Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(9): 1119-1133, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32719519

RESUMEN

The full neutrophil heterogeneity and differentiation landscape remains incompletely characterized. Here, we profiled >25,000 differentiating and mature mouse neutrophils using single-cell RNA sequencing to provide a comprehensive transcriptional landscape of neutrophil maturation, function and fate decision in their steady state and during bacterial infection. Eight neutrophil populations were defined by distinct molecular signatures. The three mature peripheral blood neutrophil subsets arise from distinct maturing bone marrow neutrophil subsets. Driven by both known and uncharacterized transcription factors, neutrophils gradually acquire microbicidal capability as they traverse the transcriptional landscape, representing an evolved mechanism for fine-tuned regulation of an effective but balanced neutrophil response. Bacterial infection reprograms the genetic architecture of neutrophil populations, alters dynamic transitions between subpopulations and primes neutrophils for augmented functionality without affecting overall heterogeneity. In summary, these data establish a reference model and general framework for studying neutrophil-related disease mechanisms, biomarkers and therapeutic targets at single-cell resolution.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Escherichia coli/fisiología , Neutrófilos/fisiología , Peritonitis/inmunología , Análisis de la Célula Individual/métodos , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Homeostasis , Humanos , Ratones , Análisis de Secuencia de ARN
2.
Cell ; 153(5): 1025-35, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23706740

RESUMEN

Unique among leukocytes, neutrophils follow daily cycles of release from and migration back into the bone marrow, where they are eliminated. Because removal of dying cells generates homeostatic signals, we explored whether neutrophil elimination triggers circadian events in the steady state. Here, we report that the homeostatic clearance of neutrophils provides cues that modulate the physiology of the bone marrow. We identify a population of CD62L(LO) CXCR4(HI) neutrophils that have "aged" in the circulation and are eliminated at the end of the resting period in mice. Aged neutrophils infiltrate the bone marrow and promote reductions in the size and function of the hematopoietic niche. Modulation of the niche depends on macrophages and activation of cholesterol-sensing nuclear receptors and is essential for the rhythmic egress of hematopoietic progenitors into the circulation. Our results unveil a process that synchronizes immune and hematopoietic rhythms and expand the ascribed functions of neutrophils beyond inflammation. PAPERFLICK:


Asunto(s)
Médula Ósea/fisiología , Ritmo Circadiano , Neutrófilos/citología , Neutrófilos/fisiología , Animales , Movimiento Celular , Senescencia Celular , Femenino , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Receptores Nucleares Huérfanos/metabolismo
3.
Blood ; 143(11): 967-970, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38289232

RESUMEN

ABSTRACT: The root cause of sickle cell anemia has been known for 7 decades, yet no curative therapies have been available other than allogeneic bone marrow transplantation, for which applicability is limited. Two potentially curative therapies based on gene therapy and gene editing strategies have recently received US Food and Drug Administration approval. This review surveys the nature of these therapies and the opportunities and issues raised by the prospect of definitive genetically based therapies being available in clinical practice.


Asunto(s)
Anemia de Células Falciformes , Trasplante de Células Madre Hematopoyéticas , Humanos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Trasplante de Médula Ósea , Edición Génica , Terapia Genética/métodos
4.
Immunity ; 42(1): 159-71, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25579427

RESUMEN

The cellular mechanisms controlling infection-induced emergency granulopoiesis are poorly defined. Here we found that reactive oxygen species (ROS) concentrations in the bone marrow (BM) were elevated during acute infection in a phagocytic NADPH oxidase-dependent manner in myeloid cells. Gr1(+) myeloid cells were uniformly distributed in the BM, and all c-kit(+) progenitor cells were adjacent to Gr1(+) myeloid cells. Inflammation-induced ROS production in the BM played a critical role in myeloid progenitor expansion during emergency granulopoiesis. ROS elicited oxidation and deactivation of phosphatase and tensin homolog (PTEN), resulting in upregulation of PtdIns(3,4,5)P3 signaling in BM myeloid progenitors. We further revealed that BM myeloid cell-produced ROS stimulated proliferation of myeloid progenitors via a paracrine mechanism. Taken together, our results establish that phagocytic NADPH oxidase-mediated ROS production by BM myeloid cells plays a critical role in mediating emergency granulopoiesis during acute infection.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Granulocitos/fisiología , Hematopoyesis , Células Mieloides/fisiología , Células Progenitoras Mieloides/fisiología , Enfermedad Aguda , Animales , Médula Ósea/microbiología , Médula Ósea/patología , Proliferación Celular , Células Cultivadas , Hematopoyesis/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Comunicación Paracrina , Fosfatos de Fosfatidilinositol/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
5.
Nat Rev Mol Cell Biol ; 12(2): 126-31, 2011 02.
Artículo en Inglés | MEDLINE | ID: mdl-21253000

RESUMEN

Mesenchymal stem cells (MSCs) are a diverse subset of multipotent precursors present in the stromal fraction of many adult tissues and have drawn intense interest from translational and basic investigators. MSCs have been operationally defined by their ability to differentiate into osteoblasts, adipocytes and chondrocytes after in vitro expansion. Nevertheless, their identity in vivo, heterogeneity, anatomical localization and functional roles in adult tissue homeostasis have remained enigmatic and are only just starting to be uncovered.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Adultas/citología , Animales , Diferenciación Celular , Separación Celular , Células Madre Embrionarias/citología , Humanos , Células Madre Multipotentes/citología , Células del Estroma/citología
6.
Blood ; 135(23): 2071-2084, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31990287

RESUMEN

Sickle cell disease (SCD) is a monogenic red blood cell (RBC) disorder with high morbidity and mortality. Here, we report, for the first time, the impact of SCD on the bone marrow (BM) vascular niche, which is critical for hematopoiesis. In SCD mice, we find a disorganized and structurally abnormal BM vascular network of increased numbers of highly tortuous arterioles occupying the majority of the BM cavity, as well as fragmented sinusoidal vessels filled with aggregates of erythroid and myeloid cells. By in vivo imaging, sickle and control RBCs have significantly slow intravascular flow speeds in sickle cell BM but not in control BM. In sickle cell BM, we find increased reactive oxygen species production in expanded erythroblast populations and elevated levels of HIF-1α. The SCD BM exudate exhibits increased levels of proangiogenic growth factors and soluble vascular cell adhesion molecule-1. Transplantation of SCD mouse BM cells into wild-type mice recapitulates the SCD vascular phenotype. Our data provide a model of SCD BM, in which slow RBC flow and vaso-occlusions further diminish local oxygen availability in the physiologic hypoxic BM cavity. These events trigger a milieu that is conducive to aberrant vessel growth. The distorted neovascular network is completely reversed by a 6-week blood transfusion regimen targeting hemoglobin S to <30%, highlighting the plasticity of the vascular niche. A better insight into the BM microenvironments in SCD might provide opportunities to optimize approaches toward efficient and long-term hematopoietic engraftment in the context of curative therapies.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Transfusión Sanguínea/métodos , Médula Ósea/patología , Eritrocitos Anormales/patología , Hematopoyesis , Neovascularización Patológica/prevención & control , Esplenomegalia/prevención & control , Animales , Médula Ósea/metabolismo , Eritrocitos Anormales/metabolismo , Femenino , Humanos , Masculino , Ratones , Neovascularización Patológica/etiología , Neovascularización Patológica/patología , Esplenomegalia/etiología , Esplenomegalia/patología
7.
Mol Cell ; 49(5): 934-46, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23395001

RESUMEN

To provide a lifelong supply of blood cells, hematopoietic stem cells (HSCs) need to carefully balance both self-renewing cell divisions and quiescence. Although several regulators that control this mechanism have been identified, we demonstrate that the transcription factor PU.1 acts upstream of these regulators. So far, attempts to uncover PU.1's role in HSC biology have failed because of the technical limitations of complete loss-of-function models. With the use of hypomorphic mice with decreased PU.1 levels specifically in phenotypic HSCs, we found reduced HSC long-term repopulation potential that could be rescued completely by restoring PU.1 levels. PU.1 prevented excessive HSC division and exhaustion by controlling the transcription of multiple cell-cycle regulators. Levels of PU.1 were sustained through autoregulatory PU.1 binding to an upstream enhancer that formed an active looped chromosome architecture in HSCs. These results establish that PU.1 mediates chromosome looping and functions as a master regulator of HSC proliferation.


Asunto(s)
Células Madre Adultas/metabolismo , Ciclo Celular/genética , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Células Madre Adultas/patología , Animales , Proliferación Celular , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Endogámicos , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo
8.
J Immunol ; 198(7): 2854-2864, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28235862

RESUMEN

Both microbial infection and sterile inflammation augment bone marrow (BM) neutrophil production, but whether the induced accelerated granulopoiesis is mediated by a common pathway and the nature of such a pathway are poorly defined. We recently established that BM myeloid cell-derived reactive oxygen species (ROS) externally regulate myeloid progenitor proliferation and differentiation in bacteria-elicited emergency granulopoiesis. In this article, we show that BM ROS levels are also elevated during sterile inflammation. Similar to in microbial infection, ROS were mainly generated by the phagocytic NADPH oxidase in Gr1+ myeloid cells. The myeloid cells and their ROS were uniformly distributed in the BM when visualized by multiphoton intravital microscopy, and ROS production was both required and sufficient for sterile inflammation-elicited reactive granulopoiesis. Elevated granulopoiesis was mediated by ROS-induced phosphatase and tensin homolog oxidation and deactivation, leading to upregulated PtdIns(3,4,5)P3 signaling and increased progenitor cell proliferation. Collectively, these results demonstrate that, although infection-induced emergency granulopoiesis and sterile inflammation-elicited reactive granulopoiesis are triggered by different stimuli and are mediated by distinct upstream signals, the pathways converge to NADPH oxidase-dependent ROS production by BM myeloid cells. Thus, BM Gr1+ myeloid cells represent a key hematopoietic niche that supports accelerated granulopoiesis in infective and sterile inflammation. This niche may be an excellent target in various immune-mediated pathologies or immune reconstitution after BM transplantation.


Asunto(s)
Células Precursoras de Granulocitos/metabolismo , Granulocitos/metabolismo , Hematopoyesis/inmunología , Inflamación/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Western Blotting , Diferenciación Celular/inmunología , Separación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Granulocitos/citología , Hematopoyesis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Confocal , Células Mieloides/citología , Células Mieloides/metabolismo , Nicho de Células Madre/fisiología
9.
Blood ; 127(14): 1740-1, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27056995

RESUMEN

In this issue of Blood, Arthur et al uncover that HLA alloantibodies cannot solely account for the immune mechanism in platelet refractoriness.


Asunto(s)
Plaquetas/inmunología , Linfocitos T CD8-positivos/inmunología , Transfusión de Plaquetas , Animales , Humanos
10.
N Engl J Med ; 371(15): 1407-17, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25295500

RESUMEN

BACKGROUND: In previous clinical trials involving children with X-linked severe combined immunodeficiency (SCID-X1), a Moloney murine leukemia virus-based γ-retrovirus vector expressing interleukin-2 receptor γ-chain (γc) complementary DNA successfully restored immunity in most patients but resulted in vector-induced leukemia through enhancer-mediated mutagenesis in 25% of patients. We assessed the efficacy and safety of a self-inactivating retrovirus for the treatment of SCID-X1. METHODS: We enrolled nine boys with SCID-X1 in parallel trials in Europe and the United States to evaluate treatment with a self-inactivating (SIN) γ-retrovirus vector containing deletions in viral enhancer sequences expressing γc (SIN-γc). RESULTS: All patients received bone marrow-derived CD34+ cells transduced with the SIN-γc vector, without preparative conditioning. After 12.1 to 38.7 months of follow-up, eight of the nine children were still alive. One patient died from an overwhelming adenoviral infection before reconstitution with genetically modified T cells. Of the remaining eight patients, seven had recovery of peripheral-blood T cells that were functional and led to resolution of infections. The patients remained healthy thereafter. The kinetics of CD3+ T-cell recovery was not significantly different from that observed in previous trials. Assessment of insertion sites in peripheral blood from patients in the current trial as compared with those in previous trials revealed significantly less clustering of insertion sites within LMO2, MECOM, and other lymphoid proto-oncogenes in our patients. CONCLUSIONS: This modified γ-retrovirus vector was found to retain efficacy in the treatment of SCID-X1. The long-term effect of this therapy on leukemogenesis remains unknown. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01410019, NCT01175239, and NCT01129544.).


Asunto(s)
Gammaretrovirus/genética , Terapia Genética , Vectores Genéticos , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Antígenos CD34 , ADN Complementario/uso terapéutico , Expresión Génica , Silenciador del Gen , Terapia Genética/efectos adversos , Humanos , Lactante , Subunidad gamma Común de Receptores de Interleucina/genética , Masculino , Ratones , Mutación , Linfocitos T/inmunología , Transducción Genética , Transgenes/fisiología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología
11.
Blood ; 125(20): 3105-13, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25824687

RESUMEN

Hematopoietic stem cells (HSCs) are localized within specialized microenvironments throughout the BM. Nestin-expressing (Nestin(+)) mesenchymal stromal cells (MSCs) are important in the perivascular space. Rac is critical for MSC cell shape in vitro, whereas its function in MSCs in vivo remains poorly characterized. We hypothesized that deletion of Rac in the Nestin(+) cells would perturb the perivascular space, altering HSC localization and hematopoiesis. Nestin-Cre-directed excision of Rac1 in Rac3(-/-) mice reduces Nestin(+) cells in the marrow. We observed a 2.7-fold decrease in homing of labeled wild-type hematopoietic cells into Rac1(Δ/Δ)Rac3(-/-) mice compared with control mice. Rac1(Δ/Δ)Rac3(-/-) mice demonstrated a marked decrease in arterioles and an increase in the number and volume of venous sinusoids in the marrow that was associated with a reduction in the numbers of immunophenotypically and functionally-defined long-term HSCs in the marrow, a decrease in colony-forming cells and a reduction in circulating progenitors. Rac-deleted animals demonstrated a significant increase in trabecular bone. These data demonstrate that Rac GTPases play an important role in the integrity of perivascular space. Increased trabecular bone and sinusoidal space and decreased arteriolar volume in this model were associated with decreased HSC, underscoring the complexity of regulation of hematopoiesis in the perivascular space.


Asunto(s)
Médula Ósea/metabolismo , Médula Ósea/patología , Hematopoyesis/genética , Proteínas de Unión al GTP rac/genética , Animales , Apoptosis/genética , Vasos Sanguíneos , Huesos/metabolismo , Huesos/patología , Microambiente Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Ratones , Ratones Noqueados , Nestina/genética , Nestina/metabolismo , Osteoblastos/metabolismo , Factor de Células Madre/metabolismo , Proteínas de Unión al GTP rac/deficiencia
12.
Transfusion ; 56(3): 743-54, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26634332

RESUMEN

BACKGROUND: There are 346 serologically defined red blood cell (RBC) antigens and 33 serologically defined platelet (PLT) antigens, most of which have known genetic changes in 45 RBC or six PLT genes that correlate with antigen expression. Polymorphic sites associated with antigen expression in the primary literature and reference databases are annotated according to nucleotide positions in cDNA. This makes antigen prediction from next-generation sequencing data challenging, since it uses genomic coordinates. STUDY DESIGN AND METHODS: The conventional cDNA reference sequences for all known RBC and PLT genes that correlate with antigen expression were aligned to the human reference genome. The alignments allowed conversion of conventional cDNA nucleotide positions to the corresponding genomic coordinates. RBC and PLT antigen prediction was then performed using the human reference genome and whole genome sequencing (WGS) data with serologic confirmation. RESULTS: Some major differences and alignment issues were found when attempting to convert the conventional cDNA to human reference genome sequences for the following genes: ABO, A4GALT, RHD, RHCE, FUT3, ACKR1 (previously DARC), ACHE, FUT2, CR1, GCNT2, and RHAG. However, it was possible to create usable alignments, which facilitated the prediction of all RBC and PLT antigens with a known molecular basis from WGS data. Traditional serologic typing for 18 RBC antigens were in agreement with the WGS-based antigen predictions, providing proof of principle for this approach. CONCLUSION: Detailed mapping of conventional cDNA annotated RBC and PLT alleles can enable accurate prediction of RBC and PLT antigens from whole genomic sequencing data.


Asunto(s)
Antígenos de Plaqueta Humana/genética , Antígenos de Grupos Sanguíneos/genética , Genómica , Eritrocitos/inmunología , Humanos
13.
Proc Natl Acad Sci U S A ; 110(4): 1446-51, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23302686

RESUMEN

The human heart is believed to grow by enlargement but not proliferation of cardiomyocytes (heart muscle cells) during postnatal development. However, recent studies have shown that cardiomyocyte proliferation is a mechanism of cardiac growth and regeneration in animals. Combined with evidence for cardiomyocyte turnover in adult humans, this suggests that cardiomyocyte proliferation may play an unrecognized role during the period of developmental heart growth between birth and adolescence. We tested this hypothesis by examining the cellular growth mechanisms of the left ventricle on a set of healthy hearts from humans aged 0-59 y (n = 36). The percentages of cardiomyocytes in mitosis and cytokinesis were highest in infants, decreasing to low levels by 20 y. Although cardiomyocyte mitosis was detectable throughout life, cardiomyocyte cytokinesis was not evident after 20 y. Between the first year and 20 y of life, the number of cardiomyocytes in the left ventricle increased 3.4-fold, which was consistent with our predictions based on measured cardiomyocyte cell cycle activity. Our findings show that cardiomyocyte proliferation contributes to developmental heart growth in young humans. This suggests that children and adolescents may be able to regenerate myocardium, that abnormal cardiomyocyte proliferation may be involved in myocardial diseases that affect this population, and that these diseases might be treatable through stimulation of cardiomyocyte proliferation.


Asunto(s)
Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Adolescente , Adulto , Ciclo Celular , Aumento de la Célula , Proliferación Celular , Niño , Preescolar , Femenino , Fibrosis , Corazón/fisiología , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/patología , Ploidias , Regeneración , Adulto Joven
14.
Blood ; 122(8): 1334-5, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23970354

RESUMEN

In this issue of Blood, Stowell et al describe a novel mouse model of hemolytic disease of the fetus and newborn (HDFN) that recapitulates many of the key features of human disease.1 Recently, this same group of researchers described a transgenic mouse that expresses the human KEL2 (Chellano) red cell surface protein from the Kell system on red cells, and subsequently demonstrated that Kell differences on transfused blood induce antibody responses and hemolytic transfusion reactions similar to those seen in patients. In this latest report, Stowell et al demonstrate that similar to some patients, Kell differences between mother and father can lead to maternal antibody generation and hemolytic disease in utero. In so doing, they provide experimental confirmation of a long sought after animal model of HDFN.


Asunto(s)
Anemia Hemolítica/inmunología , Eritrocitos/citología , Isoanticuerpos/inmunología , Sistema del Grupo Sanguíneo de Kell/inmunología , Modelos Animales , Animales , Femenino , Masculino , Embarazo
15.
Blood ; 121(8): 1255-64, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23212524

RESUMEN

Lineage-restricted cells can be reprogrammed to a pluripotent state known as induced pluripotent stem (iPS) cells through overexpression of 4 transcription factors. iPS cells are similar to human embryonic stem (hES) cells and have the same ability to generate all the cells of the human body, including blood cells. However, this process is extremely inefficient and to date has been unsuccessful at differentiating iPS into hematopoietic stem cells (HSCs). We hypothesized that iPS cells, injected into NOD.Cg-Prkdc(scid) Il2rg(tm1Wjl)/SzJ immunocompromised (NSG) mice could give rise to hematopoietic stem/progenitor cells (HSPCs) during teratoma formation. Here, we report a novel in vivo system in which human iPS cells differentiate within teratomas to derive functional myeloid and lymphoid cells. Similarly, HSPCs can be isolated from teratoma parenchyma and reconstitute a human immune system when transplanted into immunodeficient mice. Our data provide evidence that in vivo generation of patient customized cells is feasible, providing materials that could be useful for transplantation, human antibody generation, and drug screening applications.


Asunto(s)
Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes Inducidas/citología , Teratoma/patología , Animales , Linfocitos B/citología , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Queratinocitos/fisiología , Linfocitos/citología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Células Mieloides/citología , Trasplante de Neoplasias , Células del Estroma/citología , Células del Estroma/fisiología , Células del Estroma/trasplante , Linfocitos T/citología , Teratoma/genética , Trasplante Heterólogo , Células Tumorales Cultivadas
16.
Blood ; 122(17): 3074-81, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-23996087

