Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 200(2): 170-174, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36263593

RESUMEN

Although a growing body of evidence demonstrates that altered mtDNA content (mtDNAc) has clinical implications in several types of solid tumours, its prognostic relevance in acute promyelocytic leukaemia (APL) patients remains largely unknown. Here, we show that patients with higher-than-normal mtDNAc had better outcomes regardless of tumour burden. These results were more evident in patients with low-risk of relapse. The multivariate Cox proportional hazard model demonstrated that high mtDNAc was independently associated with a decreased cumulative incidence of relapse. Altogether, our data highlights the possible role of mitochondrial metabolism in APL patients treated with ATRA.


Asunto(s)
Leucemia Promielocítica Aguda , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/genética , Tretinoina/uso terapéutico , ADN Mitocondrial/genética , Relevancia Clínica , Recurrencia Local de Neoplasia/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resultado del Tratamiento
2.
Cytotherapy ; 24(7): 699-710, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35473998

RESUMEN

Pancreatic cancer is a highly lethal cancer characterized by local invasiveness, early metastasis, recurrence and high resistance to current therapies. Extensive stroma or desmoplasia is a key histological feature of the disease, and interactions between cancer and stromal cells are critical for pancreatic cancer development and progression. Mesenchymal stromal cells [MSCs] exhibit preferential tropism to primary and metastatic tumor sites and may either suppress or support tumor growth. Although MSCs represent a potential source of pancreatic cancer stroma, their contribution to pancreatic tumor growth remains poorly known. Here, we show that bone marrow MSCs significantly contribute to pancreatic cancer growth in vitro and in vivo. Furthermore, MSCs create a pro-carcinogenic microenvironment through the release of key factors mediating growth and angiogenesis, including interleukin (IL)-6, IL-8, vascular endothelial growth factor and activation of STAT3 signaling in tumor cells. IL-6 released by MSCs was largely responsible for the pro-tumorigenic effects of MSCs. Knockdown of IL-6 expression in MSCs by small interfering RNA (siRNA) abolished the MSC growth-promoting effect in vitro, reducing tumor cell proliferation and clonogenic potential. In addition, in a heterotopic nude mouse model of human pancreatic tumor xenografts, blockade of IL-6 with the anti-IL-6 receptor antibody, tocilizumab, or of its downstream effector STAT3 with the small molecule STAT3 inhibitor S3I-201, abrogated MSC-mediated tumor promotion and delayed tumor formation significantly. Our data demonstrate that MSCs promote pancreatic cancer growth, with IL-6 produced by MSCs playing a pivotal role.


Asunto(s)
Interleucina-6/metabolismo , Células Madre Mesenquimatosas , Neoplasias Pancreáticas , Animales , Médula Ósea/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neoplasias Pancreáticas
3.
Am J Hematol ; 97(12): 1538-1547, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36087071

RESUMEN

Autologous stem cell transplantation (ASCT) remains a key therapeutic strategy for treating patients with relapsed or refractory non-Hodgkin and Hodgkin lymphoma. Clonal hematopoiesis (CH) has been proposed as a major contributor not only to the development of therapy-related myeloid neoplasms but also to inferior overall survival (OS) in patients who had undergone ASCT. Herein, we aimed to investigate the prognostic implications of CH after ASCT in a cohort of 420 lymphoma patients using ultra-deep, highly sensitive error-correction sequencing. CH was identified in the stem cell product samples of 181 patients (43.1%) and was most common in those with T-cell lymphoma (72.2%). The presence of CH was associated with a longer time to neutrophil and platelet recovery. Moreover, patients with evidence of CH had inferior 5-year OS from the time of first relapse (39.4% vs. 45.8%, p = .043) and from the time of ASCT (51.8% vs. 59.3%, p = .018). The adverse prognostic impact of CH was not due to therapy-related myeloid neoplasms, the incidence of which was low in our cohort (10-year cumulative incidence of 3.3% vs. 3.0% in those with and without CH, p = .445). In terms of specific-gene mutations, adverse OS was mostly associated with PPM1D mutations (hazard ratio (HR) 1.74, 95% confidence interval (CI) 1.13-2.67, p = .011). In summary, we found that CH is associated with an increased risk of non-lymphoma-related death after ASCT, which suggests that lymphoma survivors with CH may need intensified surveillance strategies to prevent and treat late complications.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Enfermedad de Hodgkin , Linfoma , Neoplasias Primarias Secundarias , Humanos , Trasplante Autólogo/efectos adversos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Hematopoyesis Clonal , Linfoma/terapia , Linfoma/complicaciones , Enfermedad de Hodgkin/complicaciones , Neoplasias Primarias Secundarias/terapia , Neoplasias Primarias Secundarias/genética , Trasplante de Células Madre/efectos adversos , Estudios Retrospectivos
4.
Exp Cell Res ; 399(2): 112473, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33428902

RESUMEN

Sepsis is a complicated multi-system disorder characterized by a dysregulated host response to infection. Despite substantial progress in the understanding of mechanisms of sepsis, translation of these advances into clinically effective therapies remains challenging. Mesenchymal Stromal Cells (MSCs) possess immunomodulatory properties that have shown therapeutic promise in preclinical models of sepsis. The therapeutic effects of MSCs may vary depending on the source and type of these cells. In this comparative study, the gene expression pattern and surface markers of bone marrow-derived MSCs (BM-MSCs) and umbilical cord-derived MSCs (UC-MSCs) as well as their therapeutic effects in a clinically relevant mouse model of polymicrobial sepsis, cecal ligation and puncture (CLP), were investigated. The results showed remarkable differences in gene expression profile, surface markers and therapeutic potency in terms of enhancing survival and pro/anti-inflammatory responses between the two MSC types. BM-MSCs improved survival concomitant with an enhanced systemic bacterial clearance and improved inflammatory profile post CLP surgery. Despite some improvement in the inflammatory profile of the septic animals, treatment with UC-MSCs did not enhance survival or bacterial clearance. Overall, the beneficial therapeutic effects of BM-MSCs over UC-MSCs may likely be attributed to their pro-inflammatory function, and to some extent anti-inflammatory features, reflected in their gene expression pattern enhancing macrophage polarization to M1/M2 phenotypes resulting in a balanced pro- and anti-inflammatory response against polymicrobial sepsis.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Sepsis/terapia , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea/métodos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Inmunofenotipificación , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Sepsis/genética , Sepsis/inmunología , Sepsis/patología
5.
Mol Ther ; 29(5): 1716-1728, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33831557

RESUMEN

The inclusion of genes that control cell fate (so-called suicide, or kill-switch, genes) into gene therapy vectors is based on a compelling rationale for the safe and selective elimination of aberrant transfected cells. Prodrug-activated systems were developed in the 1980s and 1990s and rely on the enzymatic conversion of non-active prodrugs to active metabolites that lead to cell death. Although considerable effort and ingenuity has gone into vector design for gene therapy, less attention has been directed at the efficacy or associated adverse effects of the prodrug systems employed. In this review, we discuss prodrug systems employed in clinical trials and consider their role in the field of gene therapy. We highlight potential drawbacks associated with the use of specific prodrugs, such as systemic toxicity of the activated compound, the paucity of data on biodistribution of prodrugs, bystander effects, and destruction of genetically modified cells, and how these can inform future advances in cell therapies.


Asunto(s)
Terapia Genética/métodos , Neoplasias/terapia , Profármacos/uso terapéutico , Terapia Combinada , Humanos , Neoplasias/genética , Profármacos/farmacocinética , Distribución Tisular
6.
Blood ; 134(12): 951-959, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31292112

RESUMEN

By combining the analysis of mutations with aberrant expression of genes previously related to poorer prognosis in both acute promyelocytic leukemia (APL) and acute myeloid leukemia, we arrived at an integrative score in APL (ISAPL) and demonstrated its relationship with clinical outcomes of patients treated with all-trans retinoic acid (ATRA) in combination with anthracycline-based chemotherapy. Based on fms-like tyrosine kinase-3-internal tandem duplication mutational status; the ΔNp73/TAp73 expression ratio; and ID1, BAALC, ERG, and KMT2E gene expression levels, we modeled ISAPL in 159 patients (median ISAPL score, 3; range, 0-10). ISAPL modeling identified 2 distinct groups of patients, with significant differences in early mortality (P < .001), remission (P = .004), overall survival (P < .001), cumulative incidence of relapse (P = .028), disease-free survival (P = .03), and event-free survival (P < .001). These data were internally validated by using a bootstrap resampling procedure. At least for patients treated with ATRA and anthracycline-based chemotherapy, ISAPL modeling may identify those who need to be treated differently to maximize their chances for a cure.


Asunto(s)
Biomarcadores de Tumor/genética , Análisis Mutacional de ADN , Perfilación de la Expresión Génica , Leucemia Promielocítica Aguda/diagnóstico , Leucemia Promielocítica Aguda/genética , Adolescente , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores Farmacológicos/análisis , Biomarcadores de Tumor/análisis , Estudios de Cohortes , Análisis Mutacional de ADN/métodos , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/patología , Masculino , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular/métodos , Mutación , Pronóstico , Secuencias Repetidas en Tándem/genética , Transcriptoma , Resultado del Tratamiento , Tretinoina/administración & dosificación , Adulto Joven , Tirosina Quinasa 3 Similar a fms/genética
7.
Biol Blood Marrow Transplant ; 26(4): 672-682, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31676338

RESUMEN

Killer immunoglobulin-like receptor (KIR) and KIR ligand (KIRL) interactions play an important role in natural killer (NK) cell-mediated graft-versus-leukemia effect following hematopoietic cell transplantation (HCT). However, there is considerable heterogeneity in the KIR gene and KIRL content in individuals, making it difficult to estimate the full clinical impact of NK cell reconstitution following HCT. Here we present a novel adaptive mathematical model designed to quantify these interactions to better assess the influence of NK cell-mediated alloreactivity on transplant outcomes. Ninety-eight HLA- matched unrelated donor (URD) HCT recipients were studied retrospectively. The KIR-KIRL interactions were quantified using a system of matrix equations. Unit values were ascribed to each KIR-KIRL interaction, and the directionality of interactions was denoted by either a positive (activating) or negative (inhibition) symbol; these interactions were then summed. The absolute values of both the missing KIRL and inhibitory KIR-KIRL interactions were significantly associated with overall survival and relapse. These score components were initially used to develop a weighted score (w-KIR score) and subsequently a simplified, nonweighted KIR-KIRL interaction score (IM-KIR score). Increased w-KIR score and IM-KIR score were predictive of all-cause mortality and relapse (w-KIR score: hazard ratio [HR], .37 [P = .001] and .44 [P = .044], respectively; IM-KIR score: HR, .5 [P = .049] and .44 [P = .002], respectively). IM-KIR score was also associated with NK cell reconstitution post-HCT. KIR-KIRL interactions as reflected by the w-KIR and IM-KIR scores influence both relapse risk and survival in recipients of HLA-matched URD HCT with hematologic malignancies.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Donante no Emparentado , Humanos , Ligandos , Receptores KIR/genética , Estudios Retrospectivos
8.
Biol Blood Marrow Transplant ; 25(5): 868-882, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30677510

RESUMEN

High-throughput sequencing (HTS) of human T cell receptors has revealed a high level of complexity in the T cell repertoire, which makes it difficult to correlate T cell reconstitution with clinical outcomes. The associations identified thus far are of a broadly statistical nature, precluding precise modeling of outcomes based on T cell repertoire development following bone marrow transplantation (BMT). Previous work has demonstrated an inherent, mathematically definable order observed in the T cells from a diverse group of donors, which is perturbed in recipients following BMT. In this study, T cell receptor (TCR)-ß sequences from HLA-matched related donor and recipient pairs are analyzed to further develop this methodology. TCR-ß sequencing from unsorted and sorted T cell subsets isolated from the peripheral blood samples of BMT donors and recipients show conservation and symmetry of VJ segment usage in the clonal frequencies, linked to the organization of the gene segments along the TCR locus. This TCR-ß VJ segment translational symmetry is preserved post-transplantation and even in cases of acute graft-versus-host disease (aGVHD), suggesting that GVHD occurrence represents a polyclonal donor T cell response to recipient antigens. The complexity of the repertoire is significantly diminished after BMT, and the T cell clonal hierarchy is altered post-transplantation. Low-frequency donor clones tended to take on a higher rank in the recipients following BMT, especially in patients with aGVHD. Over time, the repertoire evolves to a more donor-like state in the recipients who did not develop GVHD as opposed to those who did. The results presented here support new methods of quantifying and characterizing post-transplantation T cell repertoire reconstitution.


Asunto(s)
Trasplante de Médula Ósea , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/inmunología , Adulto , Enfermedad Injerto contra Huésped/inmunología , Histocompatibilidad , Humanos , Donantes de Tejidos , Receptores de Trasplantes , Trasplante Homólogo
9.
Stem Cells ; 36(11): 1655-1662, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30171669

RESUMEN

Mesenchymal stromal cells (MSCs) deploy Toll-like receptors (TLRs) to respond to exogenous and endogenous signals. Activation of TLR pathways in MSCs alters their inflammatory profile and immunomodulatory effects on cells from both the innate and adaptive immune systems. Micro-RNAs (miRNAs), whose expression is modulated by TLR activation, can regulate inflammatory responses by targeting components of the TLR signaling pathways either in MSCs or in the cells with which they interact. Here, we review how the miRNA-TLR pathway axis can regulate the immunomodulatory functions of MSCs, including their interactions with monocytes/macrophages and natural killer cells, and discuss the therapeutic implications for MSC-based therapies. Stem Cells 2018;36:1655-1662.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Receptores Toll-Like/metabolismo , Diferenciación Celular , Humanos
10.
Gerontology ; 65(3): 294-298, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30897578

RESUMEN

Osteoarthritis (OA) is designated the 11th highest contributor of 291 diseases of global disability and the most common cause of chronic disability in elderly people. OA has a devastating impact on quality of life and represents an enormous socio-economic burden. Currently, OA is incurable, and no approved medications, biological therapy, or procedure prevents the progressive destruction of the osteoarthritic knee joint. All current treatments provide symptomatic relief rather than preventative or regenerative results. There is an urgent and compelling need to find, validate, and test new biological therapeutics. Cell-based therapies involving the delivery of mesenchymal stromal cells (MSCs) to the os-teoarthritic knee joint have emerged as a potential solution to overcome this clinical shortcoming. In this review, we address the clinical evidence, challenges, and recent advances surrounding MSC treatment in knee OA.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Osteoartritis de la Rodilla/terapia , Anciano , Anciano de 80 o más Años , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Femenino , Geriatría , Humanos , Masculino , Medicina Regenerativa
11.
Stem Cells ; 35(1): 265-275, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27571579

RESUMEN

Mesenchymal stromal cells (MSCs) are the subject of numerous clinical trials, largely due to their immunomodulatory and tissue regenerative properties. Toll-like receptors (TLRs), especially TLR3 and TLR4, are highly expressed on MSCs and their activation can significantly modulate the immunosuppressive and anti-inflammatory functions of MSCs. While MSCs can recruit and promote the generation of regulatory T cells (Tregs), the effect of TLR activation on MSC-mediated Treg induction is unknown. In this study, we investigated the effect of ligand-mediated activation of TLR3 and TLR4 on Treg induction by human MSCs. We found that generation of Tregs in human CD4(+) lymphocyte and MSC cocultures was enhanced by either TLR3 or TLR4 activation of MSCs and that the increase was abolished by TLR3 and TLR4 gene-silencing. Augmented Treg induction by TLR-activated MSCs was cell contact-dependent and associated with increased gene expression of the Notch ligand, Delta-like 1. Moreover, inhibition of Notch signaling abrogated the augmented Treg levels in the MSC cocultures. Our data show that TLR3 or TLR4 activation of MSCs increases Treg induction via the Notch pathway and suggest new means to enhance the potency of MSCs for treating disorders with an underlying immune dysfunction, including steroid resistant acute graft-versus-host disease. Stem Cells 2017;35:265-275.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Antígenos CD/metabolismo , Comunicación Celular , Silenciador del Gen , Humanos , Terapia de Inmunosupresión , Ligandos , Linfocitos T Reguladores , Trasplante Homólogo
12.
Haematologica ; 103(10): 1720-1729, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29976748

RESUMEN

Patients with acute myeloid leukemia (AML) often relapse after initial therapy because of persistence of leukemic stem cells that frequently express the IL-3 receptor alpha chain CD123. Natural killer (NK) cell-based therapeutic strategies for AML show promise and we explore the NK cell lines, NK-92 and CD16+ NK-92, as a treatment for AML. NK-92 has been tested in phase I clinical trials with minimal toxicity; irradiation prior to infusion prevents risk of engraftment. The CD16 negative NK-92 parental line was genetically modified to express the high affinity Fc gamma receptor, enabling antibody-dependent cell-mediated cytotoxicity, which we utilized in combination with an anti-CD123 antibody to target leukemic stem cells. NK-92 was preferentially cytotoxic against leukemic stem and progenitor cells compared with bulk leukemia in in vitro assays, while CD16+ NK-92 in combination with an anti-CD123 mAb mediated antibody-dependent cell-mediated cytotoxicity against CD123+ leukemic targets. Furthermore, NK-92 infusions (with or without prior irradiation) improved survival in a primary AML xenograft model. Mice xenografted with primary human AML cells had a superior survival when treated with irradiated CD16+NK-92 cells and an anti-CD123 monoclonal antibody (7G3) versus treatment with irradiated CD16+NK-92 cells combined with an isotype control antibody. In this proof-of-principle study, we show for the first time that a CD16+NK-92 cell line combined with an antibody that targets a leukemic stem cell antigen can lead to improved survival in a relevant pre-clinical model of AML.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Subunidad alfa del Receptor de Interleucina-3/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Receptores de IgG/antagonistas & inhibidores , Animales , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Células K562 , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Neoplasias/metabolismo , Receptores de IgG/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cytotherapy ; 20(8): 1001-1012, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30076069

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) promote wound healing, including after radiotherapy (RT) and surgery. The use of MSCs in regenerative medicine in the context of malignancy, such as to enhance wound healing post-RT/surgery in patients with soft tissue sarcomas (STSs), requires safety validation. The aim of this study was to determine the effects of human MSCs on STS growth in vitro and local recurrence and metastasis in vivo. METHODS: Human primary STS and HT-1080 fibrosarcoma lines were transduced to express luciferase/eGFP (enhanced green fluorescent protein). Sarcoma cells were co-cultured or co-injected with bone marrow-derived MSCs for growth studies. Xenograft tumor models were established with STS lines in NOD/SCID/γcnull mice. To emulate a clinical scenario, subcutaneous tumors were treated with RT/surgery prior to MSC injection into the tumor bed. Local and distant tumor recurrence was studied using histology and bioluminescence imaging. RESULTS: MSCs did not promote STS proliferation upon co-culture in vitro, which was consistent among MSCs from different donors. Co-injection of MSCs with sarcoma cells in mice exhibited no significant tumor-stimulating effect, compared with control mice injected with sarcoma cells alone. MSC administration after RT/surgery had no effect on local recurrence or metastasis of STS. DISCUSSION: These studies are important for the establishment of a safety profile for MSC administration in patients with STS. Our data suggest that MSCs are safe in STS management after standard of care RT/surgery, which can be further investigated in early-phase clinical trials to also determine the efficacy of MSCs in reducing morbidity and to mitigate wound complications in these patients.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Radioterapia , Sarcoma/patología , Sarcoma/terapia , Procedimientos Quirúrgicos Operativos , Adulto , Animales , Técnicas de Cocultivo , Terapia Combinada , Células HEK293 , Xenoinjertos , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Recurrencia Local de Neoplasia/etiología , Recurrencia Local de Neoplasia/patología , Radioterapia/efectos adversos , Radioterapia/métodos , Procedimientos Quirúrgicos Operativos/efectos adversos , Procedimientos Quirúrgicos Operativos/métodos , Células Tumorales Cultivadas , Cicatrización de Heridas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Biol Blood Marrow Transplant ; 23(3): 405-411, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28007665

RESUMEN

Daclizumab, a humanized monoclonal antibody, binds CD25 and blocks formation of the IL-2 receptor on T cells. A study of daclizumab as acute graft-versus-host disease (GVHD) prophylaxis after unrelated bone marrow transplantation was conducted before the importance of CD25+FOXP3+ regulatory T cells (Tregs) was recognized. Tregs can abrogate the onset of GVHD. The relation between Tregs and a graft-versus-malignancy effect is not fully understood. An international, multicenter, double-blind clinical trial randomized 210 adult or pediatric patients to receive 5 weekly doses of daclizumab at 0.3 mg/kg (n = 69) or 1.2 mg/kg (n = 76) or placebo (n = 65) after unrelated marrow transplantation for treatment of hematologic malignancies or severe aplastic anemia. The risk of acute GVHD did not differ among the groups (P = .68). Long-term follow-up of clinical outcomes and correlative analysis of peripheral blood T cell phenotype suggested that the patients treated with daclizumab had an increased risk of chronic GVHD (hazard ratio [HR], 1.49; 95% confidence interval [CI], 1.0 to 2.3; P = .08) and a decreased risk of relapse (HR, 0.57; 95% CI, 0.3 to 1.0; P = .05), but similar survival (HR, 0.89; 95% CI, 0.6 to 1.3; P = .53). T cells from a subset of patients (n = 107) were analyzed by flow cytometry. Compared with placebo, treatment with daclizumab decreased the proportion of Tregs among CD4 T cells at days 11-35 and increased the proportion of central memory cells among CD4 T cells at 1 year. Prophylactic administration of daclizumab does not prevent acute GVHD, but may increase the risk of chronic GVHD and decrease the risk of relapse. By delaying Treg reconstitution and promoting immunologic memory, anti-CD25 therapy may augment alloreactivity and antitumor immunity.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunoglobulina G/farmacología , Subunidad alfa del Receptor de Interleucina-2/antagonistas & inhibidores , Linfocitos T Reguladores/efectos de los fármacos , Adolescente , Adulto , Anticuerpos Monoclonales Humanizados/uso terapéutico , Recuento de Linfocito CD4 , Niño , Preescolar , Daclizumab , Método Doble Ciego , Femenino , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Inmunoglobulina G/uso terapéutico , Lactante , Masculino , Persona de Mediana Edad , Linfocitos T Reguladores/citología , Trasplante Homólogo , Adulto Joven
15.
J Immunol ; 194(9): 4298-308, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25825436

RESUMEN

Mesenchymal stem/stromal cells (MSCs) can influence the destiny of hematopoietic stem/progenitor cells (HSCs) and exert broadly immunomodulatory effects on immune cells. However, how MSCs regulate the differentiation of regulatory dendritic cells (regDCs) from HSCs remains incompletely understood. In this study, we show that mouse bone marrow-derived Sca-1(+)Lin(-)CD117(-) MSCs can drive HSCs to differentiate into a novel IFN regulatory factor (IRF)8-controlled regDC population (Sca(+) BM-MSC-driven DC [sBM-DCs]) when cocultured without exogenous cytokines. The Notch pathway plays a critical role in the generation of the sBM-DCs by controlling IRF8 expression in an RBP-J-dependent way. We observed a high level of H3K27me3 methylation and a low level of H3K4me3 methylation at the Irf8 promoter during sBM-DC induction. Importantly, infusion of sBM-DCs could alleviate colitis in mice with inflammatory bowel disease by inhibiting lymphocyte proliferation and increasing the numbers of CD4(+)CD25(+) regulatory T cells. Thus, these data infer a possible mechanism for the development of regDCs and further support the role of MSCs in treating immune disorders.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Factores Reguladores del Interferón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Antígenos Ly/metabolismo , Diferenciación Celular , Citocinas/metabolismo , Células Dendríticas/citología , Modelos Animales de Enfermedad , Expresión Génica , Histonas/metabolismo , Inmunomodulación , Mediadores de Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Factores Reguladores del Interferón/genética , Proteínas de la Membrana/metabolismo , Ratones , Modelos Biológicos , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo
16.
Curr Opin Organ Transplant ; 22(1): 86-96, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28005573

RESUMEN

PURPOSE OF REVIEW: This review focuses on articles published from January 2015 to June 2016 on mesenchymal stromal cell (MSC) therapy for cardiac regeneration and repair. RECENT FINDINGS: During this period, reports published on MSCs address the best MSC tissue source for cellular therapy, mechanisms of MSC activity and improving MSC longevity, and homing in vivo. Currently, there is no definitive therapeutic advantage of any one tissue-derived MSC over another, and even combination therapies struggle with conflicting outcomes. MSC activity, persistence in vivo, or homing can be improved by priming strategies, genetic modification, or biomaterials. Despite numerous studies showing improvement in heart function after acute cardiac injury, the reproducibility and efficacy of the therapy remains elusive and falls short of expectations in clinical trials. Although the safety of MSCs is undisputed, the success of MSC preparations in improving cardiac function clinically remains uncertain due to challenges in correlating MSC potency with clinical outcomes, donor-related variation in MSC function, and a profusion of culture methodologies. SUMMARY: Several strategies are available to advance MSC cell therapy for acute cardiac injury to promote cardiac regeneration and repair in rigorous preclinical and clinical studies.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Corazón/fisiopatología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Regeneración
17.
Stem Cells ; 33(11): 3212-27, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26086629

RESUMEN

Chronic heart failure is a leading cause of death. The demand for new therapies and the potential regenerative capacity of bone marrow-derived cells has led to numerous clinical trials. We critically discuss current knowledge of the biology and clinical application of bone marrow cells. It appears unlikely that bone marrow cells can develop into functional cardiomyocyte after infusion but may have favorable paracrine effects. Most, but not all, clinical trials report a modest short- but not long-term benefit of infusing bone marrow-derived cells. Effect size appears to correlate with stringency of study-design: the most stringent trials report the smallest effect-sizes. We conclude there may be short- but not substantial long-term benefit of infusing bone marrow-derived cells into persons with chronic heart failure and any benefit observed is unlikely to result from trans-differentiation of bone marrow-derived cells into functioning cardiomyocytes.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea/métodos , Cardiopatías/terapia , Regeneración/fisiología , Animales , Trasplante de Médula Ósea/tendencias , Diferenciación Celular/fisiología , Enfermedad Crónica , Ensayos Clínicos como Asunto/métodos , Cardiopatías/diagnóstico , Cardiopatías/fisiopatología , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Humanos , Miocitos Cardíacos/fisiología
18.
Cytotherapy ; 18(6): 729-39, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27173749

RESUMEN

BACKGROUND AIMS: The use of bone marrow-derived mesenchymal stromal cells (MSCs) in cell-based therapies is currently being developed for a number of diseases. Thus far, the clinical results have been inconclusive and variable, in part because of the variety of cell isolation procedures and culture conditions used in each study. A new isolation technique that streamlines the method of concentration and demands less time and attention could provide clinical and economic advantages compared with current methodologies. In this study, we evaluated the concentrating capability of an integrated centrifuge-based technology compared with standard Ficoll isolation. METHODS: MSCs were concentrated from bone marrow aspirate using the new device and the Ficoll method. The isolation capabilities of the device and the growth characteristics, secretome production, and differentiation capacity of the derived cells were determined. RESULTS: The new MSC isolation device concentrated the bone marrow in 90 seconds and resulted in a mononuclear cell yield 10-fold higher and with a twofold increase in cell retention compared with Ficoll. The cells isolated using the device were shown to exhibit similar morphology and functional activity as assessed by growth curves and secretome production compared to the Ficoll-isolated cells. The surface marker and trilineage differentiation profile of the device-isolated cells was consistent with the known profile of MSCs. DISCUSSION: The faster time to isolation and greater cell yield of the integrated centrifuge-based technology may make this an improved approach for MSC isolation from bone marrow aspirates.


Asunto(s)
Células de la Médula Ósea/citología , Separación Celular/métodos , Centrifugación/métodos , Células Madre Mesenquimatosas/citología , Médula Ósea , Diferenciación Celular/fisiología , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ficoll , Humanos , Osteoblastos/citología
19.
Mol Ther ; 23(3): 561-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25409745

RESUMEN

The contribution of bone marrow cells (BMC) in lung repair is controversial. We previously reported a subpopulation of BMC that express Clara cell secretory protein (CCSP). To determine the contribution of endogenous CCSP(+) BMC to airway regeneration, we performed bone marrow transplantation studies using the CCtk mouse, which expresses a thymidine kinase suicide gene under regulation of the CCSP promoter. Mice were transplanted with wild-type or CCtk BMC and treated with ganciclovir to eliminate CCSP(+) cells. After airway injury using naphthalene, mice depleted of CCSP(+) BMC had more inflammatory cells in lung and decreased levels of oxygen in arterial blood. They also had reduced expression of airway epithelial genes and less Clara cells compared to control mice that had intact CCSP(+) BMC and bone marrow derived CCSP(+) cells in the airways. After naphthalene injury, administration of CCSP reproduced the beneficial effect of CCSP(+) BMC by improving recovery of airway epithelium, reducing lung inflammation and increasing oxygen in arterial blood from mice depleted of CCSP(+) BMC. Our data demonstrate that ablation of CCSP(+) BMC delays airway recovery and suggests the beneficial effect of CCSP(+) BMC in lung recovery is in part due to production of CCSP itself.


Asunto(s)
Lesión Pulmonar Aguda/genética , Células de la Médula Ósea/metabolismo , Células Epiteliales/metabolismo , Pulmón/metabolismo , Regeneración/genética , Uteroglobina/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/terapia , Animales , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica , Genes Letales , Humanos , Pulmón/patología , Masculino , Ratones Transgénicos , Naftalenos , Oxígeno/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Transgenes , Uteroglobina/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
20.
Br J Haematol ; 168(3): 384-94, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25303497

RESUMEN

Acute myeloid leukaemia (AML) patients with hyperleucocytosis have higher early mortality, lower complete remission (CR) and overall survival (OS). Whether different pre-induction leucoreduction strategies can improve outcome is unknown. A single centre retrospective cohort study was conducted on AML patients with a white blood cell count (WBC) >100 × 10(9) /l between 1987 and 1997, and on all AML patients between 1998 and 2006, to determine (a) the effect of four different leucoreductive strategies (leukapheresis, hydroxycarbamide, leukapheresis and hydroxycarbamide or no pre-induction leucoreduction) on early (day 28) mortality, CR, and OS; and (b) whether a high presenting WBC remains a negative predictor of OS in patients surviving induction (first 28 d). In the 1998-2006 cohort (n = 702), higher WBC was associated with higher early mortality and lower OS but its effects were greatly diminished in patients who survived the first 28 d (Hazard Ratio 1·094 vs. 1·002). A WBC of 34·1 × 10(9) /l had the highest sensitivity (75·6%) and specificity (67·4%) for early mortality. None of the four leucoreduction strategies differed significantly in early mortality, CR, or OS in patients with WBC>100 × 10(9) /l (n = 166). The number of leucostatic signs was a significant predictor of early mortality (P < 0·0001) and OS (P = 0·0007). The results suggest that AML patients with hyperleucocytosis should be induced, if eligible, without pre-induction leucoreduction.


Asunto(s)
Antineoplásicos/uso terapéutico , Hidroxiurea/uso terapéutico , Leucaféresis/métodos , Leucemia Mieloide Aguda/terapia , Leucocitosis/terapia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Terapia Combinada , Femenino , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/complicaciones , Recuento de Leucocitos , Leucocitosis/etiología , Masculino , Persona de Mediana Edad , Pronóstico , Inducción de Remisión , Estudios Retrospectivos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA