Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell ; 172(1-2): 191-204.e10, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29224778

RESUMEN

Hematopoietic stem cell transplantation is a potential curative therapy for malignant and nonmalignant diseases. Improving the efficiency of stem cell collection and the quality of the cells acquired can broaden the donor pool and improve patient outcomes. We developed a rapid stem cell mobilization regimen utilizing a unique CXCR2 agonist, GROß, and the CXCR4 antagonist AMD3100. A single injection of both agents resulted in stem cell mobilization peaking within 15 min that was equivalent in magnitude to a standard multi-day regimen of granulocyte colony-stimulating factor (G-CSF). Mechanistic studies determined that rapid mobilization results from synergistic signaling on neutrophils, resulting in enhanced MMP-9 release, and unexpectedly revealed genetic polymorphisms in MMP-9 that alter activity. This mobilization regimen results in preferential trafficking of stem cells that demonstrate a higher engraftment efficiency than those mobilized by G-CSF. Our studies suggest a potential new strategy for the rapid collection of an improved hematopoietic graft.


Asunto(s)
Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Adulto , Animales , Bencilaminas , Quimiocina CXCL2/farmacología , Ciclamas , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Polimorfismo Genético
2.
Cell ; 166(2): 263, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27419862

RESUMEN

Adenosine deaminase (ADA) deficiency results in the accumulation of toxic metabolites that destroy the immune system, causing severe combined immunodeficiency (ADA-SCID), often referred to as the "bubble boy" disease. Strimvelis is a European Medicines Agency approved gene therapy for ADA-SCID patients without a suitable bone marrow donor.

3.
Nature ; 495(7441): 365-9, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23485965

RESUMEN

To maintain lifelong production of blood cells, haematopoietic stem cells (HSCs) are tightly regulated by inherent programs and extrinsic regulatory signals received from their microenvironmental niche. Long-term repopulating HSCs reside in several, perhaps overlapping, niches that produce regulatory molecules and signals necessary for homeostasis and for increased output after stress or injury. Despite considerable advances in the specific cellular or molecular mechanisms governing HSC-niche interactions, little is known about the regulatory function in the intact mammalian haematopoietic niche. Recently, we and others described a positive regulatory role for prostaglandin E2 (PGE2) on HSC function ex vivo. Here we show that inhibition of endogenous PGE2 by non-steroidal anti-inflammatory drug (NSAID) treatment in mice results in modest HSC egress from the bone marrow. Surprisingly, this was independent of the SDF-1-CXCR4 axis implicated in stem-cell migration. Stem and progenitor cells were found to have differing mechanisms of egress, with HSC transit to the periphery dependent on niche attenuation and reduction in the retentive molecule osteopontin. Haematopoietic grafts mobilized with NSAIDs had superior repopulating ability and long-term engraftment. Treatment of non-human primates and healthy human volunteers confirmed NSAID-mediated egress in other species. PGE2 receptor knockout mice demonstrated that progenitor expansion and stem/progenitor egress resulted from reduced E-prostanoid 4 (EP4) receptor signalling. These results not only uncover unique regulatory roles for EP4 signalling in HSC retention in the niche, but also define a rapidly translatable strategy to enhance transplantation therapeutically.


Asunto(s)
Dinoprostona/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre/citología , Animales , Antiinflamatorios no Esteroideos/farmacología , Bencilaminas , Recuento de Células , Movimiento Celular/fisiología , Células Cultivadas , Ciclamas , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Humanos , Meloxicam , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteopontina/genética , Papio , Subtipo EP4 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Células Madre/efectos de los fármacos , Tiazinas/farmacología , Tiazoles/farmacología
4.
Blood ; 123(2): 203-7, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24167196