RESUMEN

Umbilical cord blood (UCB) is a valuable source of hematopoietic stem cells (HSCs) for use in allogeneic transplantation. Key advantages of UCB are rapid availability and less stringent requirements for HLA matching. However, UCB contains an inherently limited HSC count, which is associated with delayed time to engraftment, high graft failure rates, and early mortality. 16,16-Dimethyl prostaglandin E2 (dmPGE2) was previously identified to be a critical regulator of HSC homeostasis, and we hypothesized that brief ex vivo modulation with dmPGE2 could improve patient outcomes by increasing the "effective dose" of HSCs. Molecular profiling approaches were used to determine the optimal ex vivo modulation conditions (temperature, time, concentration, and media) for use in the clinical setting. A phase 1 trial was performed to evaluate the safety and therapeutic potential of ex vivo modulation of a single UCB unit using dmPGE2 before reduced-intensity, double UCB transplantation. Results from this study demonstrated clear safety with durable, multilineage engraftment of dmPGE2-treated UCB units. We observed encouraging trends in efficacy, with accelerated neutrophil recovery (17.5 vs 21 days, P = .045), coupled with preferential, long-term engraftment of the dmPGE2-treated UCB unit in 10 of 12 treated participants.


Asunto(s)
16,16-Dimetilprostaglandina E2/farmacología , Plaquetas/efectos de los fármacos , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Sangre Fetal/efectos de los fármacos , Supervivencia de Injerto/inmunología , Neoplasias Hematológicas/terapia , Adulto , Anciano , Plaquetas/citología , Plaquetas/inmunología , Células Cultivadas , Criopreservación , Femenino , Sangre Fetal/citología , Sangre Fetal/inmunología , Sangre Fetal/trasplante , Perfilación de la Expresión Génica , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Humanos , Masculino , Persona de Mediana Edad , Quimera por Trasplante , Trasplante Homólogo , Resultado del Tratamiento
17.
J Immunol ; 190(3): 1094-102, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23264658

RESUMEN

Progenitor B cells reside in complex bone marrow (BM) microenvironments where they receive signals for growth and maturation. We reported previously that the CXCL12-focal adhesion kinase (FAK)-VLA4 pathway plays an important role in progenitor B cell adhesion and migration. In this study, we have conditionally targeted in B cells FAK, and found that the numbers of progenitor pro-B, pre-B, and immature B cells are reduced by 30-40% in B cell-specific FAK knockout mice. When cultured in methylcellulose with IL-7 ± CXCL12, Fak-deleted pro-B cells yield significantly fewer cells and colonies. Using in situ quantitative imaging cytometry, we establish that in longitudinal femoral BM sections, pro-B cells are preferentially localized in close proximity to the endosteum of the metaphyses and the diaphysis. Fak deletion disrupts the nonrandom distribution of pro-B cells and induces the mobilization of pro-B cells to the periphery in vivo. These effects of Fak deletion on pro-B cell mobilization and localization in BM are amplified under inflammatory stress, that is, after immunization with nitrophenol-conjugated chicken γ-globulin in alum. Collectively, these studies suggest the importance of FAK in regulating pro-B cell homeostasis and maintenance of their spatial distribution in BM niches.


Asunto(s)
Linfocitos B/citología , Médula Ósea/ultraestructura , Quinasa 1 de Adhesión Focal/fisiología , Células Madre Hematopoyéticas/enzimología , Linfopoyesis/fisiología , Animales , Apoptosis , Linfocitos B/trasplante , Médula Ósea/inmunología , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Microambiente Celular , Quimiocina CXCL12/fisiología , Quimiotaxis de Leucocito/fisiología , Ensayo de Unidades Formadoras de Colonias , Femenino , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Homeostasis , Integrina alfa4beta1/fisiología , Interleucina-7/farmacología , Linfopenia/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
Proc Natl Acad Sci U S A ; 108(16): 6486-91, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21464312

RESUMEN

The phosphatidylinositol-3,4,5-triphosphate (PIP3) binding function of pleckstrin homology (PH) domain is essential for the activation of oncogenic Akt/PKB kinase. Following the PIP3-mediated activation at the membrane, the activated Akt is subjected to other regulatory events, including ubiquitination-mediated deactivation. Here, by identifying and characterizing an allosteric inhibitor, SC66, we show that the facilitated ubiquitination effectively terminates Akt signaling. Mechanistically, SC66 manifests a dual inhibitory activity that directly interferes with the PH domain binding to PIP3 and facilitates Akt ubiquitination. A known PH domain-dependent allosteric inhibitor, which stabilizes Akt, prevents the SC66-induced Akt ubiquitination. A cancer-relevant Akt1 (e17k) mutant is unstable, making it intrinsically sensitive to functional inhibition by SC66 in cellular contexts in which the PI3K inhibition has little inhibitory effect. As a result of its dual inhibitory activity, SC66 manifests a more effective growth suppression of transformed cells that contain a high level of Akt signaling, compared with other inhibitors of PIP3/Akt pathway. Finally, we show the anticancer activity of SC66 by using a soft agar assay as well as a mouse xenograft tumor model. In conclusion, in this study, we not only identify a dual-function Akt inhibitor, but also demonstrate that Akt ubiquitination could be chemically exploited to effectively facilitate its deactivation, thus identifying an avenue for pharmacological intervention in Akt signaling.


Asunto(s)
Antineoplásicos/farmacología , Ciclohexanonas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Ubiquitinación/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , Sustitución de Aminoácidos , Animales , Antineoplásicos/química , Ciclohexanonas/química , Femenino , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación Missense , Fosfatidilinositoles/genética , Fosfatidilinositoles/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Inhibidores de Proteínas Quinasas/química , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/genética , Piridinas/química , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Ubiquitinación/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Biol Chem ; 287(35): 29979-87, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22791709

RESUMEN

Mechanisms that regulate proliferation and expansion of human hematopoietic stem/multipotent progenitor cells (HSC/MPPs) are targets of intensive investigations. Several cell intrinsic factors and signaling pathways have been implicated in the proliferation and differentiation of human HSC/MPPs. Nevertheless, expansion of human HSC/MPPs for clinical application remains a critical challenge. VentX is a human homeobox transcription factor that was recently identified as an anti-proliferation and pro-differentiation factor in human hematopoietic cells. Here, we report that VentX expression is up-regulated during ontogenesis of human hematopoietic cells. Strikingly, suppression of VentX expression led to significant expansion of HSC/MPPs ex vivo and a 20-fold increase in engraftment potential in the NOD/SCID/IL2Rγ2(null) mouse model. VentX suppression helped preserve the HSC/MPP pools and promote clonogenicity of hematopoietic progenitor cells. Mechanistically, we show that VentX regulates critical cell cycle regulators and Wnt downstream genes previously implicated in HSC/MPP proliferation and expansion.


Asunto(s)
Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Multipotentes/metabolismo , Factores de Transcripción/metabolismo , Animales , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Multipotentes/citología , Factores de Transcripción/genética , Trasplante Heterólogo , Regulación hacia Arriba/fisiología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
20.
Blood ; 117(24): 6702-13, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21521784

RESUMEN

The clinical outcome of granulocyte transfusion therapy is often hampered by short ex vivo shelf life, inefficiency of recruitment to sites of inflammation, and poor pathogen-killing capability of transplanted neutrophils. Here, using a recently developed mouse granulocyte transfusion model, we revealed that the efficacy of granulocyte transfusion can be significantly increased by elevating intracellular phosphatidylinositol (3,4,5)-trisphosphate signaling with a specific phosphatase and tensin homolog deleted on chromosome 10 (PTEN) inhibitor SF1670. Neutrophils treated with SF1670 were much sensitive to chemoattractant stimulation. Neutrophil functions, such as phagocytosis, oxidative burst, polarization, and chemotaxis, were augmented after SF1670 treatment. The recruitment of SF1670-pretreated transfused neutrophils to the inflamed peritoneal cavity and lungs was significantly elevated. In addition, transfusion with SF1670-treated neutrophils led to augmented bacteria-killing capability (decreased bacterial burden) in neutropenic recipient mice in both peritonitis and bacterial pneumonia. Consequently, this alleviated the severity of and decreased the mortality of neutropenia-related pneumonia. Together, these observations demonstrate that the innate immune responses can be enhanced and the severity of neutropenia-related infection can be alleviated by augmenting phosphatidylinositol (3,4,5)-trisphosphate in transfused neutrophils with PTEN inhibitor SF1670, providing a therapeutic strategy for improving the efficacy of granulocyte transfusion.


Asunto(s)
Inhibidores Enzimáticos/administración & dosificación , Granulocitos/trasplante , Fosfohidrolasa PTEN/antagonistas & inhibidores , Peritonitis/terapia , Neumonía Bacteriana/terapia , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Terapia Combinada , Modelos Animales de Enfermedad , Esquema de Medicación , Inhibidores Enzimáticos/farmacología , Granulocitos/efectos de los fármacos , Transfusión de Leucocitos/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Neutropenia/patología , Neutropenia/terapia , Peritonitis/patología , Neumonía Bacteriana/patología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA