Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cytotherapy ; 25(8): 810-814, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36931996

RESUMEN

The International Society for Cell & Gene Therapy Scientific Signature Series event "Therapeutic Advances With Native and Engineered Human EVs" took place as part of the International Society for Cell & Gene Therapy 2022 Annual Meeting, held from May 4 to 7, 2022, in San Francisco, California, USA. This was the first signature series event on extracellular vesicles (EVs) and a timely reflection of the growing interest in EVs, including both native and engineered human EVs, for therapeutic applications. The event successfully gathered academic and industrial key opinion leaders to discuss the current state of the art in developing and understanding native and engineered EVs and applying our knowledge toward advancing EV therapeutics. Latest advancements in understanding the mechanisms by which native and engineered EVs exert their therapeutic effects against different diseases in animal models were presented, with some diseases such as psoriasis and osteoarthritis already reaching clinical testing of EVs. The discussion also covered various aspects relevant to advancing the clinical translation of EV therapies, including EV preparation, manufacturing, consistency, site(s) of action, route(s) of administration, and luminal cargo delivery of RNA and other compounds.


Asunto(s)
Vesículas Extracelulares , Animales , Humanos , Tratamiento Basado en Trasplante de Células y Tejidos , Terapia Genética
2.
Biomacromolecules ; 24(1): 150-165, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36542545

RESUMEN

The increasing demand for tissue replacement has encouraged scientists worldwide to focus on developing new biofabrication technologies. Multimaterials/cells printed with stringent resolutions are necessary to address the high complexity of tissues. Advanced inkjet 3D printing can use multimaterials and attain high resolution and complexity of printed structures. However, a decisive yet limiting aspect of translational 3D bioprinting is selecting the befitting material to be used as bioink; there is a complete lack of cytoactive bioinks with adequate rheological, mechanical, and reactive properties. This work strives to achieve the right balance between resolution and cell support through methacrylamide functionalization of a psychrophilic gelatin and new fluorosurfactants used to engineer a photo-cross-linkable and immunoevasive bioink. The syntonized parameters following optimal formulation conditions allow proficient printability in a PolyJet 3D printer comparable in resolution to a commercial synthetic ink (∼150 µm). The bioink formulation achieved the desired viability (∼80%) and proliferation of co-printed cells while demonstrating in vivo immune tolerance of printed structures. The practical usage of existing high-resolution 3D printing systems using a novel bioink is shown here, allowing 3D bioprinted structures with potentially unprecedented complexity.


Asunto(s)
Bioimpresión , Bioimpresión/métodos , Impresión Tridimensional , Gelatina/química , Reología , Andamios del Tejido/química , Ingeniería de Tejidos/métodos
3.
Cell Mol Life Sci ; 79(3): 177, 2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35247083

RESUMEN

There is a steadily growing interest in the use of mitochondria as therapeutic agents. The use of mitochondria derived from mesenchymal stem/stromal cells (MSCs) for therapeutic purposes represents an innovative approach to treat many diseases (immune deregulation, inflammation-related disorders, wound healing, ischemic events, and aging) with an increasing amount of promising evidence, ranging from preclinical to clinical research. Furthermore, the eventual reversal, induced by the intercellular mitochondrial transfer, of the metabolic and pro-inflammatory profile, opens new avenues to the understanding of diseases' etiology, their relation to both systemic and local risk factors, and also leads to new therapeutic tools for the control of inflammatory and degenerative diseases. To this end, we illustrate in this review, the triggers and mechanisms behind the transfer of mitochondria employed by MSCs and the underlying benefits as well as the possible adverse effects of MSCs mitochondrial exchange. We relay the rationale and opportunities for the use of these organelles in the clinic as cell-based product.


