Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cells ; 11(6)2022 03 17.
Article in English | MEDLINE | ID: mdl-35326470

ABSTRACT

Spinal cord injury (SCI) remains an important public health problem which often causes permanent loss of muscle strength, sensation, and function below the site of the injury, generating physical, psychological, and social impacts throughout the lives of the affected individuals, since there are no effective treatments available. The use of stem cells has been investigated as a therapeutic approach for the treatment of SCI. Although a significant number of studies have been conducted in pre-clinical and clinical settings, so far there is no established cell therapy for the treatment of SCI. One aspect that makes it difficult to evaluate the efficacy is the heterogeneity of experimental designs in the clinical trials that have been published. Cell transplantation methods vary widely among the trials, and there are still no standardized protocols or recommendations for the therapeutic use of stem cells in SCI. Among the different cell types, mesenchymal stem/stromal cells (MSCs) are the most frequently tested in clinical trials for SCI treatment. This study reviews the clinical applications of MSCs for SCI, focusing on the critical analysis of 17 clinical trials published thus far, with emphasis on their design and quality. Moreover, it highlights the need for more evidence-based studies designed as randomized controlled trials and potential challenges to be addressed in context of stem cell therapies for SCI.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Spinal Cord Injuries/metabolism , Treatment Outcome
2.
Stem Cell Res ; 41: 101630, 2019 12.
Article in English | MEDLINE | ID: mdl-31706097

ABSTRACT

Human-induced pluripotent stem cell (hiPSC) CBTCi001-A line was generated from a healthy 30-year old male dermal fibroblasts using non-integrative reprogramming method using episomal-based plasmids expressing OCT4, SOX2, KLF4, and MYCL. Characterization of CBTCi001-A was confirmed by the expression of typical markers of pluripotency and differentiation potential in vitro.


Subject(s)
Cell Culture Techniques/methods , Cell Line/cytology , Dermis/cytology , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Tissue Donors , Adult , Cell Differentiation , Humans , Kruppel-Like Factor 4 , Male , Reproducibility of Results
3.
Stem Cell Res ; 39: 101488, 2019 08.
Article in English | MEDLINE | ID: mdl-31272037

ABSTRACT

Autism spectrum disorders (ASDs) are a group of diseases that affect social interaction, communication and behavior. Molecular mechanisms involved in the pathogenesis of ASDs are complex due to genetic heterogeneity. Recently, pathogenic variants of SCN2A have been strongly associated with ASDs. Here, we generated iPSCs from a patient with ASD and a heterozygous nonsense mutation in SCN2A, by reprogramming mesenchymal stromal cells with non-integrating vectors. The generated iPSC line expresses pluripotency markers, presents a normal karyotype and is able to differentiate into the three germ layers. This iPSC line is a useful tool for modeling ASD and drug screening studies.


Subject(s)
Autism Spectrum Disorder/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , NAV1.2 Voltage-Gated Sodium Channel/genetics , Autism Spectrum Disorder/genetics , Cell Line , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Flow Cytometry , Haploinsufficiency/genetics , Haploinsufficiency/physiology , Humans , Karyotype , Microsatellite Repeats/genetics , Mutation/genetics , Real-Time Polymerase Chain Reaction
4.
Stem Cell Res ; 38: 101454, 2019 07.
Article in English | MEDLINE | ID: mdl-31078962

ABSTRACT

Sickle cell disease (SCD) is one of the most prevalent and severe monogenetic disorders. Previously, we generated iPS cell lines from SCD patients. Here, we generated iPS cell lines from three age-, ethnicity- and gender-matched healthy individuals as control cell lines. Cell reprogramming was performed using erythroblasts expanded from PBMC by a non-integrative method. SCD-iPSC controls expressed pluripotency markers, presented a normal karyotype, were able to differentiate into the three germ layers in embryoid body spontaneous differentiation and confirmed to be integration-free. The cell lines generated here may be used as matched healthy controls for SCD studies.


Subject(s)
Anemia, Sickle Cell , Cellular Reprogramming Techniques , Erythroblasts , Induced Pluripotent Stem Cells/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Cell Culture Techniques , Cell Line , Erythroblasts/metabolism , Erythroblasts/pathology , Humans , Induced Pluripotent Stem Cells/pathology
6.
Am J Pathol ; 187(5): 1134-1146, 2017 May.
Article in English | MEDLINE | ID: mdl-28322201

ABSTRACT

Chronic Chagas disease cardiomyopathy, caused by Trypanosoma cruzi infection, is a major cause of heart failure in Latin America. Galectin-3 (Gal-3) has been linked to cardiac remodeling and poor prognosis in heart failure of different etiologies. Herein, we investigated the involvement of Gal-3 in the disease pathogenesis and its role as a target for disease intervention. Gal-3 expression in mouse hearts was evaluated during T. cruzi infection by confocal microscopy and flow cytometry analysis, showing a high expression in macrophages, T cells, and fibroblasts. In vitro studies using Gal-3 knockdown in cardiac fibroblasts demonstrated that Gal-3 regulates cell survival, proliferation, and type I collagen synthesis. In vivo blockade of Gal-3 with N-acetyl-d-lactosamine in T. cruzi-infected mice led to a significant reduction of cardiac fibrosis and inflammation in the heart. Moreover, a modulation in the expression of proinflammatory genes in the heart was observed. Finally, histological analysis in human heart samples obtained from subjects with Chagas disease who underwent heart transplantation showed the expression of Gal-3 in areas of inflammation, similar to the mouse model. Our results indicate that Gal-3 plays a role in the pathogenesis of experimental chronic Chagas disease, favoring inflammation and fibrogenesis. Moreover, by demonstrating Gal-3 expression in human hearts, our finding reinforces that this protein could be a novel target for drug development for Chagas cardiomyopathy.


Subject(s)
Chagas Cardiomyopathy/metabolism , Galectin 3/metabolism , Myocarditis/metabolism , Myocardium/pathology , Acetylgalactosamine/pharmacology , Animals , Cell Proliferation/physiology , Cell Survival/physiology , Chronic Disease , Collagen Type I/biosynthesis , Fibrosis/etiology , Fibrosis/metabolism , Galectin 3/antagonists & inhibitors , Heart Transplantation , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myocarditis/etiology , Myocardium/metabolism , Myofibroblasts/metabolism , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...