Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3075, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594286

ABSTRACT

Immune checkpoint blockade (ICB) has improved outcome for patients with metastatic melanoma but not all benefit from treatment. Several immune- and tumor intrinsic features are associated with clinical response at baseline. However, we need to further understand the molecular changes occurring during development of ICB resistance. Here, we collect biopsies from a cohort of 44 patients with melanoma after progression on anti-CTLA4 or anti-PD1 monotherapy. Genetic alterations of antigen presentation and interferon gamma signaling pathways are observed in approximately 25% of ICB resistant cases. Anti-CTLA4 resistant lesions have a sustained immune response, including immune-regulatory features, as suggested by multiplex spatial and T cell receptor (TCR) clonality analyses. One anti-PD1 resistant lesion harbors a distinct immune cell niche, however, anti-PD1 resistant tumors are generally immune poor with non-expanded TCR clones. Such immune poor microenvironments are associated with melanoma cells having a de-differentiated phenotype lacking expression of MHC-I molecules. In addition, anti-PD1 resistant tumors have reduced fractions of PD1+ CD8+ T cells as compared to ICB naïve metastases. Collectively, these data show the complexity of ICB resistance and highlight differences between anti-CTLA4 and anti-PD1 resistance that may underlie differential clinical outcomes of therapy sequence and combination.


Subject(s)
Melanoma , Humans , Melanoma/drug therapy , Melanoma/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Receptors, Antigen, T-Cell , Tumor Microenvironment
2.
Front Immunol ; 14: 1130930, 2023.
Article in English | MEDLINE | ID: mdl-37138883

ABSTRACT

The LIN28B RNA binding protein exhibits an ontogenically restricted expression pattern and is a key molecular regulator of fetal and neonatal B lymphopoiesis. It enhances the positive selection of CD5+ immature B cells early in life through amplifying the CD19/PI3K/c-MYC pathway and is sufficient to reinitiate self-reactive B-1a cell output when ectopically expressed in the adult. In this study, interactome analysis in primary B cell precursors showed direct binding by LIN28B to numerous ribosomal protein transcripts, consistent with a regulatory role in cellular protein synthesis. Induction of LIN28B expression in the adult setting is sufficient to promote enhanced protein synthesis during the small Pre-B and immature B cell stages, but not during the Pro-B cell stage. This stage dependent effect was dictated by IL-7 mediated signaling, which masked the impact of LIN28B through an overpowering stimulation on the c-MYC/protein synthesis axis in Pro-B cells. Importantly, elevated protein synthesis was a distinguishing feature between neonatal and adult B cell development that was critically supported by endogenous Lin28b expression early in life. Finally, we used a ribosomal hypomorphic mouse model to demonstrate that subdued protein synthesis is specifically detrimental for neonatal B lymphopoiesis and the output of B-1a cells, without affecting B cell development in the adult. Taken together, we identify elevated protein synthesis as a defining requirement for early-life B cell development that critically depends on Lin28b. Our findings offer new mechanistic insights into the layered formation of the complex adult B cell repertoire.


Subject(s)
B-Lymphocytes , Precursor Cells, B-Lymphoid , Mice , Animals
3.
Cell Rep ; 42(2): 112099, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36763502

ABSTRACT

MLL-rearrangements (MLL-r) are recurrent genetic events in acute myeloid leukemia (AML) and frequently associate with poor prognosis. In infants, MLL-r can be sufficient to drive transformation. However, despite the prenatal origin of MLL-r in these patients, congenital leukemia is very rare with transformation usually occurring postnatally. The influence of prenatal signals on leukemogenesis, such as those mediated by the fetal-specific protein LIN28B, remains controversial. Here, using a dual-transgenic mouse model that co-expresses MLL-ENL and LIN28B, we investigate the impact of LIN28B on AML. LIN28B impedes the progression of MLL-r AML through compromised leukemia-initiating cell activity and suppression of MYB signaling. Mechanistically, LIN28B directly binds to MYBBP1A mRNA, resulting in elevated protein levels of this MYB co-repressor. Functionally, overexpression of MYBBP1A phenocopies the tumor-suppressor effects of LIN28B, while its perturbation omits it. Thereby, we propose that developmentally restricted expression of LIN28B provides a layer of protection against MYB-dependent AML.


Subject(s)
Leukemia, Myeloid, Acute , Myeloid-Lymphoid Leukemia Protein , Humans , Mice , Animals , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Gene Rearrangement , Mice, Transgenic , Cell Transformation, Neoplastic/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , RNA-Binding Proteins/genetics
4.
Immunity ; 55(10): 1829-1842.e6, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36115337

ABSTRACT

The adult immune system consists of cells that emerged at various times during ontogeny. We aimed to define the relationship between developmental origin and composition of the adult B cell pool during unperturbed hematopoiesis. Lineage tracing stratified murine adult B cells based on the timing of output, revealing that a substantial portion originated within a restricted neonatal window. In addition to B-1a cells, early-life time-stamped B cells included clonally interrelated IgA plasma cells in the gut and bone marrow. These were actively maintained by B cell memory within gut chronic germinal centers and contained commensal microbiota reactivity. Neonatal rotavirus infection recruited recurrent IgA clones that were distinct from those arising by infection with the same antigen in adults. Finally, gut IgA plasma cells arose from the same hematopoietic progenitors as B-1a cells during ontogeny. Thus, a complex layer of neonatally imprinted B cells confer unique antibody responses later in life.


Subject(s)
Immunoglobulin A , Microbiota , Animals , B-Lymphocytes , Germinal Center , Mice , Plasma Cells
5.
Blood Adv ; 6(24): 6228-6241, 2022 12 27.
Article in English | MEDLINE | ID: mdl-35584393

ABSTRACT

The fetal-to-adult switch in hematopoietic stem cell (HSC) behavior is characterized by alterations in lineage output and entry into deep quiescence. Here we identify the emergence of megakaryocyte (Mk)-biased HSCs as an event coinciding with this developmental switch. Single-cell chromatin accessibility analysis reveals a ubiquitous acquisition of Mk lineage priming signatures in HSCs during the fetal-to-adult transition. These molecular changes functionally coincide with increased amplitude of early Mk differentiation events after acute inflammatory insult. Importantly, we identify LIN28B, known for its role in promoting fetal-like self-renewal, as an insulator against the establishment of an Mk-biased HSC pool. LIN28B protein is developmentally silenced in the third week of life, and its prolonged expression delays emergency platelet output in young adult mice. We propose that developmental regulation of Mk priming may represent a switch for HSCs to toggle between prioritizing self-renewal in the fetus and increased host protection in postnatal life.


Subject(s)
Cues , Megakaryocytes , Animals , Mice , Megakaryocytes/metabolism , Hematopoietic Stem Cells/metabolism , Blood Platelets/metabolism , Hematopoiesis
6.
Immunol Rev ; 300(1): 194-202, 2021 03.
Article in English | MEDLINE | ID: mdl-33501672

ABSTRACT

The autoimmune checkpoint during B cell maturation eliminates self-antigen reactive specificities from the mature B cell repertoire. However, an exception to this rule is illustrated by B-1 cells, an innate-like self-reactive B cell subset that is positively selected into the mature B cell pool in a self-antigen-driven fashion. The mechanisms by which B-1 cells escape central tolerance have puzzled the field for decades. A key clue comes from their restricted developmental window during fetal and neonatal life. Here we use B-1 cells as a prototypic early life derived B cell subset to explore developmental changes in the constraints of B cell selection. We discuss recent advancements in the understanding of the molecular program, centered around the RNA binding protein Lin28b, that licenses self-reactive B-1 cell output during ontogeny. Finally, we speculate on the possible link between the unique rules of early life B cell tolerance and the establishment of B cell - microbial mutualism to propose an integrated model for how developmental and environmental cues come together to create a protective layer of B cell memory involved in neonatal immune imprinting.


Subject(s)
B-Lymphocyte Subsets , Antibody Specificity , Autoantigens , B-Lymphocytes , Immune Tolerance
7.
Home Healthc Now ; 38(5): 238, 2020.
Article in English | MEDLINE | ID: mdl-32889990
8.
Immunity ; 53(1): 11-13, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32668222

ABSTRACT

Group A Streptococcus is a common pathogen that elicits a protective humoral response against the cell wall component GlcNAc. In this issue of Immunity, New et al. demonstrate the ability of long-lived B-1 cells to be programmed by microbial colonization and early life immunization to uniquely incorporate GlcNAc reactivity in mice, establishing their critical role in mediating neonatal immune imprinting.


Subject(s)
B-Lymphocyte Subsets , Animals , B-Lymphocyte Subsets/immunology , Bacteria , Immunization , Mice , Polysaccharides , Vaccination
9.
Sci Immunol ; 4(39)2019 09 27.
Article in English | MEDLINE | ID: mdl-31562190

ABSTRACT

The ability of B-1 cells to become positively selected into the mature B cell pool, despite being weakly self-reactive, has puzzled the field since its initial discovery. Here, we explore changes in B cell positive selection as a function of developmental time by exploiting a link between CD5 surface levels and the natural occurrence of self-reactive B cell receptors (BCRs) in BCR wild-type mice. We show that the heterochronic RNA binding protein Lin28b potentiates a neonatal mode of B cell selection characterized by enhanced overall positive selection in general and the developmental progression of CD5+ immature B cells in particular. Lin28b achieves this by amplifying the CD19/PI3K/c-Myc positive feedback loop, and ectopic Lin28b expression restores both positive selection and mature B cell numbers in CD19-/- adult mice. Thus, the temporally restricted expression of Lin28b relaxes the rules for B cell selection during ontogeny by modulating tonic signaling. We propose that this neonatal mode of B cell selection represents a cell-intrinsic cue to accelerate the de novo establishment of the adaptive immune system and incorporate a layer of natural antibody-mediated immunity throughout life.


Subject(s)
B-Lymphocytes/immunology , RNA-Binding Proteins/immunology , Animals , Mice , Mice, Knockout
10.
Elife ; 72018 12 18.
Article in English | MEDLINE | ID: mdl-30561324

ABSTRACT

A hallmark of adult hematopoiesis is the continuous replacement of blood cells with limited lifespans. While active hematopoietic stem cell (HSC) contribution to multilineage hematopoiesis is the foundation of clinical HSC transplantation, recent reports have questioned the physiological contribution of HSCs to normal/steady-state adult hematopoiesis. Here, we use inducible lineage tracing from genetically marked adult HSCs and reveal robust HSC-derived multilineage hematopoiesis. This commences via defined progenitor cells, but varies substantially in between different hematopoietic lineages. By contrast, adult HSC contribution to hematopoietic cells with proposed fetal origins is neglible. Finally, we establish that the HSC contribution to multilineage hematopoiesis declines with increasing age. Therefore, while HSCs are active contributors to native adult hematopoiesis, it appears that the numerical increase of HSCs is a physiologically relevant compensatory mechanism to account for their reduced differentiation capacity with age.


Subject(s)
Aging/physiology , Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/physiology , Age Factors , Animals , Cell Lineage , Mice , Staining and Labeling
11.
Br J Haematol ; 183(4): 588-600, 2018 11.
Article in English | MEDLINE | ID: mdl-30596405

ABSTRACT

Given that FLT3 expression is highly restricted on lymphoid progenitors, it is possible that the established role of FLT3 in the regulation of B and T lymphopoiesis reflects its high expression and role in regulation of lymphoid-primed multipotent progenitors (LMPPs) or common lymphoid progenitors (CLPs). We generated a Flt3 conditional knock-out (Flt3fl/fl) mouse model to address the direct role of FLT3 in regulation of lymphoid-restricted progenitors, subsequent to turning on Rag1 expression, as well as potentially ontogeny-specific roles in B and T lymphopoiesis. Our studies establish a prominent and direct role of FLT3, independently of the established role of FLT3 in regulation of LMPPs and CLPs, in regulation of fetal as well as adult early B cell progenitors, and the early thymic progenitors (ETPs) in adult mice but not in the fetus. Our findings highlight the potential benefit of targeting poor prognosis acute B-cell progenitor leukaemia and ETP leukaemia with recurrent FLT3 mutations using clinical FLT3 inhibitors.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Bone Marrow Cells/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lymphoid Progenitor Cells/pathology , Mice , Mice, Knockout , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thymus Gland/metabolism , Thymus Gland/pathology , fms-Like Tyrosine Kinase 3/genetics
12.
Curr Opin Immunol ; 51: 7-13, 2018 04.
Article in English | MEDLINE | ID: mdl-29272734

ABSTRACT

The adult adaptive immune system is comprised of a wide spectrum of lymphocyte subsets with distinct antigen receptor repertoire profiles, effector functions, turnover times and anatomical locations, acting in concert to provide optimal host protection and self-regulation. While some lymphocyte populations are replenished by bone marrow hematopoietic stem cells (HSCs) through adulthood, others emerge during a limited window of time during fetal and postnatal life and sustain through self-replenishment. Despite fundamental implications in immune regeneration, early life immunity and leukemogenesis, the impact of developmental timing on lymphocyte output remains an under explored frontier in immunology. In this review, we spotlight recent insights into the developmental changes in B cell output in mice and explore how several age specific cellular and molecular factors may shape the formation of a diverse adaptive immune system.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Differentiation , Age Factors , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Biomarkers , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement , Clonal Selection, Antigen-Mediated/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Phenotype , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
13.
Cell Rep ; 21(11): 3285-3297, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241553

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) in the fetus and adult possess distinct molecular landscapes that regulate cell fate and change their susceptibility to initiation and progression of hematopoietic malignancies. Here, we applied in-depth quantitative proteomics to comprehensively describe and compare the proteome of fetal and adult HSPCs. Our data uncover a striking difference in complexity of the cellular proteomes, with more diverse adult-specific HSPC proteomic signatures. The differential protein content in fetal and adult HSPCs indicate distinct metabolic profiles and protein complex stoichiometries. Additionally, adult characteristics include an arsenal of proteins linked to viral and bacterial defense, as well as protection against ROS-induced protein oxidation. Further analyses show that interferon α, as well as Neutrophil elastase, has distinct functional effects in fetal and adult HSPCs. This study provides a rich resource aimed toward an enhanced mechanistic understanding of normal and malignant hematopoiesis during fetal and adult life.


Subject(s)
Aging/genetics , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Proteome/genetics , Age Factors , Aging/metabolism , Animals , Cell Differentiation , Cell Proliferation , Embryo, Mammalian , Fetus , Gene Expression Profiling , Gene Regulatory Networks , Hematopoietic Stem Cells/cytology , Interferon-alpha/genetics , Interferon-alpha/metabolism , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Mice, Inbred C57BL , Proteome/metabolism , Proteomics/methods
14.
Proc Natl Acad Sci U S A ; 114(44): E9328-E9337, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29078319

ABSTRACT

B cell receptor signaling and downstream NF-κB activity are crucial for the maturation and functionality of all major B cell subsets, yet the molecular players in these signaling events are not fully understood. Here we use several genetically modified mouse models to demonstrate that expression of the multifunctional BRCT (BRCA1 C-terminal) domain-containing PTIP (Pax transactivation domain-interacting protein) chromatin regulator is controlled by B cell activation and potentiates steady-state and postimmune antibody production in vivo. By examining the effects of PTIP deficiency in mice at various ages during ontogeny, we demonstrate that PTIP promotes bone marrow B cell development as well as the neonatal establishment and subsequent long-term maintenance of self-reactive B-1 B cells. Furthermore, we find that PTIP is required for B cell receptor- and T:B interaction-induced proliferation, differentiation of follicular B cells during germinal center formation, and normal signaling through the classical NF-κB pathway. Together with the previously identified role for PTIP in promoting sterile transcription at the Igh locus, the present results establish PTIP as a licensing factor for humoral immunity that acts at several junctures of B lineage maturation and effector cell differentiation by controlling B cell activation.


Subject(s)
B-Lymphocyte Subsets/immunology , Carrier Proteins/immunology , Chromatin/immunology , Immunity, Humoral/immunology , Nuclear Proteins/immunology , Animals , Bone Marrow/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Cell Proliferation/physiology , Cells, Cultured , DNA-Binding Proteins , Lymphocyte Activation/immunology , Mice , NF-kappa B/immunology , Signal Transduction/immunology
15.
Nat Commun ; 8: 14533, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28224997

ABSTRACT

Ageing associates with significant alterations in somatic/adult stem cells and therapies to counteract these might have profound benefits for health. In the blood, haematopoietic stem cell (HSC) ageing is linked to several functional shortcomings. However, besides the recent realization that individual HSCs might be preset differentially already from young age, HSCs might also age asynchronously. Evaluating the prospects for HSC rejuvenation therefore ultimately requires approaching those HSCs that are functionally affected by age. Here we combine genetic barcoding of aged murine HSCs with the generation of induced pluripotent stem (iPS) cells. This allows us to specifically focus on aged HSCs presenting with a pronounced lineage skewing, a hallmark of HSC ageing. Functional and molecular evaluations reveal haematopoiesis from these iPS clones to be indistinguishable from that associating with young mice. Our data thereby provide direct support to the notion that several key functional attributes of HSC ageing can be reversed.


Subject(s)
Aging/physiology , Cell Lineage , Cellular Senescence , Clone Cells/cytology , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Lineage/genetics , Cellular Reprogramming/genetics , Cellular Senescence/genetics , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice, Inbred C57BL , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
16.
Bio Protoc ; 7(8): e2242, 2017 Apr 20.
Article in English | MEDLINE | ID: mdl-34541235

ABSTRACT

Cellular barcoding enables the dissection of clonal dynamics in heterogeneous cell populations through single cell lineage tracing. The labeling of hematopoietic stem and progenitor cells (HSPCs) with unique and heritable DNA barcodes, makes it possible to resolve donor cell heterogeneity in terms of differentiation potential and lineage bias at the single cell level, through subsequent transplantation and high-throughput sequencing. Furthermore, cellular barcoding allows for bona fide hematopoietic stem cells (HSCs) to be defined based on functional rather than immunophenotypic parameters. This protocol describes the work flow of lentiviral cellular barcoding, tracking 14.5 days post coitum (d.p.c.) fetal liver (FL) Lineage-Sca+cKit+ (LSK) HSPCs following long-term reconstitution (Figure 1) ( Kristiansen et al., 2016 ), but can be adapted to the cell type or time frame of choice. Figure 1.Summary of experimental workflow ( Naik et al., 2013 ).

17.
Cell Stem Cell ; 19(6): 673-674, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27912084

ABSTRACT

Blood development relies on discrete stem and progenitor cell populations with unclear lineage relationships and distinct functional characteristics that change during ontogeny. In this issue of Cell Stem Cell, Beaudin et al. (2016) identify a hematopoietic stem cell population with fetal characteristics that is developmentally restricted yet capable of long-term multi-lineage reconstitution upon transplantation into adult recipients.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cell Transplantation , Fetal Blood , Fetus , Hematopoietic Stem Cells
18.
Immunity ; 45(2): 346-57, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27533015

ABSTRACT

Hematopoietic stem cells (HSCs) undergo a functional switch in neonatal mice hallmarked by a decrease in self-renewing divisions and entry into quiescence. Here, we investigated whether the developmental attenuation of B-1a cell output is a consequence of a shift in stem cell state during ontogeny. Using cellular barcoding for in vivo single-cell fate analyses, we found that fetal liver definitive HSCs gave rise to both B-1a and B-2 cells. Whereas B-1a potential diminished in all HSCs with time, B-2 output was maintained. B-1a and B-2 plasticity could be reinitiated in a subset of adult HSCs by ectopic expression of the RNA binding protein LIN28B, a key regulator of fetal hematopoiesis, and this coincided with the clonal reversal to fetal-like elevated self-renewal and repopulation potential. These results anchor the attenuation of B-1a cell output to fetal HSC behavior and demonstrate that the developmental decline in regenerative potential represents a reversible HSC state.


Subject(s)
B-Lymphocytes/physiology , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/physiology , Liver/physiology , Lymphocyte Subsets/physiology , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Plasticity , Cell Self Renewal , Clone Cells , DNA-Binding Proteins/genetics , Female , Hematopoiesis/genetics , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA-Binding Proteins , Single-Cell Analysis
19.
Genes Dev ; 30(2): 149-63, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26744420

ABSTRACT

Class switch recombination (CSR) diversifies antibodies for productive immune responses while maintaining stability of the B-cell genome. Transcription at the immunoglobulin heavy chain (Igh) locus targets CSR-associated DNA damage and is promoted by the BRCT domain-containing PTIP (Pax transactivation domain-interacting protein). Although PTIP is a unique component of the mixed-lineage leukemia 3 (MLL3)/MLL4 chromatin-modifying complex, the mechanisms for how PTIP promotes transcription remain unclear. Here we dissected the minimal structural requirements of PTIP and its different protein complexes using quantitative proteomics in primary lymphocytes. We found that PTIP functions in transcription and CSR separately from its association with the MLL3/MLL4 complex and from its localization to sites of DNA damage. We identified a tandem BRCT domain of PTIP that is sufficient for CSR and identified PA1 as its main functional protein partner. Collectively, we provide genetic and biochemical evidence that a PTIP-PA1 subcomplex functions independently from the MLL3/MLL4 complex to mediate transcription during CSR. These results further our understanding of how multifunctional chromatin-modifying complexes are organized by subcomplexes that harbor unique and distinct activities.


Subject(s)
Carrier Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Immunoglobulin Class Switching/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Nuclear Proteins/metabolism , DNA Damage , DNA-Binding Proteins , Gene Expression Regulation/immunology , Molecular Structure , Protein Structure, Tertiary , Protein Transport
20.
Blood ; 122(6): 1034-41, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23798711

ABSTRACT

Reactivation of fetal hemoglobin (HbF) holds therapeutic potential for sickle cell disease and ß-thalassemias. In human erythroid cells and hematopoietic organs, LIN28B and its targeted let-7 microRNA family, demonstrate regulated expression during the fetal-to-adult developmental transition. To explore the effects of LIN28B in human erythroid cell development, lentiviral transduction was used to knockdown LIN28B expression in erythroblasts cultured from human umbilical cord CD34+ cells. The subsequent reduction in LIN28B expression caused increased expression of let-7 and significantly reduced HbF expression. Conversely, LIN28B overexpression in cultured adult erythroblasts reduced the expression of let-7 and significantly increased HbF expression. Cellular maturation was maintained including enucleation. LIN28B expression in adult erythroblasts increased the expression of γ-globin, and the HbF content of the cells rose to levels >30% of their hemoglobin. Expression of carbonic anhydrase I, glucosaminyl (N-acetyl) transferase 2, and miR-96 (three additional genes marking the transition from fetal-to-adult erythropoiesis) were reduced by LIN28B expression. The transcription factor BCL11A, a well-characterized repressor of γ-globin expression, was significantly down-regulated. Independent of LIN28B, experimental suppression of let-7 also reduced BCL11A expression and significantly increased HbF expression. LIN28B expression regulates HbF levels and causes adult human erythroblasts to differentiate with a more fetal-like phenotype.


Subject(s)
DNA-Binding Proteins/metabolism , Erythroblasts/cytology , Erythrocytes/cytology , Fetal Hemoglobin/metabolism , Gene Expression Regulation , Antigens, CD34/metabolism , Carbonic Anhydrase I/metabolism , Cell Culture Techniques , Fetal Blood/cytology , Hemoglobin A/metabolism , Humans , MicroRNAs/metabolism , N-Acetylglucosaminyltransferases/metabolism , Phenotype , RNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL