Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
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.
Stem Cells Transl Med ; 4(10): 1173-86, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26273065

ABSTRACT

UNLABELLED: This pilot feasibility study aimed to determine the outcome of canine epidermal neural crest stem cell (cEPI-NCSC) grafts in the normal spinal cords of healthy bred-for-research dogs. This included developing novel protocols for (a) the ex vivo expansion of cEPI-NCSCs, (b) the delivery of cEPI-NCSCs into the spinal cord, and (c) the labeling of the cells and subsequent tracing of the graft in the live animal by magnetic resonance imaging. A total of four million cEPI-NCSCs were injected into the spinal cord divided in two locations. Differences in locomotion at baseline and post-treatment were evaluated by gait analysis and compared with neurological outcome and behavioral exams. Histopathological analyses of the spinal cords and cEPI-NCSC grafts were performed at 3 weeks post-transplantation. Neurological and gait parameters were minimally affected by the stem cell injection. cEPI-NCSCs survived in the canine spinal cord for the entire period of investigation and did not migrate or proliferate. Subsets of cEPI-NCSCs expressed the neural crest stem cell marker Sox10. There was no detectable expression of markers for glial cells or neurons. The tissue reaction to the cell graft was predominantly vascular in addition to a degree of reactive astrogliosis and microglial activation. In the present study, we demonstrated that cEPI-NCSC grafts survive in the spinal cords of healthy dogs without major adverse effects. They persist locally in the normal spinal cord, may promote angiogenesis and tissue remodeling, and elicit a tissue response that may be beneficial in patients with spinal cord injury. SIGNIFICANCE: It has been established that mouse and human epidermal neural crest stem cells are somatic multipotent stem cells with proved innovative potential in a mouse model of spinal cord injury (SCI) offering promise of a valid treatment for SCI. Traumatic SCI is a common neurological problem in dogs with marked similarities, clinically and pathologically, to the syndrome in people. For this reason, dogs provide a readily accessible, clinically realistic, spontaneous model for evaluation of epidermal neural crest stem cells therapeutic intervention. The results of this study are expected to give the baseline data for a future clinical trial in dogs with traumatic SCI.


Subject(s)
Neural Crest/transplantation , Neural Stem Cells/transplantation , Spinal Cord/cytology , Stem Cell Transplantation/methods , Animals , Behavior, Animal , Cell Survival , Dogs , Epidermal Cells , Feasibility Studies , Gait , Injections, Spinal , Magnetic Resonance Imaging , Mice , Mice, Knockout , Neurogenesis , Pilot Projects , Stem Cell Transplantation/adverse effects , Teratoma , Walking
3.
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
4.
Stem Cells Transl Med ; 2(6): 409-19, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23681950

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) hold great potential for use in regenerative medicine, novel drug development, and disease progression/developmental studies. Here, we report highly efficient differentiation of hiPSCs toward a relatively homogeneous population of functional hepatocytes. hiPSC-derived hepatocytes (hiHs) not only showed a high expression of hepatocyte-specific proteins and liver-specific functions, but they also developed a functional biotransformation system including phase I and II metabolizing enzymes and phase III transporters. Nuclear receptors, which are critical for regulating the expression of metabolizing enzymes, were also expressed in hiHs. hiHs also responded to different compounds/inducers of cytochrome P450 as mature hepatocytes do. To follow up on this observation, we analyzed the drug metabolizing capacity of hiHs in real time using a novel ultra performance liquid chromatography-tandem mass spectrometry. We found that, like freshly isolated primary human hepatocytes, the seven major metabolic pathways of the drug bufuralol were found in hiHs. In addition, transplanted hiHs engrafted, integrated, and proliferated in livers of an immune-deficient mouse model, and secreted human albumin, indicating that hiHs also function in vivo. In conclusion, we have generated a method for the efficient generation of hepatocytes from induced pluripotent stem cells in vitro and in vivo, and it appears that the cells function similarly to primary human hepatocytes, including developing a complete metabolic function. These results represent a significant step toward using patient/disease-specific hepatocytes for cell-based therapeutics as well as for pharmacology and toxicology studies.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/metabolism , Hepatocytes/transplantation , Induced Pluripotent Stem Cells/cytology , Metabolic Detoxication, Phase II/physiology , Metabolic Detoxication, Phase I/physiology , Adrenergic beta-Antagonists/metabolism , Albumins/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Chromatography, Liquid , Cytochrome P-450 Enzyme System/genetics , Ethanolamines/metabolism , Gene Expression , Hepatocytes/cytology , Humans , Immunocompromised Host , Induced Pluripotent Stem Cells/metabolism , Liver , Mice , Mice, SCID , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Tandem Mass Spectrometry , Transplantation, Heterologous
5.
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
6.
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
7.
Invest Ophthalmol Vis Sci ; 53(2): 986-94, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22247454

ABSTRACT

PURPOSE: To determine long-term safety of intravitreal administration of good manufacturing practice (GMP)-grade human bone-marrow-derived CD34(+) cells in NOD-SCID (nonobese diabetic-severe combined immunodeficiency) mice with acute retinal ischemia-reperfusion injury, a model for retinal vasculopathy. METHOD: Acute ischemia-reperfusion injury was induced in the right eye of adult NOD-SCID mice (n = 23) by transient elevation of intraocular pressure. Seven days later, 12 injured eyes and 5 normal contralateral eyes were injected each intravitreally with 5 × 10(4) CD34(+) cells isolated under GMP conditions from a healthy human donor bone marrow using an immunomagnetic cell isolation system. The remaining 11 injured eyes were not treated and served as controls. Mice were euthanized 1 day, 4 months, and 8 months later. Both eyes were enucleated and examined by immunohistochemical analysis and hematoxylin and eosin staining. Among mice followed for 8 months, electroretinography (ERG) was performed on both eyes before euthanization. All major organs were examined grossly and histologically after serial sectioning. RESULTS: Immunohistochemical staining 4 months after injection showed detectable CD34(+) cells in the retinal vasculature. ERG at 8 months after CD34(+) cell injection showed signals that were similar in untreated eyes. Histology of the enucleated eyes injected with CD34(+) cells showed no intraocular tumor or abnormal tissue growth after 8 months. Histologic analysis of all major organs showed no abnormal proliferation of human cells. CONCLUSIONS: Intravitreal administration of GMP-grade human bone-marrow-derived CD34(+) cells appears to be well tolerated long-term in eyes with acute retinal ischemic injury. A clinical trial will start to further explore this therapy.


Subject(s)
Antigens, CD34 , Bone Marrow Cells/immunology , Hematopoietic Stem Cells/immunology , Reperfusion Injury/drug therapy , Retinal Diseases/drug therapy , Stem Cell Transplantation/methods , Acute Disease , Animals , Bone Marrow Cells/cytology , Electroretinography , Follow-Up Studies , Graft Survival , Humans , Intravitreal Injections , Mice , Mice, Inbred NOD , Mice, SCID , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Retinal Diseases/pathology , Retinal Diseases/physiopathology , Time Factors , Transplantation, Heterologous
8.
Mol Neurobiol ; 45(1): 87-98, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22161544

ABSTRACT

There is much interest in the use of mesenchymal stem cells/marrow stromal cells (MSC) to treat neurodegenerative disorders, in particular those that are fatal and difficult to treat, such as Huntington's disease. MSC present a promising tool for cell therapy and are currently being tested in FDA-approved phase I-III clinical trials for many disorders. In preclinical studies of neurodegenerative disorders, MSC have demonstrated efficacy, when used as delivery vehicles for neural growth factors. A number of investigators have examined the potential benefits of innate MSC-secreted trophic support and augmented growth factors to support injured neurons. These include overexpression of brain-derived neurotrophic factor and glial-derived neurotrophic factor, using genetically engineered MSC as a vehicle to deliver the cytokines directly into the microenvironment. Proposed regenerative approaches to neurological diseases using MSC include cell therapies in which cells are delivered via intracerebral or intrathecal injection. Upon transplantation, MSC in the brain promote endogenous neuronal growth, encourage synaptic connection from damaged neurons, decrease apoptosis, reduce levels of free radicals, and regulate inflammation. These abilities are primarily modulated through paracrine actions. Clinical trials for MSC injection into the central nervous system to treat amyotrophic lateral sclerosis, traumatic brain injury, and stroke are currently ongoing. The current data in support of applying MSC-based cellular therapies to the treatment of Huntington's disease is discussed.


Subject(s)
Genetic Engineering/methods , Huntington Disease/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Nerve Growth Factors/metabolism , Animals , Disease Models, Animal , Genetic Engineering/trends , Humans , Huntington Disease/pathology , Huntington Disease/physiopathology , Mesenchymal Stem Cell Transplantation/trends , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics
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
10.
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
11.
Semin Immunopathol ; 31(3): 411-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19533133

ABSTRACT

Immune-deficient mouse models of liver damage allow examination of human stem cell migration to sites of damage and subsequent contribution to repair and survival. In our studies, in the absence of a selective advantage, transplanted human stem cells from adult sources did not robustly become hepatocytes, although some level of fusion or hepatic differentiation was documented. However, injected stem cells did home to the injured liver tissue and release paracrine factors that hastened endogenous repair and enhanced survival. There were significantly higher levels of survival in mice with a toxic liver insult that had been transplanted with human stem cells but not in those transplanted with committed progenitors. Transplantation of autologous adult stem cells without conditioning is a relatively safe therapy. Adult stem cells are known to secrete bioactive factors that suppress the local immune system, inhibit fibrosis (scar formation) and apoptosis, enhance angiogenesis, and stimulate recruitment, retention, mitosis, and differentiation of tissue-residing stem cells. These paracrine effects are distinct from the direct differentiation of stem cells to repair tissue. In patients at high risk while waiting for a liver transplant, autologous stem cell therapy could be considered, as it could delay the decline in liver function.


Subject(s)
Hematopoietic Stem Cells/immunology , Liver Diseases/immunology , Liver Regeneration/physiology , Liver/immunology , Animals , Cell Movement/immunology , Cell Movement/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hepatocyte Growth Factor/immunology , Hepatocyte Growth Factor/metabolism , Humans , Liver/pathology , Liver Diseases/therapy , Liver Regeneration/immunology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL