Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38682338

RESUMEN

BACKGROUND: Most organs are maintained lifelong by resident stem/progenitor cells. During development and regeneration, lineage-specific stem/progenitor cells can contribute to the growth or maintenance of different organs, whereas fully differentiated mature cells have less regenerative potential. However, it is unclear whether vascular endothelial cells (ECs) are also replenished by stem/progenitor cells with EC-repopulating potential residing in blood vessels. It has been reported recently that some EC populations possess higher clonal proliferative potential and vessel-forming capacity compared with mature ECs. Nevertheless, a marker to identify vascular clonal repopulating ECs (CRECs) in murine and human individuals is lacking, and, hence, the mechanism for the proliferative, self-renewal, and vessel-forming potential of CRECs is elusive. METHODS: We analyzed colony-forming, self-renewal, and vessel-forming potential of ABCG2 (ATP binding cassette subfamily G member 2)-expressing ECs in human umbilical vessels. To study the contribution of Abcg2-expressing ECs to vessel development and regeneration, we developed Abcg2CreErt2;ROSA TdTomato mice and performed lineage tracing during mouse development and during tissue regeneration after myocardial infarction injury. RNA sequencing and chromatin methylation chromatin immunoprecipitation followed by sequencing were conducted to study the gene regulation in Abcg2-expressing ECs. RESULTS: In human and mouse vessels, ECs with higher ABCG2 expression (ABCECs) possess higher clonal proliferative potential and in vivo vessel-forming potential compared with mature ECs. These cells could clonally contribute to vessel formation in primary and secondary recipients after transplantation. These features of ABCECs meet the criteria of CRECs. Results from lineage tracing experiments confirm that Abcg2-expressing CRECs (AbcCRECs) contribute to arteries, veins, and capillaries in cardiac tissue development and vascular tissue regeneration after myocardial infarction. Transcriptome and epigenetic analyses reveal that a gene expression signature involved in angiogenesis and vessel development is enriched in AbcCRECs. In addition, various angiogenic genes, such as Notch2 and Hey2, are bivalently modified by trimethylation at the 4th and 27th lysine residue of histone H3 (H3K4me3 and H3K27me3) in AbcCRECs. CONCLUSIONS: These results are the first to establish that a single prospective marker identifies CRECs in mice and human individuals, which holds promise to provide new cell therapies for repair of damaged vessels in patients with endothelial dysfunction.

2.
Mol Ther ; 31(2): 454-470, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36114673

RESUMEN

Fetal cutaneous wound closure and repair differ from that in adulthood. In this work, we identify an oxidant stress sensor protein, nonselenocysteine-containing phospholipid hydroperoxide glutathione peroxidase (NPGPx), that is abundantly expressed in normal fetal epidermis (and required for fetal wound closure), though not in adult epidermis, but is variably re-induced upon adult tissue wounding. NPGPx is a direct target of the miR-29 family. Following injury, abundance of miR-29 is lowered, permitting a prompt increase in NPGPx transcripts and protein expression in adult wound-edge tissue. NPGPx expression was required to mediate increased keratinocyte migration induced by miR-29 inhibition in vitro and in vivo. Increased NPGPx expression induced increased SOX2 expression and ß-catenin nuclear localization in keratinocytes. Augmenting physiologic NPGPx expression via experimentally induced miR-29 suppression, using cutaneous tissue nanotransfection or targeted lipid nanoparticle delivery of anti-sense oligonucleotides, proved to be sufficient to overcome the deleterious effects of diabetes on this specific pathway to enhance tissue repair.


Asunto(s)
MicroARNs , Cicatrización de Heridas , Embarazo , Humanos , Femenino , Cicatrización de Heridas/genética , Piel/metabolismo , Queratinocitos/metabolismo , Movimiento Celular , MicroARNs/metabolismo
3.
J Infect Dis ; 219(7): 1076-1083, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30239747

RESUMEN

BACKGROUND: Human immunodeficiency virus (HIV) may be related to cardiovascular disease through monocyte activation-associated endothelial dysfunction. METHODS: Blood samples from 15 HIV-negative participants (the uninfected group), 8 HIV-positive participants who were not receiving antiretroviral therapy (ART) (the infected, untreated group), and 15 HIV-positive participants who were receiving ART (the infected, treated group) underwent flow cytometry of endothelial colony-forming cells (ECFCs) and monocyte proportions. IncuCyte live cell imaging of 8 capillary proliferative capacity parameters were obtained from cord blood ECFCs treated with participant plasma. RESULTS: The ECFC percentage determined by flow cytometry was not different between the study groups; however, values of the majority of capillary proliferative capacity parameters (ie, cell area, network length, network branch points, number of networks, and average tube width uniformity) were significantly lower in infected, untreated participants as compared to values for uninfected participants or infected, treated participants (P < .00625 for all comparisons). CD14+CD16+ intermediate monocytes and soluble CD163 were significantly and negatively correlated with several plasma-treated, cord blood ECFC proliferative capacity parameters in the combined HIV-positive groups but not in the uninfected group. CONCLUSIONS: Cord blood ECFC proliferative capacity was significantly impaired by plasma from infected, untreated patients, compared with plasma from uninfected participants and from infected, treated participants. Several ECFC functional parameters were adversely associated with monocyte activation in the HIV-positive groups, thereby suggesting a mechanism by which HIV-related inflammation may impair vascular reparative potential and consequently increase the risk of cardiovascular disease during HIV infection.


Asunto(s)
Endotelio/inmunología , Seronegatividad para VIH/inmunología , Seropositividad para VIH/inmunología , Monocitos , Células Madre , Adulto , Alquinos , Fármacos Anti-VIH/uso terapéutico , Benzoxazinas/uso terapéutico , Proliferación Celular , Quimiocina CCL5/sangre , Ciclopropanos , Endotelio/patología , Femenino , Sangre Fetal , Citometría de Flujo , Proteínas Ligadas a GPI/metabolismo , Seropositividad para VIH/sangre , Seropositividad para VIH/tratamiento farmacológico , Humanos , Receptores de Lipopolisacáridos/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Neovascularización Fisiológica , Plasma/inmunología , Estudios Prospectivos , Receptores de IgG/metabolismo , Células Madre/fisiología , Molécula 1 de Adhesión Celular Vascular/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre
5.
Stem Cells ; 35(4): 1053-1064, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28009085

RESUMEN

The molecular pathways regulating lymphoid priming, fate, and development of multipotent bone marrow hematopoietic stem and progenitor cells (HSPCs) that continuously feed thymic progenitors remain largely unknown. While Notch signal is indispensable for T cell specification and differentiation, the downstream effectors are not well understood. PRL2, a protein tyrosine phosphatase that regulates hematopoietic stem cell proliferation and self-renewal, is highly expressed in murine thymocyte progenitors. Here we demonstrate that protein tyrosine phosphatase PRL2 and receptor tyrosine kinase c-Kit are critical downstream targets and effectors of the canonical Notch/RBPJ pathway in early T cell progenitors. While PRL2 deficiency resulted in moderate defects of thymopoiesis in the steady state, de novo generation of T cells from Prl2 null hematopoietic stem cells was significantly reduced following transplantation. Prl2 null HSPCs also showed impaired T cell differentiation in vitro. We found that Notch/RBPJ signaling upregulated PRL2 as well as c-Kit expression in T cell progenitors. Further, PRL2 sustains Notch-mediated c-Kit expression and enhances stem cell factor/c-Kit signaling in T cell progenitors, promoting effective DN1-DN2 transition. Thus, we have identified a critical role for PRL2 phosphatase in mediating Notch and c-Kit signals in early T cell progenitors. Stem Cells 2017;35:1053-1064.


Asunto(s)
Proteínas Inmediatas-Precoces/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores Notch/metabolismo , Células Madre/citología , Células Madre/metabolismo , Linfocitos T/citología , Animales , Animales Recién Nacidos , Diferenciación Celular , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Transducción de Señal , Timo/metabolismo , Regulación hacia Arriba
6.
Stem Cells ; 35(5): 1303-1315, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28299842

RESUMEN

Electroacupuncture (EA) performed in rats and humans using limb acupuncture sites, LI-4 and LI-11, and GV-14 and GV-20 (humans) and Bai-hui (rats) increased functional connectivity between the anterior hypothalamus and the amygdala and mobilized mesenchymal stem cells (MSCs) into the systemic circulation. In human subjects, the source of the MSC was found to be primarily adipose tissue, whereas in rodents the tissue sources were considered more heterogeneous. Pharmacological disinhibition of rat hypothalamus enhanced sympathetic nervous system (SNS) activation and similarly resulted in a release of MSC into the circulation. EA-mediated SNS activation was further supported by browning of white adipose tissue in rats. EA treatment of rats undergoing partial rupture of the Achilles tendon resulted in reduced mechanical hyperalgesia, increased serum interleukin-10 levels and tendon remodeling, effects blocked in propranolol-treated rodents. To distinguish the afferent role of the peripheral nervous system, phosphoinositide-interacting regulator of transient receptor potential channels (Pirt)-GCaMP3 (genetically encoded calcium sensor) mice were treated with EA acupuncture points, ST-36 and LIV-3, and GV-14 and Bai-hui and resulted in a rapid activation of primary sensory neurons. EA activated sensory ganglia and SNS centers to mediate the release of MSC that can enhance tissue repair, increase anti-inflammatory cytokine production and provide pronounced analgesic relief. Stem Cells 2017;35:1303-1315.


Asunto(s)
Sistema Nervioso Central/citología , Electroacupuntura , Células Madre Mesenquimatosas/citología , Tendón Calcáneo/patología , Puntos de Acupuntura , Adipocitos/citología , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Antígenos CD/metabolismo , Miembro Anterior/fisiología , Miembro Posterior/fisiología , Humanos , Hiperalgesia/terapia , Hipotálamo/citología , Interleucina-10/sangre , Macrófagos/citología , Ratones , Red Nerviosa/fisiología , Ratas , Rotura , Células Receptoras Sensoriales/metabolismo , Proteína Desacopladora 1/metabolismo
7.
Pediatr Res ; 83(1-2): 283-290, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28915234

RESUMEN

Repairing and rebuilding damaged tissue in diseased human subjects remains a daunting challenge for clinical medicine. Proper vascular formation that serves to deliver blood-borne nutrients and adequate levels of oxygen and to remove wastes is critical for successful tissue regeneration. Endothelial colony-forming cells (ECFC) represent a promising cell source for revascularization of damaged tissue. ECFCs are identified by displaying a hierarchy of clonal proliferative potential and by pronounced postnatal vascularization ability in vivo. In this review, we provide a brief overview of human ECFC isolation and characterization, a survey of a number of animal models of human disease in which ECFCs have been shown to have prominent roles in tissue repair, and a summary of current challenges that must be overcome before moving ECFC into human subjects as a cell therapy.


Asunto(s)
Células Endoteliales/citología , Endotelio Vascular/metabolismo , Regeneración , Medicina Regenerativa/métodos , Animales , Encefalopatías/terapia , Proliferación Celular , Células Cultivadas , Ensayos Clínicos como Asunto , Sangre Fetal/citología , Humanos , Inflamación , Enfermedades Renales/terapia , Enfermedades Pulmonares/terapia , Modelos Animales , Neoplasias/terapia , Neovascularización Fisiológica , Enfermedades de la Retina/terapia , Células Madre/citología , Cordón Umbilical/citología
8.
Arterioscler Thromb Vasc Biol ; 37(11): 2014-2025, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29025705

RESUMEN

The emergence of induced pluripotent stem cell (iPSC) technology paves the way to generate large numbers of patient-specific endothelial cells (ECs) that can be potentially delivered for regenerative medicine in patients with cardiovascular disease. In the last decade, numerous protocols that differentiate EC from iPSC have been developed by many groups. In this review, we will discuss several common strategies that have been optimized for human iPSC-EC differentiation and subsequent studies that have evaluated the potential of human iPSC-EC as a cell therapy or as a tool in disease modeling. In addition, we will emphasize the importance of using in vivo vessel-forming ability and in vitro clonogenic colony-forming potential as a gold standard with which to evaluate the quality of human iPSC-EC derived from various protocols.


Asunto(s)
Diferenciación Celular , Células Endoteliales/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Animales , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/cirugía , Línea Celular , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/trasplante , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Fenotipo , Medicina Regenerativa
9.
Nature ; 549(7672): 340-342, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28854170
10.
J Am Soc Nephrol ; 28(8): 2420-2430, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28250053

RESUMEN

In the live animal, tissue autofluorescence arises from a number of biologically important metabolites, such as the reduced form of nicotinamide adenine dinucleotide. Because autofluorescence changes with metabolic state, it can be harnessed as a label-free imaging tool with which to study metabolism in vivo Here, we used the combination of intravital two-photon microscopy and frequency-domain fluorescence lifetime imaging microscopy (FLIM) to map cell-specific metabolic signatures in the kidneys of live animals. The FLIM images are analyzed using the phasor approach, which requires no prior knowledge of metabolite species and can provide unbiased metabolic fingerprints for each pixel of the lifetime image. Intravital FLIM revealed the metabolic signatures of S1 and S2 proximal tubules to be distinct and resolvable at the subcellular level. Notably, S1 and distal tubules exhibited similar metabolic profiles despite apparent differences in morphology and autofluorescence emission with traditional two-photon microscopy. Time-lapse imaging revealed dynamic changes in the metabolic profiles of the interstitium, urinary lumen, and glomerulus-areas that are not resolved by traditional intensity-based two-photon microscopy. Finally, using a model of endotoxemia, we present examples of the way in which intravital FLIM can be applied to study kidney diseases and metabolism. In conclusion, intravital FLIM of intrinsic metabolites is a bias-free approach with which to characterize and monitor metabolism in vivo, and offers the unique opportunity to uncover dynamic metabolic changes in living animals with subcellular resolution.


Asunto(s)
Microscopía Intravital , Riñón/citología , Riñón/metabolismo , Microscopía de Fluorescencia por Excitación Multifotónica , Animales , Riñón/diagnóstico por imagen , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Dev Dyn ; 246(12): 1001-1014, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28975680

RESUMEN

BACKGROUND: Accumulating evidence suggests the origin of juvenile myelomonocytic leukemia (JMML) is closely associated with fetal development. Nevertheless, the contribution of embryonic progenitors to JMML pathogenesis remains unexplored. We hypothesized that expression of JMML-initiating PTPN11 mutations in HSC-independent yolk sac erythromyeloid progenitors (YS EMPs) would result in a mouse model of pediatric myeloproliferative neoplasm (MPN). RESULTS: E9.5 YS EMPs from VavCre+;PTPN11D61Y embryos demonstrated growth hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF) and hyperactive RAS-ERK signaling. Mutant EMPs engrafted the spleens of neonatal recipients, but did not cause disease. To assess MPN development during unperturbed hematopoiesis we generated CSF1R-MCM+;PTPN11E76K ;ROSAYFP mice in which oncogene expression was restricted to EMPs. Yellow fluorescent protein-positive progeny of mutant EMPs persisted in tissues one year after birth and demonstrated hyperactive RAS-ERK signaling. Nevertheless, these mice had normal survival and did not demonstrate features of MPN. CONCLUSIONS: YS EMPs expressing mutant PTPN11 demonstrate functional and molecular features of JMML but do not cause disease following transplantation nor following unperturbed development. Developmental Dynamics 246:1001-1014, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Células Precursoras Eritroides/enzimología , Mutación con Ganancia de Función , Leucemia Mielomonocítica Juvenil/enzimología , Sistema de Señalización de MAP Quinasas , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Saco Vitelino/metabolismo , Animales , Células Precursoras Eritroides/patología , Células Precursoras Eritroides/trasplante , Leucemia Mielomonocítica Juvenil/embriología , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Saco Vitelino/patología
12.
Am J Physiol Renal Physiol ; 312(5): F897-F907, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28228404

RESUMEN

Damage to endothelial cells contributes to acute kidney injury (AKI) by leading to impaired perfusion. Endothelial colony-forming cells (ECFC) are endothelial precursor cells with high proliferative capacity, pro-angiogenic activity, and in vivo vessel forming potential. We hypothesized that ECFC may ameliorate the degree of AKI and/or promote repair of the renal vasculature following ischemia-reperfusion (I/R). Rat pulmonary microvascular endothelial cells (PMVEC) with high proliferative potential were compared with pulmonary artery endothelial cells (PAEC) with low proliferative potential in rats subjected to renal I/R. PMVEC administration reduced renal injury and hastened recovery as indicated by serum creatinine and tubular injury scores, while PAEC did not. Vehicle-treated control animals showed consistent reductions in renal medullary blood flow (MBF) within 2 h of reperfusion, while PMVEC protected against loss in MBF as measured by laser Doppler. Interestingly, PMVEC mediated protection occurred in the absence of homing to the kidney. Conditioned medium (CM) from human cultured cord blood ECFC also conveyed beneficial effects against I/R injury and loss of MBF. Moreover, ECFC-CM significantly reduced the expression of ICAM-1 and decreased the number of differentiated lymphocytes typically recruited into the kidney following renal ischemia. Taken together, these data suggest that ECFC secrete factors that preserve renal function post ischemia, in part, by preserving microvascular function.


Asunto(s)
Lesión Renal Aguda/cirugía , Proliferación Celular , Células Progenitoras Endoteliales/trasplante , Endotelio Vascular/trasplante , Riñón/irrigación sanguínea , Neovascularización Fisiológica , Daño por Reperfusión/cirugía , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Velocidad del Flujo Sanguíneo , Comunicación Celular , Células Cultivadas , Quimiotaxis de Leucocito , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Sangre Fetal/citología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Riñón/metabolismo , Riñón/patología , Masculino , Fenotipo , Ratas Sprague-Dawley , Circulación Renal , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Transducción de Señal , Factores de Tiempo
14.
Development ; 140(9): 1946-57, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23571217

RESUMEN

Trabeculation and compaction of the embryonic myocardium are morphogenetic events crucial for the formation and function of the ventricular walls. Fkbp1a (FKBP12) is a ubiquitously expressed cis-trans peptidyl-prolyl isomerase. Fkbp1a-deficient mice develop ventricular hypertrabeculation and noncompaction. To determine the physiological function of Fkbp1a in regulating the intercellular and intracellular signaling pathways involved in ventricular trabeculation and compaction, we generated a series of Fkbp1a conditional knockouts. Surprisingly, cardiomyocyte-restricted ablation of Fkbp1a did not give rise to the ventricular developmental defect, whereas endothelial cell-restricted ablation of Fkbp1a recapitulated the ventricular hypertrabeculation and noncompaction observed in Fkbp1a systemically deficient mice, suggesting an important contribution of Fkbp1a within the developing endocardia in regulating the morphogenesis of ventricular trabeculation and compaction. Further analysis demonstrated that Fkbp1a is a novel negative modulator of activated Notch1. Activated Notch1 (N1ICD) was significantly upregulated in Fkbp1a-ablated endothelial cells in vivo and in vitro. Overexpression of Fkbp1a significantly reduced the stability of N1ICD and direct inhibition of Notch signaling significantly reduced hypertrabeculation in Fkbp1a-deficient mice. Our findings suggest that Fkbp1a-mediated regulation of Notch1 plays an important role in intercellular communication between endocardium and myocardium, which is crucial in controlling the formation of the ventricular walls.


Asunto(s)
Endocardio/metabolismo , Ventrículos Cardíacos/patología , Miocardio/metabolismo , Receptor Notch1/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Linaje de la Célula , Células Cultivadas , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Desarrollo Embrionario , Endocardio/embriología , Endocardio/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados/embriología , Ratones Noqueados/metabolismo , Miocardio/patología , Cresta Neural/metabolismo , Cresta Neural/patología , Fenotipo , Receptor Notch1/genética , Transducción de Señal , Proteínas de Unión a Tacrolimus/genética , Transfección
15.
Stem Cells ; 33(3): 925-38, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25385494

RESUMEN

While Polycomb group protein Bmi1 is important for stem cell maintenance, its role in lineage commitment is largely unknown. We have identified Bmi1 as a novel regulator of erythroid development. Bmi1 is highly expressed in mouse erythroid progenitor cells and its deficiency impairs erythroid differentiation. BMI1 is also important for human erythroid development. Furthermore, we discovered that loss of Bmi1 in erythroid progenitor cells results in decreased transcription of multiple ribosomal protein genes and impaired ribosome biogenesis. Bmi1 deficiency stabilizes p53 protein, leading to upregulation of p21 expression and subsequent G0/G1 cell cycle arrest. Genetic inhibition of p53 activity rescues the erythroid defects seen in the Bmi1 null mice, demonstrating that a p53-dependent mechanism underlies the pathophysiology of the anemia. Mechanistically, Bmi1 is associated with multiple ribosomal protein genes and may positively regulate their expression in erythroid progenitor cells. Thus, Bmi1 promotes erythroid development, at least in part through regulating ribosome biogenesis. Ribosomopathies are human disorders of ribosome dysfunction, including Diamond-Blackfan anemia (DBA) and 5q- syndrome, in which genetic abnormalities cause impaired ribosome biogenesis, resulting in specific clinical phenotypes. We observed that BMI1 expression in human hematopoietic stem and progenitor cells from patients with DBA is correlated with the expression of some ribosomal protein genes, suggesting that BMI1 deficiency may play a pathological role in DBA and other ribosomopathies.


Asunto(s)
Células Eritroides/citología , Células Eritroides/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ribosomas/metabolismo , Animales , Diferenciación Celular/fisiología , Eritropoyesis/fisiología , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Ribosómicas/biosíntesis , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/genética
16.
Curr Opin Hematol ; 22(3): 252-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25767955

RESUMEN

PURPOSE OF REVIEW: Methods to isolate endothelial cells from murine and human pluripotent stem cells continue to evolve and increasingly diverse endothelial cell populations have been generated. This review provides an update of key articles published within the past year that report on some of those advances. RECENT FINDINGS: Cooperative interactions among microRNA (miRNA), transcription factors and some downstream interacting proteins have been reported to enhance endothelial specification from embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Endothelial cell differentiation can also be modulated by various growth factor additions, Notch pathway activation or inhibition, and modulation of the microenvironment of the differentiating ESC and iPSC. Functionality of the derived endothelium has been demonstrated by a variety of in-vitro and in-vivo assays. Finally, two recent reports have identified endothelial progenitor populations with robust proliferative potential. SUMMARY: Progress in differentiating endothelial cells from ESC and iPSC has been made. The recent report of formation of endothelial colony forming cells from human ESC and iPSC provides a protocol that can generate clinically relevant numbers of cells for human cell therapy.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Endoteliales/metabolismo , Células Madre Pluripotentes/citología , Diferenciación Celular , Humanos
17.
Am J Pathol ; 184(2): 397-408, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24287405

RESUMEN

Lysosomal acid lipase (LAL) is essential for the hydrolysis of cholesteryl esters and triglycerides to generate cholesterol and free fatty acids in cellular lysosomes. Ablation of the lal gene (lal(-/-)) systemically increased expansion of cluster of differentiation molecule 11b (CD11b), lymphocyte antigen 6G (Ly6G) myeloid-derived suppressor cells (MDSCs) that caused myeloproliferative neoplasms in mice. Study of lal(-/-) bone marrow Ly6G(+) MDSCs via transcriptional profiling showed increases in mammalian target of rapamycin (mTOR) signaling pathway transcripts. Injection of mTOR pharmacologic inhibitors into lal(-/-) mice significantly reduced bone marrow myelopoiesis and systemic CD11b(+)Ly6G(+) cell expansion. Rapamycin treatment of lal(-/-) mice stimulated a shift from immature CD11b(+)Ly6G(+) cells to CD11b(+) single-positive cells in marrow and tissues and partially reversed the increased cell proliferation, decreased apoptosis, increased ATP synthesis, and increased cell cycling of bone marrow CD11b(+)Ly6G(+) cells obtained from lal(-/-) mice. Pharmacologic and siRNA suppression of mTOR, regulatory-associated protein of mTOR, rapamycin-insensitive companion of mTOR, and Akt1 function corrected CD11b(+)Ly6G(+) cell in lal(-/-) mice development from Lin(-) progenitor cells and reversed the immune suppression on T-cell proliferation and function in association with decreased reactive oxygen species production, and recovery from impairment of mitochondrial membrane potential compared with control mutant cells. These results indicate a crucial role of LAL-regulated mTOR signaling in the production and function of CD11b(+)Ly6G(+) cells. The mTOR pathway may serve as a novel target to modulate the emergence of MDSCs in those pathophysiologic states in which these cells play an immunosuppressive role.


Asunto(s)
Células Mieloides/citología , Transducción de Señal , Esterol Esterasa/deficiencia , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos Ly/metabolismo , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Antígeno CD11b/metabolismo , Proteínas Portadoras/metabolismo , Proliferación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Terapia de Inmunosupresión , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Especies Reactivas de Oxígeno/metabolismo , Proteína Reguladora Asociada a mTOR , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Esterol Esterasa/metabolismo
18.
Blood ; 121(21): 4252-4, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23704046

RESUMEN

In this issue of Blood, Sperandio and colleagues report the use of a unique intravital microscopic system to characterize an ontogenic process of blood cell and yolk sac endothelial maturation that is required to display full adult-type inflammation-induced leukocyte recruitment.(1) They report that murine fetal blood neutrophil rolling, adhesion, and extravasation from inflamed yolk sac vessels is apparent late in development, but that before embryonic day (E) 15, fetal blood neutrophils display little ability to roll or adhere to inflamed vascular endothelial cells. Similar behavior was displayed when fetal blood cells were tested in vitro on immobilized recombinant adhesion molecules.


Asunto(s)
Movimiento Celular/inmunología , Sistema Inmunológico/embriología , Leucocitos/citología , Microvasos/embriología , Saco Vitelino/embriología , Animales , Femenino , Embarazo
19.
Blood ; 121(24): 4815-7, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23766458

RESUMEN

In this issue of Blood, Gandre-Babbe et al have, in part, overcome the obstacle of validating the molecular underpinnings of juvenile myelomonocytic leukemia (JMML) with the generation of induced pluripotent stem cells (iPSCs) from individuals with JMML.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Leucemia Mielomonocítica Juvenil/metabolismo , Mutación Missense , Células Madre Neoplásicas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Femenino , Humanos , Masculino
20.
Microvasc Res ; 101: 72-81, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26122935

RESUMEN

Human cord blood (CB) is enriched in circulating endothelial colony forming cells (ECFCs) that display high proliferative potential and in vivo vessel forming ability. Since diminished ECFC survival is known to dampen the vasculogenic response in vivo, we tested how long implanted ECFC survive and generate vessels in three-dimensional (3D) type I collagen matrices in vitro and in vivo. We hypothesized that human platelet lysate (HPL) would promote cell survival and enhance vasculogenesis in the 3D collagen matrices. We report that the percentage of ECFC co-cultured with HPL that were alive was significantly enhanced on days 1 and 3 post-matrix formation, compared to ECFC alone containing matrices. Also, co-culture of ECFC with HPL displayed significantly more vasculogenic activity compared to ECFC alone and expressed significantly more pro-survival molecules (pAkt, p-Bad and Bcl-xL) in the 3D collagen matrices in vitro. Treatment with Akt1 inhibitor (A-674563), Akt2 inhibitor (CCT128930) and Bcl-xL inhibitor (ABT-263/Navitoclax) significantly decreased the cell survival and vasculogenesis of ECFC co-cultured with or without HPL and implicated activation of the Akt1 pathway as the critical mediator of the HPL effect on ECFC in vitro. A significantly greater average vessel number and total vascular area of human CD31(+) vessels were present in implants containing ECFC and HPL, compared to the ECFC alone implants in vivo. We conclude that implantation of ECFC with HPL in vivo promotes vasculogenesis and augments blood vessel formation via diminishing apoptosis of the implanted ECFC.


Asunto(s)
Plaquetas/citología , Plaquetas/metabolismo , Colágeno/química , Trasplante de Células Madre de Sangre del Cordón Umbilical , Sangre Fetal/citología , Neovascularización Fisiológica/fisiología , Animales , Apoptosis , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA