Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Oral Radiol ; 38(3): 349-355, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34505258

RESUMEN

PURPOSE: The purpose of this study was to assess quantitatively the mandibular bone marrow of patients with and without diabetes mellitus (DM) using the apparent diffusion coefficient (ADC) values on diffusion-weighted imaging (DWI). METHODS: 65 DM patients (28 men, 37 women, 29-84 years of age, mean age 55.7 ± 15.7 years) and age-, sex- and periodontitis stage-matched 65 non-DM patients who had underwent MRI between April 2006 and March 2018 were included in this study. The ADC was calculated using the ADC visualization tool implemented in a dedicated off-line workstation. The regions of interest (ROI) were manually placed on the ADC map on which the mandibular bone marrow from the lower first molar to the lower second molar was observed in patients with and without DM. Statistical analysis was performed using the Mann-Whitney U test and receiver operating characteristic (ROC) curve analysis. P values < 0.05 were considered statistically significant. RESULTS: The mean ADC values of the mandibular bone marrow of patients with and without DM were 1.18 ± 0.21 × 10-3 mm2/s and 0.83 ± 0.14 × 10-3 mm2/s, respectively. The ADC values of DM patients were significantly higher than those of patients without DM. CONCLUSION: The ADC values allowed the quantitative evaluation of the mandibular bone marrow of DM patients. DWI might serve as a new and noninvasive method to assess the presence of DM.


Asunto(s)
Médula Ósea , Diabetes Mellitus , Adulto , Anciano , Médula Ósea/diagnóstico por imagen , Médula Ósea/patología , Diabetes Mellitus/diagnóstico por imagen , Diabetes Mellitus/patología , Imagen de Difusión por Resonancia Magnética/métodos , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Curva ROC
2.
NPJ Regen Med ; 6(1): 81, 2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34824291

RESUMEN

Stem cell-based regenerative therapy has opened an avenue for functional recovery of patients with spinal cord injury (SCI). Regenerative rehabilitation is attracting wide attention owing to its synergistic effects, feasibility, non-invasiveness, and diverse and systemic properties. In this review article, we summarize the features of rehabilitation, describe the mechanism of combinatorial treatment, and discuss regenerative rehabilitation in the context of SCI. Although conventional rehabilitative methods have commonly been implemented alone, especially in studies of acute-to-subacute SCI, the combinatorial effects of intensive and advanced methods, including various neurorehabilitative approaches, have also been reported. Separating the concept of combined rehabilitation from regenerative rehabilitation, we suggest that the main roles of regenerative rehabilitation can be categorized as conditioning/reconditioning, functional training, and physical exercise, all of which are indispensable for enhancing functional recovery achieved using stem cell therapies.

3.
Stem Cells Transl Med ; 8(3): 260-270, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30485733

RESUMEN

Tumorigenesis is an important problem that needs to be addressed in the field of human stem/progenitor cell transplantation for the treatment of subacute spinal cord injury (SCI). When certain "tumorigenic" cell lines are transplanted into the spinal cord of SCI mice model, there is initial improvement of motor function, followed by abrupt deterioration secondary to the effect of tumor growth. A significant proportion of the transplanted cells remains undifferentiated after transplantation and is thought to increase the risk of tumorigenesis. In this study, using lentiviral vectors, we introduced the herpes simplex virus type 1 thymidine kinase (HSVtk) gene into a human induced pluripotent stem cell-derived neural stem/progenitor cell (hiPSC-NS/PC) line that is known to undergo tumorigenic transformation. Such approach enables selective ablation of the immature proliferating cells and thereby prevents subsequent tumor formation. In vitro, the HSVtk system successfully ablated the immature proliferative neural cells while preserving mature postmitotic neuronal cells. Similar results were observed in vivo following transplantation into the injured spinal cords of immune-deficient (nonobese diabetic-severe combined immune-deficient) mice. Ablation of the proliferating cells exerted a protective effect on the motor function which was regained after transplantation, simultaneously defending the spinal cord from the harmful tumor growth. These results suggest a potentially promising role of suicide genes in opposing tumorigenesis during stem cell therapy. This system allows both preventing and treating tumorigenesis following hiPSC-NS/PC transplantation without sacrificing the improved motor function. Stem Cells Translational Medicine 2019;8:260&270.


Asunto(s)
Carcinogénesis/patología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Traumatismos de la Médula Espinal/terapia , Médula Espinal/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neuronas/fisiología , Recuperación de la Función/fisiología , Trasplante de Células Madre/métodos
4.
Stem Cells ; 37(1): 6-13, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30371964

RESUMEN

There have been numerous attempts to develop stem cell transplantation approaches to promote the regeneration of spinal cord injury (SCI). Our multicenter team is currently planning to launch a first-in-human clinical study of an induced pluripotent stem cell (iPSC)-based cell transplant intervention for subacute SCI. This trial was conducted as class I regenerative medicine protocol as provided for under Japan's Act on the Safety of Regenerative Medicine, using neural stem/progenitor cells derived from a clinical-grade, integration-free human "iPSC stock" generated by the Kyoto University Center for iPS Cell Research and Application. In the present article, we describe how we are preparing to initiate this clinical study, including addressing the issues of safety and tumorigenesis as well as practical problems that must be overcome to enable the development of therapeutic interventions for patients with chronic SCI. Stem Cells 2019;37:6-13.


Asunto(s)
Células Madre Pluripotentes Inducidas/trasplante , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/métodos , Humanos
5.
Sci Rep ; 7(1): 12996, 2017 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-29021610

RESUMEN

Resolving the immunogenicity of cells derived from induced pluripotent stem cells (iPSCs) remains an important challenge for cell transplant strategies that use banked allogeneic cells. Thus, we evaluated the immunogenicity of mouse fetal neural stem/progenitor cells (fetus-NSPCs) and iPSC-derived neural stem/progenitor cells (iPSC-NSPCs) both in vitro and in vivo. Flow cytometry revealed the low expression of immunological surface antigens, and these cells survived in all mice when transplanted syngeneically into subcutaneous tissue and the spinal cord. In contrast, an allogeneic transplantation into subcutaneous tissue was rejected in all mice, and allogeneic cells transplanted into intact and injured spinal cords survived for 3 months in approximately 20% of mice. In addition, cell survival was increased after co-treatment with an immunosuppressive agent. Thus, the immunogenicity and post-transplantation immunological dynamics of iPSC-NSPCs resemble those of fetus-NSPCs.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/inmunología , Animales , Proliferación Celular , Supervivencia Celular , Feto/citología , Regulación del Desarrollo de la Expresión Génica , Inflamación/patología , Lentivirus/genética , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Mediciones Luminiscentes , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Médula Espinal/patología , Traumatismos de la Médula Espinal/patología , Transducción Genética
6.
Stem Cell Reports ; 8(3): 673-684, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28262544

RESUMEN

Human induced pluripotent stem cells (iPSCs) are promising in regenerative medicine. However, the risks of teratoma formation and the overgrowth of the transplanted cells continue to be major hurdles that must be overcome. Here, we examined the efficacy of the inducible caspase-9 (iCaspase9) gene as a fail-safe against undesired tumorigenic transformation of iPSC-derived somatic cells. We used a lentiviral vector to transduce iCaspase9 into two iPSC lines and assessed its efficacy in vitro and in vivo. In vitro, the iCaspase9 system induced apoptosis in approximately 95% of both iPSCs and iPSC-derived neural stem/progenitor cells (iPSC-NS/PCs). To determine in vivo function, we transplanted iPSC-NS/PCs into the injured spinal cord of NOD/SCID mice. All transplanted cells whose mass effect was hindering motor function recovery were ablated upon transduction of iCaspase9. Our results suggest that the iCaspase9 system may serve as an important countermeasure against post-transplantation adverse events in stem cell transplant therapies.


Asunto(s)
Transformación Celular Neoplásica , Células Madre Pluripotentes Inducidas/citología , Trasplante de Células Madre/efectos adversos , Animales , Apoptosis/genética , Diferenciación Celular , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Femenino , Expresión Génica , Genes Reporteros , Humanos , Ratones , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/terapia , Teratoma/etiología , Teratoma/patología
7.
Stem Cell Res ; 19: 128-138, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28135684

RESUMEN

To achieve the goal of a first-in-human trial for human induced pluripotent stem cell (hiPSC)-based transplantation for the treatment of various diseases, allogeneic human leukocyte antigen (HLA)-matched hiPSC cell banks represent a realistic tool from the perspective of quality control and cost performance. Furthermore, considering the limited therapeutic time-window for acute injuries, including neurotraumatic injuries, an iPS cell bank is of potential interest. However, due to the relatively immunoprivileged environment of the central nervous system, it is unclear whether HLA matching is required in hiPSC-derived neural stem/progenitor cell (hiPSC-NS/PC) transplantation for the treatment of neurodegenerative diseases and neurotraumatic injuries. In this study, we evaluated the significance of HLA matching in hiPSC-NS/PC transplantation by performing modified mixed lymphocyte reaction (MLR) assays with hiPSC-NS/PCs. Compared to fetus-derived NS/PCs, the expression levels of human leukocyte antigen-antigen D related (HLA-DR) and co-stimulatory molecules on hiPSC-NS/PCs were significantly low, even with the addition of tumor necrosis factor-α (TNFα) and/or interferon-γ (IFNγ) to mimic the inflammatory environment surrounding transplanted hiPSC-NS/PCs in injured tissues. Interestingly, both the allogeneic HLA-matched and the HLA-mismatched responses were similarly low in the modified MLR assay. Furthermore, the autologous response was also similar to the allogeneic response. hiPSC-NS/PCs suppressed the proliferative responses of allogeneic HLA-mismatched peripheral blood mononuclear cells (PBMCs) in a dose-dependent manner. Thus, the low antigen-presenting function and immunosuppressive effects of hiPSC-NS/PCs result in a depressed immune response, even in an allogeneic HLA-mismatched setting. It is crucial to verify whether these in vitro results are reproducible in a clinical setting.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/inmunología , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Feto/citología , Expresión Génica/efectos de los fármacos , Genotipo , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Interferón gamma/farmacología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
8.
Stem Cell Reports ; 7(4): 649-663, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27666789

RESUMEN

Neural stem/progenitor cells (NS/PCs) derived from human induced pluripotent stem cells (hiPSCs) are considered to be a promising cell source for cell-based interventions that target CNS disorders. We previously reported that transplanting certain hiPSC-NS/PCs in the spinal cord results in tumor-like overgrowth of hiPSC-NS/PCs and subsequent deterioration of motor function. Remnant immature cells should be removed or induced into more mature cell types to avoid adverse effects of hiPSC-NS/PC transplantation. Because Notch signaling plays a role in maintaining NS/PCs, we evaluated the effects of γ-secretase inhibitor (GSI) and found that pretreating hiPSC-NS/PCs with GSI promoted neuronal differentiation and maturation in vitro, and GSI pretreatment also reduced the overgrowth of transplanted hiPSC-NS/PCs and inhibited the deterioration of motor function in vivo. These results indicate that pretreatment with hiPSC-NS/PCs decreases the proliferative capacity of transplanted hiPSC-NS/PCs, triggers neuronal commitment, and improves the safety of hiPSC-based approaches in regenerative medicine.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Transformación Celular Neoplásica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Trasplante de Células Madre , Animales , Biomarcadores , Proliferación Celular , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Perfilación de la Expresión Génica , Humanos , Locomoción , Ratones , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/métodos
9.
PLoS One ; 11(4): e0153957, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27099926

RESUMEN

The spatiotemporal distribution of drugs in the inner ear cannot be precisely evaluated because of its small area and complex structure. In the present study, we used hyaluronic acid (HA)-dispersed luciferin to image transgenic mice and to determine the effect of HA on controlled drug delivery to the cochlea. GFAP-luc mice, which express luciferase in cochlear spiral ganglion cells, were subcutaneously administered HA-luciferin (HA-sc) or luciferin dissolved in saline (NS-sc) or intraperitoneally administered luciferin dissolved in saline (NS-ip). The bioluminescence of luciferin was monitored in vivo in real time. The peak time and half-life of fluorescence emission were significantly increased in HA-sc-treated mice compared with those in NS-sc- and NS-ip-treated mice; however, significant differences were not observed in peak photon counts. We detected differences in the pharmacokinetics of luciferin in the inner ear, including its sustained release, in the presence of HA. The results indicate the clinical potential of using HA for controlled drug delivery to the cochlea.


Asunto(s)
Cóclea/efectos de los fármacos , Ácido Hialurónico/administración & dosificación , Ganglio Espiral de la Cóclea/efectos de los fármacos , Animales , Cóclea/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Oído Interno/efectos de los fármacos , Oído Interno/metabolismo , Semivida , Inyecciones Intraperitoneales/métodos , Luciferasas/metabolismo , Ratones , Ratones Transgénicos , Ganglio Espiral de la Cóclea/metabolismo
10.
Stem Cell Reports ; 6(1): 1-8, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26724902

RESUMEN

Murine- and human-induced pluripotent stem cell-derived neural stem/progenitor cells (iPSC-NS/PCs) promote functional recovery following transplantation into the injured spinal cord in rodents and primates. Although remyelination of spared demyelinated axons is a critical mechanism in the regeneration of the injured spinal cord, human iPSC-NS/PCs predominantly differentiate into neurons both in vitro and in vivo. We therefore took advantage of our recently developed protocol to obtain human-induced pluripotent stem cell-derived oligodendrocyte precursor cell-enriched neural stem/progenitor cells and report the benefits of transplanting these cells in a spinal cord injury (SCI) model. We describe how this approach contributes to the robust remyelination of demyelinated axons and facilitates functional recovery after SCI.


Asunto(s)
Axones/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Traumatismos de la Médula Espinal/terapia , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Miembro Posterior/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones Endogámicos NOD , Ratones SCID , Regeneración Nerviosa/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Neuronas/citología , Neuronas/fisiología , Oligodendroglía/citología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Trasplante de Células Madre/métodos , Trasplante Heterólogo
11.
Neurosci Res ; 107: 20-9, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26804710

RESUMEN

Stem cells represent a potential cellular resource in the development of regenerative medicine approaches to the treatment of pathologies in which specific cells are degenerated or damaged by genetic abnormality, disease, or injury. Securing sufficient supplies of cells suited to the demands of cell transplantation, however, remains challenging, and the establishment of safe and efficient cell banking procedures is an important goal. Cryopreservation allows the storage of stem cells for prolonged time periods while maintaining them in adequate condition for use in clinical settings. Conventional cryopreservation systems include slow-freezing and vitrification both have advantages and disadvantages in terms of cell viability and/or scalability. In the present study, we developed an advanced slow-freezing technique using a programmed freezer with a magnetic field called Cells Alive System (CAS) and examined its effectiveness on human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs). This system significantly increased cell viability after thawing and had less impact on cellular proliferation and differentiation. We further found that frozen-thawed hiPSC-NS/PCs were comparable with non-frozen ones at the transcriptome level. Given these findings, we suggest that the CAS is useful for hiPSC-NS/PCs banking for clinical uses involving neural disorders and may open new avenues for future regenerative medicine.


Asunto(s)
Criopreservación/instrumentación , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Supervivencia Celular , Células Cultivadas , Criopreservación/métodos , Humanos , Campos Magnéticos
12.
Stem Cells Transl Med ; 4(7): 708-19, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26019226

RESUMEN

UNLABELLED: : Previous studies have demonstrated that neural stem/progenitor cells (NS/PCs) promote functional recovery in rodent animal models of spinal cord injury (SCI). Because distinct differences exist in the neuroanatomy and immunological responses between rodents and primates, it is critical to determine the effectiveness and safety of allografted embryonic stem cell (ESC)-derived NS/PCs (ESC-NS/PCs) in a nonhuman primate SCI model. In the present study, common marmoset ESC-NS/PCs were grafted into the lesion epicenter 14 days after contusive SCI in adult marmosets (transplantation group). In the control group, phosphate-buffered saline was injected instead of cells. In the presence of a low-dose of tacrolimus, several grafted cells survived without tumorigenicity and differentiated into neurons, astrocytes, or oligodendrocytes. Significant differences were found in the transverse areas of luxol fast blue-positive myelin sheaths, neurofilament-positive axons, corticospinal tract fibers, and platelet endothelial cell adhesion molecule-1-positive vessels at the lesion epicenter between the transplantation and control groups. Immunoelectron microscopic examination demonstrated that the grafted ESC-NS/PC-derived oligodendrocytes contributed to the remyelination of demyelinated axons. In addition, some grafted neurons formed synaptic connections with host cells, and some transplanted neurons were myelinated by host cells. Eventually, motor functional recovery significantly improved in the transplantation group compared with the control group. In addition, a mixed lymphocyte reaction assay indicated that ESC-NS/PCs modulated the allogeneic immune rejection. Taken together, our results indicate that allogeneic transplantation of ESC-NS/PCs from a nonhuman primate promoted functional recovery after SCI without tumorigenicity. SIGNIFICANCE: This study demonstrates that allogeneic embryonic stem cell (ESC)-derived neural stem/progenitor cells (NS/PCs) promoted functional recovery after transplantation into the injured spinal cord in nonhuman primates. ESC-NS/PCs were chosen because ESC-NS/PCs are one of the controls for induced pluripotent stem cell-derived NS/PCs and because ESC derivatives are possible candidates for clinical use. This translational research using an allograft model of a nonhuman primate is critical for clinical application of grafting NS/PCs derived from various allogeneic pluripotent stem cells, especially induced pluripotent stem cells, into injured spinal cord at the subacute phase.

13.
PLoS One ; 10(2): e0116413, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25706286

RESUMEN

Our previous work reported functional recovery after transplantation of mouse and human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs) into rodent models of spinal cord injury (SCI). Although hiPSC-NS/PCs proved useful for the treatment of SCI, the tumorigenicity of the transplanted cells must be resolved before they can be used in clinical applications. The current study sought to determine the feasibility of ablation of the tumors formed after hiPSC-NS/PC transplantation through immunoregulation. Tumorigenic hiPSC-NS/PCs were transplanted into the intact spinal cords of immunocompetent BALB/cA mice with or without immunosuppressant treatment. In vivo bioluminescence imaging was used to evaluate the chronological survival and growth of the transplanted cells. The graft survival rate was 0% in the group without immunosuppressants versus 100% in the group with immunosuppressants. Most of the mice that received immunosuppressants exhibited hind-limb paralysis owing to tumor growth at 3 months after iPSC-NS/PC transplantation. Histological analysis showed that the tumors shared certain characteristics with low-grade gliomas rather than with teratomas. After confirming the progression of the tumors in immunosuppressed mice, the immunosuppressant agents were discontinued, resulting in the complete rejection of iPSC-NS/PC-derived masses within 42 days after drug cessation. In accordance with the tumor rejection, hind-limb motor function was recovered in all of the mice. Moreover, infiltration of microglia and lymphocytes was observed during the course of tumor rejection, along with apoptosis of iPSC-NS/PC-generated cells. Thus, immune rejection can be used as a fail-safe system against potential tumorigenicity after transplantation of iPSC-NS/PCs to treat SCI.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Rechazo de Injerto/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/trasplante , Médula Espinal/efectos de los fármacos , Trasplante de Células Madre/métodos , Animales , Rechazo de Injerto/inmunología , Humanos , Inmunosupresores/farmacología , Ratones , Médula Espinal/inmunología , Médula Espinal/patología
14.
Stem Cell Reports ; 4(3): 360-73, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25684226

RESUMEN

Previously, we described the safety and therapeutic potential of neurospheres (NSs) derived from a human induced pluripotent stem cell (iPSC) clone, 201B7, in a spinal cord injury (SCI) mouse model. However, several safety issues concerning iPSC-based cell therapy remain unresolved. Here, we investigated another iPSC clone, 253G1, that we established by transducing OCT4, SOX2, and KLF4 into adult human dermal fibroblasts collected from the same donor who provided the 201B7 clone. The grafted 253G1-NSs survived, differentiated into three neural lineages, and promoted functional recovery accompanied by stimulated synapse formation 47 days after transplantation. However, long-term observation (for up to 103 days) revealed deteriorated motor function accompanied by tumor formation. The tumors consisted of Nestin(+) undifferentiated neural cells and exhibited activation of the OCT4 transgene. Transcriptome analysis revealed that a heightened mesenchymal transition may have contributed to the progression of tumors derived from grafted cells.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre , Animales , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular , Linaje de la Célula , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Análisis por Conglomerados , Biología Computacional , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Células Madre Pluripotentes Inducidas/patología , Factor 4 Similar a Kruppel , Ratones , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Transducción de Señal , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Trasplante de Células Madre/efectos adversos , Transcriptoma
15.
Cell Transplant ; 24(7): 1299-311, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24818989

RESUMEN

Recent studies have demonstrated that transplantation of induced pluripotent stem cell-derived neurospheres can promote functional recovery after spinal cord injury in rodents, as well as in nonhuman primates. However, the potential tumorigenicity of the transplanted cells remains a matter of apprehension prior to clinical applications. As a first step to overcome this concern, this study established a glioblastoma multiforme xenograft model mouse. The feasibility of controlling immune suppression to ablate the grafted cells was then investigated. The human glioblastoma multiforme cell line U251 MG was transplanted into the intact spinal cords of immunodeficient NOD/SCID mice or into those of immunocompetent C57BL/6J H-2kb mice treated with or without immunosuppressants [FK506 plus anticluster of differentiation (CD) 4 antibody (Ab), or FK506 alone]. In vivo bioluminescent imaging was used to evaluate the chronological survival of the transplanted cells. The graft survival rate was 100% (n = 9/9) in NOD/SCID mice, 0% (n = 6/6) in C57BL/6J mice without immunosuppressant treatment, and 100% (n = 37/37) in C57BL6/J mice with immunosuppressant treatment. After confirming the growth of the grafted cells in the C57/BL6J mice treated with immunosuppressants, immune suppression was discontinued. The grafted cells were subsequently rejected within 3 days in C57BL/6J mice treated with FK506 alone, as opposed to 26 days in C57BL/6J mice treated with FK506 plus anti-CD4 Ab. Histological evaluation confirmed the ablation of the grafted cells. Although this work describes a xenograft setting, the results suggest that this immunomodulatory strategy could provide a safety lock against tumor formation stemming from transplanted cells.


Asunto(s)
Glioblastoma/cirugía , Médula Espinal/cirugía , Animales , Diferenciación Celular , Trasplante de Células , Modelos Animales de Enfermedad , Femenino , Glioblastoma/tratamiento farmacológico , Rechazo de Injerto , Humanos , Terapia de Inmunosupresión , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Médula Espinal/patología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Exp Neurol ; 261: 171-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24873731

RESUMEN

Spinal cord injury (SCI) is a devastating condition with no established treatment. To better understand the pathology and develop a treatment modality for SCI, an understanding of the physiological changes following SCI at the molecular level is essential. However, studies on SCI have primarily used rodent models, and few studies have examined SCI in non-human primates. In this study, we analyzed the temporal changes in gene expression patterns following SCI in common marmosets (Callithrix jacchus) using microarray analysis and mRNA deep sequencing. This analysis revealed that, although the sequence of events is comparable between primates and rodents, the inflammatory response following SCI is significantly prolonged and the onset of glial scar formation is temporally delayed in primates compared with rodents. These observations indicate that the optimal time window to treat SCI significantly differs among different species. This study provides the first extensive analysis of gene expression following SCI in non-human primates and will serve as a valuable resource in understanding the pathology of SCI.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Expresión Génica/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Animales , Callithrix , Modelos Animales de Enfermedad , Femenino , Redes Reguladoras de Genes , Análisis por Micromatrices , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Traumatismos de la Médula Espinal/patología
17.
PLoS One ; 7(12): e52787, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300777

RESUMEN

Murine and human iPSC-NS/PCs (induced pluripotent stem cell-derived neural stem/progenitor cells) promote functional recovery following transplantation into the injured spinal cord in rodents. However, for clinical applicability, it is critical to obtain proof of the concept regarding the efficacy of grafted human iPSC-NS/PCs (hiPSC-NS/PCs) for the repair of spinal cord injury (SCI) in a non-human primate model. This study used a pre-evaluated "safe" hiPSC-NS/PC clone and an adult common marmoset (Callithrix jacchus) model of contusive SCI. SCI was induced at the fifth cervical level (C5), followed by transplantation of hiPSC-NS/PCs at 9 days after injury. Behavioral analyses were performed from the time of the initial injury until 12 weeks after SCI. Grafted hiPSC-NS/PCs survived and differentiated into all three neural lineages. Furthermore, transplantation of hiPSC-NS/PCs enhanced axonal sparing/regrowth and angiogenesis, and prevented the demyelination after SCI compared with that in vehicle control animals. Notably, no tumor formation occurred for at least 12 weeks after transplantation. Quantitative RT-PCR showed that mRNA expression levels of human neurotrophic factors were significantly higher in cultured hiPSC-NS/PCs than in human dermal fibroblasts (hDFs). Finally, behavioral tests showed that hiPSC-NS/PCs promoted functional recovery after SCI in the common marmoset. Taken together, these results indicate that pre-evaluated safe hiPSC-NS/PCs are a potential source of cells for the treatment of SCI in the clinic.


Asunto(s)
Transformación Celular Neoplásica/patología , Células Madre Pluripotentes Inducidas/trasplante , Células-Madre Neurales/trasplante , Traumatismos de la Médula Espinal/terapia , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Callithrix , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Cicatriz/patología , Enfermedades Desmielinizantes/prevención & control , Femenino , Fuerza de la Mano , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Locomoción , Microglía/fisiología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Neovascularización Fisiológica , Factores de Crecimiento Nervioso/metabolismo , Regeneración Nerviosa , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Recuperación de la Función , Médula Espinal/irrigación sanguínea , Médula Espinal/patología , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/patología , Trasplante de Células Madre/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA