Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
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
5.
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.
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
8.
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.

9.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA