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

2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Cell Stem Cell ; 19(4): 530-543, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27524439

RESUMEN

Physiological stem cell function is regulated by secreted factors produced by niche cells. In this study, we describe an unbiased approach based on the differential single-cell gene expression analysis of mesenchymal osteolineage cells close to, and further removed from, hematopoietic stem/progenitor cells (HSPCs) to identify candidate niche factors. Mesenchymal cells displayed distinct molecular profiles based on their relative location. We functionally examined, among the genes that were preferentially expressed in proximal cells, three secreted or cell-surface molecules not previously connected to HSPC biology-the secreted RNase angiogenin, the cytokine IL18, and the adhesion molecule Embigin-and discovered that all of these factors are HSPC quiescence regulators. Therefore, our proximity-based differential single-cell approach reveals molecular heterogeneity within niche cells and can be used to identify novel extrinsic stem/progenitor cell regulators. Similar approaches could also be applied to other stem cell/niche pairs to advance the understanding of microenvironmental regulation of stem cell function.


Asunto(s)
Células Madre Hematopoyéticas/citología , Análisis de la Célula Individual/métodos , Nicho de Células Madre , Animales , Células de la Médula Ósea/citología , Huesos/citología , Linaje de la Célula/genética , Autorrenovación de las Células/genética , Separación Celular , Eliminación de Gen , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Interleucina-18/metabolismo , Glicoproteínas de Membrana/metabolismo , Ribonucleasa Pancreática/metabolismo , Factores de Tiempo , Transcripción Genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
9.
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.

10.
Nat Biotechnol ; 34(7): 738-45, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27272386

RESUMEN

Hematopoietic stem cell transplantation (HSCT) offers curative therapy for patients with hemoglobinopathies, congenital immunodeficiencies, and other conditions, possibly including AIDS. Autologous HSCT using genetically corrected cells would avoid the risk of graft-versus-host disease (GVHD), but the genotoxicity of conditioning remains a substantial barrier to the development of this approach. Here we report an internalizing immunotoxin targeting the hematopoietic-cell-restricted CD45 receptor that effectively conditions immunocompetent mice. A single dose of the immunotoxin, CD45-saporin (SAP), enabled efficient (>90%) engraftment of donor cells and full correction of a sickle-cell anemia model. In contrast to irradiation, CD45-SAP completely avoided neutropenia and anemia, spared bone marrow and thymic niches, enabling rapid recovery of T and B cells, preserved anti-fungal immunity, and had minimal overall toxicity. This non-genotoxic conditioning method may provide an attractive alternative to current conditioning regimens for HSCT in the treatment of non-malignant blood diseases.


Asunto(s)
Daño del ADN/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Antígenos Comunes de Leucocito/inmunología , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Daño del ADN/genética , Femenino , Mejoramiento Genético/métodos , Fenómenos Inmunogenéticos/genética , Inmunotoxinas , Ratones , Ratones Endogámicos C57BL , Saporinas
11.
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
12.
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
13.
Int J Nanomedicine ; 10: 2647-52, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25878498

RESUMEN

Chemotherapy, irradiation, and other agents are widely used to target the process of cell division in neoplastic cells. However, while these therapies are effective against most cancers, the high proliferative rate of the cells of the hematopoietic system that produce billions of blood cells needed daily throughout life is extremely sensitive to these agents, resulting in loss of blood cell populations, which can be life threatening. Neutropenia is the most serious hematologic toxicity of chemotherapy, which can result in patient morbidity and mortality due to opportunistic infection and often is the limiting factor in dose escalation or duration of chemotherapeutic administration. Neutropenic patients often require hospitalization and incur substantial medical costs associated with anti-infective therapy. Treatment of iatrogenic and congenic neutropenia was changed in the early 1990s with the introduction of filgrastim (Neupogen(®)) and pegfilgrastim (Neulasta(®)). With the expiration of patent lives of both of these drugs, biosimilars have begun to emerge. In this review, we will summarize the chemical characteristics, pharmacokinetics, safety and efficacy of lipegfilgrastim (Lonquex(®)), the first long-acting biosimilar filgrastim to receive regulatory approval and enter the marketplace.


Asunto(s)
Neutropenia Febril Inducida por Quimioterapia/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos , Filgrastim , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Factor Estimulante de Colonias de Granulocitos/química , Factor Estimulante de Colonias de Granulocitos/farmacocinética , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Polietilenglicoles , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico
14.
Methods Mol Biol ; 1185: 43-64, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25062621

RESUMEN

Hematopoietic stem cell transplantation (HSCT) can be performed with hematopoietic stem and progenitor cells (HSPC) acquired directly from bone marrow, from umbilical cord blood or placental tissue, or from the peripheral blood after treatment of the donor with agents that enhance egress of HSPC into the circulation, a process known as "mobilization." Mobilized peripheral blood stem cells (PBSC) have become the predominate hematopoietic graft for HSCT, particularly for autologous transplants. Despite the success of PBSC transplant, many patients and donors do not achieve optimal levels of mobilization. Thus, accurate animal models and basic laboratory investigations are needed to further investigate the mechanisms that lead to PBSC mobilization and define improved or new mobilizing agents and/or strategies to enhance PBSC mobilization and transplant. This chapter outlines assays and techniques for exploration of hematopoietic mobilization using mice as a model organism.


Asunto(s)
Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/citología , Animales , Recuento de Células Sanguíneas , Vías de Administración de Medicamentos , Eritrocitos/citología , Femenino , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/farmacología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Cinética , Leucocitos Mononucleares/citología , Masculino , Ratones
15.
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
16.
Expert Opin Investig Drugs ; 23(1): 21-35, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24073859

RESUMEN

INTRODUCTION: Granulocyte colony-stimulating factor (G-CSF; filgrastim) and its pegylated form (pegfilgrastim) are widely used to treat neutropenia associated with myelosuppressive chemotherapy and bone marrow transplantation, AIDS-associated or drug-induced neutropenia, and neutropenic diseases. G-CSF facilitates restoration of neutrophil counts, decreases incidence of infection/febrile neutropenia and reduces resource utilization. G-CSF is also widely used to mobilize peripheral blood stem cells for hematopoietic transplant. AREAS COVERED: We review the therapeutic use, cost effectiveness and disease impact of G-CSF for neutropenia, development of G-CSF biosimilars and current next-generation discovery efforts. EXPERT OPINION: G-CSF has impacted the treatment and survival of patients with congenital neutropenias. For chemotherapy-associated neutropenia, cost effectiveness and impact on survival are still unclear. G-CSFs are expensive and require systemic administration. Market entry of new biosimilars, some with enhanced half-life profiles, will probably reduce cost and increase cost effectiveness. There is no evidence that marketed or late development biosimilars display effectiveness superior to current G-CSFs. Second-generation compounds that mimic the activity of G-CSF at its receptor, induce endogenous ligand(s) or offer adjunct activity have been reported and represent attractive G-CSF alternatives, but are in preclinical stages. A significant therapeutic advance will require reduced depth and duration of neutropenia compared to current G-CSFs.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/agonistas , Neutropenia/tratamiento farmacológico , Animales , Biosimilares Farmacéuticos/uso terapéutico , Humanos
17.
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
18.
Expert Opin Pharmacother ; 14(18): 2453-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24138174

RESUMEN

For a number of malignant hematologic diseases, including leukemias, lymphomas and myelomas, hematopoietic stem cell transplantation remains the only curative option. The stem cell sources for these life-saving transplants come from bone marrow, umbilical cord blood, or from the peripheral blood of patients or donors treated with mobilizing agents. Recently, the use of nonsteroidal anti-inflammatory drugs (NSAIDs) including aspirin, ibuprofen and meloxicam has been reported to enhance the ability to acquire stem cells from mobilized peripheral blood, resulting in a superior stem cell graft. The addition of NSAIDs, notably meloxicam, to current mobilization strategies is convenient, cost effective, and given the long track record of NSAID use, presumably safe. This article discusses the potential to translate these findings to clinical practice and addresses unanswered questions regarding the use of NSAIDs in stem cell transplantation.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Hematopoyéticas , Tiazinas/uso terapéutico , Tiazoles/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/efectos adversos , Humanos , Meloxicam , Tiazinas/administración & dosificación , Tiazinas/efectos adversos , Tiazoles/administración & dosificación , Tiazoles/efectos adversos
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA