Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 151(3): 559-75, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23084400

RESUMEN

ETS transcription factors ETV2, FLI1, and ERG1 specify pluripotent stem cells into induced vascular endothelial cells (iVECs). However, iVECs are unstable and drift toward nonvascular cells. We show that human midgestation c-Kit(-) lineage-committed amniotic cells (ACs) can be reprogrammed into vascular endothelial cells (rAC-VECs) without transitioning through a pluripotent state. Transient ETV2 expression in ACs generates immature rAC-VECs, whereas coexpression with FLI1/ERG1 endows rAC-VECs with a vascular repertoire and morphology matching mature endothelial cells (ECs). Brief TGFß-inhibition functionalizes VEGFR2 signaling, augmenting specification of ACs into rAC-VECs. Genome-wide transcriptional analyses showed that rAC-VECs are similar to adult ECs in which vascular-specific genes are expressed and nonvascular genes are silenced. Functionally, rAC-VECs form stable vasculature in Matrigel plugs and regenerating livers. Therefore, short-term ETV2 expression and TGFß inhibition with constitutive ERG1/FLI1 coexpression reprogram mature ACs into durable rAC-VECs with clinical-scale expansion potential. Banking of HLA-typed rAC-VECs establishes a vascular inventory for treatment of diverse disorders.


Asunto(s)
Líquido Amniótico/citología , Diferenciación Celular , Células Endoteliales/citología , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Oncogénicas de Retroviridae/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos
2.
Blood ; 143(17): 1713-1725, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38194692

RESUMEN

ABSTRACT: Tisagenlecleucel is approved for adults with relapsed/refractory (r/r) follicular lymphoma (FL) in the third- or later-line setting. The primary analysis (median follow-up, 17 months) of the phase 2 ELARA trial reported high response rates and excellent safety profile in patients with extensively pretreated r/r FL. Here, we report longer-term efficacy, safety, pharmacokinetic, and exploratory biomarker analyses after median follow-up of 29 months (interquartile range, 22.2-37.7). As of 29 March 2022, 97 patients with r/r FL (grades 1-3A) received tisagenlecleucel infusion (0.6 × 108-6 × 108 chimeric antigen receptor-positive viable T cells). Bridging chemotherapy was allowed. Baseline clinical factors, tumor microenvironment, blood soluble factors, and circulating blood cells were correlated with clinical response. Cellular kinetics were assessed by quantitative polymerase chain reaction. Median progression-free survival (PFS), duration of response (DOR), and overall survival (OS) were not reached. Estimated 24-month PFS, DOR, and OS rates in all patients were 57.4% (95% confidence interval [CI], 46.2-67), 66.4% (95% CI, 54.3-76), and 87.7% (95% CI, 78.3-93.2), respectively. Complete response rate and overall response rate were 68.1% (95% CI, 57.7-77.3) and 86.2% (95% CI, 77.5-92.4), respectively. No new safety signals or treatment-related deaths were reported. Low levels of tumor-infiltrating LAG3+CD3+ exhausted T cells and higher baseline levels of naïve CD8+ T cells were associated with improved outcomes. Tisagenlecleucel continued to demonstrate highly durable efficacy and a favorable safety profile in this extended follow-up of 29 months in patients with r/r FL enrolled in ELARA. This trial was registered at www.clinicaltrials.gov as #NCT03568461.


Asunto(s)
Linfoma Folicular , Humanos , Linfoma Folicular/tratamiento farmacológico , Linfoma Folicular/mortalidad , Persona de Mediana Edad , Masculino , Femenino , Anciano , Adulto , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T/uso terapéutico , Estudios de Seguimiento , Resultado del Tratamiento
3.
Nature ; 569(7755): 222-228, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30971824