Asunto(s)
Mitocondrias/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Enfermedades Pulmonares/terapia , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/trasplante , Dinámicas Mitocondriales , Comunicación Paracrina
4.
EMBO Rep ; 21(2): e48052, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31984629

RESUMEN

Mesenchymal stem cells (MSCs) have fueled ample translation for the treatment of immune-mediated diseases. They exert immunoregulatory and tissue-restoring effects. MSC-mediated transfer of mitochondria (MitoT) has been demonstrated to rescue target organs from tissue damage, yet the mechanism remains to be fully resolved. Therefore, we explored the effect of MitoT on lymphoid cells. Here, we describe dose-dependent MitoT from mitochondria-labeled MSCs mainly to CD4+ T cells, rather than CD8+ T cells or CD19+ B cells. Artificial transfer of isolated MSC-derived mitochondria increases the expression of mRNA transcripts involved in T-cell activation and T regulatory cell differentiation including FOXP3, IL2RA, CTLA4, and TGFß1, leading to an increase in a highly suppressive CD25+ FoxP3+ population. In a GVHD mouse model, transplantation of MitoT-induced human T cells leads to significant improvement in survival and reduction in tissue damage and organ T CD4+ , CD8+ , and IFN-γ+ expressing cell infiltration. These findings point to a unique CD4+ T-cell reprogramming mechanism with pre-clinical proof-of-concept data that pave the way for the exploration of organelle-based therapies in immune diseases.


Asunto(s)
Células Madre Mesenquimatosas , Linfocitos T CD8-positivos , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Mitocondrias , Linfocitos T Reguladores
5.
Crit Care Med ; 49(9): e880-e890, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33870913

RESUMEN

OBJECTIVES: To ascertain whether systemic administration of mitochondria-rich fraction isolated from mesenchymal stromal cells would reduce lung, kidney, and liver injury in experimental sepsis. DESIGN: Animal study. SETTING: Laboratory investigation. SUBJECTS: Sixty C57BL/6 male mice. INTERVENTIONS: Sepsis was induced by cecal ligation and puncture; sham-operated animals were used as control. At 24 hours after surgery, cecal ligation and puncture and Sham animals were further randomized to receive saline or mitochondria-rich fraction isolated from mesenchymal stromal cells (3 × 106) IV. At 48 hours, survival, peritoneal bacterial load, lung, kidney, and liver injury were analyzed. Furthermore, the effects of mitochondria on oxygen consumption rate and reactive oxygen species production of lung epithelial and endothelial cells were evaluated in vitro. MEASUREMENTS AND MAIN RESULTS: In vitro exposure of lung epithelial and endothelial cells from cecal ligation and puncture animals to mitochondria-rich fraction isolated from mesenchymal stromal cells restored oxygen consumption rate and reduced total reactive oxygen species production. Infusion of exogenous mitochondria-rich fraction from mesenchymal stromal cells (mitotherapy) reduced peritoneal bacterial load, improved lung mechanics and histology, and decreased the expression of interleukin-1ß, keratinocyte chemoattractant, indoleamine 2,3-dioxygenase-2, and programmed cell death protein 1 in lung tissue, while increasing keratinocyte growth factor expression and survival rate in cecal ligation and puncture-induced sepsis. Mitotherapy also reduced kidney and liver injury, plasma creatinine levels, and messenger RNA expressions of interleukin-18 in kidney, interleukin-6, indoleamine 2,3-dioxygenase-2, and programmed cell death protein 1 in liver, while increasing nuclear factor erythroid 2-related factor-2 and superoxide dismutase-2 in kidney and interleukin-10 in liver. CONCLUSIONS: Mitotherapy decreased lung, liver, and kidney injury and increased survival rate in cecal ligation and puncture-induced sepsis.


Asunto(s)
Células Madre Mesenquimatosas/patología , Mitocondrias/metabolismo , Sepsis/complicaciones , Animales , Modelos Animales de Enfermedad , Hígado/metabolismo , Hígado/patología , Pulmón/metabolismo , Pulmón/patología , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL/metabolismo , Insuficiencia Multiorgánica
6.
FASEB J ; 34(6): 8250-8264, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32333618

RESUMEN

Hypoxia-inducible factor 1 α (HIF1α), a regulator of metabolic change, is required for the survival and differentiation potential of mesenchymal stem/stromal cells (MSC). Its role in MSC immunoregulatory activity, however, has not been completely elucidated. In the present study, we evaluate the role of HIF1α on MSC immunosuppressive potential. We show that HIF1α silencing in MSC decreases their inhibitory potential on Th1 and Th17 cell generation and limits their capacity to generate regulatory T cells. This reduced immunosuppressive potential of MSC is associated with a metabolic switch from glycolysis to OXPHOS and a reduced capacity to express or produce some immunosuppressive mediators including Intercellular Adhesion Molecule (ICAM), IL-6, and nitric oxide (NO). Moreover, using the Delayed-Type Hypersensitivity murine model (DTH), we confirm, in vivo, the critical role of HIF1α on MSC immunosuppressive effect. Indeed, we show that HIF1α silencing impairs MSC capacity to reduce inflammation and inhibit the generation of pro-inflammatory T cells. This study reveals the pivotal role of HIF1α on MSC immunosuppressive activity through the regulation of their metabolic status and identifies HIF1α as a novel mediator of MSC immunotherapeutic potential.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Inmunosupresores/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Citocinas/metabolismo , Tolerancia Inmunológica/fisiología , Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Reguladores/metabolismo , Células TH1 , Células Th17/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Gene Ther ; 27(1-2): 2-5, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30962535

RESUMEN

Rigorous testing of cell therapies in South America struggles with emerging opportunities and regulatory deficiencies. As in other continents, these tend to be permissive with commercial opportunism but stifling for research. We describe a successful biotechnological entrepreneurship, born from within an academic institution, to foster science and promote translational research. Sustainability, however, requires a more complex niche, and realistic contributions from investors, state agencies, and legislators. An added level of complexity is required to enable multicentric studies. Herein we succinctly describe some of the most urgent challenges that the deployment of cell therapy faces in Chile. If this is truly an aspiration, fantasy should not be allowed to direct regulatory agents or legislators, and our Latin American Magic realism should remain within the realm of literary fiction.


Asunto(s)
Investigación/tendencias , Investigación con Células Madre/ética , Investigación Biomédica Traslacional/tendencias , Diferenciación Celular , Humanos , América Latina/epidemiología , Células Madre/metabolismo
8.
Eur Respir J ; 55(6)2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32265310

RESUMEN

The severe respiratory consequences of the coronavirus disease 2019 (COVID-19) pandemic have prompted urgent need for novel therapies. Cell-based approaches, primarily using mesenchymal stem (stromal) cells (MSCs), have demonstrated safety and possible efficacy in patients with acute respiratory distress syndrome (ARDS), although they are not yet well studied in respiratory virus-induced ARDS. Limited pre-clinical data suggest that systemic MSC administration can significantly reduce respiratory virus (influenza strains H5N1 and H9N2)-induced lung injury; however, there are no available data in models of coronavirus respiratory infection.There is a rapidly increasing number of clinical investigations of cell-based therapy approaches for COVID-19. These utilise a range of different cell sources, doses, dosing strategies and targeted patient populations. To provide a rational strategy to maximise potential therapeutic use, it is critically important to understand the relevant pre-clinical studies and postulated mechanisms of MSC actions in respiratory virus-induced lung injuries. This review presents these, along with consideration of current clinical investigations.


Asunto(s)
Infecciones por Coronavirus/terapia , Medios de Cultivo Condicionados , Gripe Humana/terapia , Lesión Pulmonar/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Neumonía Viral/terapia , Síndrome de Dificultad Respiratoria/terapia , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus , COVID-19 , Tratamiento Basado en Trasplante de Células y Tejidos , Vesículas Extracelulares/trasplante , Humanos , Subtipo H5N1 del Virus de la Influenza A , Subtipo H9N2 del Virus de la Influenza A , Lesión Pulmonar/virología , Células Madre Mesenquimatosas/metabolismo , Infecciones por Orthomyxoviridae/terapia , Pandemias , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/metabolismo
9.
Cytotherapy ; 22(11): 602-605, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32933835

RESUMEN

The serious consequences of the global coronavirus disease 2019 (COVID-19) pandemic have prompted a rapid global response to develop effective therapies that can lessen disease severity in infected patients. Cell-based approaches, primarily using mesenchymal stromal cells (MSCs), have demonstrated a strong safety profile and possible efficacy in patients with acute respiratory distress syndrome (ARDS), but whether these therapies are effective for treating respiratory virus-induced ARDS is unknown. According to the World Health Organization International Clinical Trials Registry Platform and the National Institutes of Health ClinicalTrials.gov databases, 27 clinical investigations of MSC-based cell therapy approaches have begun in China since the onset of the COVID-19 outbreak, with a growing number of academic and industry trials elsewhere as well. Several recent published reports have suggested potential efficacy; however, the available data presented are either anecdotal or from incomplete, poorly controlled investigations. Therefore, although there may be a potential role for MSCs and other cell-based therapies in treatment of COVID-19, these need to be investigated in a rationally designed, controlled approach if safety and efficacy are to be demonstrated accurately. The authors urge that the field proceed by finding a balance between swift experimentation and communication of results and scientifically coherent generation and analysis of clinical data.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Infecciones por Coronavirus/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Neumonía Viral/terapia , Síndrome de Dificultad Respiratoria/terapia , Betacoronavirus , COVID-19 , China , Humanos , Células Madre Mesenquimatosas/citología , Pandemias , SARS-CoV-2
10.
BMC Biotechnol ; 19(1): 42, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31253149

RESUMEN

BACKGROUND: Artificial Mitochondrial Transfer or Transplant (AMT/T) can be used to reduce the stress and loss of viability of damaged cells. In MitoCeption, a type of AMT/T, the isolated mitochondria and recipient cells are centrifuged together at 4 °C and then co-incubated at 37 °C in normal culture conditions, inducing the transfer. Ultraviolet radiation (UVR) can affect mitochondria and other cell structures, resulting in tissue stress, aging, and immunosuppression. AMT/T could be used to repair UVR cellular and mitochondrial damage. We studied if a mitochondrial mix from different donors (Primary Allogeneic Mitochondrial Mix, PAMM) can repair UVR damage and promote cell survival. RESULTS: Using a simplified adaption of the MitoCeption protocol, we used peripheral blood mononuclear cells (PBMCs) as the recipient cell model of the PAMM in order to determine if this protocol could repair UVR damage. Our results showed that when PBMCs are exposed to UVR, there is a decrease in metabolic activity, mitochondrial mass, and mtDNA sequence stability as well as an increase in p53 expression and the percentage of dead cells. When PAMM MitoCeption was used on UVR-damaged cells, it successfully transferred mitochondria from different donors to distinct PBMCs populations and repaired the observed UVR damage. CONCLUSION: Our results represent an advancement in the applications of MitoCeption and other AMT/T. We showed that PBMCs could be used as a PAMM source of mitochondria. We also showed that these mitochondria can be transferred in a mix from different donors (PAMM) to UVR-damaged, non-adherent primary cells. Additionally, we decreased the duration of the MitoCeption protocol.


Asunto(s)
Daño del ADN , Leucocitos Mononucleares/metabolismo , Mitocondrias/metabolismo , Mitocondrias/trasplante , Rayos Ultravioleta , Adulto , Supervivencia Celular/genética , Células Cultivadas , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Femenino , Humanos , Leucocitos Mononucleares/efectos de la radiación , Masculino , Mitocondrias/genética , Especies Reactivas de Oxígeno/metabolismo , Trasplante Homólogo/métodos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
J Exp Biol ; 222(Pt 20)2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31537653

RESUMEN

Microvesicles are key players in cellular communication. As glandular secretions present a rich source of active exosomes, we hypothesized that exosome-like vesicles are present in Apis mellifera hypopharyngeal gland secretomal products (honey, royal jelly and bee pollen), and participate in their known antibacterial and pro-regenerative effects. We developed an isolation protocol based on serial centrifugation and ultracentrifugation steps and demonstrated the presence of protein-containing exosome-like vesicles in all three bee-derived products. Assessing their antibacterial properties, we found that exosome-like vesicles had bacteriostatic, bactericidal and biofilm-inhibiting effects on Staphylococcus aureus Furthermore, we demonstrated that mesenchymal stem cells (MSCs) internalize bee-derived exosome-like vesicles and that these vesicles influence the migration potential of the MSCs. In an in vitro wound-healing assay, honey and royal jelly exosome-like vesicles increased migration of human MSCs, demonstrating their inter-kingdom activity. In summary, we have discovered exosome-like vesicles as a new, active compound in bee pollen, honey and royal jelly.


Asunto(s)
Antibacterianos/farmacología , Abejas/metabolismo , Exosomas/metabolismo , Ácidos Grasos/química , Miel , Polen/química , Regeneración/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Exosomas/efectos de los fármacos , Humanos , Polen/ultraestructura
12.
Circ Res ; 121(10): 1192-1204, 2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-28974553

RESUMEN

RATIONALE: Umbilical cord-derived mesenchymal stem cells (UC-MSC) are easily accessible and expanded in vitro, possess distinct properties, and improve myocardial remodeling and function in experimental models of cardiovascular disease. Although bone marrow-derived mesenchymal stem cells have been previously assessed for their therapeutic potential in individuals with heart failure and reduced ejection fraction, no clinical trial has evaluated intravenous infusion of UC-MSCs in these patients. OBJECTIVE: Evaluate the safety and efficacy of the intravenous infusion of UC-MSC in patients with chronic stable heart failure and reduced ejection fraction. METHODS AND RESULTS: Patients with heart failure and reduced ejection fraction under optimal medical treatment were randomized to intravenous infusion of allogenic UC-MSCs (Cellistem, Cells for Cells S.A., Santiago, Chile; 1×106 cells/kg) or placebo (n=15 per group). UC-MSCs in vitro, compared with bone marrow-derived mesenchymal stem cells, displayed a 55-fold increase in the expression of hepatocyte growth factor, known to be involved in myogenesis, cell migration, and immunoregulation. UC-MSC-treated patients presented no adverse events related to the cell infusion, and none of the patients tested at 0, 15, and 90 days presented alloantibodies to the UC-MSCs (n=7). Only the UC-MSC-treated group exhibited significant improvements in left ventricular ejection fraction at 3, 6, and 12 months of follow-up assessed both through transthoracic echocardiography (P=0.0167 versus baseline) and cardiac MRI (P=0.025 versus baseline). Echocardiographic left ventricular ejection fraction change from baseline to month 12 differed significantly between groups (+7.07±6.22% versus +1.85±5.60%; P=0.028). In addition, at all follow-up time points, UC-MSC-treated patients displayed improvements of New York Heart Association functional class (P=0.0167 versus baseline) and Minnesota Living with Heart Failure Questionnaire (P<0.05 versus baseline). At study completion, groups did not differ in mortality, heart failure admissions, arrhythmias, or incident malignancy. CONCLUSIONS: Intravenous infusion of UC-MSC was safe in this group of patients with stable heart failure and reduced ejection fraction under optimal medical treatment. Improvements in left ventricular function, functional status, and quality of life were observed in patients treated with UC-MSCs. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov/ct2/show/NCT01739777. Unique identifier: NCT01739777.


Asunto(s)
Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Cordón Umbilical/trasplante , Anciano , Movimiento Celular/fisiología , Método Doble Ciego , Femenino , Humanos , Infusiones Intravenosas , Masculino , Células Madre Mesenquimatosas/fisiología , Persona de Mediana Edad , Resultado del Tratamiento
13.
Gynecol Obstet Invest ; 84(4): 390-395, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30712040

RESUMEN

OBJECTIVE: To determine the normal limits of menstrual fluid volume during reproductive life, quantified by direct measurement. METHODS: This was an observational, prospective clinical trial of healthy women aged 20-49 years old, with normal menstrual periods, recruited in a Natural Family Planning Unit. Women collected their menstrual fluid for at least 3 menstrual periods using a vaginal cup. Menstrual volume and different covariables were evaluated using a multilevel mixed-effects linear regression. RESULTS: Ninety-six cycles from 28 patients between 24 and 49 years old were analyzed. The average menstrual volume was 86.7 mL with a range from 15 to 271 mL. The 50th percentile of all samples was 81 mL and the 95th percentile was 162 mL. For multiparous patients the 50th percentile was 93 mL and the 95th was 169 mL. Menstrual fluid volume was higher in multigravida (99.1 mL) than in nulliparous women (45.9 Ml; p < 0.02). No statistically significant associations were identified between different variables and menstrual volume. CONCLUSION: A menstrual volume over 169 mL should be considered abnormal on multiparous patients. Age was not associated with changes on menstrual fluid volume.


Asunto(s)
Secreciones Corporales , Menstruación , Hemorragia Uterina/diagnóstico , Adulto , Femenino , Humanos , Modelos Lineales , Ciclo Menstrual , Persona de Mediana Edad , Análisis Multinivel , Estudios Prospectivos , Valores de Referencia , Reproducción , Vagina , Adulto Joven
14.
Stem Cells ; 34(2): 456-69, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26528946

RESUMEN

Recently, a noninvasive and highly proliferative stem cell population from menstrual blood called MenSCs has been identified. Despite their use in clinical studies, their immunomodulatory properties have not yet been investigated. In this context, we studied the immunosuppressive properties of MenSCs in comparison with the well-characterized bone marrow derived-MSCs (BM-MSCs). Using an in vitro proliferation assays, we showed that MenSCs displayed a lower suppressive effect on peripheral blood mononuclear cells and in particular on the proinflammatory CD4(+) IFN-γ(+) and CD8(+) IFNγ(+) cells than BM-MSCs. Moreover, compared to BM-MSCs, MenSCs activated with IFN-γ and IL-1ß produced lower amounts of immunosuppressive factors such as IDO, PDL-1, PGE2, and Activin A and exhibited a substantial lower expression level of IFN-γ receptor subunits. In the collagen induced arthritis model, while BM-MSCs administration resulted in a potent therapeutic effect associated with a significant decrease of proinflammatory T cell frequency in the lymph nodes, MenSCs injection did not. In contrast, in the xeno-GVHD model, only MenSCs administration significantly increased the survival of mice. This beneficial effect mediated by MenSCs was associated with a higher capacity to migrate into the intestine and liver and not to their anti-inflammatory capacities. All together our results demonstrate for the first time that the therapeutic potential of MSC in the experimental xeno-GVHD model is independent of their immunosuppressive properties. These findings should be taken into consideration for the development of safe and effective cell therapies.


Asunto(s)
Artritis Experimental/terapia , Enfermedad Injerto contra Huésped/terapia , Ciclo Menstrual , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Tolerancia al Trasplante , Adolescente , Adulto , Artritis Experimental/inmunología , Artritis Experimental/patología , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Xenoinjertos , Humanos , Masculino , Persona de Mediana Edad
16.
Int J Mol Sci ; 16(8): 16953-65, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26225955

RESUMEN

MicroRNAs control the differentiation and function of B cells, which are considered key elements in the pathogenesis of systemic lupus erythematosus (SLE). However, a common micro(mi)RNA signature has not emerged since published data includes patients of variable ethnic background, type of disease, and organ involvement, as well as heterogeneous cell populations. Here, we aimed at identifying a miRNA signature of purified B cells from renal and non-renal severe SLE patients of Latin American background, a population known to express severe disease. Genome-wide miRNA expression analyses were performed on naive and memory B cells and revealed two categories of miRNA signatures. The first signature represents B cell subset-specific miRNAs deregulated in SLE: 11 and six miRNAs discriminating naive and memory B cells of SLE patients from healthy controls (HC), respectively. Whether the miRNA was up or down-regulated in memory B cells as compared with naive B cells in HC, this difference was abolished in SLE patients, and vice versa. The second signature identifies six miRNAs associated with specific pathologic features affecting renal outcome, providing a further understanding for SLE pathogenesis. Overall, the present work provided promising biomarkers in molecular diagnostics for disease severity as well as potential new targets for therapeutic intervention in SLE.


Asunto(s)
Subgrupos de Linfocitos B/metabolismo , Biomarcadores/metabolismo , Perfilación de la Expresión Génica , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , MicroARNs/genética , Adulto , Estudios de Casos y Controles , Chile , Análisis por Conglomerados , Francia , Humanos , Lupus Eritematoso Sistémico/diagnóstico , Nefritis Lúpica/diagnóstico , Nefritis Lúpica/genética , MicroARNs/metabolismo
17.
Rev Med Chil ; 143(4): 415-23, 2015 Apr.
Artículo en Español | MEDLINE | ID: mdl-26204531

RESUMEN

BACKGROUND: Intracoronary delivery of autologous bone marrow mononuclear cells is an interesting therapeutic promise for patients with heart failure of different etiologies. AIM: To evaluate the long-term safety and efficacy of this therapy in patients with dilated cardiomyopathy of different etiologies under optimal medical treatment. PATIENTS AND METHODS: Prospective, open-label, controlled clinical trial. Of 23 consecutive patients, 12 were assigned to autologous bone marrow mononuclear cell intracoronary transplantation, receiving a mean dose of 8.19 ± 4.43 x 10(6) CD34+ cells. Mortality, cardiovascular readmissions and cancer incidence rate, changes in functional capacity, quality of life questionnaires and echocardiographic measures from baseline, were assessed at long-term follow-up (37.7 ± 9.7 months) in patients receiving or not the cells. RESULTS: No significant differences were observed in mortality, cardiovascular readmissions or cancer incidence rate amongst groups. An improvement in functional class and quality of life questionnaires in the transplanted group was observed (p < 0.01). The treated group showed a non-significant increase in left ventricular ejection fraction at long-term follow-up (from 26.75 ± 4.85% to 34.90 ± 8.57%, p = 0.059 compared to baseline). There were no changes in left ventricular volumes. We observed no improvement of these variables in the control group. CONCLUSIONS: Intracoronary transplantation of autologous bone marrow mononuclear cells is feasible and safe in patients with dilated cardiomyopathy of diverse etiologies. This therapy was associated to persistent improvements in functional class and quality of life. There was also a non-significant long-term improvement of left ventricular function.


Asunto(s)
Trasplante de Médula Ósea/métodos , Cardiomiopatía Dilatada/cirugía , Trasplante de Médula Ósea/mortalidad , Volumen Cardíaco/fisiología , Cardiomiopatía Dilatada/diagnóstico por imagen , Cardiomiopatía Dilatada/mortalidad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Readmisión del Paciente/estadística & datos numéricos , Estudios Prospectivos , Calidad de Vida , Volumen Sistólico/fisiología , Encuestas y Cuestionarios , Factores de Tiempo , Trasplante Autólogo , Resultado del Tratamiento , Ultrasonografía , Función Ventricular/fisiología
18.
Stem Cells ; 31(6): 1160-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23404852

RESUMEN

Much controversy surrounds the identity and origin of human hepatic stem and progenitor cells in part because of a lack of small animal models in which the developmental potential of isolated candidate cell populations can be functionally evaluated. We show here that adoptive transfer of CD34(+) cells from human fetal liver into sublethally irradiated NOD-SCID Il2rg(-/-) (NSG) mice leads to an efficient development of not only human hematopoietic cells but also human hepatocyte-like cells in the liver of the recipient mice. Using this simple in vivo assay in combination with cell fractionation, we show that CD34(+) fetal liver cells can be separated into three distinct subpopulations: CD34(hi) CD133(hi), CD34(lo) CD133(lo), and CD34(hi) CD133(neg). The CD34(hi) CD133(hi) population contains hematopoietic stem/progenitor cells (HSPCs) as they give rise to T cells, B cells, NK cells, dendritic cells, and monocytes/macrophages in NSG mice and colony-forming unit (CFU)-GEMM cells in vitro. The CD34(lo) CD133(lo) population does not give rise to hematopoietic cells, but reproducibly generates hepatocyte-like cells in NSG mice and in vitro. The CD34(hi) CD133(neg) population only gives rise to CFU-GM and burst-forming unit-erythroid in vitro. Furthermore, we show that the CD34(lo) CD133(lo) cells express hematopoietic, hepatic, and mesenchymal markers, including CD34, CD133, CD117, epithelial cell adhesion molecule, CD73, albumin, α-fetal protein, and vimentin and transcriptionally are more closely related to HSPCs than to mature hepatocytes. These results show that CD34(lo) CD133(lo) fetal liver cells possess the hepatic progenitor cell properties and that human hepatic and hematopoietic progenitor cells are distinct, although they may originate from the same precursors in the fetal liver.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Hepatocitos/fisiología , Hígado/citología , Células Madre/fisiología , Antígeno AC133 , Albúminas/genética , Albúminas/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Dendríticas/metabolismo , Células Dendríticas/fisiología , Molécula de Adhesión Celular Epitelial , Glicoproteínas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/embriología , Hígado/metabolismo , Linfocitos/metabolismo , Linfocitos/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Monocitos/metabolismo , Monocitos/fisiología , Péptidos/metabolismo , Células Madre/metabolismo , Transcripción Genética , Vimentina/genética , Vimentina/metabolismo , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo
19.
Rev Med Chil ; 142(8): 1034-46, 2014 Aug.
Artículo en Español | MEDLINE | ID: mdl-25424676

RESUMEN

Available medical therapy is unable to completely prevent or revert the pathological cardiac remodeling secondary to ischemia or other injuries, which is responsible for the development of heart failure. Regenerative medicine through stem cells had an explosive development in the cardiovascular area during the past decade. Stem cells possess the capacity to regenerate, repair or substitute damaged tissue, allowing the reestablishment of its function. Stem cells can also modulate apoptosis, angiogenesis, fibrosis and inflammation, favoring the endogenous regenerative process initiated by the damaged tissue. These capacities have been corroborated in several animal models of cardiovascular diseases with positive results. In humans, therapies with bone marrow mononuclear stem cells, mesenchymal stem cells and cardiac stem cells are safe. Most randomized clinical trials in patients with myocardial infarction or cardiomyopathies of different etiologies have reported benefits on ventricular function, quality of life and even over mortality of treated patients. This article reviews the state of art of stem cell therapy in cardiovascular diseases, focusing on the most common cellular types used in patients with acute myocardial infarction and chronic cardiomyopathies of different etiologies.


Asunto(s)
Enfermedades Cardiovasculares/cirugía , Trasplante de Células Madre/métodos , Transdiferenciación Celular , Enfermedad Crónica , Cardiopatías/cirugía , Humanos , Células Madre Multipotentes/fisiología , Células Madre Multipotentes/trasplante , Infarto del Miocardio/cirugía
20.
J Endod ; 50(5): 596-601, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38387795

RESUMEN

INTRODUCTION: Modern tissue engineering strategies have elucidated the potential of regenerative endodontic treatment (RET) as an alternative for treating mature teeth. METHODS: Here, we report two cases in which cell-based RET (CB-RET) using encapsulated allogeneic umbilical cord mesenchymal stem cells (UC-MSCs) in a platelet-poor plasma (PPP)-based scaffold was used in two mature teeth with pulp necrosis and apical periodontitis. RESULTS: After 5 years of follow-up, the healing response was satisfactory in both cases, with evidence of pulp revitalization. CONCLUSIONS: This is the first study to report the success of an extended, 5-year follow-up for allogeneic CB-RET. This report presents an innovative and sustainable solution to challenging endodontic scenarios.


Asunto(s)
Necrosis de la Pulpa Dental , Periodontitis Periapical , Endodoncia Regenerativa , Humanos , Necrosis de la Pulpa Dental/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Periodontitis Periapical/terapia , Endodoncia Regenerativa/métodos , Tratamiento del Conducto Radicular/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido , Ápice del Diente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA