Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Mol Ther ; 24(5): 965-77, 2016 05.
Article in English | MEDLINE | ID: mdl-26765769

ABSTRACT

Huntington's disease (HD) is a fatal degenerative autosomal dominant neuropsychiatric disease that causes neuronal death and is characterized by progressive striatal and then widespread brain atrophy. Brain-derived neurotrophic factor (BDNF) is a lead candidate for the treatment of HD, as it has been shown to prevent cell death and to stimulate the growth and migration of new neurons in the brain in transgenic mouse models. BDNF levels are reduced in HD postmortem human brain. Previous studies have shown efficacy of mesenchymal stem/stromal cells (MSC)/BDNF using murine MSCs, and the present study used human MSCs to advance the therapeutic potential of the MSC/BDNF platform for clinical application. Double-blinded studies were performed to examine the effects of intrastriatally transplanted human MSC/BDNF on disease progression in two strains of immune-suppressed HD transgenic mice: YAC128 and R6/2. MSC/BDNF treatment decreased striatal atrophy in YAC128 mice. MSC/BDNF treatment also significantly reduced anxiety as measured in the open-field assay. Both MSC and MSC/BDNF treatments induced a significant increase in neurogenesis-like activity in R6/2 mice. MSC/BDNF treatment also increased the mean lifespan of the R6/2 mice. Our genetically modified MSC/BDNF cells set a precedent for stem cell-based neurotherapeutics and could potentially be modified for other neurodegenerative disorders such as amyotrophic lateral sclerosis, Alzheimer's disease, and some forms of Parkinson's disease. These cells provide a platform delivery system for future studies involving corrective gene-editing strategies.

2.
Hum Mol Genet ; 21(17): 3795-805, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22641815

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a leading monogenic neurodegenerative disorder affecting premutation carriers of the fragile X (FMR1) gene. To investigate the underlying cellular neuropathology, we produced induced pluripotent stem cell-derived neurons from isogenic subclones of primary fibroblasts of a female premutation carrier, with each subclone bearing exclusively either the normal or the expanded (premutation) form of the FMR1 gene as the active allele. We show that neurons harboring the stably-active, expanded allele (EX-Xa) have reduced postsynaptic density protein 95 protein expression, reduced synaptic puncta density and reduced neurite length. Importantly, such neurons are also functionally abnormal, with calcium transients of higher amplitude and increased frequency than for neurons harboring the normal-active allele. Moreover, a sustained calcium elevation was found in the EX-Xa neurons after glutamate application. By excluding the individual genetic background variation, we have demonstrated neuronal phenotypes directly linked to the FMR1 premutation. Our approach represents a unique isogenic, X-chromosomal epigenetic model to aid the development of targeted therapeutics for FXTAS, and more broadly as a model for the study of common neurodevelopmental (e.g. autism) and neurodegenerative (e.g. Parkinsonism, dementias) disorders.


Subject(s)
Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Neurons/metabolism , Neurons/pathology , Signal Transduction/genetics , Action Potentials , Alleles , Calcium/metabolism , Cell Differentiation , Clone Cells , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/physiopathology , Gene Expression Regulation , Humans , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synapses/metabolism , Synapses/pathology , Tissue Donors , X Chromosome Inactivation/genetics
3.
J Virol ; 86(10): 5719-29, 2012 May.
Article in English | MEDLINE | ID: mdl-22398281

ABSTRACT

HIV gene therapy has the potential to offer an alternative to the use of current small-molecule antiretroviral drugs as a treatment strategy for HIV-infected individuals. Therapies designed to administer HIV-resistant stem cells to an infected patient may also provide a functional cure, as observed in a bone marrow transplant performed with hematopoietic stem cells (HSCs) homozygous for the CCR5-Δ32-bp allele. In our current studies, preclinical evaluation of a combination anti-HIV lentiviral vector was performed, in vivo, in humanized NOD-RAG1(-/-) IL2rγ(-/-) knockout mice. This combination vector, which displays strong preintegration inhibition of HIV-1 infection in vitro, contains a human/rhesus macaque TRIM5α isoform, a CCR5 short hairpin RNA (shRNA), and a TAR decoy. Multilineage hematopoiesis from anti-HIV lentiviral vector-transduced human CD34(+) HSCs was observed in the peripheral blood and in various lymphoid organs, including the thymus, spleen, and bone marrow, of engrafted mice. Anti-HIV vector-transduced CD34(+) cells displayed normal development of immune cells, including T cells, B cells, and macrophages. The anti-HIV vector-transduced cells also displayed knockdown of cell surface CCR5 due to the expression of the CCR5 shRNA. After in vivo challenge with either an R5-tropic BaL-1 or X4-tropic NL4-3 strain of HIV-1, maintenance of human CD4(+) cell levels and a selective survival advantage of anti-HIV gene-modified cells were observed in engrafted mice. The data provided from our study confirm the safety and efficacy of this combination anti-HIV lentiviral vector in a hematopoietic stem cell gene therapy setting for HIV and validates its potential application in future clinical trials.


Subject(s)
Cell- and Tissue-Based Therapy , HIV Infections/immunology , HIV Infections/therapy , HIV-1/immunology , Hematopoietic Stem Cells/immunology , Transduction, Genetic , Animals , Antigens, CD34/immunology , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , HIV Infections/genetics , HIV-1/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/virology , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice
4.
Mol Cell Neurosci ; 49(3): 271-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22198539

ABSTRACT

Huntington's disease (HD) is a fatal, autosomal dominant neurodegenerative disorder caused by an expanded trinucleotide (CAG) repeat in exon 1 of the huntingtin gene (Htt). This expansion creates a toxic polyglutamine tract in the huntingtin protein (HTT). Currently, there is no treatment for either the progression or prevention of the disease. RNA interference (RNAi) technology has shown promise in transgenic mouse models of HD by reducing expression of mutant HTT and slowing disease progression. The advancement of RNAi therapies to human clinical trials is hampered by problems delivering RNAi to affected neurons in a robust and sustainable manner. Mesenchymal stem cells (MSC) have demonstrated a strong safety profile in both completed and numerous ongoing clinical trials. MSC exhibit a number of innate therapeutic effects, such as immune system modulation, homing to injury, and cytokine release into damaged microenvironments. The ability of MSC to transfer larger molecules and even organelles suggested their potential usefulness as delivery vehicles for therapeutic RNA inhibition. In a series of model systems we have found evidence that MSC can transfer RNAi targeting both reporter genes and mutant huntingtin in neural cell lines. MSC expressing shRNA antisense to GFP were found to decrease expression of GFP in SH-SY5Y cells after co-culture when assayed by flow cytometry. Additionally MSC expressing shRNA antisense to HTT were able to decrease levels of mutant HTT expressed in both U87 and SH-SY5Y target cells when assayed by Western blot and densitometry. These results are encouraging for expanding the therapeutic abilities of both RNAi and MSC for future treatments of Huntington's disease.


Subject(s)
Genetic Vectors , Mesenchymal Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , RNA Interference/physiology , Cell Line , Coculture Techniques , Down-Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Lentivirus/genetics
5.
Mol Ther ; 19(3): 584-93, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21119622

ABSTRACT

Induced pluripotent stem cells (iPSCs) have radically advanced the field of regenerative medicine by making possible the production of patient-specific pluripotent stem cells from adult individuals. By developing iPSCs to treat HIV, there is the potential for generating a continuous supply of therapeutic cells for transplantation into HIV-infected patients. In this study, we have used human hematopoietic stem cells (HSCs) to generate anti-HIV gene expressing iPSCs for HIV gene therapy. HSCs were dedifferentiated into continuously growing iPSC lines with four reprogramming factors and a combination anti-HIV lentiviral vector containing a CCR5 short hairpin RNA (shRNA) and a human/rhesus chimeric TRIM5α gene. Upon directed differentiation of the anti-HIV iPSCs toward the hematopoietic lineage, a robust quantity of colony-forming CD133(+) HSCs were obtained. These cells were further differentiated into functional end-stage macrophages which displayed a normal phenotypic profile. Upon viral challenge, the anti-HIV iPSC-derived macrophages exhibited strong protection from HIV-1 infection. Here, we demonstrate the ability of iPSCs to develop into HIV-1 resistant immune cells and highlight the potential use of iPSCs for HIV gene and cellular therapies.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Macrophages/cytology , Macrophages/immunology , AC133 Antigen , Adult , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Glycoproteins/metabolism , HEK293 Cells , HIV Infections/virology , Humans , Induced Pluripotent Stem Cells/cytology , Peptides/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, CCR5/genetics , Receptors, CCR5/metabolism
6.
Mol Ther Methods Clin Dev ; 3: 16053, 2016.
Article in English | MEDLINE | ID: mdl-27610394

ABSTRACT

Numerous clinical trials are utilizing mesenchymal stem cells (MSC) to treat critical limb ischemia, primarily for their ability to secrete signals that promote revascularization. These cells have demonstrated clinical safety, but their efficacy has been limited, possibly because these paracrine signals are secreted at subtherapeutic levels. In these studies the combination of cell and gene therapy was evaluated by engineering MSC with a lentivirus to overexpress vascular endothelial growth factor (VEGF). To achieve clinical compliance, the number of viral insertions was limited to 1-2 copies/cell and a constitutive promoter with demonstrated clinical safety was used. MSC/VEGF showed statistically significant increases in blood flow restoration as compared with sham controls, and more consistent improvements as compared with nontransduced MSC. Safety of MSC/VEGF was assessed in terms of genomic stability, rule-out tumorigenicity, and absence of edema or hemangiomas in vivo. In terms of retention, injected MSC/VEGF showed a steady decline over time, with a very small fraction of MSC/VEGF remaining for up to 4.5 months. Additional safety studies completed include absence of replication competent lentivirus, sterility tests, and absence of VSV-G viral envelope coding plasmid. These preclinical studies are directed toward a planned phase 1 clinical trial to treat critical limb ischemia.

7.
Hum Gene Ther Methods ; 26(6): 193-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26544924

ABSTRACT

Muscle-derived progenitor cell (myoblast) therapy has promise for the treatment of denervated, weakened, and fibrotic muscle. The best methods for injecting myoblasts to promote fusion and retention have yet to be determined, however. Mesenchymal stem/stromal cells have also been reported to have beneficial effects in restoring damaged tissue, through increasing vascularization and reducing inflammation. The interactions between human primary skeletal myoblasts and bone marrow-derived mesenchymal stem/stromal cells were examined using time-lapse images put into video format. Of interest, there is a high degree of cell-to-cell interaction with microparticles transferring between both cell types, and formation of nanotubules to bridge cytoplasmic contents between the two types of cell. This model provides an in vitro platform for examining mechanisms for cell-to-cell interaction preceding myoblast fusion.


Subject(s)
Cell Communication , Mesenchymal Stem Cells/metabolism , Microscopy, Video , Myoblasts/metabolism , Cells, Cultured , Coculture Techniques , Gene Expression , Genes, Reporter , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Transduction, Genetic
8.
World Neurosurg ; 84(5): 1256-66.e1, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26074438

ABSTRACT

BACKGROUND: The dura mater can be easily biopsied during most cranial neurosurgical operations. We describe a protocol that allows for robust generation of induced pluripotent stem cells (iPSCs) and neural progenitors from acutely harvested dura mater. OBJECTIVE: To generate iPSCs and neural progenitor cells from dura mater obtained during ventriculoperitoneal shunt surgery. METHODS: Dura was obtained during ventriculoperitoneal shunt surgery for normal pressure hydrocephalus from a 60-year-old patient with severe cognitive impairment. Fibroblasts were isolated from the dural matrix and transduced with nonintegrating Sendai virus for iPSC induction. A subset of successfully generated iPSC clones underwent immunocytochemical analysis, teratoma assay, karyotyping, and targeted neural differentiation. RESULTS: Eleven iPSC clones were obtained from the transduction of an estimated 600,000 dural fibroblasts after 3 passages. Three clones underwent immunocytochemical analysis and were shown to express the transcription factors OCT-4, SOX2, and the embryonic cell markers SSEA-4, TRA-1-60, and Nanog. Two clones were tested for pluripotency and formed teratomas at the injection site in immunodeficient mice. Three clones underwent chromosomal analysis and were found to have a normal metaphase spread and karyotype. One clone underwent targeted neural differentiation and formed neural rosettes as well as TuJ1/SOX1-positive neural progenitor cells. CONCLUSIONS: IPSCs and neural progenitor cells can be efficiently derived from the dura of patients who need to undergo cranial neurosurgical operations. IPSCs were obtained with a nonintegrating virus and exhibited a normal karyotype, making them candidates for future autotransplantation after targeted differentiation to treat functional deficits.


Subject(s)
Dura Mater/cytology , Induced Pluripotent Stem Cells , Neural Stem Cells , Ventriculoperitoneal Shunt , Animals , Cell Differentiation , Cellular Reprogramming , Clone Cells , Cognition Disorders/etiology , Dura Mater/surgery , Fibroblasts , Humans , Hydrocephalus, Normal Pressure/surgery , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Male , Mice , Middle Aged , Neural Stem Cells/metabolism , Sendai virus , Teratoma/pathology , Transcription Factors/metabolism
9.
Tissue Eng Part A ; 17(11-12): 1517-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21275830

ABSTRACT

Mesenchymal stem cells (MSCs) have been shown to contribute to the recovery of tissues through homing to injured areas, especially to hypoxic, apoptotic, or inflamed areas and releasing factors that hasten endogenous repair. In some cases genetic engineering of the MSC is desired, since they are excellent delivery vehicles. We have derived MSCs from the human embryonic stem cell (hESC) line H9 (H9-MSCs). They expressed CD105, CD90, CD73, and CD146, and lacked expression of CD45, CD34, CD14, CD31, and HLA-DR, the hESC pluripotency markers SSEA-4 and Tra-1-81, and the hESC early differentiation marker SSEA-1. Marrow-derived MSCs showed a similar phenotype. H9-MSCs did not form teratoma in our initial studies, whereas the parent H9 line did so robustly. H9-MSCs differentiated into bone, cartilage, and adipocytes in vitro, and displayed increased migration under hypoxic conditions. Finally, using a hindlimb ischemia model, H9-MSCs were shown to home to the hypoxic muscle, but not the contralateral limb, by 48 h after IV injection. In summary, we have defined methods for differentiation of hESCs into MSCs and have defined their characteristics and in vivo migratory properties.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Animals , Cell Differentiation , Cell Hypoxia , Cell Line , Cell Lineage , Cell Movement , Cell Shape , Cells, Cultured , Flow Cytometry , Hindlimb/blood supply , Hindlimb/pathology , Humans , Ischemia/pathology , Ischemia/therapy , Karyotyping , Mesenchymal Stem Cell Transplantation , Mice , Teratoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL