Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Mol Neurobiol ; 43(7): 3211-3250, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37356043

RESUMO

Bone marrow-derived mononuclear cells (BMMNCs) have been used for decades in preclinical and clinical studies to treat various neurological diseases. However, there is still a knowledge gap in the understanding of the underlying mechanisms of BMMNCs in the treatment of neurological diseases. In addition, prerequisite factors for the efficacy of BMMNC administration, such as the optimal route, dose, and number of administrations, remain unclear. In this review, we discuss known and unknown aspects of BMMNCs, including the cell harvesting, administration route and dose; mechanisms of action; and their applications in neurological diseases, including stroke, cerebral palsy, spinal cord injury, traumatic brain injury, amyotrophic lateral sclerosis, autism spectrum disorder, and epilepsy. Furthermore, recommendations on indications for BMMNC administration and the advantages and limitations of BMMNC applications for neurological diseases are discussed. BMMNCs in the treatment of neurological diseases. BMMNCs have been applied in several neurological diseases. Proposed mechanisms for the action of BMMNCs include homing, differentiation and paracrine effects (angiogenesis, neuroprotection, and anti-inflammation). Further studies should be performed to determine the optimal cell dose and administration route, the roles of BMMNC subtypes, and the indications for the use of BMMNCs in neurological conditions with and without genetic abnormalities.


Assuntos
Transtorno do Espectro Autista , Acidente Vascular Cerebral , Humanos , Medula Óssea , Acidente Vascular Cerebral/terapia , Células da Medula Óssea
2.
Signal Transduct Target Ther ; 7(1): 272, 2022 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-35933430

RESUMO

Recent advancements in stem cell technology open a new door for patients suffering from diseases and disorders that have yet to be treated. Stem cell-based therapy, including human pluripotent stem cells (hPSCs) and multipotent mesenchymal stem cells (MSCs), has recently emerged as a key player in regenerative medicine. hPSCs are defined as self-renewable cell types conferring the ability to differentiate into various cellular phenotypes of the human body, including three germ layers. MSCs are multipotent progenitor cells possessing self-renewal ability (limited in vitro) and differentiation potential into mesenchymal lineages, according to the International Society for Cell and Gene Therapy (ISCT). This review provides an update on recent clinical applications using either hPSCs or MSCs derived from bone marrow (BM), adipose tissue (AT), or the umbilical cord (UC) for the treatment of human diseases, including neurological disorders, pulmonary dysfunctions, metabolic/endocrine-related diseases, reproductive disorders, skin burns, and cardiovascular conditions. Moreover, we discuss our own clinical trial experiences on targeted therapies using MSCs in a clinical setting, and we propose and discuss the MSC tissue origin concept and how MSC origin may contribute to the role of MSCs in downstream applications, with the ultimate objective of facilitating translational research in regenerative medicine into clinical applications. The mechanisms discussed here support the proposed hypothesis that BM-MSCs are potentially good candidates for brain and spinal cord injury treatment, AT-MSCs are potentially good candidates for reproductive disorder treatment and skin regeneration, and UC-MSCs are potentially good candidates for pulmonary disease and acute respiratory distress syndrome treatment.


Assuntos
Células-Tronco Mesenquimais , Tecido Adiposo , Diferenciação Celular/genética , Humanos , Medicina Regenerativa , Cordão Umbilical
3.
Cell Transplant ; 31: 9636897221110876, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35815930

RESUMO

Anti-N-methyl-d-aspartate (NMDA) receptor encephalitis is caused by altered patient immune reactions. This study reports the first patient with severe neurologic sequelae after NMDA receptor encephalitis treated with allogeneic umbilical cord-derived mesenchymal stem/stromal cells (UC-MSCs). A 5-year-old girl was diagnosed with NMDA receptor encephalitis and treated with immunosuppressive medicaments and intravenous immunoglobulin (IVIG). Despite intensive therapy, the patient's condition worsened so that allogenic UC-MSC therapy was contemplated. The patient received three intrathecal infusions of xeno- and serum-free cultured UC-MSCs at a dose of 106 cells/kg. At baseline and after each UC-MSC administration, the patient was examined by the German Coma Recovery Scale (CRS), the Gross Motor Function Classification System (GMFCS), the Gross Motor Function Measure-88 (GMFM-88), the Manual Ability Classification System (MACS), the Modified Ashworth Scale, and the Denver II test. Before cell therapy, she was in a permanent vegetative state with diffuse cerebral atrophy. Her cognition and motor functions improved progressively after three UC-MSC infusions. At the last visit, she was capable of walking, writing, and counting numbers. Control of urinary and bowel functions was completely recovered. Cerebral atrophy was reduced on brain magnetic resonance imaging (MRI). Overall, the outcomes of this patient suggest a potential cell therapy for autoimmune encephalitis and its neurological consequences.


