy for underactive bladder (UAB) in an FDA-approved, compassionate-use IND trial to evaluate treatment safety and potential clinical efficacy of autologous muscle-derived stem cells (AMDC) on a patient with UAB. No study-related adverse events or side effects were reported. In the 1-year follow-up period, the subject denied any gross hematuria, urgency, frequency or infection. A reduction in maximum cystometric capacity from 844 to 663 mL was observed, and the patient was able to void small amounts but continues to require self-catheterization 1 year after AMDC injection. Intradetrusor injection of AMDC is safe, minimally invasive and a promising treatment option for the UAB.


Subject(s)
Myoblasts, Skeletal/transplantation , Urinary Bladder Diseases/therapy , Aged , Chronic Disease , Compassionate Use Trials , Humans , Injections, Intramuscular/methods , Intermittent Urethral Catheterization , Male , Pilot Projects , Urinary Bladder , Urinary Bladder Diseases/complications , Urinary Retention/etiology
4.
Urology ; 73(2): 437-41, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18701145

ABSTRACT

OBJECTIVES: Lidocaine cytotoxicity has been reported in some cell types, which could affect its use as a local anesthetic in cell-based therapy. We evaluated the in vitro and in vivo effect of lidocaine on rat muscle-derived progenitor cells (MDCs). METHODS: MDCs were isolated from rat skeletal muscle and purified using the preplate technique. For in vitro tests, the MDCs underwent either 2 hours of, or continuous, exposure to lidocaine (50 microM-5 mM). After 72 hours of incubation, cell viability was measured using the methylthiazololetetrazolium assay. For the in vivo tests, periurethral injection of either phosphate-buffered saline, MDCs (1 x 10(6) cells/20 microL), or 2% lidocaine plus MDCs was performed in pudendal nerve-transected rats. The leak point pressure (LPP) was measured at 4 weeks after the injection. RESULTS: Lidocaine concentrations of < or = 500 microM had no effect on MDCs with continuous exposure. MDCs in 1 mM lidocaine showed decreased survival and no MDCs in 5 mM lidocaine survived. With a 2-hour exposure, only MDCs in the 5-mM lidocaine group showed decreased survival. Rats with nerve transection and phosphate-buffered saline injection showed significantly lower LPPs than the controls. The LPP was restored to a significantly greater level after MDCs only or lidocaine plus MDC injection. No statistically significant difference in LPP restoration was found between the MDC-only and lidocaine plus MDC injections. CONCLUSIONS: Cytotoxicity to lidocaine was minimal at a physiologic concentration in vitro. The functional recovery of LPP by MDC treatment was not affected by lidocaine preinfiltration. Taken together, our data indicate that lidocaine can be applied as a local anesthetic in periurethral MDC injection without decreasing the efficacy of the therapy.


Subject(s)
Anesthetics, Local/pharmacology , Lidocaine/pharmacology , Muscle Cells/drug effects , Muscle Cells/transplantation , Urinary Incontinence, Stress/surgery , Anesthetics, Local/adverse effects , Animals , Cells, Cultured , Female , Lidocaine/adverse effects , Rats , Rats, Sprague-Dawley
5.
Int Urogynecol J Pelvic Floor Dysfunct ; 19(9): 1229-34, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18421407

ABSTRACT

The physiological effects of human muscle-derived stem cell (MDSC) implantation on urethral smooth muscle function were investigated in pudendal nerve-transected nude rats with human MDSC (TM) or saline (TS) injection into the proximal urethra compared with sham-operated, saline-injected nude rats (SS). Leak point pressure (LPP) before and after hexamethonium application, which can block autonomic efferent nerves, and proximal urethral contractile responses to carbachol and phenylephrine in muscle strip study were examined 6 weeks after the implantation. There was no significant difference between the LPPs in SS and TM. Following hexamethonium application, the LPP in TM was, however, significantly decreased compared with SS. The contractile responses to phenylephrine, but not to carbachol, in TM were significantly increased compared with SS and TS. These results suggest that the restorative effects of MDSCs are mediated by autonomic nerves and that increased sensitivity of alpha(1)-adrenoceptors may be related to restore the deficient urethral function.


Subject(s)
Muscle, Smooth/physiology , Stem Cell Transplantation , Urethra/physiology , Urinary Incontinence/therapy , Animals , Female , Humans , Immunohistochemistry , Muscle Contraction/drug effects , Muscle Contraction/physiology , Rats , Rats, Nude , Rats, Sprague-Dawley
6.
Expert Opin Biol Ther ; 7(10): 1483-6, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17916041

ABSTRACT

The suburethral sling procedures, such as transvaginal tape (TVT), have recently gained popularity for the treatment of stress urinary incontinence (SUI). This TVT procedure can reinforce the weakness of pelvic floor muscles but urethral sphincter deficiency remains. Adult stem cell injection therapy for SUI has recently been at the forefront of the repair of deficient urethral function. Muscle-derived stem cells and adipose-derived stem cells are regarded as candidates for the treatment of SUI because these stem cells can be easily obtained in large quantities under local anesthesia, they have the potential to undergo long-term proliferation, self-renewal and multipotent differentiation, and can serve as a vehicle of releasing neurotrophins, such as nerve growth factor, to repair the deficient urethra.


Subject(s)
Adipocytes/transplantation , Adult Stem Cells/transplantation , Mesenchymal Stem Cell Transplantation , Myocytes, Smooth Muscle/transplantation , Urinary Incontinence, Stress/surgery , Urologic Surgical Procedures , Adipocytes/metabolism , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Humans , Myocytes, Smooth Muscle/metabolism , Nerve Growth Factors/metabolism , Urinary Incontinence, Stress/metabolism , Urinary Incontinence, Stress/pathology
7.
Curr Urol Rep ; 8(5): 373-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17880836

ABSTRACT

The promise of stem cell therapy for the treatment of stress urinary incontinence is that transplanted stem cells may undergo self-renewal and potential multipotent differentiation, leading to urethral sphincter regeneration. Cell-based therapies are most often associated with the use of autologous multipotent stem cells, such as bone marrow cells. However, harvesting bone marrow stromal stem cells is difficult, painful, and may yield low numbers of stem cells. Alternatively, autologous adult stem cells, such as muscle-derived stem cells, can be obtained in large quantities and with minimal discomfort. Not all cells and cellular therapies are the same, however, and proper placement of cells into target structures may be critical to eventual treatment success. In particular, restoration and repair of the damaged urethral sphincter is crucial to maintain urinary continence because active urethral closure is largely mediated by pudendal nerves that innervate the striated muscles and rhabdosphincter of the middle urethra.


Subject(s)
Adult Stem Cells/transplantation , Stem Cell Transplantation/methods , Urinary Incontinence/surgery , Clinical Trials as Topic , Female , Follow-Up Studies , Humans , Male , Risk Assessment , Sensitivity and Specificity , Treatment Outcome , Urinary Incontinence/prevention & control , Urologic Surgical Procedures/methods
8.
Muscle Nerve ; 36(3): 391-3, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17617803

ABSTRACT

We investigated the use of human muscle-derived cells (hMDCs) for the treatment of stress urinary incontinence (SUI) in a nude rat model. hMDCs were isolated from adult skeletal muscle. Three groups of six animals consisting of controls, animals undergoing sciatic nerve transection (SNT) with periurethral sham-injection, and SNT with hMDCs (1 x 10(6) cells/20 microl saline) were utilized. Leak point pressure (LPP) was measured 4 weeks following injection. Bilateral SNT resulted in a significantly lower LPP that was significantly higher following hMDCs than sham injection. The results demonstrate the efficacy of human muscle cell therapy alone in improving physiologic outcomes in an animal model of SUI.


Subject(s)
Muscle Fibers, Skeletal/transplantation , Urinary Incontinence, Stress/therapy , Animals , Disease Models, Animal , Female , Humans , Injections , Rats , Rats, Nude , Urinary Incontinence, Stress/pathology
9.
Neurourol Urodyn ; 26(7): 966-71, 2007.
Article in English | MEDLINE | ID: mdl-17580339

ABSTRACT

AIMS: This review aims to discuss: 1) the neurophysiology, highlighting the importance of the middle urethra, and treatment of stress urinary incontinence (SUI); 2) current injectable cell sources for minimally-invasive treatment; and 3) the potential of muscle-derived stem cells (MDSCs) for the delivery of neurotrophic factors. METHODS: A PUB-MED search was conducted using combinations of heading terms: urinary incontinence, urethral sphincter, stem cells, muscle, adipose, neurotrophins. In addition, we will update the recent work from our laboratory. RESULTS: In anatomical and functional studies of human and animal urethra, the middle urethra containing rhabdosphincter, is critical for maintaining continence. Cell-based therapies are most often associated with the use of autologous multipotent stem cells, such as the bone marrow stromal cells. However, harvesting bone marrow stromal stem cells is difficult, painful, and may yield low numbers of stem cells upon processing. In contrast, alternative autologous adult stem cells such as MDSCs and adipose-derived stem cells can be easily obtained in large quantities and with minimal discomfort. Not all cellular therapies are the same, as demonstrated by the differences in safety and efficacy from muscle-sourced MDSCs versus myoblasts versus fibroblasts. CONCLUSIONS: Transplanted stem cells may have the ability to undergo self-renewal and multipotent differentiation, leading to sphincter regeneration. In addition, such cells may release, or be engineered to release, neurotrophins with subsequent paracrine recruitment of endogenous host cells to concomitantly promote a regenerative response of nerve-integrated muscle. The dawn of a new paradigm in the treatment of SUI may be near.


Subject(s)
Myoblasts/transplantation , Stem Cell Transplantation/trends , Urinary Incontinence, Stress/surgery , Animals , Humans , Myoblasts/pathology , Urethra/pathology , Urinary Incontinence, Stress/pathology , Urologic Surgical Procedures
10.
Urology ; 68(2): 449-54, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16904482

ABSTRACT

OBJECTIVES: To compare muscle-derived cells (MDCs) and fibroblasts with regard to their potential for restoration of urethral function on injection in a previously established animal model of stress urinary incontinence. METHODS: The animals were divided into four (dosage) or five (cell concentration) experimental groups: normal, nontreated controls (normal group) or bilateral sciatic nerve transection with either periurethral injection of saline (saline group), MDCs (MDC group), fibroblasts (fibroblast group), or MDC/fibroblast mixture (mixed group). At 4 weeks after injection, the leak point pressure (LPP) was measured and contractility testing and histologic analysis were performed. RESULTS: The histologic examination demonstrated muscular atrophy in the saline group and new striated muscle fibers at the sites of MDC injection in the MDC group, but not in the fibroblast group. Denervation of the urethra resulted in a significant decrease of maximal fast-twitch muscle contraction amplitude to only 9% of normal. MDC injection into the denervated urethra significantly improved the fast-twitch muscle contraction amplitude to 73% of normal. The LPP of the normal, saline, MDC, fibroblast, and mixed groups at 4 weeks after treatment was 43.3 +/- 2.5, 25.8 +/- 1.4, 38.2 +/- 4.2, 38.3 +/- 1.2, and 34.5 +/- 3.3 cm H2O, respectively. In the cell dosage experiment, the LPP increased with increases in the injected cell number. Evidence of obstruction was observed in the high-dose (1 x 10(7) cells) fibroblast group. CONCLUSIONS: Although both MDCs and fibroblast injection increased the LPP in a stress urinary incontinence rat model, only MDCs significantly improved urethral muscle strip contractility. Moreover, urinary retention developed with high-dose fibroblast injection, but not with MDC injection.


Subject(s)
Fibroblasts , Muscle Contraction , Stem Cells , Urinary Incontinence, Stress/physiopathology , Urinary Incontinence, Stress/therapy , Animals , Disease Models, Animal , Female , Injections , Rats , Rats, Sprague-Dawley
11.
Biomaterials ; 26(4): 443-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15275818

ABSTRACT

Bladder wall replacement remains a challenging problem for urological surgery due to leakage, infection, stone formation, and extensive time needed for tissue regeneration. To explore the feasibility of producing a more functional biomaterial for bladder reconstitution, we incorporated muscle-derived cells (MDC) into small intestinal submucosa (SIS) scaffolds. MDC were harvested from mice hindleg muscle, transfected with a plasmid encoding for beta-galactosidase, and placed into single-layer SIS cell culture inserts. Twenty-five MDC and/or SIS specimens were incubated at 37 degrees C for either 10 or 20 days. After harvesting, mechanical properties were characterized using biaxial testing, and the areal strain under 1 MPa peak stress used to quantify tissue compliance. Histological results indicated that MDC migrated throughout the SIS after 20 days. The mean (+/-SE) areal strain of the 0 day control group was 0.182 +/- 0.027 (n=5). After 10 days incubation, the mean (+/-SE) areal strain in MDC/SIS was 0.247 +/- 0.014 (n=5) compared to 10 day control SIS 0.200 +/- 0.024 (n=6). After 20 days incubation, the mean areal strain of MDC/SIS was 0.255 +/- 0.019 (n=5) compared to control SIS 0.170 +/- 0.025 (n=5). Both 10 and 20 days seeded groups were significantly different (p=0.027) than that of incubated SIS alone, but were not different from each other. These results suggest that MDC growth was supported by SIS and that initial remodeling of the SIS ECM had occurred within the first 10 days of incubation, but may have slowed once the MDC had grown to confluence within the SIS.


Subject(s)
Bioprosthesis , Guided Tissue Regeneration/methods , Intestinal Mucosa/physiology , Myoblasts/physiology , Tissue Engineering/methods , Urinary Bladder/growth & development , Animals , Biomechanical Phenomena/methods , Cell Differentiation , Cell Proliferation , Cells, Cultured , Compressive Strength , Elasticity , Feasibility Studies , Intestinal Mucosa/cytology , Intestinal Mucosa/transplantation , Intestine, Small/cytology , Intestine, Small/physiology , Mice , Myoblasts/cytology , Myoblasts/transplantation , Stress, Mechanical , Tensile Strength , Urinary Bladder/cytology , Urinary Bladder/surgery
12.
Am J Physiol Endocrinol Metab ; 287(5): E857-62, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15226098

ABSTRACT

Intramyocellular lipid (IMCL) has been associated with insulin resistance. However, an association between IMCL and insulin resistance might be modulated by oxidative capacity in skeletal muscle. We examined the hypothesis that 12 wk of exercise training would increase both IMCL and the oxidative capacity of skeletal muscle in older (67.3 +/- 0.7 yr), previously sedentary subjects (n = 13; 5 men and 8 women). Maximal aerobic capacity (Vo(2 max)) increased from 1.65 +/- 0.20 to 1.85 +/- 0.14 l/min (P < 0.05), and systemic fat oxidation induced by 1 h of cycle exercise at 45% of Vo(2 max) increased (P < 0.05) from 15.03 +/- 40 to 19.29 +/- 0.80 (micromol.min(-1).kg fat-free mass(-1)). IMCL, determined by quantitative histological staining in vastus lateralis biopsies, increased (P < 0.05) from 22.9 +/- 1.9 to 25.9 +/- 2.6 arbitrary units (AU). The oxidative capacity of muscle, determined by succinate dehydrogenase staining intensity, significantly increased (P < 0.05) from 75.2 +/- 5.2 to 83.9 +/- 3.6 AU. The percentage of type I fibers significantly increased (P < 0.05) from 35.4 +/- 2.1 to 40.1 +/- 2.3%. In conclusion, exercise training increases IMCL in older persons in parallel with an enhanced capacity for fat oxidation.


Subject(s)
Aged/physiology , Exercise/physiology , Lipid Metabolism , Muscle Cells/metabolism , Oxygen Consumption , Body Composition , Female , Humans , Insulin Resistance , Lipids/analysis , Male , Muscle Cells/chemistry , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Oxidation-Reduction , Physical Fitness/physiology , Reference Values
13.
J Gene Med ; 6(2): 155-65, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14978769

ABSTRACT

BACKGROUND: Adenoviral gene transfer to adult skeletal muscle is hindered by several major limitations, including host immune responses and maturation-dependent loss of myofiber infectivity. Ex vivo gene delivery is more efficient than direct viral injection in surmounting maturation-dependent adenoviral transduction. Here we investigated the use of helper cells to improve the efficiency of ex vivo gene transfer to adult mouse skeletal muscle. METHODS: New producer cells carrying the E1 gene of adenovirus type 5 (E32 cells) were developed using primary myoblasts from mdx mice. The E32 cells and 293 cells were infected with an E1-deleted first-generation adenovirus carrying the LacZ gene. These transduced helper cells were injected into the skeletal muscle of adult mdx and SCID mice. RESULTS: LacZ-positive mature myofibers were detected in the skeletal muscle of adult mice sacrificed 5 days post-injection. The gene transfer efficiency using 293 cells and E32 cells was 6.2 and 3.6 times higher than myoblast-mediated gene transfer, respectively. Ex vivo gene transfer of these cell types led to a better outcome than did direct adenoviral injection. CONCLUSIONS: We achieved more efficient adenoviral gene transduction by using 293 and E32 helper cells than by myoblast-mediated gene transfer and direct viral injection. These helper cells also enabled adenoviral gene transfer to mature myofibers. The mechanisms by which this method facilitated adenoviral gene transfer to mature myofibers remains unclear; however, we hypothesize that the in vivo occurrence of cytopathic effects (CPE) in the transduced 293 and E32 helper cell populations facilitated the improved adenoviral transduction of myofibers.


Subject(s)
Adenoviridae , Genetic Therapy , Genetic Vectors , Helper Viruses , Muscle, Skeletal/metabolism , Animals , Cell Fusion , Genes, Reporter , In Vitro Techniques , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/therapy
14.
Rev Urol ; 6(2): 51-7, 2004.
Article in English | MEDLINE | ID: mdl-16985578

ABSTRACT

Suburethral pubovaginal sling placement is a common surgical procedure for the treatment of stress urinary incontinence. A wide variety of graft materials is available, each associated with inherent desirable and undesirable characteristics and complications. In this article, we discuss the rationale for and application of small intestinal submucosa (SIS) in lower urinary tract tissue engineering, with emphasis on the use of SIS as a suitable and biologically compatible sling material. In addition, we discuss exciting research regarding the engineering of true functional sphincter reconstruction using this biologic scaffold and pre-seeded muscle cells.

15.
Urology ; 62(5): 958-63, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14624934

ABSTRACT

OBJECTIVES: To study the physiologic outcome of allogenic transplant of muscle-derived progenitor cells (MDPCs) in the denervated female rat urethra. METHODS: MDPCs were isolated from muscle biopsies of normal 6-week-old Sprague-Dawley rats and purified using the preplate technique. Sciatic nerve-transected rats were used as a model of stress urinary incontinence. The experimental group was divided into three subgroups: control, denervated plus 20 microL saline injection, and denervated plus allogenic MDPCs (1 to 1.5 x 10(6) cells) injection. Two weeks after injection, urethral muscle strips were prepared and underwent electrical field stimulation. The pharmacologic effects of d-tubocurare, phentolamine, and tetrodotoxin on the urethral strips were assessed by contractions induced by electrical field stimulation. The urethral tissues also underwent immunohistochemical staining for fast myosin heavy chain and CD4-activated lymphocytes. RESULTS: Urethral denervation resulted in a significant decrease of the maximal fast-twitch muscle contraction amplitude to only 8.77% of the normal urethra and partial impairment of smooth muscle contractility. Injection of MDPCs into the denervated sphincter significantly improved the fast-twitch muscle contraction amplitude to 87.02% of normal animals. Immunohistochemistry revealed a large amount of new skeletal muscle fiber formation at the injection site of the urethra with minimal inflammation. CD4 staining showed minimal lymphocyte infiltration around the MDPC injection sites. CONCLUSIONS: Urethral denervation resulted in near-total abolishment of the skeletal muscle and partial impairment of smooth muscle contractility. Allogenic MDPCs survived 2 weeks in sciatic nerve-transected urethra with minimal inflammation. This is the first report of the restoration of deficient urethral sphincter function through muscle-derived progenitor cell tissue engineering. MDPC-mediated cellular urethral myoplasty warrants additional investigation as a new method to treat stress urinary incontinence.


Subject(s)
Mesenchymal Stem Cells , Muscle Contraction , Muscle, Smooth/surgery , Stem Cell Transplantation , Urethra/surgery , Animals , CD4-Positive T-Lymphocytes/immunology , Denervation , Electric Stimulation , Female , Lymphocyte Activation , Models, Animal , Muscle Contraction/drug effects , Muscle, Skeletal/cytology , Muscle, Smooth/physiopathology , Myosin Heavy Chains/analysis , Phentolamine/pharmacology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/injuries , Tetrodotoxin/pharmacology , Transplantation, Homologous , Tubocurarine/pharmacology , Urethra/drug effects , Urethra/innervation , Urinary Incontinence, Stress/surgery
16.
Hum Gene Ther ; 14(16): 1535-46, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14577915

ABSTRACT

Duchenne's muscular dystrophy (DMD) is a lethal muscle disease caused by a lack of dystrophin expression at the sarcolemma of muscle fibers. We investigated retroviral vector delivery of dystrophin in dystrophin-deficient DMD(mdx) (hereafter referred to as mdx) mice via an ex vivo approach using mdx muscle-derived stem cells (MDSCs). We generated a retrovirus carrying a functional human mini-dystrophin (RetroDys3999) and used it to stably transduce mdx MDSCs obtained by the preplate technique (MD3999). These MD3999 cells expressed dystrophin and continued to express stem cell markers, including CD34 and Sca-1. MD3999 cells injected into mdx mouse skeletal muscle were able to deliver dystrophin. Though a relatively low number of dystrophin-positive myofibers was generated within the gastrocnemius muscle, these fibers persisted for up to 24 weeks postinjection. The injection of cells from additional MDSC/Dys3999 clones into mdx skeletal muscle resulted in varying numbers of dystrophin-positive myofibers, suggesting a differential regenerating capacity among the clones. At 2 and 4 weeks postinjection, the infiltration of CD4- and CD8-positive lymphocytes and a variety of cytokines was detected within the injected site. These data suggest that the transplantation of retrovirally transduced mdx MDSCs can enable persistent dystrophin restoration in mdx skeletal muscle; however, the differential regenerating capacity observed among the MDSC/Dys3999 clones and the postinjection immune response are potential challenges facing this technology.


Subject(s)
Dystrophin/genetics , Gene Transfer Techniques , Genetic Vectors , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Stem Cell Transplantation , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Transplantation , Cells, Cultured , Dystrophin/deficiency , Gene Expression , Genetic Therapy , Injections, Intramuscular , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/immunology , Retroviridae/genetics , Transplantation, Isogeneic
17.
Urology ; 61(6): 1285-91, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12809930

ABSTRACT

OBJECTIVES: To explore the contractile activity and physiologic properties of muscle-derived stem cells (MDSCs) incorporated into small intestinal submucosa (SIS) scaffolds. METHODS: MDSCs were harvested from mice hind leg muscles using the preplate technique and stably transfected with a plasmid to express the LacZ reporter gene. Fifty different preparations of SIS cultured with MDSCs (MDSC/SIS) or SIS alone were incubated at 37 degrees C for 1, 4, and 8 weeks and also were mounted in a bath to measure the isometric contractions. RESULTS: LacZ and Masson-trichrome staining revealed MDSCs could migrate into and distribute throughout the SIS and form myotubes. In MDSC/SIS, spontaneous contractile activities were noted in the 4-week (five of six specimens) and 8-week (eight of eight specimens) cultures, but not in 1-week cultures (n = 11). All SIS control groups after 1 (n = 11), 4 (n = 6), and 8 (n = 8) weeks of incubation did not show any activity. In most of the 4-week, and all of the 8-week, MDSC/SIS cultures, the frequency and amplitude of spontaneous contractile activities were decreased by succinylcholine 10 microM and 20 microM. Electrical field stimulation, carbachol, and KCl did not alter the frequency, amplitude, or pattern of spontaneous contractile activities in MDSC/SIS. Spontaneous contractile activities were blocked by Ca(32+)-free Krebs solution with ethyleneglycoltetraacetic acid 200 microM and distilled water. CONCLUSIONS: MDSCs could be incorporated into SIS-forming myotubes capable of contracting. The contractile activity of this three-dimensional construct is Ca(2+) dependent and is modulated by nicotinic receptors. MDSC seeding of an acellular matrix may become a functional sling to reengineer the deficient sphincter or as contractile bladder augmentation.


Subject(s)
Calcium/physiology , Multipotent Stem Cells/physiology , Muscle Contraction/physiology , Receptors, Nicotinic/physiology , Animals , Calcium/antagonists & inhibitors , Cell Separation , Cells, Cultured , Chick Embryo , Dose-Response Relationship, Drug , Hindlimb , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Mice , Mice, Inbred mdx , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Muscle Contraction/drug effects , Muscle, Skeletal , Nicotinic Antagonists/pharmacology , Succinylcholine/pharmacology
18.
Urol Res ; 30(5): 310-6, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12389119

ABSTRACT

Gene therapy is a novel form of molecular medicine that may have a major impact on the future of human health care. We explored the efficacy of skeletal muscle derived cells (MDC) transduced with four viruses for ex vivo gene transfer into the lower urinary tract. Primary MDC were isolated from normal neonatal rats and transduced with: (1). adenovirus, (2). herpes simplex virus type-1 (HSV-1), (3). retrovirus or (4). adeno-associated virus (AAV), all of which express the beta-galactosidase reporter gene. Adult Sprague Dawley rats ( n=4 each group-time) were used. The MDC were injected into the right and left lateral bladder walls. The number of injected MDC ranged from 1 to 1.5 x 10(6). The tissues were harvested after 1, 4, 7, and 15 days, sectioned and assayed for beta-galactosidase expression. In the bladder wall, we noted cells expressing beta-galactosidase for each viral group. Adenoviral and HSV-1 transduced cells showed strong expression at 1 and 4 days post-injection, but the expression decreased gradually and was not detectable at 15 days post-injection. Retroviral transduced cells were detected at each time point with a strong expression persisting for 15 days but decreasing gradually over time. Although expression of the AAV transduced cells was initially weak, the later time points exhibited a much stronger expression, especially at day 7 post-injection. This expression persisted for at least 15 days post-injection. In conclusion, successful MDC mediated ex vivo gene transfer into the lower urinary tract was achieved with all four viral vectors. Our results suggest that the ex vivo approach may lead to an efficient and persistent viral gene delivery to the lower urinary tract while minimizing exposure of the host to virus.


Subject(s)
Cell Transplantation , Gene Transfer Techniques , Genetic Vectors , Muscle, Skeletal/cytology , Muscle, Skeletal/virology , Urinary Tract , Adenoviridae/genetics , Animals , Dependovirus/genetics , Gene Expression , Genes, Reporter , Herpesvirus 1, Human/genetics , Injections , Rats , Rats, Sprague-Dawley , Retroviridae/genetics , Time Factors , Urinary Tract Physiological Phenomena , beta-Galactosidase/genetics
19.
J Cell Biol ; 157(5): 851-64, 2002 May 27.
Article in English | MEDLINE | ID: mdl-12021255

ABSTRACT

Three populations of myogenic cells were isolated from normal mouse skeletal muscle based on their adhesion characteristics and proliferation behaviors. Although two of these populations displayed satellite cell characteristics, a third population of long-time proliferating cells expressing hematopoietic stem cell markers was also identified. This third population comprises cells that retain their phenotype for more than 30 passages with normal karyotype and can differentiate into muscle, neural, and endothelial lineages both in vitro and in vivo. In contrast to the other two populations of myogenic cells, the transplantation of the long-time proliferating cells improved the efficiency of muscle regeneration and dystrophin delivery to dystrophic muscle. The long-time proliferating cells' ability to proliferate in vivo for an extended period of time, combined with their strong capacity for self-renewal, their multipotent differentiation, and their immune-privileged behavior, reveals, at least in part, the basis for the improvement of cell transplantation. Our results suggest that this novel population of muscle-derived stem cells will significantly improve muscle cell-mediated therapies.


Subject(s)
Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Regeneration/physiology , Stem Cell Transplantation , Stem Cells/cytology , Animals , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Separation , Dystrophin/physiology , Endothelial Growth Factors/pharmacology , Hematopoietic Stem Cell Transplantation , In Vitro Techniques , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/immunology , Muscular Dystrophy, Animal/pathology , Nerve Growth Factor/pharmacology , Stem Cells/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...