Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
1.
Pharmaceuticals (Basel) ; 16(1)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36678621

RESUMEN

The main cause of erectile dysfunction (ED) is the damage in penile cavernous endothelial cells (EC). Murine primary ECs have a limited growth potential, and the easy availability of murine ECs will facilitate the study of cavernous endothelial dysfunction in rats. This study was performed to establish immortalized rat penile cavernous ECs (rEC) and investigate how they could repair erectile dysfunction in rats with cavernous nerve injury (CNI). rEC was isolated enzymatically by collagenase digestion and were cultured. An amphotropic replication-incompetent retroviral vector encoding v-myc oncogene was used to transfect rEC for immortalization (vREC). Morphological and immunohistochemical properties of vREC were examined. Eight-week-old male Sprague-Dawley rats were divided into three groups of five rats each, including group 1 = sham operation, group 2 = bilateral CN injury, group 3 = vREC (1 × 106 cells) treatment after CNI. Erectile response was assessed at 2, 4 weeks after transplantation of vREC., Penile tissue were harvested at 4 weeks after transplantation and immune−histochemical examination was performed. vREC showed the expression of CD31, vWF, cell type-specific markers for EC by RT-PCR and flowcytometry. At 2, 4 weeks after transplantation, rats with CNI had significantly lower erectile function than control group (p < 0.05). The group transplanted with vREC showed higher erectile function than the group without vRECs (p < 0.05). vREC was established and repaired erectile dysfunction in rats with CNI. This cell line may be useful for studying mechanisms and drug screening of erectile dysfunction of rats.

2.
Basic Clin Androl ; 32(1): 21, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36451096

RESUMEN

BACKGROUND: Because of limited differentiation to endothelium from mesenchymal stem cells, it has been strongly recommended to use endothelial progenitor cells for the regeneration of the damaged endothelium of corpora cavernosa. This study was performed to investigate the immortalized human cerebral endothelial cells and their capability for repairing erectile dysfunction in a rat model of cavernous nerve injury. Circulating endothelial progenitor cells were isolated from human fetal brain vasculature at the periventricular region of telencephalic tissues. Over 95% of CD 31-positive cells were sorted and cultured for 10 days. Human cerebral endothelial progenitor cells were injected into the cavernosa of rats with cavernous nerve injury. Erectile response was then assessed. In in vivo assays, rats were divided into three groups: group 1, sham operation: group 2, bilateral cavernous nerve injury: and group 3, treatment with human cerebral endothelial cells after cavernous nerve injury. RESULTS: Established immortalized circulating endothelial progenitor cells showed expression of human telomerase reverse transcriptase transcript by RT-PCR. They also showed the expression of vascular endothelial growth factor, von Willebrand factor, vascular endothelial growth factor receptor, and CD31, cell type-specific markers for endothelial cells by RT-PCR. In in vitro angiogenesis assays, they demonstrated tube formation that suggested morphological properties of endothelial progenitor cells. In in vivo assays, impaired erectile function of rat with cavernous nerve injury recovered at 2, 4, and 12 weeks after transplantation of human cerebral endothelial cells into the cavernosa. CONCLUSIONS: Telomerase reverse transcriptase-circulating endothelial progenitor cells from fetal brain vasculature could repair erectile dysfunction of rats with cavernous nerve injury.


RéSUMé: CONTEXTE: En raison de la différenciation limitée de l'endothélium à partir de cellules souches mésenchymateuses, il a été fortement recommandé d'utiliser des cellules progénitrices endothéliales pour la régénération de l'endothélium endommagé des corps caverneux. Cette étude a été réalisée pour étudier les cellules endothéliales cérébrales humaines immortalisées, et leur capacité à réparer la dysfonction érectile dans un modèle de rat avec lésion du nerf caverneux. Les cellules progénitrices endothéliales circulantes ont été isolées du système vasculaire cérébral fœtal humain dans la région périventriculaire des tissus télencéphaliques. Plus de 95% des cellules CD31 positives ont été sélectionnées et cultivées pendant 10 jours. Des cellules progénitrices endothéliales cérébrales humaines ont été injectées dans les corps caverneux de rats présentant une lésion nerveuse des corps caverneux. La réponse érectile a ensuite été évaluée. Dans les essais in vivo, les rats ont été divisés en trois groupes: groupe 1, opération simulée; groupe 2, lésion bilatérale du nerf caverneux; et groupe 3, traitement par cellules endothéliales cérébrales humaines après lésion du nerf caverneux. RéSULTATS: Les cellules progénitrices endothéliales circulantes immortalisées établies ont montré l'expression de la transcription de la transcriptase inverse de la télomérase humaine par RT-PCR. Elles ont également montré l'expression du facteur de croissance de l'endothélium vasculaire, du facteur de von Willebrand, du récepteur du facteur de croissance de l'endothélium vasculaire, et de CD31, marqueurs spécifiques du type cellulaire par RT-PCR pour les cellules endothéliales. Dans les essais in vivo, la fonction érectile altérée des rats avec lésion du nerf caverneux s'est rétablie à 2, 4 et 12 semaines après transplantation de cellules endothéliales cérébrales humaines dans les corps caverneux. CONCLUSIONS: Les cellules progénitrices endothéliales circulantes exprimant la transcriptase inverse de la télomérase, provenant du système vasculaire cérébral fœtal humain, pourraient réparer la dysfonction érectile de rats atteints de lésions des nerfs caverneux. MOTS-CLéS: Dysfonction érectile; Cellules endothéliales humaines; Transcriptase inverse de la Télomérase humaine.

3.
PLoS One ; 16(12): e0261402, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34936660

RESUMEN

INTRODUCTION: An underactive bladder can lead to difficulty in voiding that causes incomplete emptying of the bladder, suggesting the need for a new strategy to increase bladder contractility in such patients. This study was performed to investigate whether human mesenchymal stem cells (hMSCs) were capable of restoring bladder contractility in rats with underactive bladder due to bladder outlet obstruction (BOO) and enhancing their effects by overexpressing hepatocyte growth factor (HGF) in hMSCs. MATERIALS AND METHODS: The hMSCs were transplanted into the bladder wall of rats. Fifty female Sprague-Dawley rats at six weeks of age were divided into five groups: group 1: control; group 2: sham intervention; group 3: eight-week BOO; group 4: BOO rats transplanted with hMSCs; and group 5: BOO rats transplanted with hMSCs overexpressing HGF. Two weeks after the onset of BOO in groups 4 and 5, hMSCs were injected into the bladder wall. Cystometry evaluation was followed by Masson's trichrome staining of bladder tissues. Realtime PCR and immunohistochemical staining were performed to determine for hypoxia, apoptosis, and angiogenesis. RESULTS: Collagen deposition of bladder increased in BOO but decreased after transplantation of hMSCs. The increased inter-contraction interval and residual urine volume after BOO was reversed after hMSCs transplantation. The decreased maximal voiding pressure after BOO was restored by hMSCs treatment. The mRNA expression of bladder collagen1 and TGF-ß1 increased in BOO but decreased after hMSCs transplantation. The decrease in vWF-positive cells in the bladder following BOO was increased after hMSCs transplantation. Caspase 3 and TUNEL-positive apoptosis of bladder cells increased in BOO but decreased after transplantation of hMSCs. These effects were enhanced by overexpressing HGF in hMSCs. CONCLUSION: Transplantation of hMSCs into bladder wall increased the number of micro-vessels, decreased collagen deposition and apoptosis of detrusor muscle, and improved bladder underactivity. The effects were enhanced by overexpressing HGF in hMSCs. Our findings suggest that the restoration of underactive bladder using hMSCs may be used to rectify micturition disorders in patients following resolution of BOO. Further studies are needed before hMSCs can be used in clinical applications.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Trasplante de Células Madre Mesenquimatosas , Obstrucción del Cuello de la Vejiga Urinaria/cirugía , Vejiga Urinaria de Baja Actividad/cirugía , Vejiga Urinaria/fisiopatología , Animales , Línea Celular , Colágeno/genética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Factor de Crecimiento de Hepatocito/biosíntesis , Factor de Crecimiento de Hepatocito/genética , Humanos , Células Madre Mesenquimatosas/fisiología , Contracción Muscular/fisiología , Neovascularización Fisiológica/fisiología , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Regeneración/fisiología , Micción/fisiología
4.
Mol Biol Rep ; 48(4): 3549-3559, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33866496

RESUMEN

This study investigated the differentiation of transplanted transplanted mesenchymal stem cells MSCs into neuron-like cells, repair of erectile dysfunction (ED), and synergy of MSCs seeded to nanofibrous scaffolds with after transplantation around the injured cavernous nerve (CN) of rats. The synthesized polymer was electrospun in a rotating drum to prepare nanofiber meshes (NMs). Human MSCs were prepared and confirmed. Eight-week-old male Sprague-Dawley rats were divided into five groups of six each: group 1-sham operation; group 2-CN injury; group 3-MSCs treatment after CN injury; group 4-nanofibrous scaffold treatment after CN injury; and group 5-post-CN injury treatment combining a nanofibrous scaffold and MSCs (nano-MSCs). In the latter group, the damaged CN was instantly surrounded by an MSC-containing a nanofibrous scaffold in the aftermath of injury. Morphological analysis and immuno-histochemical staining in relation to nerves (Tuj1, NF, MAP2, MBP and peripherin), endothelium (vWF), smooth muscle (SMA), neurofilament (NF), and apoptosis (TUNEL) were performed. We evaluated the mean proportion expressed as a percentage of the ratio of muscle to collagen of penile cavernous smooth-muscle cells as well as the expression of cavernous SMA, NF, vWF, and TUNEL makers. Compared to the group free of CN injury, erectile function was markedly reduced in the group with CN injury at 2 and 4 weeks (p < 0.05). By contrast, compared to the sham operation group, erectile function was better in the group with MSC transplantation (p < 0.05). Similarly, by comparison to the group solely with hMSCs, erectile function was better in the group with nano-MSC transplantation (p < 0.05). Transplantation of MSCs demonstrated the neuronal differentiation. By contrast to MSCs on their own, neuronal differentiation was more significantly expressed in nano-MSCs. The mean proportion expressed as a percentage of the ratio of muscle to collagen of penile cavernous smooth-muscle cells, the expression of cavernous SMA, NF, vWF, and apoptosis improved in the cavernosum after transplantation. NMs showed synergy with MSCs for the repair of erectile dysfunction. Transplanted MSCs differentiated into neuron-like cells and repaired erectile dysfunction in the rats with CN injury. Transplanted MSCs increased the mean percentage of the collagen area of the caversnosum as well as the expression levels of cavernous neuronal, endothelial, smooth-muscle markers, and apoptosis.


Asunto(s)
Diferenciación Celular , Disfunción Eréctil/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/terapia , Actinas/genética , Actinas/metabolismo , Animales , Apoptosis , Células Cultivadas , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Pene/inervación , Poliésteres/química , Ratas , Ratas Sprague-Dawley , Andamios del Tejido/química , Tubulina (Proteína)/metabolismo
5.
Biofabrication ; 13(3)2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33849004

RESUMEN

The neurovascular unit (NVU) comprises multiple types of brain cells, including brain endothelial cells, astrocytes, pericytes, neurons, microglia, and oligodendrocytes. Each cell type contributes to the maintenance of the molecular transport barrier and brain tissue homeostasis. Several disorders and diseases of the central nervous system, including neuroinflammation, Alzheimer's disease, stroke, and multiple sclerosis, have been associated with dysfunction of the NVU. As a result, there has been increased demand for the development of NVUin vitromodels. Here, we present a three-dimensional (3D) immortalized human cell-based NVU model generated by organizing the brain microvasculature in a collagen matrix embedded with six different types of cells that comprise the NVU. By surrounding a perfusable brain endothelium with six types of NVU-composing cells, we demonstrated a significant impact of the 3D co-culture on the maturation of barrier function, which is supported by cytokines secreted from NVU-composing cells. Furthermore, NVU-composing cells alleviated the inflammatory responses induced by lipopolysaccharides. Our human cell-based NVUin vitromodel could enable elucidation of both physiological and pathological mechanisms in the human brain and evaluation of safety and efficacy in the context of high-content analysis during the process of drug development.


Asunto(s)
Barrera Hematoencefálica , Células Endoteliales , Inflamación , Neuronas , Astrocitos , Humanos , Pericitos
6.
EBioMedicine ; 60: 102989, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32920368

RESUMEN

BACKGROUND: Stem cells` (SC) functional heterogeneity and its poorly understood aetiology impedes clinical development of cell-based therapies in regenerative medicine and oncology. Recent studies suggest a strong correlation between the SC migration potential and their therapeutic efficacy in humans. Designating SC migration as a denominator of functional SC heterogeneity, we sought to identify highly migrating subpopulations within different SC classes and evaluate their therapeutic properties in comparison to the parental non-selected cells. METHODS: We selected highly migrating subpopulations from mesenchymal and neural SC (sMSC and sNSC), characterized their features including but not limited to migratory potential, trophic factor release and transcriptomic signature. To assess lesion-targeted migration and therapeutic properties of isolated subpopulations in vivo, surgical transplantation and intranasal administration of MSCs in mouse models of glioblastoma and Alzheimer's disease respectively were performed. FINDINGS: Comparison of parental non-selected cells with isolated subpopulations revealed superior motility and migratory potential of sMSC and sNSC in vitro. We identified podoplanin as a major regulator of migratory features of sMSC/sNSC. Podoplanin engineering improved oncovirolytic activity of virus-loaded NSC on distantly located glioblastoma cells. Finally, sMSC displayed more targeted migration to the tumour site in a mouse glioblastoma model and remarkably higher potency to reduce pathological hallmarks and memory deficits in transgenic Alzheimer's disease mice. INTERPRETATION: Functional heterogeneity of SC is associated with their motility and migration potential which can serve as predictors of SC therapeutic efficacy. FUNDING: This work was supported in part by the Robert Bosch Stiftung (Stuttgart, Germany) and by the IZEPHA grant.


Asunto(s)
Movimiento Celular , Células Madre/fisiología , Enfermedad de Alzheimer/terapia , Animales , Biomarcadores , Supervivencia Celular , Rastreo Celular/métodos , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Transgénicos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Viroterapia Oncolítica , Trasplante de Células Madre , Células Madre/citología , Resultado del Tratamiento
7.
Liver Int ; 40(7): 1736-1743, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32239602

RESUMEN

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) risk in chronic hepatitis B (CHB) substantially decreased in the era of potent antiviral therapy. We developed an optimized HCC risk prediction model for CHB with well-controlled viremia by nucelos(t)ide analogs (NUCs). METHOD: We analysed those who achieved virological response (VR; serum HBV-DNA < 2000 IU/mL on two consecutive assessments) by NUCs. Liver stiffness by transient elastography, ultrasonography and laboratory tests was performed at the time of confirmed VR. Patients with decompensated cirrhosis or HCC at baseline were excluded. Multivariate Cox-regression analysis was used to determine key variables to construct a novel risk-scoring model. RESULTS: Among 1511 patients, 9.5% developed HCC. Cirrhosis on ultrasonography (adjusted HR [aHR] 2.47), age (aHR 1.04), male (aHR 1.90), platelet count <135 000/uL (aHR 1.57), albumin <4.5 g/dL (aHR 1.77) and liver stiffness ≥11 kPa (aHR 6.09) were independently associated with HCC. Using these, CAMPAS model was developed with c-index of 0.874. The predicted and observed HCC probabilities were calibrated with a reliable agreement. Such results were reproduced from internal validation and external validation among the independent cohort (n = 252). The intermediate-risk (CAMPAS model score 75 ~ 161) and high-risk (score >161) groups were more likely to develop HCC compared with the low-risk group (score ≤75) with statistical significances (HRs; 4.43 and 47.693 respectively; both P < .001). CONCLUSION: CAMPAS model derived through comprehensive clinical evaluation of liver disease allowed the more delicate HCC prediction for CHB patients with well-controlled viremia by NUCs.


Asunto(s)
Carcinoma Hepatocelular , Hepatitis B Crónica , Neoplasias Hepáticas , Antivirales/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Virus de la Hepatitis B , Hepatitis B Crónica/complicaciones , Hepatitis B Crónica/tratamiento farmacológico , Humanos , Cirrosis Hepática/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Masculino , Factores de Riesgo , Viremia/tratamiento farmacológico
8.
Oncol Rep ; 43(6): 2045-2052, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32236604

RESUMEN

Although the effects of stem cells expressing anticancer genes on tumor growth have been demonstrated by many researchers in various types of cancer, relatively few studies have investigated their inhibitory effects on cancer metastasis. In the present study, we examined the inhibitory effects of cytosine deaminase (CD)/5­fluorocytosine (5­FC) and interferon­ß (IFN­ß) using genetically engineered neural stem cells (hNSCs) in a cellular and metastasis model of renal cell carcinoma (RCC). The CD/5­FC method has the advantage of minimizing damage to normal tissues since it selectively targets cancer cells by the CD gene, which converts prodrug 5­FC to the drug 5­fluorouracil. Moreover, we used hNSCs as a tool to effectively deliver the anticancer genes to the tumor site. These stem cells are known to possess tumor­tropism because of chemoattractant factors expressed in cancer cells. Therefore, we ascertained the expression of these factors in A498 cells, a cell line of RCC, and identified the A498­specific migration ability of hNSCs. We also confirmed that the proliferation of A498 cells was significantly reduced by therapeutic hNSCs in the presence of 5­FC. Furthermore, we established an A498 metastasis model. In the animal experiment, the weight of the lungs increased in response to cancer metastasis, but was normalized by hNSCs expressing CD and/or IFN­ß genes, while the incidence of liver metastasis was suppressed by the hNSCs. Overall, the results of this study demonstrate that stem cells expressing anticancer genes have the potential for use as an alternative to conventional therapy for metastatic cancer.


Asunto(s)
Carcinoma de Células Renales/terapia , Citosina Desaminasa/genética , Interferón beta/genética , Neoplasias Hepáticas/terapia , Células-Madre Neurales/citología , Trasplante de Células Madre/métodos , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Citosina Desaminasa/metabolismo , Femenino , Ingeniería Genética , Humanos , Interferón beta/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Metástasis de la Neoplasia , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Cell Biochem ; 121(2): 1586-1598, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31512776

RESUMEN

Stem cells that express therapeutic proteins have been identified to have an anticancer effects on various types of cancer. In the present study study, human neural stem cells (hNSCs) that were genetically engineered to express cytosine deaminase (CD) and human interferon-ß (IFN-ß) were used for anaplastic thyroid cancer (ATC) treatment owing to their tumor-tropic properties and therapeutic effects. CD is an enzyme that converts 5-fluorocytosine (5-FC), a prodrug, to 5-fluorouracil (5-FU) which is a medication to suppress tumor growth through DNA synthesis inhibition. Also, IFN-ß suppresses tumor growth by the induction of apoptotic process. In water soluble tetrazolium salt (WST) assay, SNU-80 cells which are human female ATC cells were cocultured with three cell types including engineered hNSCs such as HB1.F3, HB1.F3.CD, and HB1.F3.CD.IFN-ß cells on transwells and treated with 5-FC for 72 hours. Finally, the SNU-80 cell viability was reduced by the coculture with HB1.F3.CD and HB1.F3.CD.IFN-ß cells. In dichlorofluorescein diacetate (DCF-DA) and TdT-mediated dUTP nick-end labeling (TUNEL) assays, the production of reactive oxygen species (ROS) and the number of apoptotic cells were increased by HB1.F3.CD and HB1.F3.CD.IFN-ß cells in the presence of 5-FC. In Western blot assay, ROS, and apoptosis-related genes were increased in SNU-80 cells when they were cocultured with HB1.F3.CD and HB1.F3.CD.IFN-ß cells. In transwell migration assay, hNSCs selectively migrated to SNU-80 cells because hNSCs interacted with chemoattractant factors like SDF-1α, uPAR, and CCR2 secreted by SNU-80 cells. Taken together, engineered hNSCs were revealed to selectively migrate to ATC cells and to inhibit growth as well as to induce apoptosis of ATC cells via ROS production through the actions of transgenes such as CD and IFN-ß. Therefore, these engineered hNSCs can be promising candidates for the treatment of metastatic ATC.


Asunto(s)
Citosina Desaminasa/metabolismo , Flucitosina , Expresión Génica , Células-Madre Neurales/enzimología , Carcinoma Anaplásico de Tiroides , Línea Celular Tumoral , Técnicas de Cocultivo , Citosina Desaminasa/genética , Flucitosina/farmacocinética , Flucitosina/farmacología , Humanos , Profármacos/farmacocinética , Profármacos/farmacología , Carcinoma Anaplásico de Tiroides/metabolismo , Carcinoma Anaplásico de Tiroides/patología , Carcinoma Anaplásico de Tiroides/terapia , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/terapia
10.
Glia ; 67(9): 1667-1679, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31050055

RESUMEN

During postnatal neurodevelopment, excessive synapses must be eliminated by microglia to complete the establishment of neural circuits in the brain. The lack of synaptic regulation by microglia has been implicated in neurodevelopmental disorders such as autism, schizophrenia, and intellectual disability. Here we suggest that vaccinia-related kinase 2 (VRK2), which is expressed in microglia, may stimulate synaptic elimination by microglia. In VRK2-deficient mice (VRK2KO ), reduced numbers of presynaptic puncta within microglia were observed. Moreover, the numbers of presynaptic puncta and synapses were abnormally increased in VRK2KO mice by the second postnatal week. These differences did not persist into adulthood. Even though an increase in the number of synapses was normalized, adult VRK2KO mice showed behavioral defects in social behaviors, contextual fear memory, and spatial memory.


Asunto(s)
Encéfalo/enzimología , Encéfalo/crecimiento & desarrollo , Microglía/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Sinapsis/enzimología , Animales , Encéfalo/citología , Células Cultivadas , Potenciales Postsinápticos Excitadores/fisiología , Miedo/fisiología , Humanos , Masculino , Memoria/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Potenciales Postsinápticos Miniatura/fisiología , Proteínas Serina-Treonina Quinasas/genética , Conducta Social , Técnicas de Cultivo de Tejidos
11.
Cancer Res Treat ; 51(2): 797-811, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30213181

RESUMEN

PURPOSE: In the present study, human neural stem cells (hNSCs) with tumor-tropic behavior were used as drug delivery vehicle to selectively target melanoma. A hNSC line (HB1.F3) was transduced into two types: one expressed only the cytosine deaminase (CD) gene (HB1.F3. CD) and the other expressed both CD and human interferon-ß (IFN-ß) genes (HB1.F3.CD. IFN-ß). MATERIALS AND METHODS: This study verified the tumor-tropic migratory competence of engineered hNSCs on melanoma (A375SM) using a modified Boyden chamber assay in vitro and CM-DiI staining in vivo. The antitumor effect of HB1.F3.CD and HB1.F3.CD.IFN-ß on melanoma was also confirmed using an MTT assay in vitro and xenograft mouse models. RESULTS: A secreted form of IFN-ß from the HB1.F3.CD.IFN-ß cells modified the epithelial-mesenchymal transition (EMT) process and metastasis of melanoma. 5-Fluorouracil treatment also accelerated the expression of the pro-apoptotic protein BAX and decelerated the expression of the anti-apoptotic protein Bcl-xL on melanoma cell line. CONCLUSION: Our results illustrate that engineered hNSCs prevented malignant melanoma cells from proliferating in the presence of the prodrug, and the form that secreted IFN-ß intervened in the EMT process and melanoma metastasis. Hence, neural stem cell-directed enzyme/prodrug therapy is a plausible treatment for malignant melanoma.


Asunto(s)
Melanoma/terapia , Trasplante de Células Madre , Células Madre/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Fluorouracilo/farmacología , Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ingeniería Genética , Humanos , Interferón beta/genética , Melanoma/prevención & control , Ratones , Células-Madre Neurales/metabolismo , Trasplante de Células Madre/métodos , Células Madre/efectos de los fármacos , Transducción Genética , Transgenes , Resultado del Tratamiento , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cytotherapy ; 20(9): 1191-1201, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30078654

RESUMEN

To confirm the anti-tumor effect of engineered neural stem cells (NSCs) expressing cytosine deaminase (CD) and interferon-ß (IFN-ß) with prodrug 5-fluorocytosine (FC), K562 chronic myeloid leukemia (CML) cells were co-cultured with the neural stem cell lines HB1.F3.CD and HB1.F3.CD.IFN-ß in 5-FC containing media. A significant decrease in the viability of K562 cells was observed by the treatment of the NSC lines, HB1.F3.CD and HB1.F3.CD.IFN-ß, compared with the control. A modified trans-well assay showed that engineered human NSCs significantly migrated toward K562 CML cells more than human normal lung cells. In addition, the important chemoattractant factors involved in the specific migration ability of stem cells were found to be expressed in K562 CML cells. In a xenograft mouse model, NSC treatments via subcutaneous and intravenous injections resulted in significant inhibitions of tumor mass growth and extended survival dates of the mice. Taken together, these results suggest that gene therapy using genetically engineered stem cells expressing CD and IFN-ß may be effective for treating CML in these mouse models.


Asunto(s)
Células-Madre Neurales/trasplante , Animales , Técnicas de Cocultivo , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Femenino , Flucitosina/farmacología , Ingeniería Genética , Terapia Genética/métodos , Humanos , Interferón beta/genética , Interferón beta/metabolismo , Células K562 , Leucemia/terapia , Ratones Desnudos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Profármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Transl Oncol ; 11(1): 74-85, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29202279

RESUMEN

Cancer treatments using stem cells expressing therapeutic genes have been identified for various types of cancers. In this study, we investigated inhibitory effects of HB1.F3.CD and HB1.F3.CD.IFN-ß cells expressing Escherichia coli cytosine deaminase (CD) and human interferon-ß (IFN-ß) genes in intravenously (i.v.) injected mice with a metastasis model. In this treatment, pro-drug 5-fluorocytosine (5-FC) is converted to cytotoxic drug 5-fluorouracil by hNSCs expressing the CD gene, which inhibits DNA synthesis in cancer cells. Moreover, IFN-ß induces apoptosis and reduces the growth of cancer cells. Upon MTT assay, proliferation of choriocarcinoma (JEG-3) cells decreased when co-cultured with hNSCs expressing CD and IFN-ß genes. To confirm the cancer-tropic effect of these stem cells, chemoattractant factors (VEGF, CXCR4, and C-kit) secreted from JEG-3 cells were identified by polymerase chain reaction. hNSCs migrate toward JEG-3 cells due to ligand-receptor interactions of these factors. Accordingly, the migration capability of hNSCs toward JEG-3 cells was confirmed using an in vitro Trans-well assay, in vivo subcutaneously (s.c.) injected mice groups (xenograft model), and metastasis model. Intravenously injected hNSCs migrated freely to other organs when compared to s.c. injected hNSCs. Thus, we confirmed the inhibition of lung and ovarian metastasis of choriocarcinoma by i.v. injected HB1.F3.CD or HB1.F3.CD.IFN-ß cells in the presence of 5-FC. Treatment of these stem cells also increased the survival rates of mice. In conclusion, this study showed that metastatic cancer was diminished by genetically engineered hNSCs and noncytotoxic drug 5-FC. This is the first report of the therapeutic potential of i.v. injected hNSCs in a metastasis model; therefore, the results indicate that this stem cell therapy can be used as an alternative novel tool to treat metastatic choriocarcinoma.

14.
Brain Behav Immun ; 66: 347-358, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28760504

RESUMEN

Alzheimer's disease (AD), which is the most commonly encountered neurodegenerative disease, causes synaptic dysfunction and neuronal loss due to various pathological processes that include tau abnormality and amyloid beta (Aß) accumulation. Aß stimulates the secretion and the synthesis of Receptor for Advanced Glycation End products (RAGE) ligand by activating microglial cells, and has been reported to cause neuronal cell death in Aß1-42 treated rats and in mice with neurotoxin-induced Parkinson's disease. The soluble form of RAGE (sRAGE) is known to reduce inflammation, and to decrease microglial cell activation and Aß deposition, and thus, it protects from neuronal cell death in AD. However, sRAGE protein has too a short half-life for therapeutic purposes. We developed sRAGE-secreting umbilical cord derived mesenchymal stem cells (sRAGE-MSCs) to enhance the inhibitory effects of sRAGE on Aß deposition and to reduce the secretion and synthesis of RAGE ligands in 5xFAD mice. In addition, these cells improved the viability of injected MSCs, and enhanced the protective effects of sRAGE by inhibiting the binding of RAGE and RAGE ligands in 5xFAD mice. These findings suggest sRAGE protein from sRAGE-MSCs has better protection against neuronal cell death than sRAGE protein or single MSC treatment by inhibiting the RAGE cell death cascade and RAGE-induce inflammation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apoptosis , Encéfalo/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Trasplante de Células Madre de Sangre del Cordón Umbilical , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Ratones Transgénicos , Microglía/metabolismo , Receptor para Productos Finales de Glicación Avanzada/genética
15.
Cell Transplant ; 26(2): 271-281, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-27524466

RESUMEN

Despite the increasing importance of minipigs in biomedical research, there has been relatively little research concerning minipig-derived adult stem cells as a promising research tool that could be used to develop stem cell-based therapies. We first generated immortalized neural stem cells (iNSCs) from primary minipig olfactory bulb cells (pmpOBCs) and defined the characteristics of the cell line. Primary neural cells were prepared from minipig neonate olfactory bulbs and immortalized by infection with retrovirus carrying the v-myc gene. The minipig iNSCs (mpiNSCs) had normal karyotypes and expressed NSC-specific markers, including nestin, vimentin, Musashi1, and SOX2, suggesting a similarity to human NSCs. On the basis of the global gene expression profiles from the microarray analysis, neurogenesis-associated transcript levels were predominantly altered in mpiNSCs compared with pmpOBCs. These findings increase our understanding of minipig stem cells and contribute to the utility of mpiNSCs as resources for immortalized stem cell experiments.


Asunto(s)
Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Bulbo Olfatorio/citología , Animales , Técnicas de Cultivo de Célula , Proteínas del Tejido Nervioso/metabolismo , Nestina/metabolismo , Factores de Transcripción SOXB1/metabolismo , Porcinos , Porcinos Enanos , Vimentina/metabolismo
16.
J Cell Physiol ; 232(5): 1123-1134, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27580405

RESUMEN

Vascular inflammation is characteristic feature of diabetic retinopathy. In diabetic retina, a variety of the pro-inflammatory cytokines are elevated and involved in endothelial dysfunction. STAT3 transcription factor has been implicated in mediating cytokine signaling during vascular inflammation. However, whether and how STAT3 is involved in the direct regulation of the endothelial permeability is currently undefined. Our studies revealed that IL-6-induced STAT3 activation increases retinal endothelial permeability and vascular leakage in retinas of mice through the reduced expression of the tight junction proteins ZO-1 and occludin. In a co-culture model with microglia and endothelial cells under a high glucose condition, the microglia-derived IL-6 induced STAT3 activation in the retinal endothelial cells, leading to increasing endothelial permeability. In addition, IL-6-induced STAT3 activation was independent of ROS generation in the retinal endothelial cells. Moreover, we demonstrated that STAT3 activation downregulates the ZO-1 and occludin levels and increases the endothelial permeability through the induction of VEGF production in retinal endothelial cells. These results suggest the potential importance of IL-6/STAT3 signaling in regulating endothelial permeability and provide a therapeutic target to prevent the pathology of diabetic retinopathy. J. Cell. Physiol. 232: 1123-1134, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Regulación hacia Abajo , Células Endoteliales/metabolismo , Ocludina/metabolismo , Vasos Retinianos/patología , Factor de Transcripción STAT3/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Animales , Línea Celular , Permeabilidad de la Membrana Celular , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Glucosa/toxicidad , Humanos , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Retina/efectos de los fármacos , Retina/patología , Vasos Retinianos/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Cancer Res Treat ; 49(1): 79-91, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27188205

RESUMEN

PURPOSE: Genetically engineered stem cells may be advantageous for gene therapy against various human cancers due to their inherent tumor-tropic properties. In this study, genetically engineered human neural stem cells (HB1.F3) expressing Escherichia coli cytosine deaminase (CD) (HB1.F3.CD) and human interferon-ß (IFN-ß) (HB1.F3.CD.IFN-ß) were employed against lymph node-derived metastatic colorectal adenocarcinoma. MATERIALS AND METHODS: CD can convert a prodrug, 5-fluorocytosine (5-FC), to active 5-fluorouracil, which inhibits tumor growth through the inhibition of DNA synthesis,while IFN-ß also strongly inhibits tumor growth by inducing the apoptotic process. In reverse transcription polymerase chain reaction analysis, we confirmed that HB1.F3.CD cells expressed the CD gene and HB1.F3.CD.IFN-ß cells expressed both CD and IFN-ß genes. RESULTS: In results of a modified trans-well migration assay, HB1.F3.CD and HB1.F3.CD.IFN-ß cells selectively migrated toward SW-620, human lymph node-derived metastatic colorectal adenocarcinoma cells. The viability of SW-620 cells was significantly reduced when co-cultured with HB1.F3.CD or HB1.F3.CD.IFN-ß cells in the presence of 5-FC. In addition, it was found that the tumor-tropic properties of these engineered human neural stem cells (hNSCs) were attributed to chemoattractant molecules including stromal cell-derived factor 1, c-Kit, urokinase receptor, urokinase-type plasminogen activator, and C-C chemokine receptor type 2 secreted by SW-620 cells. In a xenograft mouse model, treatment with hNSC resulted in significantly inhibited growth of the tumor mass without virulent effects on the animals. CONCLUSION: The current results indicate that engineered hNSCs and a prodrug treatment inhibited the growth of SW-620 cells. Therefore, hNSC therapy may be a clinically effective tool for the treatment of lymph node metastatic colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/secundario , Citosina Desaminasa/genética , Expresión Génica , Interferón beta/genética , Células-Madre Neurales/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Neoplasias Colorrectales/terapia , Terapia Combinada , Citosina Desaminasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Flucitosina/farmacología , Ingeniería Genética , Terapia Genética , Humanos , Interferón beta/metabolismo , Ratones , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Oncotarget ; 7(46): 75319-75327, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27659534

RESUMEN

Advanced pancreatic cancer is one of the most lethal malignant human diseases lacking effective treatment. Its extremely low survival rate necessitates development of novel therapeutic approach. Human neural stem cells (NSCs) are known to have tumor-tropic effect. We genetically engineered them to express rabbit carboxyl esterase (F3.CE), which activates prodrug CPT-11(irinotecan) into potent metabolite SN-38. We found significant inhibition of the growth of BxPC3 human pancreatic cancer cell line in vitro by F3.CE in presence of CPT-11. Apoptosis was also markedly increased in BxPC3 cells treated with F3.CE and CPT-11. The ligand VEGF and receptor VEGF-1(Flt1) were identified to be the relevant tumor-tropic chemoattractant. We confirmed in vivo that in mice injected with BxPC3 on their skin, there was significant reduction of tumor size in those treated with both F3.CE and BxPC3 adjacent to the cancer mass. Administration of F3.CE in conjunction with CPT-11 could be a new possibility as an effective treatment regimen for patients suffering from advanced pancreatic cancer.


Asunto(s)
Carboxilesterasa/genética , Carboxilesterasa/metabolismo , Terapia Genética , Células-Madre Neurales/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Animales , Apoptosis/genética , Efecto Espectador/genética , Línea Celular , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Expresión Génica , Terapia Genética/métodos , Humanos , Masculino , Ratones , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Conejos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cell Transplant ; 25(12): 2129-2144, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27442084

RESUMEN

Oligodendrocytes play a crucial role in creating the myelin sheath that is an important component in neural transmission. In an animal model of transient cerebral ischemia, application of oligodendrocyte progenitor cells (OPCs) has not yet been reported. In this study, the effects of F3.Olig2 transplantation on memory and cognitive dysfunction were investigated in the aged gerbil in which ischemic stroke was induced. To investigate the possible mechanisms underlying repair, changes in the expression of myelin basic protein (MBP), oligodendrocyte-specific protein (OSP), and brain-derived neurotrophic factor (BDNF) were examined. Experimental ischemic stroke was induced by occlusion of bilateral common carotid arteries in aged gerbils. Gerbils (n=31 per group) were randomly divided into three groups: (1) vehicle sham group, (2) vehicle ischemia group, and (3) F3.Olig2 ischemia group. After 1, 3, and 7 days of ischemiareperfusion (I-R), saline or F3.Olig2 cells (1106 cells in 100 l) were injected into the gerbils intravenously. The gerbils were sacrificed 10 days after I-R for identification of grafted F3.Olig2 cells, and 15 and 30 days after I-R for tissue analysis after conducting passive avoidance and novel object recognition test. Injected F3.Olig2 cells and MBP, OSP, and BDNF were detected by specific antibodies using immunohistochemistry and/or Western blots. Memory and cognition were significantly increased in the F3.Olig2 ischemia group compared with the vehicle ischemia group. In the F3.Olig2 ischemia group, the neurons were not protected from ischemic damage; however, MBP, OSP, and BDNF expressions were significantly increased. Our results show that injection of F3.Olig2 cells significantly improved impaired memory and cognition, which might be related to increased MBP expression via increasing OSP and BDNF expression in the aged gerbil hippocampus following transient cerebral ischemia.


Asunto(s)
Ataque Isquémico Transitorio/terapia , Células-Madre Neurales/citología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Claudinas/metabolismo , Gerbillinae , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Ataque Isquémico Transitorio/metabolismo , Masculino , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Daño por Reperfusión/metabolismo , Daño por Reperfusión/terapia
20.
Stem Cells Int ; 2016: 4079863, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27429621

RESUMEN

Since multiple sclerosis (MS) is featured with widespread demyelination caused by autoimmune response, we investigated the recovery effects of F3.olig2 progenitors, established by transducing human neural stem cells (F3 NSCs) with Olig2 transcription factor, in myelin oligodendrocyte glycoprotein- (MOG-) induced experimental autoimmune encephalomyelitis (EAE) model mice. Six days after EAE induction, F3 or F3.olig2 cells (1 × 10(6)/mouse) were intravenously transplanted. MOG-injected mice displayed severe neurobehavioral deficits which were remarkably attenuated and restored by cell transplantation, in which F3.olig2 cells were superior to its parental F3 cells. Transplanted cells migrated to the injured spinal cord, matured to oligodendrocytes, and produced myelin basic proteins (MBP). The F3.olig2 cells expressed growth and neurotrophic factors including brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), ciliary neurotrophic factor (CNTF), and leukemia inhibitory factor (LIF). In addition, the transplanted cells markedly attenuated inflammatory cell infiltration, reduced cytokine levels in the spinal cord and lymph nodes, and protected host myelins. The results indicate that F3.olig2 cells restore neurobehavioral symptoms of EAE mice by regulating autoimmune inflammatory responses as well as by stimulating remyelination and that F3.olig2 progenitors could be a candidate for the cell therapy of demyelinating diseases including MS.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...