Assuntos
Encefalite Antirreceptor de N-Metil-D-Aspartato , Células-Tronco Mesenquimais , Encefalite Antirreceptor de N-Metil-D-Aspartato/complicações , Encefalite Antirreceptor de N-Metil-D-Aspartato/diagnóstico , Encefalite Antirreceptor de N-Metil-D-Aspartato/terapia , Atrofia/complicações , Pré-Escolar , Progressão da Doença , Feminino , Humanos , Receptores de N-Metil-D-Aspartato/uso terapêutico , Cordão Umbilical
4.
Stem Cells Transl Med ; 10(9): 1266-1278, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34080789

RESUMO

Human bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs) represent promising stem cell therapy for the treatment of type 2 diabetes mellitus (T2DM), but the results of autologous BM-MSC administration in T2DM patients are contradictory. The purpose of this study was to test the hypothesis that autologous BM-MSC administration in T2DM patient is safe and that the efficacy of the treatment is dependant on the quality of the autologous BM-MSC population and administration routes. T2DM patients were enrolled, randomly assigned (1:1) by a computer-based system into the intravenous and dorsal pancreatic arterial groups. The safety was assessed in all the treated patients, and the efficacy was evaluated based on the absolute changes in the hemoglobin A1c, fasting blood glucose, and C-peptide levels throughout the 12-month follow-up. Our data indicated that autologous BM-MSC administration was well tolerated in 30 T2DM patients. Short-term therapeutic effects were observed in patients with T2DM duration of <10 years and a body mass index <23, which is in line with the phenotypic analysis of the autologous BM-MSC population. T2DM duration directly altered the proliferation rate of BM-MSCs, abrogated the glycolysis and mitochondria respiration of BM-MSCs, and induced the accumulation of mitochondria DNA mutation. Our data suggest that autologous administration of BM-MSCs in the treatment of T2DM should be performed in patients with T2DM duration <10 years and no obesity. Prior to further confirming the effects of T2DM on BM-MSC biology, future work with a larger cohort focusing on patients with different T2DM history is needed to understand the mechanism underlying our observation.


Assuntos
Diabetes Mellitus Tipo 2 , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Medula Óssea , Células da Medula Óssea , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Humanos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Obesidade/metabolismo
6.
Stem Cells Transl Med ; 10(1): 14-26, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32902182

RESUMO

The aim of this study was to evaluate the safety and efficacy of autologous bone marrow mononuclear cell transplantation combined with educational intervention for children with autism spectrum disorder. An open-label clinical trial was performed from July 2017 to August 2019 at Vinmec International Hospital, Hanoi, Vietnam. Thirty children who fulfilled the autism criteria of the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, and had Childhood Autism Rating Scale (CARS) scores >37 were selected. Bone marrow was harvested by anterior iliac crest puncture under general anesthesia. The volume collected was as follows: 8 mL/kg for patients under 10 kg (80 mL + [body weight in kg - 10] × 7 mL) for patients above 10 kg. Mononuclear cells were isolated with a Ficoll gradient and then infused intrathecally. The same procedure was repeated 6 months later. After the first transplantation, all patients underwent 8 weeks of educational intervention based on the Early Start Denver Model. There were no severe adverse events associated with transplantation. The severity of autism spectrum disorder (ASD) was significantly reduced, with the median CARS score decreasing from 50 (range 40-55.5) to 46.5 (range 33.5-53.5) (P < .05). Adaptive capacity increased, with the median Vineland Adaptive Behavior Scales score rising from 53.5 to 60.5. Social communication, language, and daily skills improved markedly within 18 months after transplantation. Conversely, repetitive behaviors and hyperactivity decreased remarkably. Autologous bone marrow mononuclear cell transplantation in combination with behavioral intervention was safe and well tolerated in children with ASD (Trial registration: ClinicalTrials.gov identifier: NCT03225651).


Assuntos
Transtorno do Espectro Autista , Transplante de Medula Óssea , Leucócitos Mononucleares/transplante , Transtorno do Espectro Autista/terapia , Medula Óssea , Criança , Humanos , Vietnã
7.
Front Pediatr ; 8: 564, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33014944

RESUMO

Aim: Anoxic brain injury (ABI) due to non-fatal drowning may cause persistent vegetative state (VS) that is currently incurable. The aim of this paper is to present the safety and feasibility of autologous bone marrow-derived mononuclear cell (BMMNC) transplantation in five drowning children surviving in persistent VS. Methods: We used BMMNC as a novel candidate therapeutic tool in a pilot phase-I study for five patients affected by neurological sequelae after near-death drowning. Autologous BMMNCs were freshly isolated using Ficoll gradient centrifugation then infused intrathecally to five patients. The number of transplantation varied from two to four times depending on the motor function improvement of patient after transplantation. Clinical therapeutic effects were evaluated using gross motor function measure and muscle spasticity rating scales, cognitive assessments, and brain MRI before and after cell administrations. Results: Six months after BMMNC transplantation, no serious complications or adverse events were reported. All five patients displayed improvement across the major parameters of gross motor function, cognition, and muscle spasticity. Three patients displayed improved communication including the expression of words. In particular, one patient remarkably reduced cerebral atrophy, with nearly normal cerebral parenchyma after BMMNC transplantation. Conclusions: Autologous BMMNC transplantation for the treatment of children in persistent VS after drowning is safe, feasible, and can potentially improve motor function and cognition and reduce muscle spasticity. These results pave the way for a future phase II clinical trial to evaluate the efficacy of the therapy.

9.
PLoS One ; 9(7): e101449, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25000346

RESUMO

OBJECTIVES: This study evaluates the safety, principal feasibility and restoration potential of laser-supported CD133+ intramyocardial cell transplantation in patients with ischemic cardiomyopathy. METHODS: Forty-two patients with severe ischemic cardiomyopathy (left ventricular ejection fraction (LVEF) >15% and <35%) were included in this prospective multicenter phase I trial. They underwent coronary artery bypass grafting (CABG) with subsequent transepicardial low-energy laser treatment and autologous CD133+ cell transplantation, and were followed up for 12 months. To evaluate segmental myocardial contractility as well as perfusion and to identify the areas of scar tissue, cardiac MRI was performed at 6 months and compared to the preoperative baseline. In addition, clinical assessment comprising of CCS scoring, blood and physical examination was performed at 3, 6 and 12 months, respectively. RESULTS: Intraoperative cell isolation resulted in a mean cell count of 9.7±1.2×106. Laser treatment and subsequent CD133+ cell therapy were successfully and safely carried out in all patients and no procedure-related complications occurred. At 6 months, the LVEF was significantly increased (29.7±1.9% versus 24.6±1.5% with p = 0.004). In addition, freedom from angina was achieved, and quality of life significantly improved after therapy (p<0.0001). Interestingly, an extended area of transmural delayed enhancement (>3 myocardial segments) determined in the preoperative MRI was inversely correlated with a LVEF increase after laser-supported cell therapy (p = 0.024). CONCLUSIONS: This multicenter trial demonstrates that laser-supported CD133+ cell transplantation is safe and feasible in patients with ischemic cardiomyopathy undergoing CABG, and in most cases, it appears to significantly improve the myocardial function. Importantly, our data show that the beneficial effect was significantly related to the extent of transmural delayed enhancement, suggesting that MRI-guided selection of patients is mandatory to ensure the effectiveness of the therapy. TRIAL REGISTRATION: EudraCT 2005-004051-35) Controlled-Trials.com ISRCTN49998633.


Assuntos
Antígenos CD/metabolismo , Células da Medula Óssea/metabolismo , Cardiomiopatias/complicações , Transplante de Células/métodos , Glicoproteínas/metabolismo , Terapia a Laser , Isquemia Miocárdica/complicações , Isquemia Miocárdica/terapia , Peptídeos/metabolismo , Antígeno AC133 , Idoso , Separação Celular , Transplante de Células/efeitos adversos , Ponte de Artéria Coronária , Estudos de Viabilidade , Feminino , Humanos , Masculino , Isquemia Miocárdica/fisiopatologia , Segurança , Função Ventricular Esquerda
10.
Arch Toxicol ; 87(1): 123-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23179753

RESUMO

Developmental neurotoxicity (DNT) and many forms of reproductive toxicity (RT) often manifest themselves in functional deficits that are not necessarily based on cell death, but rather on minor changes relating to cell differentiation or communication. The fields of DNT/RT would greatly benefit from in vitro tests that allow the identification of toxicant-induced changes of the cellular proteostasis, or of its underlying transcriptome network. Therefore, the 'human embryonic stem cell (hESC)-derived novel alternative test systems (ESNATS)' European commission research project established RT tests based on defined differentiation protocols of hESC and their progeny. Valproic acid (VPA) and methylmercury (MeHg) were used as positive control compounds to address the following fundamental questions: (1) Does transcriptome analysis allow discrimination of the two compounds? (2) How does analysis of enriched transcription factor binding sites (TFBS) and of individual probe sets (PS) distinguish between test systems? (3) Can batch effects be controlled? (4) How many DNA microarrays are needed? (5) Is the highest non-cytotoxic concentration optimal and relevant for the study of transcriptome changes? VPA triggered vast transcriptional changes, whereas MeHg altered fewer transcripts. To attenuate batch effects, analysis has been focused on the 500 PS with highest variability. The test systems differed significantly in their responses (<20 % overlap). Moreover, within one test system, little overlap between the PS changed by the two compounds has been observed. However, using TFBS enrichment, a relatively large 'common response' to VPA and MeHg could be distinguished from 'compound-specific' responses. In conclusion, the ESNATS assay battery allows classification of human DNT/RT toxicants on the basis of their transcriptome profiles.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Perfilação da Expressão Gênica , Testes de Mutagenicidade/métodos , Síndromes Neurotóxicas/genética , Sítios de Ligação , Células Cultivadas , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Compostos de Metilmercúrio/toxicidade , Análise de Sequência com Séries de Oligonucleotídeos , Ácido Valproico/toxicidade
11.
Multimed Man Cardiothorac Surg ; 2010(809): mmcts.2009.003947, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24413679

RESUMO

If traditional treatment of coronary artery disease has failed, intramyocardial transplantation of CD133+ stem cells with the potential to improve myocardial function is an alternative approach to treating ischemic cardiomyopathy. The INSTEM trial aims at evaluating safety and feasibility of isolation and subsequent intramyocardial transplantation of CD133+ cells in combination with coronary artery bypass grafting (CABG). Patients suffering from severe ischemic cardiomyopathy (ejection fraction ≫15% and ≪35%) are enrolled in this trial. Bone marrow is harvested from the iliac crest and CD133+ cells are purified up to 99%. The myocardial region of interest is pre-treated by transmyocardial laser revascularization in order to trigger homing of transplanted cells. Autologous bone marrow CD133+ cells (up to 30×10(6) cells) are injected into predefined myocardial regions. Cardiac function prior to as well as three, six and 12 months after cell transplantation is assessed by echocardiography. Neither operative mortalities nor any cardiac deaths during follow-up occurred. Left ventricular ejection fraction improved from 25%±5% preoperatively to 40%±8% after six months. Our method of intraoperative CD133+ cell isolation while performing CABG and subsequent transmyocardial cell transplantation is feasible and safe. Although the follow-up has not yet been completed we propose this procedure to be a promising causal therapy of severe ischemic cardiomyopathy.

12.
BMC Genomics ; 6: 99, 2005 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-16042783

RESUMO

BACKGROUND: Microarray studies of the same phenomenon in different labs often appear at variance because the published lists of regulated transcripts have disproportionately small intersections. We demonstrate that comparing studies by intersecting lists in this manner is methodologically flawed by reanalyzing three studies of the molecular signature of "stemness" in human embryonic stem cells. There are only 7 genes common to all three published lists, suggesting disagreement. RESULTS: Carefully reanalyzing all three together from the raw data we detect 111 genes upregulated and 95 downregulated in all three studies. The upregulated list was subject to rtRTPCR analysis and 75% of the genes were confirmed. CONCLUSION: Our findings show that the three studies have a substantial core of common genes, which is missed if only the published lists are examined. Combined analysis of multiple experiments can be a powerful way to distil coherent conclusions.


Assuntos
Embrião de Mamíferos/citologia , Perfilação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Células-Tronco/citologia , Linhagem Celular , Células Cultivadas , DNA Complementar/metabolismo , Regulação para Baixo , Etiquetas de Sequências Expressas , Regulação da Expressão Gênica , Humanos , Modelos Estatísticos , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
13.
Genetics ; 165(1): 159-69, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14504224

RESUMO

Rap1 belongs to the highly conserved Ras subfamily of small GTPases. In Drosophila, Rap1 plays a critical role in many different morphogenetic processes, but the molecular mechanisms executing its function are unknown. Here, we demonstrate that Canoe (Cno), the Drosophila homolog of mammalian junctional protein AF-6, acts as an effector of Rap1 in vivo. Cno binds to the activated form of Rap1 in a yeast two-hybrid assay, the two molecules colocalize to the adherens junction, and they display very similar phenotypes in embryonic dorsal closure (DC), a process that relies on the elongation and migration of epithelial cell sheets. Genetic interaction experiments show that Rap1 and Cno act in the same molecular pathway during DC and that the function of both molecules in DC depends on their ability to interact. We further show that Rap1 acts upstream of Cno, but that Rap1, unlike Cno, is not involved in the stimulation of JNK pathway activity, indicating that Cno has both a Rap1-dependent and a Rap1-independent function in the DC process.


Assuntos
Drosophila/metabolismo , Cinesinas/metabolismo , Miosinas/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Animais , Drosophila/embriologia , Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Tirosina Quinases/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Proteínas rap1 de Ligação ao GTP/fisiologia
14.
Dev Biol ; 260(2): 404-13, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12921741

RESUMO

The molecular mechanism underlying pluripotency is largely unknown. Here, we provide the first global transcriptional profile of the state of "stemness" in human embryonic stem cells (HESCs). We have identified a set of 918 genes enriched in undifferentiated HESCs compared with their differentiated counterparts. These include ligand/receptor pairs and secreted inhibitors of the FGF, TGFbeta/BMP, and Wnt pathways, highlighting a prevalent role for these pathways in HESCs. Importantly, a significant number of HESCs-enriched genes, including several signaling components, are found to be intersected with published mouse embryonic stem cell data, indicating that a "core molecular program" is shared between the two pluripotent stem cells.


Assuntos
Células-Tronco/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Embrião de Mamíferos/citologia , Etiquetas de Sequências Expressas , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos , Células-Tronco Pluripotentes/fisiologia , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
15.
Dev Biol ; 248(1): 118-27, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12142025

RESUMO

The latent TGF-beta binding proteins (LTBP) are believed to control the availability of TGF-beta in the extracellular milieu. To gain insight into the potential roles of LTBP in early development, we isolated the Xenopus LTBP-1 (xLTBP-1) cDNA. The cDNA encodes a protein similar to the mammalian LTBP-1 in both size and domain structure. In addition, we found a novel longer splice isoform of xLTBP. The RNAs for both forms of xLTBP displayed temporal regulation and the shorter transcript is expressed maternally. Both transcripts also display spatial regulation and are found in the dorsal mesoderm of the organizer. In animal cap experiments, LTBP-1 potentiates the activity of activin and nodal. The activity of LTBP-1 did not appear to require covalent association with activin as the addition of medium containing activin and LTBP-1 to animal caps enhanced the activin effect. These results indicate that LTBP-1 may be part of the regulatory system that establishes the threshold of morphogen activity for activins and nodals in the dorsal side of the embryo during gastrulation.


Assuntos
Ativinas/metabolismo , Proteínas de Transporte/biossíntese , Proteínas de Transporte/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Clonagem Molecular , Cricetinae , DNA Complementar/metabolismo , Humanos , Hibridização In Situ , Proteínas de Ligação a TGF-beta Latente , Mesoderma/metabolismo , Dados de Sequência Molecular , Região Organizadora do Nucléolo , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Xenopus , Proteínas de Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA