Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 21(2): 241-255.e6, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28777945

ABSTRACT

Hematopoietic stem cell (HSC) transplantation represents a curative treatment for various hematological disorders. However, delayed reconstitution of innate and adaptive immunity often causes fatal complications. HSC maintenance and lineage differentiation are supported by stromal niches, and we now find that bone marrow stroma cells (BMSCs) are severely and permanently damaged by the pre-conditioning irradiation required for efficient HSC transplantation. Using mouse models, we show that stromal insufficiency limits the number of donor-derived HSCs and B lymphopoiesis. Intra-bone transplantation of primary, but not cultured, BMSCs quantitatively reconstitutes stroma function in vivo, which is mediated by a multipotent NT5E+ (CD73)+ ENG- (CD105)- LY6A+ (SCA1)+ BMSC subpopulation. BMSC co-transplantation doubles the number of functional, donor-derived HSCs and significantly reduces clinically relevant side effects associated with HSC transplantation including neutropenia and humoral immunodeficiency. These data demonstrate the potential of stroma recovery to improve HSC transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Stem Cell Niche , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Cell Count , Cells, Cultured , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Lymphopoiesis/radiation effects , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/radiation effects , Mice, Inbred C57BL , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/radiation effects , Phenotype , Radiation, Ionizing , Stem Cell Niche/radiation effects , Time Factors
2.
PLoS One ; 10(8): e0134046, 2015.
Article in English | MEDLINE | ID: mdl-26252014

ABSTRACT

Human exposure to high-linear energy transfer α-particles includes environmental (e.g. radon gas and its decay progeny), medical (e.g. radiopharmaceuticals) and occupational (nuclear industry) sources. The associated health risks of α-particle exposure for lung cancer are well documented however the risk estimates for leukaemia remain uncertain. To further our understanding of α-particle effects in target cells for leukaemogenesis and also to seek general markers of individual exposure to α-particles, this study assessed the transmission of chromosomal damage initially-induced in human haemopoietic stem and progenitor cells after exposure to high-LET α-particles. Cells surviving exposure were differentiated into mature T-cells by extra-thymic T-cell differentiation in vitro. Multiplex fluorescence in situ hybridisation (M-FISH) analysis of naïve T-cell populations showed the occurrence of stable (clonal) complex chromosome aberrations consistent with those that are characteristically induced in spherical cells by the traversal of a single α-particle track. Additionally, complex chromosome exchanges were observed in the progeny of irradiated mature T-cell populations. In addition to this, newly arising de novo chromosome aberrations were detected in cells which possessed clonal markers of α-particle exposure and also in cells which did not show any evidence of previous exposure, suggesting ongoing genomic instability in these populations. Our findings support the usefulness and reliability of employing complex chromosome exchanges as indicators of past or ongoing exposure to high-LET radiation and demonstrate the potential applicability to evaluate health risks associated with α-particle exposure.


Subject(s)
Alpha Particles , Chromosomes, Human/radiation effects , Genomic Instability/radiation effects , Lymphopoiesis/radiation effects , Adult , Bone Marrow Cells/radiation effects , CD3 Complex/metabolism , Cell Differentiation/radiation effects , Cells, Cultured , Chromosome Aberrations , Clone Cells , Flow Cytometry , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Lymphocyte Depletion , Time Factors
3.
J Allergy Clin Immunol ; 135(3): 818-20.e4, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25420683
4.
Blood ; 123(18): 2797-805, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24652996

ABSTRACT

T-cell deficiency related to disease, medical treatment, or aging represents a major clinical challenge and is associated with significant morbidity and mortality in cancer and bone marrow transplantation recipients. This study describes several innovative and clinically relevant strategies to manipulate thymic function based on an interventional radiology technique for intrathymic injection of cells or drugs. We show that intrathymic injection of multipotent hematopoietic stem/progenitor cells into irradiated syngeneic or allogeneic young or aged recipients resulted in efficient and long-lasting generation of functional donor T cells. Persistence of intrathymic donor cells was associated with intrathymic presence of cells resembling long-term hematopoietic stem cells, suggesting a self-renewal capacity of the intrathymically injected cells. Furthermore, our approach enabled the induction of long-term antigen-specific T-cell-mediated antitumor immunity following intrathymic injection of progenitor cells harboring a transgenic T-cell receptor gene. The intrathymic injection of interleukin-7 prior to irradiation conferred radioprotection. In addition, thymopoiesis of aged mice improved with a single intrathymic administration of low-dose keratinocyte growth factor, an effect that was sustained even in the setting of radiation-induced injury. Taken together, we established a preclinical framework for the development of novel clinical protocols to establish lifelong antigen-specific T-cell immunity.


Subject(s)
Immunity, Cellular , Immunotherapy , Multipotent Stem Cells/cytology , Stem Cell Transplantation , T-Lymphocytes/immunology , Thymus Gland/immunology , Age Factors , Animals , Antigens/immunology , Bone Marrow Transplantation , Disease Models, Animal , Female , Hematopoietic Stem Cells/cytology , Immunophenotyping , Lymphopoiesis/drug effects , Lymphopoiesis/immunology , Lymphopoiesis/radiation effects , Mice , Multipotent Stem Cells/metabolism , Neoplasms/immunology , Neoplasms/therapy , Phenotype , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Whole-Body Irradiation
5.
Health Phys ; 102(4): 425-36, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22378204

ABSTRACT

A biomathematical model of lymphopoiesis is described and used to analyze the lymphocyte changes observed in the blood of exposed victims in radiation accidents. The coarse-grained architecture of cellular replication and production and implicit cellular regulation mechanisms used in this model make it straightforward to incorporate various radiation conditions. Model simulations with reported absorbed doses as inputs are shown to qualitatively and quantitatively describe a wide range of accidental data in vastly different scenarios. In addition, the absolute lymphocyte counts and the depletion rate constants calculated by this model show good correlation with two widely recognized empirical methods for early dose assessment. This demonstrates the potential to use the biophysical model as an alternative method for the assessment of radiation injury in the case of large-scale radiation disaster. The physiological assumptions underlying the model are also discussed, which may provide a putative mechanism for some biodosimetric tools that use the peripheral blood cell counts as markers of radiation impairment.


Subject(s)
Lymphopoiesis/radiation effects , Models, Biological , Radioactive Hazard Release , Humans , Lymphocyte Count , Lymphocytes/cytology , Lymphocytes/radiation effects , Radiation Dosage , Radiometry
6.
Blood ; 119(3): 717-26, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22101896

ABSTRACT

Alterations of the BM microenvironment have been shown to occur after chemoradiotherapy, during aging, and after genetic manipulations of telomere length. Nevertheless, whether BM stromal cells adopt senescent features in response to these events is unknown. In the present study, we provide evidence that exposure to ionizing radiation (IR) leads murine stromal BM cells to express senescence markers, namely senescence-associated ß-galactosidase and increased p16(INK4a)/p19(ARF) expression. Long (8 weeks) after exposure of mice to IR, we observed a reduction in the number of stromal cells derived from BM aspirates, an effect that we found to be absent in irradiated Ink4a/arf-knockout mice and to be mostly independent of the CFU potential of the stroma. Such a reduction in the number of BM stromal cells was specific, because stromal cells isolated from collagenase-treated bones were not reduced after IR. Surprisingly, we found that exposure to IR leads to a cellular nonautonomous and Ink4a/arf-dependent effect on lymphopoiesis. Overall, our results reveal the distinct sensitivity of BM stromal cell populations to IR and suggest that long-term residual damage to the BM microenvironment can influence hematopoiesis in an Ink4a/arf-dependent manner.


Subject(s)
ADP-Ribosylation Factor 1/physiology , Bone Marrow/radiation effects , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/physiology , Homeostasis/radiation effects , Radiation, Ionizing , Stromal Cells/radiation effects , Animals , Apoptosis , Blotting, Western , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Differentiation , Cell Proliferation , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Lymphopoiesis/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Stromal Cells/metabolism , Stromal Cells/pathology
7.
Biochem Biophys Res Commun ; 377(1): 41-5, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18823940

ABSTRACT

To investigate involvement of miRNAs in radiation responses we used microRNAome profiling to analyze the sex-specific response of radiation sensitive hematopoietic lymphoid tissues. We show that radiation exposure resulted in a significant and sex-specific deregulation of microRNA expression in murine spleen and thymus tissues. Among the regulated miRNAs, we found that changes in expression of miR-34a and miR-7 may be involved in important protective mechanisms counteracting radiation cytotoxicity. We observed a significant increase in the expression of tumor-suppressor miR-34a, paralleled by a decrease in the expression of its target oncogenes NOTCH1, MYC, E2F3 and cyclin D1. Additionally, we show that miR-7 targets the lymphoid-specific helicase LSH, a pivotal regulator of DNA methylation and genome stability. While miR-7 was significantly down-regulated LSH was significantly up-regulated. These cellular changes may constitute an attempt to counteract radiation-induced hypomethylation. Tissue specificity of miRNA responses and possible regulation of miRNA expression upon irradiation are discussed.


Subject(s)
Gene Expression/radiation effects , Lymphopoiesis/radiation effects , MicroRNAs/biosynthesis , Spleen/radiation effects , Thymus Gland/radiation effects , Animals , Female , Lymphopoiesis/genetics , Male , Mice , Mice, Inbred C57BL , Sex Factors , Spleen/metabolism , Thymus Gland/metabolism , X-Rays
8.
J Exp Med ; 205(3): 523-31, 2008 Mar 17.
Article in English | MEDLINE | ID: mdl-18299401

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) requires conditioning treatments such as irradiation, which leads to a severely delayed recovery of T cell immunity and constitutes a major complication of this therapy. Currently, our understanding of the mechanisms regulating thymic recovery is limited. It is known that a subpopulation of bone marrow (BM)-derived thymic immigrant cells and the earliest intrathymic progenitors express the FMS-like tyrosine kinase 3 (Flt3) receptor; however, the functional significance of this expression in the thymus is not known. We used the BM transplant model to investigate the importance of Flt3 ligand (FL) for the regeneration of the T cell compartment. We show that FL is expressed in the adult mouse thymus on the surface of perivascular fibroblasts. These cells surround the proposed thymic entry site of Flt3 receptor-positive T cell progenitors. After irradiation, perivascular FL expression is up-regulated and results in an enhanced recovery of thymic cellularity. Thymic grafting experiments confirm an intrathymic requirement for FL. Collectively, these results show that thymic stromal cell-mediated FL-Flt3 receptor interactions are important in the reconstitution of thymopoiesis early after lethal irradiation and HSCT, and provide a functional relevance to the expression of the Flt3 receptor on intrathymic T cell progenitors.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Thymus Gland/metabolism , Thymus Gland/radiation effects , Animals , Base Sequence , Bone Marrow Transplantation , DNA Primers/genetics , Female , Gene Expression/radiation effects , Ligands , Lymphocyte Count , Lymphopoiesis/genetics , Lymphopoiesis/physiology , Lymphopoiesis/radiation effects , Male , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Stromal Cells/metabolism , Stromal Cells/radiation effects , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Thymus Gland/cytology , Transplantation Chimera , Up-Regulation/radiation effects , fms-Like Tyrosine Kinase 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...