Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Sci Adv ; 10(18): eadn6537, 2024 May 03.
Article En | MEDLINE | ID: mdl-38701219

In mammals, males and females show marked differences in immune responses. Males are globally more sensitive to infectious diseases, while females are more susceptible to systemic autoimmunity. X-chromosome inactivation (XCI), the epigenetic mechanism ensuring the silencing of one X in females, may participate in these sex biases. We perturbed the expression of the trigger of XCI, the noncoding RNA Xist, in female mice. This resulted in reactivation of genes on the inactive X, including members of the Toll-like receptor 7 (TLR7) signaling pathway, in monocyte/macrophages and dendritic and B cells. Consequently, female mice spontaneously developed inflammatory signs typical of lupus, including anti-nucleic acid autoantibodies, increased frequencies of age-associated and germinal center B cells, and expansion of monocyte/macrophages and dendritic cells. Mechanistically, TLR7 signaling is dysregulated in macrophages, leading to sustained expression of target genes upon stimulation. These findings provide a direct link between maintenance of XCI and female-biased autoimmune manifestations and highlight altered XCI as a cause of autoimmunity.


Autoimmunity , Macrophages , Toll-Like Receptor 7 , X Chromosome Inactivation , Animals , Female , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Autoimmunity/genetics , Mice , Male , Macrophages/metabolism , Macrophages/immunology , RNA, Long Noncoding/genetics , Signal Transduction , Dendritic Cells/immunology , Dendritic Cells/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology
2.
iScience ; 26(10): 107890, 2023 Oct 20.
Article En | MEDLINE | ID: mdl-37766969

The developmental cartography of human lymphopoiesis remains incompletely understood. Here, we establish a multimodal map demonstrating that lymphoid specification follows independent direct or stepwise hierarchic routes converging toward the emergence of newly characterized CD117lo multi-lymphoid progenitors (MLPs) that undergo a proliferation arrest before entering the CD127- (NK/ILC/T) or CD127+ (B) lymphoid pathways. While the differentiation of CD127- early lymphoid progenitors is mainly driven by Flt3 signaling, emergence of their CD127+ counterparts is regulated cell-intrinsically and depends exclusively on the divisional history of their upstream precursors, including hematopoietic stem cells. Further, transcriptional mapping of differentiation trajectories reveals that whereas myeloid granulomonocytic lineages follow continuous differentiation pathways, lymphoid trajectories are intrinsically discontinuous and characterized by sequential waves of cell proliferation allowing pre-commitment amplification of lymphoid progenitor pools. Besides identifying new lymphoid specification pathways and regulatory checkpoints, our results demonstrate that NK/ILC/T and B lineages are under fundamentally distinct modes of regulation. (149 words).

3.
Cell Rep ; 42(6): 112618, 2023 06 27.
Article En | MEDLINE | ID: mdl-37294633

Changes in lymphocyte production patterns occurring across human ontogeny remain poorly defined. In this study, we demonstrate that human lymphopoiesis is supported by three waves of embryonic, fetal, and postnatal multi-lymphoid progenitors (MLPs) differing in CD7 and CD10 expression and their output of CD127-/+ early lymphoid progenitors (ELPs). In addition, our results reveal that, like the fetal-to-adult switch in erythropoiesis, transition to postnatal life coincides with a shift from multilineage to B lineage-biased lymphopoiesis and an increase in production of CD127+ ELPs, which persists until puberty. A further developmental transition is observed in elderly individuals whereby B cell differentiation bypasses the CD127+ compartment and branches directly from CD10+ MLPs. Functional analyses indicate that these changes are determined at the level of hematopoietic stem cells. These findings provide insights for understanding identity and function of human MLPs and the establishment and maintenance of adaptative immunity.


Hematopoietic Stem Cells , Lymphopoiesis , Adult , Humans , Aged , Cell Differentiation , Cell Lineage , Hematopoiesis
4.
Nat Commun ; 14(1): 1830, 2023 04 01.
Article En | MEDLINE | ID: mdl-37005415

Recent data have shown that liver fibrosis can regress even at later stages of cirrhosis and shifting the immune response from pro-inflammatory towards a resolutive profile is considered as a promising option. The immune regulatory networks that govern the shift of the inflammatory phenotype and thus potential reversal of liver fibrosis are lesser known. Here we show that in precision-cut human liver slices obtained from patients with end-stage fibrosis and in mouse models, inhibiting Mucosal-Associated Invariant T (MAIT) cells using pharmacological or antibody-driven approaches, limits fibrosis progression and even regresses fibrosis, following chronic toxic- or non-alcoholic steatohepatitis (NASH)-induced liver injury. Mechanistic studies, combining RNA sequencing, in vivo functional studies (performed in male mice) and co-culture experiments indicate that disruption of the MAIT cell-monocyte/macrophage interaction results in resolution of fibrosis both by increasing the frequency of restorative Ly6Clo at the expenses of pro-fibrogenic Ly6Chi monocyte-derived macrophages and promoting an autophagic phenotype in both subsets. Thus, our data show that MAIT cell activation and the consequential phenotype shift of liver macrophages are important pathogenic features of liver fibrosis and could be targeted by anti-fibrogenic therapy.


Mucosal-Associated Invariant T Cells , Non-alcoholic Fatty Liver Disease , Humans , Male , Mice , Animals , Liver Cirrhosis/pathology , Macrophages , Liver/pathology , Non-alcoholic Fatty Liver Disease/pathology , Fibrosis , Phenotype , Mice, Inbred C57BL
5.
EJHaem ; 3(3): 970-974, 2022 Aug.
Article En | MEDLINE | ID: mdl-36051036

Chimeric antigen receptor T cells (CAR-T) have provided promising results in multiple myeloma (MM). However, many patients still relapse, pointing toward the need of improving this therapy. Here, we analyzed peripheral blood T cells from MM patients at different stages of the disease and investigated their phenotype and capacity to generate functional CAR-T directed against CS1 or B Cell Maturation antigen. We found a decrease in naive T cells and elevated frequencies of exhaustion markers in T cells from treated MM patients. Interestingly, individuals treated with daratumumab display elevated ratios of central memory T cells. CAR-T derived from patients at relapse show reduced in vitro expansion and cytotoxic capacities in response to MM cells compared to those produced at diagnosis. Of note, CAR-T from daratumumab treated patients display intermediate defects. Reduced anti-myeloma activity of CAR T cells from treated patients was also observed in a mouse model. Our findings suggest that T cell defects in MM patients, specifically during relapse, have a major impact on their capacity to generate efficient therapeutic CAR-T. Selecting naive or central memory T cell subsets to generate therapeutic T cells could improve the CAR-T therapy for MM.

7.
Life Sci Alliance ; 4(3)2021 03.
Article En | MEDLINE | ID: mdl-33446491

Accumulation of senescent cells is an important contributor to chronic inflammation upon aging. The inflammatory phenotype of senescent cells was previously shown to be driven by cytoplasmic DNA. Here, we propose that cytoplasmic double-stranded RNA has a similar effect. We find that several cell types driven into senescence by different routes share an accumulation of long promoter RNAs and 3' gene extensions rich in retrotransposon sequences. Accordingly, these cells display increased expression of genes involved in response to double stranded RNA of viral origin downstream of the interferon pathway. The RNA accumulation is associated with evidence of reduced RNA turnover, including in some cases, reduced expression of RNA exosome subunits. Reciprocally, depletion of RNA exosome subunit EXOSC3 accelerated expression of multiple senescence markers. A senescence-like RNA accumulation was also observed in cells exposed to oxidative stress, an important trigger of cellular senescence. Altogether, we propose that in a subset of senescent cells, repeat-containing transcripts stabilized by oxidative stress or reduced RNA exosome activity participate in driving and maintaining the permanent inflammatory state characterizing cellular senescence.


Cellular Senescence/genetics , RNA Stability/genetics , RNA/metabolism , Cell Line , DNA Damage , Humans , Inflammation/metabolism , Oxidative Stress/genetics , Phenotype , RNA/genetics , RNA, Double-Stranded/adverse effects , RNA, Double-Stranded/genetics , Retroelements/genetics
8.
Leukemia ; 35(6): 1710-1721, 2021 06.
Article En | MEDLINE | ID: mdl-32879426

Multiple myeloma (MM) is a currently incurable malignancy of antibody-secreting plasma cells. Long non-coding RNAs (lncRNAs) have been recognised as an important class of regulatory molecules which are increasingly implicated in tumorigenesis. While recent studies have demonstrated changes in expression of lncRNAs in MM, the functional significance and molecular pathways downstream of these changes remain poorly characterised. In this study, we have performed CRISPR-mediated deletion of the locus encoding the lncRNA Colorectal Neoplasia Differentially Expressed (CRNDE), a known oncogenic lncRNA that is overexpressed in plasma cells of MM patients and is a marker of poor prognosis. We found that CRISPR-mediated deletion of the CRNDE locus in MM cells decreases proliferation and adhesion properties, increases sensitivity to Dexamethasone and reduces tumour growth in an in vivo xenograft model. Transcriptomic profiling in CRNDE-deleted MM cells demonstrated that CRNDE activates expression of a number of genes previously implicated in the aetiology of MM, including IL6R. We further demonstrate that deletion of the CRNDE locus diminishes IL6 signalling and proliferative responses in MM cells. Altogether this study reveals the IL6 signalling pathway as a novel mechanism by which CRNDE impacts upon MM cell growth and disease progression.


Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Interleukin-6/metabolism , Multiple Myeloma/pathology , RNA, Long Noncoding/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Humans , Interleukin-6/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Acta Neuropathol ; 135(2): 267-283, 2018 02.
Article En | MEDLINE | ID: mdl-29149419

Although a growing body of evidence indicates that phenotypic plasticity exhibited by glioblastoma cells plays a central role in tumor development and post-therapy recurrence, the master drivers of their aggressiveness remain elusive. Here we mapped the changes in active (H3K4me3) and repressive (H3K27me3) histone modifications accompanying the repression of glioblastoma stem-like cells tumorigenicity. Genes with changing histone marks delineated a network of transcription factors related to cancerous behavior, stem state, and neural development, highlighting a previously unsuspected association between repression of ARNT2 and loss of cell tumorigenicity. Immunohistochemistry confirmed ARNT2 expression in cell sub-populations within proliferative zones of patients' glioblastoma. Decreased ARNT2 expression was consistently observed in non-tumorigenic glioblastoma cells, compared to tumorigenic cells. Moreover, ARNT2 expression correlated with a tumorigenic molecular signature at both the tissue level within the tumor core and at the single cell level in the patients' tumors. We found that ARNT2 knockdown decreased the expression of SOX9, POU3F2 and OLIG2, transcription factors implicated in glioblastoma cell tumorigenicity, and repressed glioblastoma stem-like cell tumorigenic properties in vivo. Our results reveal ARNT2 as a pivotal component of the glioblastoma cell tumorigenic signature, located at a node of a transcription factor network controlling glioblastoma cell aggressiveness.


Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Chromatin/metabolism , Glioblastoma/metabolism , Aged , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cells, Cultured , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/genetics , Glioblastoma/pathology , Histone Code , Homeodomain Proteins/metabolism , Humans , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oligodendrocyte Transcription Factor 2/metabolism , POU Domain Factors/metabolism , SOX9 Transcription Factor/metabolism
10.
Sci Rep ; 7(1): 5132, 2017 07 11.
Article En | MEDLINE | ID: mdl-28698620

Culture medium of mesenchymal stromal cells (MSCs) is usually supplemented with either human platelet lysate (HPL) or fetal calf serum (FCS). Many studies have demonstrated that proliferation and cellular morphology are affected by these supplements - it is therefore important to determine if they favor outgrowth of different subpopulations and thereby impact on the heterogeneous composition of MSCs. We have isolated and expanded human bone marrow-derived MSCs in parallel with HPL or FCS and demonstrated that HPL significantly increases proliferation and leads to dramatic differences in cellular morphology. Remarkably, global DNA-methylation profiles did not reveal any significant differences. Even at the transcriptomic level, there were only moderate changes in pairwise comparison. Furthermore, the effects on proliferation, cytoskeletal organization, and focal adhesions were reversible by interchanging to opposite culture conditions. These results indicate that cultivation of MSCs with HPL or FCS has no systematic bias for specific cell types.


Blood Platelets/chemistry , Culture Media/pharmacology , Mesenchymal Stem Cells/cytology , Serum/chemistry , Animals , Cattle , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Extracts/chemistry , Cell Extracts/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Culture Media/chemistry , DNA/metabolism , DNA Methylation/drug effects , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Humans , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/drug effects
11.
Stem Cell Reports ; 6(6): 970-984, 2016 06 14.
Article En | MEDLINE | ID: mdl-27304919

The capacity of hematopoietic stem cells (HSC) to generate B lymphocytes declines with age, contributing to impaired immune function in the elderly. Here we show that the histone methyltransferase SUV39H1 plays an important role in human B lymphoid differentiation and that expression of SUV39H1 decreases with age in both human and mouse HSC, leading to a global reduction in H3K9 trimethylation and perturbed heterochromatin function. Further, we demonstrate that SUV39H1 is a target of microRNA miR-125b, a known regulator of HSC function, and that expression of miR-125b increases with age in human HSC. Overexpression of miR-125b and inhibition of SUV39H1 in young HSC induced loss of B cell potential. Conversely, both inhibition of miR-125 and enforced expression of SUV39H1 improved the capacity of HSC from elderly individuals to generate B cells. Our findings highlight the importance of heterochromatin regulation in HSC aging and B lymphopoiesis.


Aging/immunology , B-Lymphocytes/cytology , Hematopoietic Stem Cells/cytology , Lymphopoiesis/immunology , Methyltransferases/genetics , MicroRNAs/genetics , Repressor Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Aging/genetics , Animals , Antagomirs/genetics , Antagomirs/metabolism , B-Lymphocytes/immunology , Base Sequence , Cell Differentiation , Female , Gene Expression Regulation , Hematopoietic Stem Cells/immunology , Heterochromatin/chemistry , Heterochromatin/metabolism , Humans , Lymphopoiesis/genetics , Male , Methyltransferases/immunology , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/immunology , Primary Cell Culture , Repressor Proteins/immunology , Signal Transduction
12.
FASEB J ; 29(2): 455-63, 2015 Feb.
Article En | MEDLINE | ID: mdl-25376832

Within the bone marrow, the endosteal niche plays a crucial role in B-cell differentiation. Because spaceflight is associated with osteoporosis, we investigated whether changes in bone microstructure induced by a ground-based model of spaceflight, hind limb unloading (HU), could affect B lymphopoiesis. To this end, we analyzed both bone parameters and the frequency of early hematopoietic precursors and cells of the B lineage after 3, 6, 13, and 21 d of HU. We found that limb disuse leads to a decrease in both bone microstructure and the frequency of B-cell progenitors in the bone marrow. Although multipotent hematopoietic progenitors were not affected by HU, a decrease in B lymphopoiesis was observed as of the common lymphoid progenitor (CLP) stage with a major block at the progenitor B (pro-B) to precursor B (pre-B) cell transition (5- to 10-fold decrease). The modifications in B lymphopoiesis were similar to those observed in aged mice and, as with aging, decreased B-cell generation in HU mice was associated with reduced expression of B-cell transcription factors, early B-cell factor (EBF) and Pax5, and an alteration in STAT5-mediated IL-7 signaling. These findings demonstrate that mechanical unloading of hind limbs results in a decrease in early B-cell differentiation resembling age-related modifications in B lymphopoiesis.


B-Lymphocytes/cytology , Hindlimb Suspension/physiology , Lymphopoiesis/physiology , Space Flight , Adrenal Cortex Hormones/metabolism , Aging , Animals , Bone Marrow Cells/cytology , Bone Remodeling , Cell Differentiation , Cell Lineage , Cytokines/metabolism , Hematopoietic Stem Cells/cytology , Immunoglobulins/metabolism , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , PAX5 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Stem Cells , Time Factors , Trans-Activators/metabolism , X-Ray Microtomography
13.
J Cell Physiol ; 230(6): 1342-51, 2015 Jun.
Article En | MEDLINE | ID: mdl-25502698

Matrix proteins of the SIBLING family interact with bone cells, extracellular matrix and mineral and are thus in a key position to regulate the microenvironment of the bone tissue, including its hematopoietic component. In this respect, osteopontin (OPN) has been implicated in the hematopoietic stem cell (HSC) niche as negative regulator of the HSC function. We investigated the impact on hematopoietic regulation of the absence of the cognate bone sialoprotein (BSP). BSP knockout (-/-) mice display increased bone marrow cellularity, and an altered commitment of hematopoietic precursors to myeloid lineages, leading in particular to an increased frequency of monocyte/macrophage cells. The B cell pool is increased in -/- bone marrow, and its composition is shifted toward more mature lymphocyte stages. BSP-null mice display a decreased HSC fraction among LSK cells and a higher percentage of more committed progenitors as compared to +/+. The fraction of proliferating LSK progenitors is higher in -/- mice, and after PTH treatment the mutant HSC pool is lower than in +/+. Strikingly, circulating levels of OPN as well as its expression in the bone tissue are much higher in the -/-. Thus, a BSP-null bone microenvironment affects the hematopoietic system both quantitatively and qualitatively, in a manner in part opposite to the OPN knockout, suggesting that the effects might in part reflect the higher OPN expression in the absence of BSP.


Bone Marrow/metabolism , Hematopoiesis/physiology , Integrin-Binding Sialoprotein/deficiency , Integrin-Binding Sialoprotein/metabolism , Osteopontin/metabolism , Animals , Bone and Bones/metabolism , Hematopoiesis/genetics , Mice , Mice, Nude , Osteogenesis/physiology , Transcriptional Activation , Up-Regulation
14.
Mol Cancer Res ; 11(7): 724-35, 2013 Jul.
Article En | MEDLINE | ID: mdl-23604035

The death domain containing TNF receptor 6 (CD95/Fas) is a direct target for the NF-κB transcription factor and is repressed in solid tumors such as colon carcinomas. Previously, we reported that the Fas death receptor, while overexpressed in low-risk myelodysplastic syndromes (MDS), becomes undetectable on CD34(+) progenitors when the disease progresses to secondary acute myeloid leukemia (AML). This study determined the interplay between NF-κB and Fas during MDS progression. We first observed that Fas was induced by TNF-α in the HL60 cell line. In these cells, p65 (RELA) was associated with the FAS promoter, and inhibition of the NF-κB pathway by an IKKα inhibitor (BAY11-7082) or lentiviral expression of a nondegradable mutant of IκBα (IκSR) blocked Fas expression. In contrast, TNF-α failed to induce Fas expression in the colon carcinoma cell line SW480, due to hypermethylation of the FAS promoter. Azacitidine rescued p65 binding on FAS promoter in vitro, and subsequently Fas expression in SW480 cells. Furthermore, inhibition of the NF-κB pathway decreased the expression of Fas in MDS CD45(lo)CD34(+) bone marrow cells. However, despite the nuclear expression of p65, Fas was often low on CD45(lo)CD34(+) AML cells. TNF-α failed to stimulate its expression, while azacitidine efficiently rescued p65 binding and Fas reexpression. Overall, these data suggest that DNA methylation at NF-κB sites is responsible for FAS gene silencing.


Disease Progression , Epigenesis, Genetic , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , NF-kappa B/genetics , Transcription, Genetic , fas Receptor/genetics , Azacitidine/pharmacology , Base Sequence , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Line, Tumor , DNA Methylation/drug effects , DNA Methylation/genetics , Epigenesis, Genetic/drug effects , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Nitriles/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , Sulfones/pharmacology , Transcription, Genetic/drug effects , fas Receptor/metabolism
15.
Blood ; 118(7): 1784-96, 2011 Aug 18.
Article En | MEDLINE | ID: mdl-21715312

The mechanisms regulating the emergence of BM prothymocytes remain poorly characterized. Genome-wide transcriptome analyses looking for genes expressed in human prothymocytes led to the identification of AF1q/MLLT11 as a candidate gene conceivably involved in this process. Analysis of AF1q protein subcellular localization and intracellular trafficking showed that despite pronounced karyophily, it was subjected to constitutive nuclear export followed by ubiquitin-mediated degradation in the centrosomal area. Using in vitro assays based on either forced expression or shRNA-mediated silencing of AF1q, we provide evidence that the protein promotes T- over B-cell differentiation in multipotent hematopoietic progenitors. At the molecular level, AF1q confers to multipotent progenitors an increased susceptibility to Delta-like/Notch-mediated signaling. Consistent with these findings, enforced AF1q expression in humanized mice fosters the emergence of BM CD34(+)CD7(+) prothymocytes, enhances subsequent thymus colonization, and accelerates intrathymic T-cell development. In contrast, AF1q silencing provokes a global shift of BM lymphopoiesis toward the B-cell lineage, hinders prothymocyte development, inhibits thymus colonization, and leads to intrathymic accumulation of B cells. Our results indicate that AF1q cooperates with the Notch signaling pathway to foster the emergence of BM prothymocytes and drive subsequent intrathymic specification toward the T-cell lineage.


Hematopoietic Stem Cells/cytology , Lymphopoiesis , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , T-Lymphocytes/cytology , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cells, Cultured , Gene Silencing , HeLa Cells , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, SCID , Molecular Sequence Data , Neoplasm Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/genetics , Sequence Alignment , Signal Transduction , T-Lymphocytes/metabolism
16.
Aging Cell ; 10(3): 542-6, 2011 Jun.
Article En | MEDLINE | ID: mdl-21418508

Adult stem cells are critical for maintaining cellular homeostasis throughout life, yet the effects of age on their regenerative capacity are poorly understood. All lymphoid and myeloid blood cell lineages are continuously generated from hematopoietic stem cells present in human bone marrow. With age, significant changes in the function and composition of mature blood cells are observed. In this study, we report that age-related changes also occur in the human hematopoietic stem cell compartment. We find that the proportion of multipotent CD34(+) CD38(-) cells increases in the bone marrow of elderly (>70 years) individuals. CD34(+) CD38(+) CD90(-) CD45RA(+/-) CD10(-) and CD34(+) CD33(+) myeloid progenitors persist at the same level in the bone marrow, while the frequency of early CD34(+) CD38(+) CD90(-) CD45RA(+) CD10(+) and committed CD34(+) CD19(+) B-lymphoid progenitors decreases with age. In contrast to mice models of aging, transplantation experiments with immunodeficient NOD/SCID/IL-2Rγ null (NSG) mice showed that the frequency of NSG repopulating cells does not change significantly with age, and there is a decrease in myeloid lineage reconstitution. An age-related decrease in the capacity of CD34(+) cells to generate myeloid cells was also seen in colony-forming assays in vitro. Thus, with increasing age, human hematopoietic stem/progenitor cells undergo quantitative changes as well as functional modifications.


Aging , Antigens, CD/analysis , Bone Marrow Cells/cytology , Bone Marrow/physiology , Hematopoietic Stem Cells/cytology , Myeloid Cells/cytology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Cell Count , Cell Differentiation , Cell Lineage , Colony-Forming Units Assay , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Humans , Male , Mice , Mice, SCID , Myeloid Cells/immunology
17.
Aging Cell ; 9(3): 410-9, 2010 Jun.
Article En | MEDLINE | ID: mdl-20331442

Aging is accompanied by a reduction in the generation of B lymphocytes leading to impaired immune responses. In this study, we have investigated whether the decline in B lymphopoiesis is due to age-related defects in the hematopoietic stem cell compartment. The ability of hematopoietic stem cells from old mice to generate B cells, as measured in vitro, is decreased 2-5-fold, while myeloid potential remains unchanged. This age-related decrease in B-cell potential is more marked in common lymphoid progenitors (CLP) and was associated with reduced expression of the B-lineage specifying factors, EBF and Pax5. Notably, retrovirus-mediated expression of EBF complemented the age-related loss of B-cell potential in CLP isolated from old mice. Furthermore, transduction of CLP from old mice with a constitutively active form of STAT5 restored both EBF and Pax5 expression and increased B-cell potential. These results are consistent with a mechanism, whereby reduced expression of EBF with age decreases the frequency with which multipotent hematopoietic progenitors commit to a B-cell fate, without altering their potential to generate myeloid cells.


Aging , B-Lymphocytes/metabolism , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Trans-Activators/metabolism , Animals , B-Lymphocytes/cytology , Cell Lineage , Down-Regulation , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Trans-Activators/genetics
18.
Stem Cells ; 27(1): 108-15, 2009 Jan.
Article En | MEDLINE | ID: mdl-18974210

To further clarify the contribution of nuclear architecture in the regulation of gene expression patterns during differentiation of human multipotent cells, we analyzed expression status, histone modifications, and subnuclear positioning relative to repressive compartments, of hematopoietic loci in multipotent and lineage-committed primary human hematopoietic progenitors. We report here that positioning of lineage-affiliated loci relative to pericentromeric heterochromatin compartments (PCH) is identical in multipotent cells from various origins and is unchanged between multipotent and lineage-committed hematopoietic progenitors. However, during differentiation of multipotent hematopoietic progenitors, changes in gene expression and histone modifications at these loci occur in committed progenitors, prior to changes in gene positioning relative to pericentromeric heterochromatin compartments, detected at later stages in precursor and mature cells. Therefore, during normal human hematopoietic differentiation, changes in gene subnuclear location relative to pericentromeric heterochromatin appear to be dictated by whether the gene will be permanently silenced or activated, rather than being predictive of commitment toward a given lineage.


Cell Compartmentation , Chromatin/metabolism , Gene Order , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Acetylation , Animals , Cell Differentiation/genetics , Cell Lineage , Erythroid Cells/metabolism , Globins/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Histones/metabolism , Humans , Immunoglobulin kappa-Chains/genetics , Infant , Mice , Multipotent Stem Cells/cytology
19.
Blood ; 112(7): 2722-9, 2008 Oct 01.
Article En | MEDLINE | ID: mdl-18625888

To address the role of chromatin structure in the establishment of hematopoietic stem cell (HSC) multilineage potential and commitment to the lymphoid lineage, we have analyzed histone modifications at a panel of lymphoid- and myeloid-affiliated genes in multipotent and lineage-committed hematopoietic cells isolated from human cord blood. Our results show that many B- and T-lymphoid genes, although silent in HSCs, are associated with acetylated histones H3 and H4. We also detected histone H3 lysine 4 methylation but not repressive lysine 9 or 27 methylation marks at these loci, indicative of an open chromatin structure. Interestingly, the relative level of H3 lysine 4 dimethylation to trimethylation at B-specific loci was high in multipotent CD34(+)CD38(lo) progenitors and decreased as they become actively transcribed in B-lineage cells. In vitro differentiation of CD34(+) cells toward the erythroid, granulocyte, and T-cell lineages resulted in a loss of histone acetylation at nonlineage-associated genes. This study provides evidence that histone modifications involved in chromatin decondensation are already in place at lymphoid-specific genes in primary human HSCs, supporting the idea that these genes are "primed" for expression before lineage commitment. This permissive chromatin structure is progressively lost as the stem cell differentiates.


Genes , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Lymphocytes/metabolism , ADP-ribosyl Cyclase 1/metabolism , Acetylation , Antigens, CD19/metabolism , Antigens, CD34/metabolism , Cell Differentiation , Cell Lineage , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Humans , Lymphocytes/cytology , Myeloid Cells/cytology , Myeloid Cells/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism
20.
J Immunol ; 176(9): 5409-17, 2006 May 01.
Article En | MEDLINE | ID: mdl-16622008

IgH genes are assembled during early B cell development by a series of regulated DNA recombination reactions in which DH and JH segments are first joined followed by V(H) to DJH rearrangement. Recent studies have highlighted the role of chromatin structure in the control of V(D)J recombination. In this study, we show that, in murine pro-B cell precursors, the JH segments are located within a 6-kb DNase I-sensitive chromatin domain containing acetylated histones H3 and H4, which is delimited 5' by the DQ52 promoter element and 3' by the intronic enhancer. Within this domain, the JH segments are covered by phased nucleosomes. High-resolution mapping of nucleosomes reveals that, in pro-B cells, unlike recombination refractory nonlymphoid cells, the recombination signal sequences flanking the four JH segments are located in regions of enhanced micrococcal nuclease and restriction enzyme accessibility, corresponding to either nucleosome-free regions or DNA rendered accessible within a nucleosome. These results support the idea that nucleosome remodeling provides an additional level of control in the regulation of Ig locus accessibility to recombination factors in B cell precursors.


Chromatin/genetics , DNA, Recombinant/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Joining Region/genetics , Nucleosomes/genetics , Acetylation , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Gene Rearrangement , Histones/metabolism , Mice , Mice, Knockout , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics
...