RESUMEN

Hematopoietic stem cell (HSC) transplantation is a lifesaving therapy for a number of immunologic disorders. For effective transplant, HSCs must traffic from the peripheral blood to supportive bone marrow niches. We previously showed that HSC trafficking can be enhanced by ex vivo treatment of hematopoietic grafts with 16-16 dimethyl prostaglandin E2 (dmPGE2). While exploring regulatory molecules involved in dmPGE2 enhancement, we found that transiently increasing the transcription factor hypoxia-inducible factor 1-α (HIF1α) is required for dmPGE2-enhanced CXCR4 upregulation and enhanced migration and homing of stem and progenitor cells and that pharmacologic manipulation of HIF1α is also capable of enhancing homing and engraftment. We also now identify the specific hypoxia response element required for CXCR4 upregulation. These data define a precise mechanism through which ex vivo pulse treatment with dmPGE2 enhances the function of hematopoietic stem and progenitor cells; these data also define a role for hypoxia and HIF1α in enhancement of hematopoietic transplantation.


Asunto(s)
Dinoprostona/farmacología , Supervivencia de Injerto/genética , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transcripción Genética
5.
Blood ; 122(17): 2997-3000, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24047650

RESUMEN

Hematopoietic stem cell (HSC) transplantation is a lifesaving therapy for malignant and nonmalignant hematologic diseases and metabolic disorders. Although successful, hematopoietic transplantation can be hindered by inadequate stem cell number or poor engrafting efficiency. To overcome these deficits, we and others have previously reported the HSC-enhancing ability of a short-term exposure of prostaglandin E2 (PGE2); this strategy has now progressed to phase 1 clinical trials in double cord blood transplantation. To further analyze the short- and long-term effects of HSC exposure to PGE2, we followed the repopulation kinetics of PGE2-treated hematopoietic grafts through 5 serial transplantations and compared inherent long-term competitiveness in a HSC head-to-head secondary transplantation model. Treatment with PGE2 did not result in a long-term increase in HSC competitiveness, lineage bias, or enhanced proliferative potential, demonstrating that pulse exposure to PGE2 results in transient increases in HSC homing and engraftment potential.


Asunto(s)
Linaje de la Célula/efectos de los fármacos , Dinoprostona/farmacología , Supervivencia de Injerto/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Recuento de Células , Linaje de la Célula/inmunología , Proliferación Celular , Células Cultivadas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Ratones , Ratones Endogámicos C57BL
6.
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
7.
Blood ; 119(7): 1671-82, 2012 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-22110249

RESUMEN

Dendritic cell (DC) homeostasis, like all mature blood cells, is maintained via hierarchal generation from hematopoietic precursors; however, little is known about the regulatory mechanisms governing DC generation. Here, we show that prostaglandin E(2) (PGE(2)) is required for optimal Flt3 ligand-mediated DC development and regulates expression of the Flt3 receptor on DC-committed progenitor cells. Inhibition of PGE(2) biosynthesis reduces Flt3-mediated activation of STAT3 and expression of the antiapoptotic protein survivin, resulting in increased apoptosis of DC-committed progenitor cells. Reduced DC development caused by diminished PGE(2) signaling is reversed by overexpression of Flt3 or survivin in DC progenitors and conversely is mimicked by STAT3 inhibition. PGE(2) regulation of DC generation is specifically mediated through the EP1 and EP3 G protein PGE(2) receptors. These studies define a novel DC progenitor regulatory pathway in which PGE(2) signaling through EP1/EP3 receptors regulates Flt3 expression and downstream STAT3 activation and survivin expression, required for optimal DC progenitor survival and DC development in vivo.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Dinoprostona/antagonistas & inhibidores , Células Madre Hematopoyéticas/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Proteínas de la Membrana/fisiología , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP3 de Receptores de Prostaglandina E/antagonistas & inhibidores , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/fisiología , Dinoprostona/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Recién Nacido , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Survivin
8.
Stem Cells ; 31(12): 2599-606, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24123398

RESUMEN

Hematopoietic stem cell transplantation is the only curative option for a number of malignant and nonmalignant diseases. As the use of hematopoietic transplant has expanded, so too has the source of stem and progenitor cells. The predominate source of stem and progenitors today, particularly in settings of autologous transplantation, is mobilized peripheral blood. This review will highlight the historical advances which led to the widespread use of peripheral blood stem cells for transplantation, with a look toward future enhancements to mobilization strategies.


Asunto(s)
Movilización de Célula Madre Hematopoyética/métodos , Animales , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Nicho de Células Madre
9.
Blood Cells Mol Dis ; 50(3): 147-53, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23206586

RESUMEN

While high dose total body irradiation (TBI) is used therapeutically, the proliferation of nuclear weapons, increasing use of nuclear power, and worldwide radical terrorism underscore the need to develop countermeasures to a radiological mass casualty event. The hematopoietic syndrome of the acute radiation syndrome (HS-ARS) results from severe compromise to the hematopoietic system, including lymphocytopenia, neutropenia, thrombocytopenia, and possible death from infection and/or hemorrhage. Given adequate time to recover, expand, and appropriately differentiate, bone marrow hematopoietic stem cells (HSC) and progenitor cells (HPC) may overcome HS-ARS and restore homeostasis of the hematopoietic system. Prostaglandin E(2) (PGE(2)) has been shown to have pleiotropic effects on hematopoiesis, acting to inhibit apoptosis and promote self-renewal of HSC, while inhibiting HPC proliferation. We assessed the radio-mitigating potential of modulating PGE(2) signaling in a mouse model of HS-ARS. Treatment with the PGE(2) analog 16,16 dimethyl PGE(2) (dmPGE(2)) 6h post-irradiation or inhibition of PGE(2) synthesis via delayed administration of the non-steroidal anti-inflammatory drug (NSAID) Meloxicam resulted in increased survival of lethally irradiated mice. Both early dmPGE(2) and delayed Meloxicam treatment were associated with increased HPC activity 35days following irradiation, demonstrating enhanced recovery of hematopoiesis. Our results define two different treatment modalities that are highly effective and safe to administer, and can be readily available.


Asunto(s)
Dinoprostona/metabolismo , Hematopoyesis/fisiología , Hematopoyesis/efectos de la radiación , Transducción de Señal , Animales , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/administración & dosificación , Dinoprostona/farmacología , Activación Enzimática/efectos de la radiación , Femenino , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Masculino , Ratones , Irradiación Corporal Total
10.
Methods Mol Biol ; 2567: 3-7, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36255691

RESUMEN

Hematopoietic stem cells (HSCs) support the lifelong production of hundreds of billions of blood cells per day. This unique, incredible ability of HSCs also creates an incredible therapeutic potential for patients. To advance this potential, effective methods to study HSCs are continually evolving. This chapter summarizes the variety of protocols and techniques covered in this book used to evaluate HSCs - modification, characterization, interaction with their niche, and in vivo function.


Asunto(s)
Células Madre Hematopoyéticas , Humanos , Terapia Genética
11.
Mol Ther Nucleic Acids ; 34: 102043, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37920545

RESUMEN

Development of factor VIII (FVIII) inhibitors is a serious complication in the treatment of hemophilia A (HemA) patients. In clinical trials, anti-CD3 antibody therapy effectively modulates the immune response of allograft rejection or autoimmune diseases without eliciting major adverse effects. In this study, we delivered mRNA-encapsulated lipid nanoparticles (LNPs) encoding therapeutic anti-CD3 antibody (αCD3 LNPs) to overcome the anti-FVIII immune responses in HemA mice. It was found that αCD3 LNPs encoding the single-chain antibodies (Fc-scFv) can efficiently deplete CD3+ and CD4+ effector T cells, whereas αCD3 LNPs encoding double-chain antibodies cannot. Concomitantly, mice treated with αCD3 (Fc-scFv) LNPs showed an increase in the CD4+CD25+Foxp3+ regulatory T cell percentages, which modulated the anti-FVIII immune responses. All T cells returned to normal levels within 2 months. HemA mice treated with αCD3 LNPs prior to hydrodynamic injection of liver-specific FVIII plasmids achieved persistent FVIII gene expression without formation of FVIII inhibitors. Furthermore, transgene expression was increased and persistent following secondary plasmid challenge, indicating induction of long-term tolerance to FVIII. Moreover, the treated mice maintained their immune competence against other antigens. In conclusion, our study established a potential new strategy to induce long-term antigen-specific tolerance using an αCD3 LNP formulation.

12.
Curr Opin Hematol ; 18(4): 231-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21537168

RESUMEN

PURPOSE OF REVIEW: Blood cell production is maintained by hematopoietic stem cells (HSCs) that reside in specialized niches within bone marrow. Treatment with granulocyte-colony stimulating factor (G-CSF) causes HSC egress from bone marrow niches and trafficking to the peripheral blood, a process termed 'mobilization'. Although the mobilization phenomenon has been known for some time and is utilized clinically to acquire HSC for transplant, the mechanisms mediating HSC release are not completely understood. We discuss recent advances and controversies in defining the mechanisms responsible for G-CSF-induced mobilization. RECENT FINDINGS: New reports define a role for resident monocytes/macrophages in maintaining niche cells, which is diminished after G-CSF treatment, suggesting a new mechanism for mobilization. Although osteoblasts have been reported to be a primary component of the HSC niche, new results suggest a unique niche composed of innervated mesenchymal stem cells. Modulating bioactive lipid signaling also facilitates mobilization, and may define a future therapeutic strategy. SUMMARY: Hematopoietic mobilization by G-CSF is primarily mediated by alterations to the bone marrow niche by both direct and indirect mechanisms, rather than directly altering HSC function. Further understanding of the processes mediating mobilization will advance our understanding on the cellular and molecular components of the HSC niche.


Asunto(s)
Movimiento Celular/fisiología , Células Madre Hematopoyéticas/citología , Nicho de Células Madre/citología , Animales , Movimiento Celular/efectos de los fármacos , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Ratones , Nicho de Células Madre/efectos de los fármacos
13.
Blood ; 113(22): 5444-55, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19324903

RESUMEN

Adult hematopoietic stem cells (HSCs) are routinely used to reconstitute hematopoiesis after myeloablation; however, transplantation efficacy and multilineage reconstitution can be limited by inadequate HSC number, or poor homing, engraftment, or self-renewal. Here we report that mouse and human HSCs express prostaglandin E2 (PGE2) receptors, and that short-term ex vivo exposure of HSCs to PGE2 enhances their homing, survival, and proliferation, resulting in increased long-term repopulating cell (LTRC) and competitive repopulating unit (CRU) frequency. HSCs pulsed with PGE2 are more competitive, as determined by head-to-head comparison in a competitive transplantation model. Enhanced HSC frequency and competitive advantage is stable and maintained upon serial transplantation, with full multilineage reconstitution. PGE2 increases HSC CXCR4 mRNA and surface expression, enhances their migration to SDF-1 in vitro and homing to bone marrow in vivo, and stimulates HSC entry into and progression through cell cycle. In addition, PGE2 enhances HSC survival, associated with an increase in Survivin mRNA and protein expression and reduction in intracellular active caspase-3. Our results define novel mechanisms of action whereby PGE2 enhances HSC function and supports a strategy to use PGE2 to facilitate hematopoietic transplantation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Dinoprostona/farmacología , Supervivencia de Injerto/efectos de los fármacos , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Receptores CXCR4/metabolismo , Receptores de Prostaglandina E/metabolismo , Acondicionamiento Pretrasplante/métodos
14.
Prostaglandins Other Lipid Mediat ; 96(1-4): 3-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21722751

RESUMEN

Eicosanoids have been implicated in the physiological regulation of hematopoiesis with pleiotropic effects on hematopoietic stem cells and various classes of lineage restricted progenitor cells. Herein we review the effects of eicosanoids on hematopoiesis, focusing on new findings implicating prostaglandin E(2) in enhancing hematopoietic stem cell engraftment by enhancing stem cell homing, survival and self-renewal. We also describe a role for cannabinoids in hematopoiesis. Lastly, we discuss the yin and yang of various eicosanoids in modulating hematopoietic stem and progenitor cell functions and summarize potential strategies to take advantage of these effects for therapeutic benefit for hematopoietic stem cell transplantation.


Asunto(s)
Cannabinoides/inmunología , Dinoprostona/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/fisiología , Transducción de Señal/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Cannabinoides/genética , Cannabinoides/metabolismo , Ciclo Celular/genética , Ciclo Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Quimiocina CXCL12/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Regulación de la Expresión Génica/inmunología , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Transducción de Señal/genética , Yin-Yang
15.
Stem Cell Rev Rep ; 17(6): 1939-1953, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34661830

RESUMEN

Hematopoietic stem cells naturally traffic out of their bone marrow niches into the peripheral blood. This natural trafficking process can be enhanced with numerous pharmacologic agents - a process termed "mobilization" - and the mobilized stem cells can be collected for transplantation. We review the current state of mobilization with an update on recent clinical trials and new biologic mechanisms regulating stem cell trafficking. We propose that hematopoietic mobilization can be used to answer questions regarding hematopoietic stem cell heterogeneity, can be used for non-toxic conditioning of patients receiving stem cell transplants, and can enhance gene editing and gene therapy strategies to cure genetic diseases.


Asunto(s)
Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Factor Estimulante de Colonias de Granulocitos , Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/metabolismo , Humanos , Trasplante de Células Madre
16.
Stem Cell Reports ; 15(2): 358-373, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32735825

RESUMEN

Ionizing radiation exposure results in acute and delayed bone marrow suppression. Treatment of mice with 16,16-dimethyl prostaglandin E2 (dmPGE2) prior to lethal ionizing radiation (IR) facilitates survival, but the cellular and molecular mechanisms are unclear. In this study we show that dmPGE2 attenuates loss and enhances recovery of bone marrow cellularity, corresponding to a less severe hematopoietic stem cell nadir, and significantly preserves long-term repopulation capacity and progenitor cell function. Mechanistically, dmPGE2 suppressed hematopoietic stem cell (HSC) proliferation through 24 h post IR, which correlated with fewer DNA double-strand breaks and attenuation of apoptosis, mitochondrial compromise, oxidative stress, and senescence. RNA sequencing of HSCs at 1 h and 24 h post IR identified a predominant interference with IR-induced p53-downstream gene expression at 1 h, and confirmed the suppression of IR-induced cell-cycle genes at 24 h. These data identify mechanisms of dmPGE2 radioprotection and its potential role as a medical countermeasure against radiation exposure.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de los fármacos , Dinoprostona/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Radiación Ionizante , Protectores contra Radiación/farmacología , Animales , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Daño del ADN , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Redes Reguladoras de Genes/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/efectos de la radiación
17.
Nat Commun ; 10(1): 617, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30728354

RESUMEN

Hematopoietic stem cell transplantation (HSCT) is a curative therapy for blood and immune diseases with potential for many settings beyond current standard-of-care. Broad HSCT application is currently precluded largely due to morbidity and mortality associated with genotoxic irradiation or chemotherapy conditioning. Here we show that a single dose of a CD117-antibody-drug-conjugate (CD117-ADC) to saporin leads to > 99% depletion of host HSCs, enabling rapid and efficient donor hematopoietic cell engraftment. Importantly, CD117-ADC selectively targets hematopoietic stem cells yet does not cause clinically significant side-effects. Blood counts and immune cell function are preserved following CD117-ADC treatment, with effective responses by recipients to both viral and fungal challenges. These results suggest that CD117-ADC-mediated HSCT pre-treatment could serve as a non-myeloablative conditioning strategy for the treatment of a wide range of non-malignant and malignant diseases, and might be especially suited to gene therapy and gene editing settings in which preservation of immunity is desired.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Inmunoconjugados/farmacología , Proteínas Proto-Oncogénicas c-kit/inmunología , Animales , Médula Ósea/efectos de los fármacos , Trasplante de Médula Ósea , Candida albicans/patogenicidad , Muerte Celular , Línea Celular , Femenino , Terapia Genética , Humanos , Inmunoconjugados/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Neoplasias , Donantes de Tejidos
18.
J Clin Invest ; 127(12): 4527-4540, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29130940

RESUMEN

Endothelial cells (ECs) are components of the hematopoietic microenvironment and regulate hematopoietic stem and progenitor cell (HSPC) homeostasis. Cytokine treatments that cause HSPC trafficking to peripheral blood are associated with an increase in dipeptidylpeptidase 4/CD26 (DPP4/CD26), an enzyme that truncates the neurotransmitter neuropeptide Y (NPY). Here, we show that enzymatically altered NPY signaling in ECs caused reduced VE-cadherin and CD31 expression along EC junctions, resulting in increased vascular permeability and HSPC egress. Moreover, selective NPY2 and NPY5 receptor antagonists restored vascular integrity and limited HSPC mobilization, demonstrating that the enzymatically controlled vascular gateway specifically opens by cleavage of NPY by CD26 signaling via NPY2 and NPY5 receptors. Mice lacking CD26 or NPY exhibited impaired HSPC trafficking that was restored by treatment with truncated NPY. Thus, our results point to ECs as gatekeepers of HSPC trafficking and identify a CD26-mediated NPY axis that has potential as a pharmacologic target to regulate hematopoietic trafficking in homeostatic and stress conditions.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Neuropéptido Y/metabolismo , Transducción de Señal/fisiología , Animales , Dipeptidil Peptidasa 4/genética , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Noqueados , Neuropéptido Y/genética , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo
19.
Annu Rev Pathol ; 11: 555-81, 2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27193455

RESUMEN

Regulation of stem cells in adult tissues is a key determinant of how well an organism can respond to the stresses of physiological challenge and disease. This is particularly true of the hematopoietic system, where demands on host defenses can call for an acute increase in cell production. Hematopoietic stem cells receive the regulatory signals for cell production in adult mammals in the bone marrow, a tissue with higher-order architectural and functional organization than previously appreciated. Here, we review the data defining particular structural components and heterologous cells in the bone marrow that participate in hematopoietic stem cell function. Further, we explore the case for stromal-hematopoietic cell interactions contributing to neoplastic myeloid disease. As the hematopoietic regulatory networks in the bone marrow are revealed, it is anticipated that strategies will emerge for how to enhance or inhibit production of specific blood cells. In that way, the control of hematopoiesis will enter the domain of therapies to modulate broad aspects of hematopoiesis, both normal and malignant.


Asunto(s)
Células Madre Adultas/citología , Células de la Médula Ósea/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Nicho de Células Madre , Animales , Humanos
20.
Exp Hematol ; 44(2): 132-137.e1, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26644183

RESUMEN

The laboratory mouse is the model most frequently used in hematologic studies and assessment of blood parameters across a broad range of disciplines. Often, analysis of blood occurs in a nonterminal manner. However, the small body size of the mouse limits collection based on volume, frequency, and accessible sites. Commonly used sites in the mouse include the retro-orbital sinus, facial vein, tail vein, saphenous vein, and heart. The method of blood acquisition varies considerably across laboratories and is often not reported in detail. In this study, we report significant alterations in blood parameters, particularly of total white blood cells, specific populations of dendritic cells and myeloid-derived suppressor cells, and hematopoietic progenitor cells, as a result of site and manner of sampling. Intriguingly, warming of mice prior to tail bleeding was found to significantly alter blood values. Our findings suggest that the same method should be used across an entire study, that mice should be warmed prior to tail bleeds to make levels uniform, and that accurate description of bleeding methods in publications should be provided to allow for interpretation of comparative reports and inter- and intralaboratory experimental variability.


Asunto(s)
Animales de Laboratorio , Sangre , Manejo de Especímenes , Animales , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA