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.
J Thorac Cardiovasc Surg ; 155(6): 2301-2313.e4, 2018 06.
Article in English | MEDLINE | ID: mdl-29559260

ABSTRACT

OBJECTIVES: Aortic aneurysms result from the degradation of multiple components represented by endothelial cells, vascular smooth muscle cells, and elastic fibers. Cells that can replenish these components are desirable for cell-based therapy. Intravenously injected multilineage-differentiating stress-enduring (Muse) cells, endogenous nontumorigenic pluripotent-like stem cells, reportedly integrate into the damaged site and repair the tissue through spontaneous differentiation into tissue-compatible cells. We evaluated the therapeutic efficacy of Muse cells in a murine aortic aneurysm model. METHODS: Human bone marrow Muse cells, isolated as stage-specific embryonic antigen-3+ from bone marrow mesenchymal stem cells, or non-Muse cells (stage-specific embryonic antigen-3- cells in mesenchymal stem cells), bone marrow mesenchymal stem cells, or vehicle was intravenously injected at day 0, day 7, and 2 weeks (20,000 cells/injection) after inducing aortic aneurysms by periaortic incubation of CaCl2 and elastase in severe combined immunodeficient mice. RESULTS: At 8 weeks, infusion of human Muse cells attenuated aneurysm dilation, and the aneurysmal size in the Muse group corresponded to approximately 62.5%, 55.6%, and 45.6% in the non-Muse, mesenchymal stem cell, and vehicle groups, respectively. Multiphoton laser confocal microscopy revealed that infused Muse cells migrated into aneurysmal tissue from the adventitial side and penetrated toward the luminal side. Histologic analysis demonstrated robust preservation of elastic fibers and spontaneous differentiation into endothelial cells and vascular smooth muscle cells. CONCLUSIONS: After intravenous injection, Muse cells homed and expanded to the aneurysm from the adventitial side. Subsequently, Muse cells differentiated spontaneously into vascular smooth muscle cells and endothelial cells, and elastic fibers were preserved. These Muse cell features together led to substantial attenuation of aneurysmal dilation.


Subject(s)
Aortic Aneurysm/surgery , Bone Marrow Transplantation , Cell Differentiation/physiology , Endothelial Cells , Animals , Aorta/cytology , Aorta/physiology , Bone Marrow Cells/cytology , Cell Movement , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/transplantation , Humans , Injections, Intravenous , Mice
2.
Stem Cells Dev ; 23(7): 717-28, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24256547

ABSTRACT

In this study, we demonstrate that a small population of pluripotent stem cells, termed adipose multilineage-differentiating stress-enduring (adipose-Muse) cells, exist in adult human adipose tissue and adipose-derived mesenchymal stem cells (adipose-MSCs). They can be identified as cells positive for both MSC markers (CD105 and CD90) and human pluripotent stem cell marker SSEA-3. They intrinsically retain lineage plasticity and the ability to self-renew. They spontaneously generate cells representative of all three germ layers from a single cell and successfully differentiate into targeted cells by cytokine induction. Cells other than adipose-Muse cells exist in adipose-MSCs, however, do not exhibit these properties and are unable to cross the boundaries from mesodermal to ectodermal or endodermal lineages even under cytokine inductions. Importantly, adipose-Muse cells demonstrate low telomerase activity and transplants do not promote teratogenesis in vivo. When compared with bone marrow (BM)- and dermal-Muse cells, adipose-Muse cells have the tendency to exhibit higher expression in mesodermal lineage markers, while BM- and dermal-Muse cells were generally higher in those of ectodermal and endodermal lineages. Adipose-Muse cells distinguish themselves as both easily obtainable and versatile in their capacity for differentiation, while low telomerase activity and lack of teratoma formation make these cells a practical cell source for potential stem cell therapies. Further, they will promote the effectiveness of currently performed adipose-MSC transplantation, particularly for ectodermal and endodermal tissues where transplanted cells need to differentiate across the lineage from mesodermal to ectodermal or endodermal in order to replenish lost cells for tissue repair.


Subject(s)
Adipocytes/cytology , Adipose Tissue/cytology , Adult Stem Cells/cytology , Germ Layers/cytology , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology , Adipocytes/metabolism , Adipose Tissue/metabolism , Adult , Adult Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage/physiology , Flow Cytometry , Germ Layers/metabolism , Humans , Immunophenotyping , Mesenchymal Stem Cells/metabolism , Mice , Pluripotent Stem Cells/metabolism , Regenerative Medicine , Stem Cell Transplantation , Telomerase/metabolism
3.
J Invest Dermatol ; 133(10): 2425-2435, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23563197

ABSTRACT

The induction of melanocytes from easily accessible stem cells has attracted attention for the treatment of melanocyte dysfunctions. We found that multilineage-differentiating stress-enduring (Muse) cells, a distinct stem cell type among human dermal fibroblasts, can be readily reprogrammed into functional melanocytes, whereas the remainder of the fibroblasts do not contribute to melanocyte differentiation. Muse cells can be isolated as cells positive for stage-specific embryonic antigen-3, a marker for undifferentiated human embryonic stem cells, and differentiate into cells representative of all three germ layers from a single cell, while also being nontumorigenic. The use of certain combinations of factors induces Muse cells to express melanocyte markers such as tyrosinase and microphthalmia-associated transcription factor and to show positivity for the 3,4-dihydroxy-L-phenylalanine reaction. When Muse cell-derived melanocytes were incorporated into three-dimensional (3D) cultured skin models, they localized themselves in the basal layer of the epidermis and produced melanin in the same manner as authentic melanocytes. They also maintained their melanin production even after the 3D cultured skin was transplanted to immunodeficient mice. This technique may be applicable to the efficient production of melanocytes from accessible human fibroblasts by using Muse cells, thereby contributing to autologous transplantation for melanocyte dysfunctions, such as vitiligo.


Subject(s)
Fibroblasts/cytology , Melanocytes/cytology , Skin Transplantation/methods , Stem Cells/cytology , Stress, Physiological/physiology , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Cells, Cultured , Dermis/cytology , Fibroblasts/metabolism , Humans , Melanins/metabolism , Melanocytes/metabolism , Mice , Mice, SCID , Stem Cells/metabolism
4.
PLoS One ; 7(12): e48677, 2012.
Article in English | MEDLINE | ID: mdl-23272044

ABSTRACT

Induced pluripotent stem (iPS) cells can be generated from somatic cells by the forced expression of four factors, Oct3/4, Sox2, Klf4, and c-Myc. While a great variety of colonies grow during induction, only a few of them develop into iPS cells. Researchers currently use visual observation to identify iPS cells and select colonies resembling embryonic stem (ES) cells, and there are no established objective criteria. Therefore, we exhaustively analyzed the morphology and gene expression of all the colonies generated from human fibroblasts after transfection with four retroviral vectors encoding individual factors (192 and 203 colonies in two experiments) and with a single polycistronic retroviral vector encoding all four factors (199 and 192 colonies in two experiments). Here we demonstrate that the morphologic features of emerged colonies can be categorized based on six parameters, and all generated colonies that could be passaged were classified into seven subtypes in colonies transfected with four retroviral vectors and six subtypes with a single polycistronic retroviral vector, both including iPS cell colonies. The essential qualifications for iPS cells were: cells with a single nucleolus; nucleus to nucleolus (N/Nls) ratio ∼2.19: cell size ∼43.5 µm(2): a nucleus to cytoplasm (N/C) ratio ∼0.87: cell density in a colony ∼5900 cells/mm(2): and number of cell layer single. Most importantly, gene expression analysis revealed for the first time that endogenous Sox2 and Cdx2 were expressed specifically in iPS cells, whereas Oct3/4 and Nanog, popularly used markers for identifying iPS cells, are expressed in colonies other than iPS cells, suggesting that Sox2 and Cdx2 are reliable markers for identifying iPS cells. Our findings indicate that morphologic parameters and the expression of endogenous Sox2 and Cdx2 can be used to accurately identify iPS cells.


Subject(s)
Cell Culture Techniques , Gene Expression Regulation , Induced Pluripotent Stem Cells/cytology , Animals , CDX2 Transcription Factor , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Factor 4 , Mice , Polymerase Chain Reaction/methods , RNA/metabolism , Retroviridae/genetics , SOXB1 Transcription Factors/metabolism
5.
Cells ; 1(4): 1045-60, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-24710542

ABSTRACT

Mesenchymal stem cells (MSCs) are easily accessible and safe for regenerative medicine. MSCs exert trophic, immunomodulatory, anti-apoptotic, and tissue regeneration effects in a variety of tissues and organs, but their entity remains an enigma. Because MSCs are generally harvested from mesenchymal tissues, such as bone marrow, adipose tissue, or umbilical cord as adherent cells, MSCs comprise crude cell populations and are heterogeneous. The specific cells responsible for each effect have not been clarified. The most interesting property of MSCs is that, despite being adult stem cells that belong to the mesenchymal tissue lineage, they are able to differentiate into a broad spectrum of cells beyond the boundary of mesodermal lineage cells into ectodermal or endodermal lineages, and repair tissues. The broad spectrum of differentiation ability and tissue-repairing effects of MSCs might be mediated in part by the presence of a novel pluripotent stem cell type recently found in adult human mesenchymal tissues, termed multilineage-differentiating stress enduring (Muse) cells. Here we review recently updated studies of the regenerative effects of MSCs and discuss their potential in regenerative medicine.

6.
Tissue Eng Part A ; 17(15-16): 1993-2004, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21457094

ABSTRACT

Bone marrow stromal cells (MSCs) are a useful source of cells because of their abundant supply and few associated ethical problems. We have previously reported that neural progenitor cells (NS-MSCs) can be effectively induced from MSCs and differentiate into neurons to contribute to functional recovery when transplanted into the rat stroke model. In this study, we attempted to enhance the therapeutic effects of NS-MSCs with a collagen sponge and basic fibroblast growth factor (bFGF) releasing microspheres. NS-MSCs were generated from MSCs by transfection of Notch-1 intracellular domain followed by culturing the cells in a free-floating culture system. The resulting NS-MSCs were transplanted into the rats with induced brain ischemia by using collagen sponges as scaffolds for transplanted cells, and with bFGF incorporated into gelatin microspheres to aid neovascularization around the transplanted region and proliferation of neural stem cells/neural progenitor cells. In culture, NS-MSCs successfully formed spheres containing cells highly expressing neural progenitor markers. Cell survival, neovascularization, and proliferation of host neural stem cells/neural progenitor cells were improved in animals that received NS-MSCs together with these biomaterials. Behavioral analysis also revealed significant functional recovery. These observations demonstrate that transplantation of NS-MSCs in combination with a collagen sponge and bFGF releasing microspheres significantly improves histological and functional recovery in the rat stroke model. When used with these biomaterials, NS-MSCs would be a promising cell source for treating stroke and neurodegenerative diseases.


Subject(s)
Bone Marrow Cells/cytology , Brain Ischemia/physiopathology , Brain Ischemia/therapy , Collagen/pharmacology , Fibroblast Growth Factor 2/pharmacology , Neural Stem Cells/transplantation , Recovery of Function/drug effects , Animals , Behavior, Animal/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Brain Ischemia/pathology , Cell Proliferation/drug effects , Immunohistochemistry , Microspheres , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Rats , Rats, Wistar , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...