RESUMEN

The bone marrow microenvironment has a key role in regulating haematopoiesis, but its molecular complexity and response to stress are incompletely understood. Here we map the transcriptional landscape of mouse bone marrow vascular, perivascular and osteoblast cell populations at single-cell resolution, both at homeostasis and under conditions of stress-induced haematopoiesis. This analysis revealed previously unappreciated levels of cellular heterogeneity within the bone marrow niche and resolved cellular sources of pro-haematopoietic growth factors, chemokines and membrane-bound ligands. Our studies demonstrate a considerable transcriptional remodelling of niche elements under stress conditions, including an adipocytic skewing of perivascular cells. Among the stress-induced changes, we observed that vascular Notch delta-like ligands (encoded by Dll1 and Dll4) were downregulated. In the absence of vascular Dll4, haematopoietic stem cells prematurely induced a myeloid transcriptional program. These findings refine our understanding of the cellular architecture of the bone marrow niche, reveal a dynamic and heterogeneous molecular landscape that is highly sensitive to stress and illustrate the utility of single-cell transcriptomic data in evaluating the regulation of haematopoiesis by discrete niche populations.


Asunto(s)
Médula Ósea/irrigación sanguínea , Microambiente Celular , Hematopoyesis , Células Madre Hematopoyéticas , Análisis de la Célula Individual , Nicho de Células Madre , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular , Linaje de la Célula , Endotelio Vascular/citología , Femenino , Regulación de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Células Mieloides/citología , Células Mieloides/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , RNA-Seq , Receptores Notch/metabolismo , Nicho de Células Madre/genética , Estrés Fisiológico/genética , Transcriptoma/genética
5.
N Engl J Med ; 382(19): 1800-1810, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32320566

RESUMEN

BACKGROUND: Acute graft-versus-host disease (GVHD) remains a major limitation of allogeneic stem-cell transplantation; not all patients have a response to standard glucocorticoid treatment. In a phase 2 trial, ruxolitinib, a selective Janus kinase (JAK1 and JAK2) inhibitor, showed potential efficacy in patients with glucocorticoid-refractory acute GVHD. METHODS: We conducted a multicenter, randomized, open-label, phase 3 trial comparing the efficacy and safety of oral ruxolitinib (10 mg twice daily) with the investigator's choice of therapy from a list of nine commonly used options (control) in patients 12 years of age or older who had glucocorticoid-refractory acute GVHD after allogeneic stem-cell transplantation. The primary end point was overall response (complete response or partial response) at day 28. The key secondary end point was durable overall response at day 56. RESULTS: A total of 309 patients underwent randomization; 154 patients were assigned to the ruxolitinib group and 155 to the control group. Overall response at day 28 was higher in the ruxolitinib group than in the control group (62% [96 patients] vs. 39% [61]; odds ratio, 2.64; 95% confidence interval [CI], 1.65 to 4.22; P<0.001). Durable overall response at day 56 was higher in the ruxolitinib group than in the control group (40% [61 patients] vs. 22% [34]; odds ratio, 2.38; 95% CI, 1.43 to 3.94; P<0.001). The estimated cumulative incidence of loss of response at 6 months was 10% in the ruxolitinib group and 39% in the control group. The median failure-free survival was considerably longer with ruxolitinib than with control (5.0 months vs. 1.0 month; hazard ratio for relapse or progression of hematologic disease, non-relapse-related death, or addition of new systemic therapy for acute GVHD, 0.46; 95% CI, 0.35 to 0.60). The median overall survival was 11.1 months in the ruxolitinib group and 6.5 months in the control group (hazard ratio for death, 0.83; 95% CI, 0.60 to 1.15). The most common adverse events up to day 28 were thrombocytopenia (in 50 of 152 patients [33%] in the ruxolitinib group and 27 of 150 [18%] in the control group), anemia (in 46 [30%] and 42 [28%], respectively), and cytomegalovirus infection (in 39 [26%] and 31 [21%]). CONCLUSIONS: Ruxolitinib therapy led to significant improvements in efficacy outcomes, with a higher incidence of thrombocytopenia, the most frequent toxic effect, than that observed with control therapy. (Funded by Novartis; REACH2 ClinicalTrials.gov number, NCT02913261.).


Asunto(s)
Enfermedad Injerto contra Huésped/tratamiento farmacológico , Inhibidores de las Cinasas Janus/uso terapéutico , Pirazoles/uso terapéutico , Trasplante de Células Madre/efectos adversos , Enfermedad Aguda , Adolescente , Adulto , Anciano , Niño , Femenino , Glucocorticoides/uso terapéutico , Humanos , Inhibidores de las Cinasas Janus/efectos adversos , Masculino , Persona de Mediana Edad , Nitrilos , Pirazoles/efectos adversos , Pirimidinas , Trombocitopenia/inducido químicamente , Trasplante Homólogo , Adulto Joven
6.
Blood ; 137(14): 1970-1979, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33512442

RESUMEN

We determined the efficacy of tocilizumab (TCZ) in preventing grade 2-4 acute graft-versus-host disease (aGVHD) in patients with acute leukemia or myelodysplasia undergoing matched sibling donor (MSD) or volunteer unrelated donor (VUD) allogeneic stem cell transplantation after myeloablative or reduced-intensity conditioning across 5 Australian centers. A total of 145 patients (50 MSD, 95 VUD) were randomly assigned to placebo or TCZ on day -1. All patients received T-cell-replete peripheral blood stem cell grafts and graft-versus-host disease (GVHD) prophylaxis with cyclosporin/methotrexate. A planned substudy analyzed the VUD cohort. With a median follow-up of 746 days, the incidence of grade 2-4 aGVHD at day 100 for the entire cohort was 36% for placebo vs 27% for TCZ (hazard ratio [HR], 0.69; 95% confidence interval [CI], 0.38-1.26; P = .23) and 45% vs 32% (HR, 0.61; 95% CI, 0.31-1.22; P = .16) for the VUD subgroup. The incidence of grade 2-4 aGVHD at day 180 for the entire cohort was 40% for placebo vs 29% for TCZ (HR, 0.68; 95% CI, 0.38-1.22; P = .19) and 48% vs 32% (HR, 0.59; 95% CI, 0.30-1.16; P = .13) for the VUD subgroup. Reductions in aGVHD were predominantly in grade 2 disease. For the entire cohort, transplant-related mortality occurred in 8% vs 11% of placebo-treated vs TCZ-treated patients, respectively (P = .56), and overall survival was 79% vs 71% (P = .27). Median day to neutrophil and platelet engraftment was delayed by 2 to 3 days in TCZ-treated patients, whereas liver toxicity and infectious complications were similar between groups. In this phase 3 randomized double-blind trial, TCZ showed nonsignificant trends toward reduced incidence of grade 2-4 aGVHD in recipients from HLA-matched VUDs but no improvements in long term-survival.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Ciclosporina/uso terapéutico , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas , Inmunosupresores/uso terapéutico , Metotrexato/uso terapéutico , Adulto , Método Doble Ciego , Femenino , Humanos , Leucemia/terapia , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/terapia , Efecto Placebo , Trasplante Homólogo , Resultado del Tratamiento
7.
Nature ; 545(7655): 439-445, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28514438

RESUMEN

Developmental pathways that orchestrate the fleeting transition of endothelial cells into haematopoietic stem cells remain undefined. Here we demonstrate a tractable approach for fully reprogramming adult mouse endothelial cells to haematopoietic stem cells (rEC-HSCs) through transient expression of the transcription-factor-encoding genes Fosb, Gfi1, Runx1, and Spi1 (collectively denoted hereafter as FGRS) and vascular-niche-derived angiocrine factors. The induction phase (days 0-8) of conversion is initiated by expression of FGRS in mature endothelial cells, which results in endogenous Runx1 expression. During the specification phase (days 8-20), RUNX1+ FGRS-transduced endothelial cells commit to a haematopoietic fate, yielding rEC-HSCs that no longer require FGRS expression. The vascular niche drives a robust self-renewal and expansion phase of rEC-HSCs (days 20-28). rEC-HSCs have a transcriptome and long-term self-renewal capacity similar to those of adult haematopoietic stem cells, and can be used for clonal engraftment and serial primary and secondary multi-lineage reconstitution, including antigen-dependent adaptive immune function. Inhibition of TGFß and CXCR7 or activation of BMP and CXCR4 signalling enhanced generation of rEC-HSCs. Pluripotency-independent conversion of endothelial cells into autologous authentic engraftable haematopoietic stem cells could aid treatment of haematological disorders.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Endotelio/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Inmunidad Adaptativa , Envejecimiento/genética , Animales , Línea Celular , Linaje de la Célula , Autorrenovación de las Células , Células Clonales/citología , Células Clonales/trasplante , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
8.
Support Care Cancer ; 32(1): 44, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38112867

RESUMEN

PURPOSE: Oral mucositis (OM) is a common complication in haematopoietic stem cell transplantation (HSCT). Polaprezinc, an anti-ulcer drug, has been shown to be effective to prevent OM in several studies when administered topically and systemically. This study aimed to evaluate the effectiveness of topical polaprezinc in patients undergoing HSCT. METHODS: This was an open-label randomised clinical trial comparing polaprezinc and sodium bicarbonate mouthwashes for the prevention of severe OM in HSCT patients. Adult patients who received conditioning regimens at moderate to high risk of developing OM were included. The primary endpoint was the incidence of severe (WHO grades 3-4) OM. The secondary endpoints included duration of grades 3-4 OM, incidence and duration of grades 2-4 OM, patient-reported pain and functional limitations. RESULTS: In total, 108 patients (55 test arm and 53 control arm) were randomised. There was no difference in the incidence of grades 3 to 4 OM (35% test arm versus 36% control arm). The secondary endpoints were not significantly different. In both arms, patients reported more throat pain compared to mouth pain. CONCLUSIONS: Topical polaprezinc had no effect in the prevention of OM in HSCT patients. Further research is required to evaluate the effects of systemic polaprezinc. The OM assessment tool needs to be reviewed as throat mucositis was a main issue in this study. TRIAL REGISTRATION: ACTRN12320001188921 (Date Registered: 10th November 2020).


Asunto(s)
Carnosina , Trasplante de Células Madre Hematopoyéticas , Estomatitis , Adulto , Humanos , Carnosina/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Dolor/etiología , Estomatitis/etiología , Estomatitis/prevención & control , Estomatitis/tratamiento farmacológico
9.
Intern Med J ; 53(3): 323-329, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35833788

RESUMEN

BACKGROUND: The COVID-19 pandemic has caused major disruption to health systems, with allogeneic haemopoietic cell transplant (alloHCT) services a particularly vulnerable area. Ongoing provision of alloHCT has required dynamic responses at national and local levels. In Australia and New Zealand (ANZ), a high reliance on unrelated donors from overseas registries has posed an additional challenge. AIMS: To describe the impact of COVID-19 on alloHCT services in ANZ in the first year of the pandemic. METHODS: Data from the national alloHCT patient and unrelated donor registries were extracted for a 2-year time frame. Comparisons were made between a pre-pandemic period of 1 March 2019 to 29 February 2020 and the corresponding dates during the pandemic, 1 March 2020 to 28 February 2021. RESULTS: There was a 13% decrease in the number of allogeneic transplants, a reversal of steady increases in previous years, with the largest decrease in unrelated donor transplants. Local donors supplied a greater proportion of unrelated stem cell products. With a switch to universal cryopreservation, the time from request of a product to infusion increased by a median of 25.5 days for overseas products and 14 days for local products. There was a significant increase in the number of products collected but not used. CONCLUSIONS: A strong public health response and coordinated transplant community activities allowed for safe provision of alloHCT in ANZ; however, our data suggest that the timely delivery of allogeneic transplants was affected by the COVID-19 pandemic. Continued dedicated efforts are required to minimise further impacts.


Asunto(s)
COVID-19 , Trasplante de Células Madre Hematopoyéticas , Humanos , Pandemias , Nueva Zelanda/epidemiología , COVID-19/epidemiología , Estudios Retrospectivos , Australia/epidemiología
10.
Intern Med J ; 53(12): 2319-2329, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38014511

RESUMEN

This position paper provides an overview of the assessment and management of both acute and chronic graft-versus-host disease (GvHD). There is a focus on the use of ruxolitinib, a selective inhibitor of Janus kinase (JAK)1 and JAK2, for the treatment of corticosteroid-refractory and corticosteroid-dependent GvHD.


Asunto(s)
Síndrome de Bronquiolitis Obliterante , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Humanos , Consenso , Esteroides/uso terapéutico , Nitrilos , Corticoesteroides/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Aguda , Enfermedad Crónica
11.
Intern Med J ; 53(1): 119-125, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36371767

RESUMEN

Patients with post-haemopoietic stem cell transplant or chimeric antigen receptor T -cell (CAR-T) therapy face a significant risk of morbidity and mortality from coronavirus disease 2019 because of their immunosuppressed state. As case numbers in Australia and New Zealand continue to rise, guidance on management in this high-risk population is needed. Whilst we have learned much from international colleagues who faced high infection rates early in the pandemic, guidance relevant to local health system structures, medication availability and emerging therapies is essential to equip physicians to manage our patients optimally.


Asunto(s)
COVID-19 , Trasplante de Células Madre Hematopoyéticas , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/uso terapéutico , Nueva Zelanda/epidemiología , Linfocitos T
12.
Nature ; 529(7586): 316-25, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26791722

RESUMEN

Endothelial cells that line capillaries are not just passive conduits for delivering blood. Tissue-specific endothelium establishes specialized vascular niches that deploy sets of growth factors, known as angiocrine factors. These cues participate actively in the induction, specification, patterning and guidance of organ regeneration, as well as in the maintainance of homeostasis and metabolism. When upregulated following injury, they orchestrate self-renewal and differentiation of tissue-specific resident stem and progenitor cells into functional organs. Uncovering the mechanisms by which organotypic endothelium distributes physiological levels of angiocrine factors both spatially and temporally will lay the foundation for clinical trials that promote organ repair without scarring.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica , Comunicación Paracrina , Animales , Capilares/citología , Diferenciación Celular , Autorrenovación de las Células , Homeostasis , Humanos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Pulmón/citología , Pulmón/patología , Especificidad de Órganos , Osteogénesis , Regeneración
13.
Nature ; 532(7599): 323-8, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27074509

RESUMEN

Bone marrow endothelial cells (BMECs) form a network of blood vessels that regulate both leukocyte trafficking and haematopoietic stem and progenitor cell (HSPC) maintenance. However, it is not clear how BMECs balance these dual roles, and whether these events occur at the same vascular site. We found that mammalian bone marrow stem cell maintenance and leukocyte trafficking are regulated by distinct blood vessel types with different permeability properties. Less permeable arterial blood vessels maintain haematopoietic stem cells in a low reactive oxygen species (ROS) state, whereas the more permeable sinusoids promote HSPC activation and are the exclusive site for immature and mature leukocyte trafficking to and from the bone marrow. A functional consequence of high permeability of blood vessels is that exposure to blood plasma increases bone marrow HSPC ROS levels, augmenting their migration and differentiation, while compromising their long-term repopulation and survival. These findings may have relevance for clinical haematopoietic stem cell transplantation and mobilization protocols.


Asunto(s)
Vasos Sanguíneos/citología , Vasos Sanguíneos/fisiología , Médula Ósea/irrigación sanguínea , Hematopoyesis , Animales , Antígenos Ly/metabolismo , Arterias/citología , Arterias/fisiología , Células de la Médula Ósea/citología , Diferenciación Celular , Movimiento Celular , Autorrenovación de las Células , Supervivencia Celular , Quimiocina CXCL12/metabolismo , Células Endoteliales/fisiología , Femenino , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Leucocitos/citología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Nestina/metabolismo , Pericitos/fisiología , Permeabilidad , Plasma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores CXCR4/metabolismo
14.
Intern Med J ; 51(8): 1321-1323, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34423546

RESUMEN

Australia and New Zealand have achieved excellent community control of COVID-19 infection. In light of the imminent COVID-19 vaccination roll out in both countries, representatives of all adult and paediatric allogeneic bone marrow transplant and cellular therapy (TCT) centres as well as representatives from autologous transplant only centres in Australia and New Zealand collaborated with infectious diseases specialists with expertise in TCT on this consensus position statement regarding COVID-19 vaccination in TCT patients in Australia and New Zealand. It is our recommendation that TCT patients, should have expedited access to high-efficacy COVID-19 vaccines given that these patients are at high risk of morbidity and mortality from COVID-19 infection. We also recommend prioritising vaccination of TCT healthcare workers and household members of TCT patients. Vaccination should not replace other public health measures in TCT patients given the effectiveness of COVID-19 vaccination in TCT patients is unknown. Furthermore, given the limited available data, prospective collection of safety and efficacy data of COVID-19 vaccination in this patient group is a priority.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Receptores de Trasplantes , Adulto , Australia/epidemiología , COVID-19/prevención & control , Niño , Consenso , Humanos , Nueva Zelanda/epidemiología , Estudios Prospectivos , Vacunación
15.
Development ; 144(13): 2469-2479, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28526753

RESUMEN

EGFL7 is a secreted angiogenic factor produced by embryonic endothelial cells. To understand its role in placental development, we established a novel Egfl7 knockout mouse. The mutant mice have gross defects in chorioallantoic branching morphogenesis and placental vascular patterning. Microangiography and 3D imaging revealed patchy perfusion of Egfl7-/- placentas marked by impeded blood conductance through sites of narrowed vessels. Consistent with poor feto-placental perfusion, Egfl7 knockout resulted in reduced placental weight and fetal growth restriction. The placentas also showed abnormal fetal vessel patterning and over 50% reduction in fetal blood space. In vitro, placental endothelial cells were deficient in migration, cord formation and sprouting. Expression of genes involved in branching morphogenesis, Gcm1, Syna and Synb, and in patterning of the extracellular matrix, Mmrn1, were temporally dysregulated in the placentas. Egfl7 knockout did not affect expression of the microRNA embedded within intron 7. Collectively, these data reveal that Egfl7 is crucial for placental vascularization and embryonic growth, and may provide insight into etiological factors underlying placental pathologies associated with intrauterine growth restriction, which is a significant cause of infant morbidity and mortality.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Neovascularización Fisiológica , Perfusión , Placenta/irrigación sanguínea , Placenta/embriología , Placentación , Proteínas/metabolismo , Animales , Secuencia de Bases , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Tipificación del Cuerpo , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Regulación hacia Abajo/genética , Familia de Proteínas EGF , Células Endoteliales/metabolismo , Femenino , Sangre Fetal/metabolismo , Feto/embriología , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Placenta/metabolismo , Embarazo
16.
Analyst ; 145(16): 5532-5538, 2020 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-32608411

RESUMEN

We report separation of genomic DNA (48 kbp) from bovine serum albumin (BSA) by the electro-hydrodynamic coupling between a pressure-driven flow and a parallel electric field. Electro-hydrodynamic extraction exploits this coupling to trap DNA molecules at the entrance of a microfluidic contraction channel, while allowing proteins and salts to be flushed from the device. Samples (10 µL) containing λ-DNA (1 ng) and BSA (0.3 mg) were injected directly into the device and convected to the contraction channel entrance by a flowing buffer solution. The DNA remains trapped in this region essentially indefinitely, while proteins and salts are eluted. The effectiveness of the concept has been assessed by fluorescence measurements of DNA and BSA concentrations. Electro-hydrodynamic extraction in a single-stage device was found to enhance the concentration of DNA 40-fold, while reducing the BSA concentration by four orders of magnitude. The relative concentrations of DNA to BSA at the contraction channel entrance can be as large as 1.5 : 1, corresponding to an A260/280 ratio of 1.9. The maximum yield of DNA from a salt-free solution is 50%, while salted (150 mM) solutions have a lower yield (38%).


Asunto(s)
Hidrodinámica , Albúmina Sérica Bovina , ADN/genética
17.
Nature ; 511(7509): 312-8, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25030167

RESUMEN

Generating engraftable human haematopoietic cells from autologous tissues is a potential route to new therapies for blood diseases. However, directed differentiation of pluripotent stem cells yields haematopoietic cells that engraft poorly. Here, we have devised a method to phenocopy the vascular-niche microenvironment of haemogenic cells, thereby enabling reprogramming of human endothelial cells into engraftable haematopoietic cells without transition through a pluripotent intermediate. Highly purified non-haemogenic human umbilical vein endothelial cells or adult dermal microvascular endothelial cells were transduced with the transcription factors FOSB, GFI1, RUNX1 and SPI1 (hereafter referred to as FGRS), and then propagated on serum-free instructive vascular niche monolayers to induce outgrowth of haematopoietic colonies containing cells with functional and immunophenotypic features of multipotent progenitor cells (MPPs). These endothelial cells that have been reprogrammed into human MPPs (rEC-hMPPs) acquire colony-forming-cell potential and durably engraft into immune-deficient mice after primary and secondary transplantation, producing long-term rEC-hMPP-derived myeloid (granulocytic/monocytic, erythroid, megakaryocytic) and lymphoid (natural killer and B cell) progenies. Conditional expression of FGRS transgenes, combined with vascular induction, activates endogenous FGRS genes, endowing rEC-hMPPs with a transcriptional and functional profile similar to that of self-renewing MPPs. Our approach underscores the role of inductive cues from the vascular niche in coordinating and sustaining haematopoietic specification and may prove useful for engineering autologous haematopoietic grafts to treat inherited and acquired blood disorders.


Asunto(s)
Microambiente Celular , Reprogramación Celular , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Células Madre Multipotentes/citología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Células Madre Adultas/trasplante , Animales , Aorta , Linaje de la Célula , Células Endoteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Gónadas , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Linfocitos/citología , Mesonefro , Ratones , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/trasplante , Células Mieloides/citología , Células Madre Pluripotentes , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transgenes/genética
18.
Electrophoresis ; 40(3): 437-446, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30229964

RESUMEN

Long strands of DNA can be trapped and concentrated near the inlet of a microfluidic channel by applying a pressure gradient and an opposing electric field. The mechanism for trapping involves a migration of DNA perpendicular to both the fluid flow and the electric field. Migration leads to a highly nonuniform distribution of DNA within a cross section of the channel, with the bulk of the DNA concentrated in a thin (10 µm) layer next to the walls of the channel. This highly concentrated layer generates an electrophoretic flux toward the inlet to the device, despite the much larger fluid flow in the opposite direction. In this paper, the extent to which DNA can be trapped and concentrated by this means has been characterized by fluorescence measurements. At short times (<2 hours) nearly all the incoming DNA remains trapped within the device until the electric field is turned off. The DNA largely accumulates near the inlet, but after 30-60 minutes additional DNA starts to accumulate deeper into the channel. Eventually DNA leaks from the device itself, but ≈80% of the incoming DNA can be retained for up to 5 hours. Optimizing the electric field strength can increase the amount of DNA that can be trapped, but the efficiency is not affected by the channel cross-section.


Asunto(s)
ADN/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento/instrumentación , Técnicas Analíticas Microfluídicas/instrumentación , ADN/química , Diseño de Equipo , Técnicas Analíticas Microfluídicas/métodos , Polielectrolitos/química
20.
Mol Cell ; 44(1): 17-28, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21924933

RESUMEN

DNA methylation has been implicated as an epigenetic component of mechanisms that stabilize cell-fate decisions. Here, we have characterized the methylomes of human female hematopoietic stem/progenitor cells (HSPCs) and mature cells from the myeloid and lymphoid lineages. Hypomethylated regions (HMRs) associated with lineage-specific genes were often methylated in the opposing lineage. In HSPCs, these sites tended to show intermediate, complex patterns that resolve to uniformity upon differentiation, by increased or decreased methylation. Promoter HMRs shared across diverse cell types typically display a constitutive core that expands and contracts in a lineage-specific manner to fine-tune the expression of associated genes. Many newly identified intergenic HMRs, both constitutive and lineage specific, were enriched for factor binding sites with an implied role in genome organization and regulation of gene expression, respectively. Overall, our studies represent an important reference data set and provide insights into directional changes in DNA methylation as cells adopt terminal fates.


Asunto(s)
Metilación de ADN , Células Madre Hematopoyéticas/citología , Adulto , Sitios de Unión , Diferenciación Celular , Linaje de la Célula , Hibridación Genómica Comparativa , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Genoma Humano , Sistema Hematopoyético , Humanos , Modelos Biológicos , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA