Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Stem Cells Transl Med ; 3(3): 334-45, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24443004

ABSTRACT

The discovery of multipotent neural crest-derived stem cells, named epidermal neural crest stem cells (EPI-NCSC), that persist postnatally in an easy-to-access location-the bulge of hair follicles-opens a spectrum of novel opportunities for patient-specific therapies. We present a detailed characterization of canine EPI-NCSC (cEPI-NCSC) from multiple dog breeds and protocols for their isolation and ex vivo expansion. Furthermore, we provide novel tools for research in canines, which currently are still scarce. In analogy to human and mouse EPI-NCSC, the neural crest origin of cEPI-NCSC is shown by their expression of the neural crest stem cell molecular signature and other neural crest-characteristic genes. Similar to human EPI-NCSC, cEPI-NCSC also expressed pluripotency genes. We demonstrated that cEPI-NCSC can generate all major neural crest derivatives. In vitro clonal analyses established multipotency and self-renewal ability of cEPI-NCSC, establishing cEPI-NCSC as multipotent somatic stem cells. A critical analysis of the literature on canine spinal cord injury (SCI) showed the need for novel treatments and suggested that cEPI-NCSC represent viable candidates for cell-based therapies in dog SCI, particularly for chondrodystrophic dogs. This notion is supported by the close ontological relationship between neural crest stem cells and spinal cord stem cells. Thus, cEPI-NCSC promise to offer not only a potential treatment for canines but also an attractive and realistic large animal model for human SCI. Taken together, we provide the groundwork for the development of a novel cell-based therapy for a condition with extremely poor prognosis and no available effective treatment.


Subject(s)
Hair Follicle/cytology , Multipotent Stem Cells/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Proliferation , Cell Separation , Cell- and Tissue-Based Therapy , Disease Models, Animal , Dogs , Female , Gene Expression , Hair Follicle/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/transplantation , Neural Crest/metabolism , Neural Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy
3.
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
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 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
6.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...