Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(21)2022 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-36362211

RESUMEN

Translation of cell therapies into clinical practice requires the adoption of robust production protocols in order to optimize and standardize the manufacture and cryopreservation of cells, in compliance with good manufacturing practice regulations. Between 2012 and 2020, we conducted two phase I clinical trials (EudraCT 2009-014484-39, EudraCT 2015-004855-37) on amyotrophic lateral sclerosis secondary progressive multiple sclerosis patients, respectively, treating them with human neural stem cells. Our production process of a hNSC-based medicinal product is the first to use brain tissue samples extracted from fetuses that died in spontaneous abortion or miscarriage. It consists of selection, isolation and expansion of hNSCs and ends with the final pharmaceutical formulation tailored to a specific patient, in compliance with the approved clinical protocol. The cells used in these clinical trials were analyzed in order to confirm their microbiological safety; each batch was also tested to assess identity, potency and safety through morphological and functional assays. Preclinical, clinical and in vitro nonclinical data have proved that our cells are safe and stable, and that the production process can provide a high level of reproducibility of the cultures. Here, we describe the quality control strategy for the characterization of the hNSCs used in the above-mentioned clinical trials.


Asunto(s)
Esclerosis Amiotrófica Lateral , Células-Madre Neurales , Humanos , Reproducibilidad de los Resultados , Criopreservación , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Control de Calidad
2.
Int J Mol Sci ; 21(5)2020 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-32182809

RESUMEN

Recent cutting-edge human genetics technology has allowed us to identify copy number variations (CNVs) and has provided new insights for understanding causative mechanisms of human diseases. A growing number of studies show that CNVs could be associated with physiological mechanisms linked to evolutionary trigger, as well as to the pathogenesis of various diseases, including cancer, autoimmune disease and mental disorders such as autism spectrum disorders, schizophrenia, intellectual disabilities or attention-deficit/hyperactivity disorder. Their incomplete penetrance and variable expressivity make diagnosis difficult and hinder comprehension of the mechanistic bases of these disorders. Additional elements such as co-presence of other CNVs, genomic background and environmental factors are involved in determining the final phenotype associated with a CNV. Genetically engineered animal models are helpful tools for understanding the behavioral consequences of CNVs. However, the genetic background and the biology of these animal model systems have sometimes led to confusing results. New cellular models obtained through somatic cellular reprogramming technology that produce induced pluripotent stem cells (iPSCs) from human subjects are being used to explore the mechanisms involved in the pathogenic consequences of CNVs. Considering the vast quantity of CNVs found in the human genome, we intend to focus on reviewing the current literature on the use of iPSCs carrying CNVs on chromosome 15, highlighting advantages and limits of this system with respect to mouse model systems.


Asunto(s)
Sistema Nervioso Central/fisiología , Cromosomas Humanos Par 15/genética , Variaciones en el Número de Copia de ADN/genética , Enfermedades Genéticas Congénitas/genética , Inestabilidad Genómica/genética , Células Madre Pluripotentes Inducidas/fisiología , Animales , Reprogramación Celular/genética , Humanos
3.
Nat Chem Biol ; 13(9): 951-955, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28671681

RESUMEN

Extracellular vesicles (EVs) are membrane particles involved in the exchange of a broad range of bioactive molecules between cells and the microenvironment. Although it has been shown that cells can traffic metabolic enzymes via EVs, much remains to be elucidated with regard to their intrinsic metabolic activity. Accordingly, herein we assessed the ability of neural stem/progenitor cell (NSC)-derived EVs to consume and produce metabolites. Our metabolomics and functional analyses both revealed that EVs harbor L-asparaginase activity, catalyzed by the enzyme asparaginase-like protein 1 (Asrgl1). Critically, we show that Asrgl1 activity is selective for asparagine and is devoid of glutaminase activity. We found that mouse and human NSC EVs traffic Asrgl1. Our results demonstrate, for the first time, that NSC EVs function as independent metabolic units that are able to modify the concentrations of critical nutrients, with the potential to affect the physiology of their microenvironment.


Asunto(s)
Asparaginasa/metabolismo , Vesículas Extracelulares/metabolismo , Modelos Biológicos
4.
J Transl Med ; 13: 17, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25889343

RESUMEN

BACKGROUND: We report the initial results from a phase I clinical trial for ALS. We transplanted GMP-grade, fetal human neural stem cells from natural in utero death (hNSCs) into the anterior horns of the spinal cord to test for the safety of both cells and neurosurgical procedures in these patients. The trial was approved by the Istituto Superiore di Sanità and the competent Ethics Committees and was monitored by an external Safety Board. METHODS: Six non-ambulatory patients were treated. Three of them received 3 unilateral hNSCs microinjections into the lumbar cord tract, while the remaining ones received bilateral (n = 3 + 3) microinjections. None manifested severe adverse events related to the treatment, even though nearly 5 times more cells were injected in the patients receiving bilateral implants and a much milder immune-suppression regimen was used as compared to previous trials. RESULTS: No increase of disease progression due to the treatment was observed for up to18 months after surgery. Rather, two patients showed a transitory improvement of the subscore ambulation on the ALS-FRS-R scale (from 1 to 2). A third patient showed improvement of the MRC score for tibialis anterior, which persisted for as long as 7 months. The latter and two additional patients refused PEG and invasive ventilation and died 8 months after surgery due to the progression of respiratory failure. The autopsies confirmed that this was related to the evolution of the disease. CONCLUSIONS: We describe a safe cell therapy approach that will allow for the treatment of larger pools of patients for later-phase ALS clinical trials, while warranting good reproducibility. These can now be carried out under more standardized conditions, based on a more homogenous repertoire of clinical grade hNSCs. The use of brain tissue from natural miscarriages eliminates the ethical concerns that may arise from the use of fetal material. TRIAL REGISTRATION: EudraCT:2009-014484-39 .


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Células-Madre Neurales/citología , Trasplante de Células Madre , Adulto , Anciano , Animales , Técnicas de Cultivo de Célula , Sistema Nervioso Central/patología , Bandeo Cromosómico , Progresión de la Enfermedad , Femenino , Humanos , Terapia de Inmunosupresión , Péptidos y Proteínas de Señalización Intercelular , Italia , Cariotipificación , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Médula Espinal/citología
5.
Cell Mol Life Sci ; 69(7): 1193-210, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22076651

RESUMEN

Cell therapy is reaching the stage of phase I clinical trials for post-traumatic, post-ischemic, or neurodegenerative disorders, and the selection of the appropriate cell source is essential. In order to assess the capacity of different human neural stem cell lines (hNSC) to contribute to neural tissue regeneration and to reduce the local inflammation after an acute injury, we transplanted GMP-grade non-immortalized hNSCs and v-myc (v-IhNSC), c-myc T58A (T-IhNSC) immortalized cells into the corpus callosum of adult rats after 5 days from focal demyelination induced by lysophosphatidylcholine. At 15 days from transplantation, hNSC and T-IhNSC migrated to the lesioned area where they promoted endogenous remyelination and differentiated into mature oligodendrocytes, while the all three cell lines were able to integrate in the SVZ. Moreover, where demyelination was accompanied by an inflammatory reaction, a significant reduction of microglial cells' activation was observed. This effect correlated with a differential migratory pattern of transplanted hNSC and IhNSC, significantly enhanced in the former, thus suggesting a specific NSC-mediated immunomodulatory effect on the local inflammation. We provide evidence that, in the subacute phase of a demyelination injury, different human immortalized and non-immortalized NSC lines, all sharing homing to the stem niche, display a differential pathotropism, both through cell-autonomous and non-cell autonomous effects. Overall, these findings promote IhNSC as an inexhaustible cell source for large-scale preclinical studies and non-immortalized GMP grade hNSC lines as an efficacious, safe, and reliable therapeutic tool for future clinical applications.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Células-Madre Neurales/metabolismo , Animales , Línea Celular , Supervivencia Celular , Trasplante de Células , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Células-Madre Neurales/citología , Fenotipo , Ratas
6.
Stem Cell Res ; 67: 103023, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36638628

RESUMEN

Familial Hypocalciuric Hypercalcemia (FHH1) is a rare autosomal dominant disease with low penetrance, caused by inactivating mutations of the calcium-sensing receptor (CaSR) gene, characterized by significant hypercalcemia, inappropriately normal serum PTH levels and a low urinary calcium level. Human induced pluripotent stem cells (hiPSCs) from a patient carrying a previously identified heterozygous mutation, a p.T972M amino acid substitution in cytoplasmic tail of CasR, were produced using a virus, xeno-free and non-integrative protocol.


Asunto(s)
Hipercalcemia , Células Madre Pluripotentes Inducidas , Humanos , Mutación Puntual , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Hipercalcemia/genética , Mutación , Calcio
7.
Cell Stem Cell ; 30(12): 1597-1609.e8, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38016468

RESUMEN

We report the analysis of 1 year of data from the first cohort of 15 patients enrolled in an open-label, first-in-human, dose-escalation phase I study (ClinicalTrials.gov: NCT03282760, EudraCT2015-004855-37) to determine the feasibility, safety, and tolerability of the transplantation of allogeneic human neural stem/progenitor cells (hNSCs) for the treatment of secondary progressive multiple sclerosis. Participants were treated with hNSCs delivered via intracerebroventricular injection in combination with an immunosuppressive regimen. No treatment-related deaths nor serious adverse events (AEs) were observed. All participants displayed stability of clinical and laboratory outcomes, as well as lesion load and brain activity (MRI), compared with the study entry. Longitudinal metabolomics and lipidomics of biological fluids identified time- and dose-dependent responses with increased levels of acyl-carnitines and fatty acids in the cerebrospinal fluid (CSF). The absence of AEs and the stability of functional and structural outcomes are reassuring and represent a milestone for the safe translation of stem cells into regenerative medicines.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Esclerosis Múltiple Crónica Progresiva , Esclerosis Múltiple , Células-Madre Neurales , Humanos , Esclerosis Múltiple Crónica Progresiva/tratamiento farmacológico , Esclerosis Múltiple/terapia , Trasplante Autólogo
8.
Methods Mol Biol ; 2389: 57-66, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34558001

RESUMEN

NSCs have been demonstrated to be very useful in grafts into the mammalian central nervous system to investigate the exploitation of NSC for the therapy of neurodegenerative disorders in animal models of neurodegenerative diseases. To push cell therapy in CNS on stage of clinical application, it is necessary to establish a continuous and standardized, clinical grade (i.e., produced following the good manufacturing practice guidelines) human neural stem cell lines.In this chapter we will illustrate some of the protocols for the production and characterization routinely used into our GMP "cell factory" for the production of "clinical grade" human neural stem cell lines already in use in clinical trials on neurodegenerative diseases, particularly amyotrophic lateral sclerosis (ALS- Clinicaltrials.gov number NCT01640067) and secondary progressive multiple sclerosis (SPMS- Clinicaltrials.gov number NCT03282760).


Asunto(s)
Células-Madre Neurales , Esclerosis Amiotrófica Lateral/terapia , Animales , Células Cultivadas , Sistema Nervioso Central , Feto , Humanos , Enfermedades Neurodegenerativas , Trasplante de Células Madre
9.
PLoS One ; 17(8): e0273679, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36040977

RESUMEN

INTRODUCTION: In central nervous system neurodegenerative disorders, stem cell-based therapies should be considered as a promising therapeutic approach. The safe use of human Neural Stem Cells (hNSCs) for the treatment of several neurological diseases is currently under evaluation of phase I/II clinical trials. Clinical application of hNSCs require the development of GMP standardized protocols capable of generating high quantities of reproducible and well characterized stem cells bearing stable functional and genetic properties. AIM: The aim of this study was to evaluate possible instabilities or modifications of the microsatellite loci in different culture passages because high culture passages represent an in vitro replicative stress leading to senescence. Experimental method: The hNSCs were characterized at different culture time points, from passage 2 to passage 25, by genetic typing at ten microsatellite loci. CONCLUSION: We showed that genetic stability at microsatellite loci is maintained by the cells even at high passages adding a further demonstration of the safety of our hNSCs GMP culture method.


Asunto(s)
Inestabilidad de Microsatélites , Células-Madre Neurales , Diferenciación Celular , Humanos , Trasplante de Células Madre
10.
Stem Cell Res ; 63: 102835, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35714448

RESUMEN

Amyotrophic Lateral Sclerosis (ALS) is a fatal disease affecting both upper and lower motoneurons. The transactive response DNA binding protein (TARDBP) gene, encoding for TDP-43, is one of the most commonly mutated gene associated with familial cases of ALS (10%). We generated a human induced pluripotent stem cell (hiPSC) line from the fibroblasts of an asymptomatic subject carrying the TARDBP p.G376D mutation. This mutation is very rare and was described in a large Apulian family, in which all ALS affected members are carriers of the mutation. The subject here described is the first identified asymptomatic carrier of the mutation.


Asunto(s)
Esclerosis Amiotrófica Lateral , Células Madre Pluripotentes Inducidas , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Heterocigoto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Mutación/genética
11.
Antioxidants (Basel) ; 11(5)2022 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-35624679

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of the upper and lower motor neurons (MNs). About 10% of patients have a family history (familial, fALS); however, most patients seem to develop the sporadic form of the disease (sALS). SOD1 (Cu/Zn superoxide dismutase-1) is the first studied gene among the ones related to ALS. Mutant SOD1 can adopt multiple misfolded conformation, lose the correct coordination of metal binding, decrease structural stability, and form aggregates. For all these reasons, it is complicated to characterize the conformational alterations of the ALS-associated mutant SOD1, and how they relate to toxicity. In this work, we performed a multilayered study on fibroblasts derived from two ALS patients, namely SOD1L145F and SOD1S135N, carrying the p.L145F and the p.S135N missense variants, respectively. The patients showed diverse symptoms and disease progression in accordance with our bioinformatic analysis, which predicted the different effects of the two mutations in terms of protein structure. Interestingly, both mutations had an effect on the fibroblast energy metabolisms. However, while the SOD1L145F fibroblasts still relied more on oxidative phosphorylation, the SOD1S135N fibroblasts showed a metabolic shift toward glycolysis. Our study suggests that SOD1 mutations might lead to alterations in the energy metabolism.

12.
Biomedicines ; 9(9)2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34572266

RESUMEN

Herein, we explored the impact of the lysosome dysfunction during the progression of Amyotrophic Lateral Sclerosis type-1 (ALS1). We conducted the study in non-neural cells, primary fibroblasts (rFFFs), and bone marrow-mesenchymal stem cells (rBM-MSCs), isolated from the animal model ratG93A for ALS1 at two stages of the disease: Pre-symptomatic-stage (ALS1-PreS) and Terminal-stage (ALS1-EndS). We documented the storage of human mutant Superoxide Dismutase 1, SOD1G93A (SOD1*) in the lysosomes of ALS1-rFFFs and ALS1-rBM-MSCs and demonstrated the hallmarks of the disease in non-neural cells as in ratG93A-ALS1-tissues. We showed that the SOD1* storage is associated with the altered glycohydrolases and proteases levels in tissues and both cell types from ALS1-PreS to ALS1-EndS. Only in ALS1-rFFFs, the lysosomes lost homeostasis, enlarge drastically, and contribute to the cell metabolic damage. Contrariwise, in ALS1-rBM-MSCs, we found a negligible metabolic dysfunction, which makes these cells' status similar to WT. We addressed this phenomenon to a safety mechanism perhaps associated with an enhanced lysosomal autophagic activity in ALS1-rBM-MSCs compared to ALS1-rFFFs, in which the lysosomal level of LC3-II/LC3I was comparable to that of WT-rFFFs. We suggested that the autophagic machinery could balance the storage of SOD1* aggregates and the lysosomal enzyme dysfunction even in ALS1-EndS-stem cells.

13.
J Immunol ; 181(8): 5711-9, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18832730

RESUMEN

The purpose of this study was to define early events during the angiogenic response of the aortic wall to injury. Rat aortic rings produced neovessels in collagen culture but lost this capacity over time. These quiescent rings responded to vascular endothelial growth factor but not to a mixture of macrophage-stimulatory cytokines and chemokines that was angiogenically active on fresh rings. Analysis of cytokine receptor expression revealed selective loss in quiescent rings of the proangiogenic chemokine receptor CXCR2, which was expressed predominantly in aortic macrophages. Pharmacologic inhibition of CXCR2 impaired angiogenesis from fresh rings but had no effect on vascular endothelial growth factor-induced angiogenesis from quiescent explants. Angiogenesis was also impaired in cultures of aortic rings from CXCR2-deficient mice. Reduced CXCR2 expression in quiescent rat aortic rings correlated with marked macrophage depletion. Pharmacologic ablation of macrophages from aortic explants blocked formation of neovessels in vitro and reduced aortic ring-induced angiogenesis in vivo. The angiogenic response of macrophage-depleted rings was completely restored by adding exogenous macrophages. Moreover, angiogenesis from fresh rings was promoted by macrophage CSF (CSF-1) and inhibited with anti-CSF-1 Ab. Thus, aortic angiogenic sprouting following injury is strongly influenced by conditions that modulate resident macrophage numbers and function.


Asunto(s)
Aorta/inmunología , Aorta/lesiones , Regulación de la Expresión Génica/inmunología , Macrófagos/inmunología , Neovascularización Fisiológica/inmunología , Receptores de Interleucina-8B/inmunología , Animales , Anticuerpos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Macrófagos/inmunología , Masculino , Ratones , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas , Ratas Endogámicas F344 , Factor A de Crecimiento Endotelial Vascular/inmunología
14.
Stem Cell Res ; 47: 101924, 2020 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32739880

RESUMEN

Among the known causative genes of familial ALS, SOD1mutation is one of the most common. It encodes for the ubiquitous detoxifying copper/zinc binding SOD1 enzyme, whose mutations selectively cause motor neuron death, although the mechanisms are not as yet clear. What is known is that mutant-mediated toxicity is not caused by loss of its detoxifying activity but by a gain-of-function. In order to better understand the pathogenic mechanisms of SOD1 mutation, a human induced pluripotent stem cell (hiPSC) line was generated from the somatic cells of a female patient carrying a missense variation in SOD1 (L145F).

15.
Cell Death Dis ; 10(5): 345, 2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-31024007

RESUMEN

Stem cells are emerging as a therapeutic option for incurable diseases, such as Amyotrophic Lateral Sclerosis (ALS). However, critical issues are related to their origin as well as to the need to deepen our knowledge of the therapeutic actions exerted by these cells. Here, we investigate the therapeutic potential of clinical-grade human neural stem cells (hNSCs) that have been successfully used in a recently concluded phase I clinical trial for ALS patients (NCT01640067). The hNSCs were transplanted bilaterally into the anterior horns of the lumbar spinal cord (four grafts each, segments L3-L4) of superoxide dismutase 1 G93A transgenic rats (SOD1 rats) at the symptomatic stage. Controls included untreated SOD1 rats (CTRL) and those treated with HBSS (HBSS). Motor symptoms and histological hallmarks of the disease were evaluated at three progressive time points: 15 and 40 days after transplant (DAT), and end stage. Animals were treated by transient immunosuppression (for 15 days, starting at time of transplantation). Under these conditions, hNSCs integrated extensively within the cord, differentiated into neural phenotypes and migrated rostro-caudally, up to 3.77 ± 0.63 cm from the injection site. The transplanted cells delayed decreases in body weight and deterioration of motor performance in the SOD1 rats. At 40DAT, the anterior horns at L3-L4 revealed a higher density of motoneurons and fewer activated astroglial and microglial cells. Accordingly, the overall survival of transplanted rats was significantly enhanced with no rejection of hNSCs observed. We demonstrated that the beneficial effects observed after stem cell transplantation arises from multiple events that counteract several aspects of the disease, a crucial feature for multifactorial diseases, such as ALS. The combination of therapeutic approaches that target different pathogenic mechanisms of the disorder, including pharmacology, molecular therapy and cell transplantation, will increase the chances of a clinically successful therapy for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Células-Madre Neurales/trasplante , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/mortalidad , Esclerosis Amiotrófica Lateral/terapia , Animales , Diferenciación Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Estimación de Kaplan-Meier , Masculino , Microglía/citología , Microglía/metabolismo , Neuronas Motoras/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Médula Espinal/patología , Superóxido Dismutasa/genética
16.
Stem Cells Transl Med ; 8(9): 887-897, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31104357

RESUMEN

The main objective of this phase I trial was to assess the feasibility and safety of microtransplanting human neural stem cell (hNSC) lines into the spinal cord of patients with amyotrophic lateral sclerosis (ALS). Eighteen patients with a definite diagnosis of ALS received microinjections of hNSCs into the gray matter tracts of the lumbar or cervical spinal cord. Patients were monitored before and after transplantation by clinical, psychological, neuroradiological, and neurophysiological assessment. For up to 60 months after surgery, none of the patients manifested severe adverse effects or increased disease progression because of the treatment. Eleven patients died, and two underwent tracheotomy as a result of the natural history of the disease. We detected a transitory decrease in progression of ALS Functional Rating Scale Revised, starting within the first month after surgery and up to 4 months after transplantation. Our results show that transplantation of hNSC is a safe procedure that causes no major deleterious effects over the short or long term. This study is the first example of medical transplantation of a highly standardized cell drug product, which can be reproducibly and stably expanded ex vivo, comprising hNSC that are not immortalized, and are derived from the forebrain of the same two donors throughout this entire study as well as across future trials. Our experimental design provides benefits in terms of enhancing both intra- and interstudy reproducibility and homogeneity. Given the potential therapeutic effects of the hNSCs, our observations support undertaking future phase II clinical studies in which increased cell dosages are studied in larger cohorts of patients. Stem Cells Translational Medicine 2019;8:887&897.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Células-Madre Neurales/trasplante , Adulto , Anciano , Esclerosis Amiotrófica Lateral/patología , Encéfalo/diagnóstico por imagen , Factor Neurotrófico Derivado del Encéfalo/análisis , Femenino , Proteína Ácida Fibrilar de la Glía/líquido cefalorraquídeo , Humanos , Inyecciones Espinales , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Dolor/etiología , Proyectos Piloto , Médula Espinal/diagnóstico por imagen , Trasplante de Células Madre/efectos adversos , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/análisis , Adulto Joven
17.
Int J Cancer ; 122(6): 1236-42, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18027853

RESUMEN

Tumor angiogenesis is a complex process that involves a series of interactions between tumor cells and endothelial cells (ECs). In vitro, glioblastoma multiforme (GBM) cells are known to induce an increase in proliferation, migration and tube formation by the ECs. We have previously shown that in human GBM specimens the proliferating ECs of the tumor vasculature express the catalytic component of telomerase, hTERT, and that telomerase can be upregulated in human ECs by exposing these cells to GBM in vitro. Here, we developed a controlled in vivo assay of tumor angiogenesis in which primary human umbilical vascular endothelial cells (HUVECs) were subcutaneously grafted with or without human GBM cells in immunocompromised mice as Matrigel implants. We found that primary HUVECs did not survive in Matrigel implants, and that telomerase upregulation had little effect on HUVEC survival. In the presence of GBM cells, however, the grafted HUVECs not only survived in Matrigel implants but developed tubule structures that integrated with murine microvessels. Telomerase upregulation in HUVECs enhanced such effect. More importantly, inhibition of telomerase in HUVECs completely abolished tubule formation and greatly reduced survival of these cells in the tumor xenografts. Our data demonstrate that telomerase upregulation by the ECs is a key requisite for GBM tumor angiogenesis.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Endotelio Vascular/enzimología , Glioblastoma/irrigación sanguínea , Neovascularización Patológica , Telomerasa/antagonistas & inhibidores , Neoplasias Encefálicas/enzimología , Línea Celular Tumoral , Citometría de Flujo , Glioblastoma/enzimología , Humanos , Inmunohistoquímica , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Results Probl Cell Differ ; 66: 307-329, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30209666

RESUMEN

Clinical trials for Parkinson's disease, which used primary brain fetal tissue, have demonstrated that neural stem cell therapy could be suitable for neurodegenerative diseases. The use of fetal tissue presents several issues that have hampered the clinical development of this approach. In addition to the ethical concerns related to the required continuous supply of fetal specimen, the necessity to use cells from multiple fetuses in a single graft greatly compounded the problem. Cell viability and composition vary in different donors, and, further, the heterogeneity in the donor cells increased the probability of immunological rejection or contamination. An ideal cell source for cell therapy is one that is renewable, thus eliminating the need for transplantation of primary fetal tissue, and that also allows for viability, sterility, cell composition, and cell maturation to be controlled, while being inherently not tumorigenic. The availability of continuous and standardized clinical grade normal human neural cells, able to combine the plasticity of fetal tissue with an extensive proliferating capacity and functional stability, would be of paramount importance for the translation of cell therapy for central nervous system (CNS) disorders into the clinic. Here we describe a well-established protocol to produce human neural stem cells following GMP guidelines that allows us to obtain "clinical grade" cell lines.


Asunto(s)
Trasplante de Tejido Fetal , Feto/citología , Células-Madre Neurales/citología , Enfermedades Neurodegenerativas/terapia , Trasplante de Células Madre , Trasplante de Tejido Encefálico , Línea Celular , Humanos , Enfermedades Neurodegenerativas/patología
19.
Cell Death Dis ; 9(10): 937, 2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-30224709

RESUMEN

Establishing specific cell lineages from human induced pluripotent stem cells (hiPSCs) is vital for cell therapy approaches in regenerative medicine, particularly for neurodegenerative disorders. While neural precursors have been induced from hiPSCs, the establishment of hiPSC-derived human neural stem cells (hiNSCs), with characteristics that match foetal hNSCs and abide by cGMP standards, thus allowing clinical applications, has not been described. We generated hiNSCs by a virus-free technique, whose properties recapitulate those of the clinical-grade hNSCs successfully used in an Amyotrophic Lateral Sclerosis (ALS) phase I clinical trial. Ex vivo, hiNSCs critically depend on exogenous mitogens for stable self-renewal and amplification and spontaneously differentiate into astrocytes, oligodendrocytes and neurons upon their removal. In the brain of immunodeficient mice, hiNSCs engraft and differentiate into neurons and glia, without tumour formation. These findings now warrant the establishment of clinical-grade, autologous and continuous hiNSC lines for clinical trials in neurological diseases such as Huntington's, Parkinson's and Alzheimer's, among others.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Adulto , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones SCID , Persona de Mediana Edad , Células-Madre Neurales/metabolismo , Enfermedades Neurodegenerativas , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Trasplante de Células Madre
20.
Expert Opin Biol Ther ; 18(8): 865-881, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30025485

RESUMEN

INTRODUCTION: Amyotrophic Lateral Sclerosis (ALS) is a progressive, incurable neurodegenerative disease that targets motoneurons. Cell-based therapies have generated widespread interest as a potential therapeutic approach but no conclusive results have yet been reported either from pre-clinical or clinical studies. AREAS COVERED: This is an integrated review of pre-clinical and clinical studies focused on the development of cell-based therapies for ALS. We analyze the biology of stem cell treatments and results obtained from pre-clinical models of ALS and examine the methods and the results obtained to date from clinical trials. We discuss scientific, clinical, and ethical issues and propose some directions for future studies. EXPERT OPINION: While data from individual studies are encouraging, stem-cell-based therapies do not yet represent a satisfactory, reliable clinical option. The field will critically benefit from the introduction of well-designed, randomized and reproducible, powered clinical trials. Comparative studies addressing key issues such as the nature, properties, and number of donor cells, the delivery mode and the selection of proper patient populations that may benefit the most from cell-based therapies are now of the essence. Multidisciplinary networks of experts should be established to empower effective translation of research into the clinic.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Trasplante de Células Madre/tendencias , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Trasplante de Células Madre/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA