Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Blood ; 143(19): 1980-1991, 2024 May 09.
Article En | MEDLINE | ID: mdl-38364109

ABSTRACT: The switch from fetal hemoglobin (γ-globin, HBG) to adult hemoglobin (ß-globin, HBB) gene transcription in erythroid cells serves as a paradigm for a complex and clinically relevant developmental gene regulatory program. We previously identified HIC2 as a regulator of the switch by inhibiting the transcription of BCL11A, a key repressor of HBG production. HIC2 is highly expressed in fetal cells, but the mechanism of its regulation is unclear. Here we report that HIC2 developmental expression is controlled by microRNAs (miRNAs), as loss of global miRNA biogenesis through DICER1 depletion leads to upregulation of HIC2 and HBG messenger RNA. We identified the adult-expressed let-7 miRNA family as a direct posttranscriptional regulator of HIC2. Ectopic expression of let-7 in fetal cells lowered HIC2 levels, whereas inhibition of let-7 in adult erythroblasts increased HIC2 production, culminating in decommissioning of a BCL11A erythroid enhancer and reduced BCL11A transcription. HIC2 depletion in let-7-inhibited cells restored BCL11A-mediated repression of HBG. Together, these data establish that fetal hemoglobin silencing in adult erythroid cells is under the control of a miRNA-mediated inhibitory pathway (let-7 ⊣ HIC2 ⊣ BCL11A ⊣ HBG).


Fetal Hemoglobin , Kruppel-Like Transcription Factors , MicroRNAs , Repressor Proteins , Humans , beta-Globins/genetics , beta-Globins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Erythroblasts/metabolism , Erythroblasts/cytology , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Transcription, Genetic
2.
bioRxiv ; 2023 Nov 29.
Article En | MEDLINE | ID: mdl-38077058

Hematopoietic stem cell (HSC) transplantation using umbilical cord blood (UCB) is a potentially life-saving treatment for leukemia and bone marrow failure but is limited by the low number of HSCs in UCB. The loss of HSCs after ex vivo manipulation is also a major obstacle to gene editing for inherited blood disorders. HSCs require a low rate of translation to maintain their capacity for self-renewal, but hematopoietic cytokines used to expand HSCs stimulate protein synthesis and impair long-term self-renewal. We previously described cytokine-free conditions that maintain but do not expand human and mouse HSCs ex vivo. Here we performed a high throughput screen and identified translation inhibitors that allow ex vivo expansion of human HSCs while minimizing cytokine exposure. Transplantation assays show a ~5-fold expansion of long-term HSCs from UCB after one week of culture in low cytokine conditions. Single cell transcriptomic analysis demonstrates maintenance of HSCs expressing mediators of the unfolded protein stress response, further supporting the importance of regulated proteostasis in HSC maintenance and expansion. This expansion method maintains and expands human HSCs after CRISPR/Cas9 editing of the BCL11A+58 enhancer, overcoming a major obstacle to ex vivo gene correction for human hemoglobinopathies.

4.
Mol Ther Nucleic Acids ; 31: 452-465, 2023 Mar 14.
Article En | MEDLINE | ID: mdl-36852088

Transcriptional enhancers can be in physical proximity of their target genes via chromatin looping. The enhancer at the ß-globin locus (locus control region [LCR]) contacts the fetal-type (HBG) and adult-type (HBB) ß-globin genes during corresponding developmental stages. We have demonstrated previously that forcing proximity between the LCR and HBG genes in cultured adult-stage erythroid cells can activate HBG transcription. Activation of HBG expression in erythroid cells is of benefit to patients with sickle cell disease. Here, using the ß-globin locus as a model, we provide proof of concept at the organismal level that forced enhancer rewiring might present a strategy to alter gene expression for therapeutic purposes. Hematopoietic stem and progenitor cells (HSPCs) from mice bearing human ß-globin genes were transduced with lentiviral vectors expressing a synthetic transcription factor (ZF-Ldb1) that fosters LCR-HBG contacts. When engrafted into host animals, HSPCs gave rise to adult-type erythroid cells with elevated HBG expression. Vectors containing ZF-Ldb1 were optimized for activity in cultured human and rhesus macaque erythroid cells. Upon transplantation into rhesus macaques, erythroid cells from HSPCs expressing ZF-Ldb1 displayed elevated HBG production. These findings in two animal models suggest that forced redirection of gene-regulatory elements may be used to alter gene expression to treat disease.

6.
Blood Cancer Discov ; 3(6): 554-567, 2022 11 02.
Article En | MEDLINE | ID: mdl-35926182

SF3B1 mutations, which occur in 20% of patients with myelodysplastic syndromes (MDS), are the hallmarks of a specific MDS subtype, MDS with ringed sideroblasts (MDS-RS), which is characterized by the accumulation of erythroid precursors in the bone marrow and primarily affects the elderly population. Here, using single-cell technologies and functional validation studies of primary SF3B1-mutant MDS-RS samples, we show that SF3B1 mutations lead to the activation of the EIF2AK1 pathway in response to heme deficiency and that targeting this pathway rescues aberrant erythroid differentiation and enables the red blood cell maturation of MDS-RS erythroblasts. These data support the development of EIF2AK1 inhibitors to overcome transfusion dependency in patients with SF3B1-mutant MDS-RS with impaired red blood cell production. SIGNIFICANCE: MDS-RS are characterized by significant anemia. Patients with MDS-RS die from a shortage of red blood cells and the side effects of iron overload due to their constant need for transfusions. Our study has implications for the development of therapies to achieve long-lasting hematologic responses. This article is highlighted in the In This Issue feature, p. 476.


Myelodysplastic Syndromes , Phosphoproteins , Humans , Aged , RNA Splicing Factors/genetics , Phosphoproteins/genetics , Myelodysplastic Syndromes/genetics , Erythroid Precursor Cells , Signal Transduction , eIF-2 Kinase
7.
Nat Genet ; 54(9): 1417-1426, 2022 09.
Article En | MEDLINE | ID: mdl-35941187

The fetal-to-adult switch in hemoglobin production is a model of developmental gene control with relevance to the treatment of hemoglobinopathies. The expression of transcription factor BCL11A, which represses fetal ß-type globin (HBG) genes in adult erythroid cells, is predominantly controlled at the transcriptional level but the underlying mechanism is unclear. We identify HIC2 as a repressor of BCL11A transcription. HIC2 and BCL11A are reciprocally expressed during development. Forced expression of HIC2 in adult erythroid cells inhibits BCL11A transcription and induces HBG expression. HIC2 binds to erythroid BCL11A enhancers to reduce chromatin accessibility and binding of transcription factor GATA1, diminishing enhancer activity and enhancer-promoter contacts. DNA-binding and crystallography studies reveal direct steric hindrance as one mechanism by which HIC2 inhibits GATA1 binding at a critical BCL11A enhancer. Conversely, loss of HIC2 in fetal erythroblasts increases enhancer accessibility, GATA1 binding and BCL11A transcription. HIC2 emerges as an evolutionarily conserved regulator of hemoglobin switching via developmental control of BCL11A.


Hemoglobins , Kruppel-Like Transcription Factors , Repressor Proteins , Tumor Suppressor Proteins , Carrier Proteins/genetics , Erythroid Cells/metabolism , Hemoglobins/genetics , Humans , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , beta-Globins/genetics , beta-Globins/metabolism , gamma-Globins/genetics
9.
Nat Genet ; 54(6): 874-884, 2022 06.
Article En | MEDLINE | ID: mdl-35618846

The mechanisms by which the fetal-type ß-globin-like genes HBG1 and HBG2 are silenced in adult erythroid precursor cells remain a fundamental question in human biology and have therapeutic relevance to sickle cell disease and ß-thalassemia. Here, we identify via a CRISPR-Cas9 genetic screen two members of the NFI transcription factor family-NFIA and NFIX-as HBG1/2 repressors. NFIA and NFIX are expressed at elevated levels in adult erythroid cells compared with fetal cells, and function cooperatively to repress HBG1/2 in cultured cells and in human-to-mouse xenotransplants. Genomic profiling, genome editing and DNA binding assays demonstrate that the potent concerted activity of NFIA and NFIX is explained in part by their ability to stimulate the expression of BCL11A, a known silencer of the HBG1/2 genes, and in part by directly repressing the HBG1/2 genes. Thus, NFI factors emerge as versatile regulators of the fetal-to-adult switch in ß-globin production.


Fetal Hemoglobin , gamma-Globins , Animals , Carrier Proteins/genetics , Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Editing , Mice , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Transcription Factors/genetics , beta-Globins/genetics , beta-Globins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism
10.
Blood Adv ; 6(23): 5956-5968, 2022 12 13.
Article En | MEDLINE | ID: mdl-35622975

The fetal-to-adult hemoglobin transition is clinically relevant because reactivation of fetal hemoglobin (HbF) significantly reduces morbidity and mortality associated with sickle cell disease (SCD) and ß-thalassemia. Most studies on the developmental regulation of the globin genes, including genome-wide genetics screens, have focused on DNA binding proteins, including BCL11A and ZBTB7A/LRF and their cofactors. Our understanding of RNA binding proteins (RBPs) in this process is much more limited. Two RBPs, LIN28B and IGF2BP1, are known posttranscriptional regulators of HbF production, but a global view of RBPs is still lacking. Here, we carried out a CRISPR/Cas9-based screen targeting RBPs harboring RNA methyltransferase and/or RNA recognition motif (RRM) domains and identified RNA binding motif 12 (RBM12) as a novel HbF suppressor. Depletion of RBM12 induced HbF expression and attenuated cell sickling in erythroid cells derived from patients with SCD with minimal detrimental effects on cell maturation. Transcriptome and proteome profiling revealed that RBM12 functions independently of major known HbF regulators. Enhanced cross-linking and immunoprecipitation followed by high-throughput sequencing revealed strong preferential binding of RBM12 to 5' untranslated regions of transcripts, narrowing down the mechanism of RBM12 action. Notably, we pinpointed the first of 5 RRM domains as essential, and, in conjunction with a linker domain, sufficient for RBM12-mediated HbF regulation. Our characterization of RBM12 as a negative regulator of HbF points to an additional regulatory layer of the fetal-to-adult hemoglobin switch and broadens the pool of potential therapeutic targets for SCD and ß-thalassemia.


Anemia, Sickle Cell , beta-Thalassemia , Adult , Humans , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , beta-Thalassemia/genetics , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , RNA-Binding Proteins/genetics
11.
Epigenetics Chromatin ; 14(1): 37, 2021 07 30.
Article En | MEDLINE | ID: mdl-34330317

BACKGROUND: Condensation of chromatin prior to enucleation is an essential component of terminal erythroid maturation, and defects in this process are associated with inefficient erythropoiesis and anemia. However, the mechanisms involved in this phenomenon are not well understood. Here, we describe a potential role for the histone variant H2A.X in erythropoiesis. RESULTS: We find in multiple model systems that this histone is essential for normal maturation, and that the loss of H2A.X in erythroid cells results in dysregulation in expression of erythroid-specific genes as well as a nuclear condensation defect. In addition, we demonstrate that erythroid maturation is characterized by phosphorylation at both S139 and Y142 on the C-terminal tail of H2A.X during late-stage erythropoiesis. Knockout of the kinase BAZ1B/WSTF results in loss of Y142 phosphorylation and a defect in nuclear condensation, but does not replicate extensive transcriptional changes to erythroid-specific genes observed in the absence of H2A.X. CONCLUSIONS: We relate these findings to Caspase-Initiated Chromatin Condensation (CICC) in terminal erythroid maturation, where aspects of the apoptotic pathway are invoked while apoptosis is specifically suppressed.


Chromatin , Erythropoiesis , Caspases , Histones/metabolism , Phosphorylation
12.
Blood Adv ; 4(18): 4560-4572, 2020 09 22.
Article En | MEDLINE | ID: mdl-32956454

Increasing fetal hemoglobin (HbF) provides clinical benefit in patients with sickle cell disease (SCD). We recently identified heme-regulated inhibitor (HRI, EIF2AK1), as a novel HbF regulator. Because HRI is an erythroid-specific protein kinase, it presents a potential target for pharmacologic intervention. We found that maximal HbF induction required >80% to 85% HRI depletion. Because it remains unclear whether this degree of HRI inhibition can be achieved pharmacologically, we explored whether HRI knockdown can be combined with pharmacologic HbF inducers to achieve greater HbF production and minimize potential adverse effects associated with treatments. Strongly cooperative HbF induction was observed when HRI depletion was combined with exposure to pomalidomide or the EHMT1/2 inhibitor UNC0638, but not to hydroxyurea. Mechanistically, reduction in the levels of the HbF repressor BCL11A reflected the cooperativity of HRI loss and pomalidomide treatment, whereas UNC0638 did not modulate BCL11A levels. In conjunction with HRI loss, pomalidomide maintained its HbF-inducing activity at 10-fold lower concentrations, in which condition there were minimal observed detrimental effects on erythroid cell maturation and viability, as well as fewer alterations in the erythroid transcriptome. When tested in cells from patients with SCD, combining HRI depletion with pomalidomide or UNC0638 achieved up to 50% to 60% HbF and 45% to 50% HbF, respectively, as measured by high-performance liquid chromatography, and markedly counteracted cell sickling. In summary, this study provides a foundation for the exploration of combining future small-molecule HRI inhibitors with additional pharmacologic HbF inducers to maximize HbF production and preserve erythroid cell functionality for the treatment of SCD and other hemoglobinopathies.


Anemia, Sickle Cell , Fetal Hemoglobin , Anemia, Sickle Cell/drug therapy , Erythrocytes, Abnormal , Erythroid Cells , Fetal Hemoglobin/genetics , Humans , Hydroxyurea/pharmacology
13.
Blood ; 135(24): 2121-2132, 2020 06 11.
Article En | MEDLINE | ID: mdl-32299090

Reactivation of fetal hemoglobin remains a critical goal in the treatment of patients with sickle cell disease and ß-thalassemia. Previously, we discovered that silencing of the fetal γ-globin gene requires the erythroid-specific eIF2α kinase heme-regulated inhibitor (HRI), suggesting that HRI might present a pharmacologic target for raising fetal hemoglobin levels. Here, via a CRISPR-Cas9-guided loss-of-function screen in human erythroblasts, we identify transcription factor ATF4, a known HRI-regulated protein, as a novel γ-globin regulator. ATF4 directly stimulates transcription of BCL11A, a repressor of γ-globin transcription, by binding to its enhancer and fostering enhancer-promoter contacts. Notably, HRI-deficient mice display normal Bcl11a levels, suggesting species-selective regulation, which we explain here by demonstrating that the analogous ATF4 motif at the murine Bcl11a enhancer is largely dispensable. Our studies uncover a linear signaling pathway from HRI to ATF4 to BCL11A to γ-globin and illustrate potential limits of murine models of globin gene regulation.


Activating Transcription Factor 4/genetics , Fetal Hemoglobin/genetics , Repressor Proteins/genetics , eIF-2 Kinase/genetics , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Animals , CRISPR-Cas Systems , Cells, Cultured , Enhancer Elements, Genetic , Erythroblasts/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Species Specificity , beta-Thalassemia/blood , beta-Thalassemia/genetics , beta-Thalassemia/therapy , gamma-Globins/biosynthesis , gamma-Globins/genetics
14.
Blood ; 135(22): 1957-1968, 2020 05 28.
Article En | MEDLINE | ID: mdl-32268371

Reversing the developmental switch from fetal hemoglobin (HbF, α2γ2) to adult hemoglobin (HbA, α2ß2) is an important therapeutic approach in sickle cell disease (SCD) and ß-thalassemia. In healthy individuals, SCD patients, and patients treated with pharmacologic HbF inducers, HbF is present only in a subset of red blood cells known as F cells. Despite more than 50 years of observations, the cause for this heterocellular HbF expression pattern, even among genetically identical cells, remains unknown. Adult F cells might represent a reversion of a given cell to a fetal-like epigenetic and transcriptional state. Alternatively, isolated transcriptional or posttranscriptional events at the γ-globin genes might underlie heterocellularity. Here, we set out to understand the heterogeneity of HbF activation by developing techniques to purify and profile differentiation stage-matched late erythroblast F cells and non-F cells (A cells) from the human HUDEP2 erythroid cell line and primary human erythroid cultures. Transcriptional and proteomic profiling of these cells demonstrated very few differences between F and A cells at the RNA level either under baseline conditions or after treatment with HbF inducers hydroxyurea or pomalidomide. Surprisingly, we did not find differences in expression of any known HbF regulators, including BCL11A or LRF, that would account for HbF activation. Our analysis shows that F erythroblasts are not significantly different from non-HbF-expressing cells and that the primary differences likely occur at the transcriptional level at the ß-globin locus.


Erythroblasts/metabolism , Fetal Hemoglobin/biosynthesis , Hemoglobin A/metabolism , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Cell Line , Cell Separation/methods , Cells, Cultured , Erythroblasts/classification , Erythroblasts/drug effects , Erythroid Cells/classification , Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Gene Expression Profiling , Hemoglobin A/genetics , Humans , Hydroxyurea/pharmacology , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thalidomide/analogs & derivatives , Thalidomide/pharmacology
15.
Blood Adv ; 3(10): 1586-1597, 2019 05 28.
Article En | MEDLINE | ID: mdl-31126914

Reactivation of fetal hemoglobin (HbF) production benefits patients with sickle cell disease and ß-thalassemia. To identify new HbF regulators that might be amenable to pharmacologic control, we screened a protein domain-focused CRISPR-Cas9 library targeting chromatin regulators, including BTB domain-containing proteins. Speckle-type POZ protein (SPOP), a substrate adaptor of the CUL3 ubiquitin ligase complex, emerged as a novel HbF repressor. Depletion of SPOP or overexpression of a dominant negative version significantly raised fetal globin messenger RNA and protein levels with minimal detrimental effects on normal erythroid maturation, as determined by transcriptome and proteome analyses. SPOP controls HbF expression independently of the major transcriptional HbF repressors BCL11A and LRF. Finally, pharmacologic HbF inducers cooperate with SPOP depletion during HbF upregulation. Our study implicates SPOP and the CUL3 ubiquitin ligase system in controlling HbF production in human erythroid cells and may offer new therapeutic strategies for the treatment of ß-hemoglobinopathies.


Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Female , Humans , Male , Nuclear Proteins/genetics , Repressor Proteins/genetics , Young Adult
16.
Curr Treat Options Oncol ; 18(12): 70, 2017 Nov 16.
Article En | MEDLINE | ID: mdl-29143887

OPINION STATEMENT: Acquired aplastic anemia (AA) is a rare, life-threatening bone marrow failure (BMF) disorder that affects patients of all ages and is caused by lymphocyte destruction of early hematopoietic cells. Diagnosis of AA requires a comprehensive approach with prompt evaluation for inherited and secondary causes of bone marrow aplasia, while providing aggressive supportive care. The choice of frontline therapy is determined by a number of factors including AA severity, age of the patient, donor availability, and access to optimal therapies. For newly diagnosed severe aplastic anemia, bone marrow transplant should be pursued in all pediatric patients and in younger adult patients when a matched sibling donor is available. Frontline therapy in older adult patients and in all patients lacking a matched sibling donor involves immunosuppressive therapy (IST) with horse antithymocyte globulin and cyclosporine A. Recent improvements in upfront therapy include encouraging results with closely matched unrelated donor transplants in younger patients and the emerging benefits of eltrombopag combined with initial IST, with randomized studies underway. In the refractory setting, several therapeutic options exist, with improving outcomes of matched unrelated donor and haploidentical bone marrow transplantation as well as the addition of eltrombopag to the non-transplant AA armamentarium. With the recent appreciation of frequent clonal hematopoiesis in AA patients and with the growing use of next-generation sequencing in the clinic, utmost caution should be exercised in interpreting the significance of somatic mutations in AA. Future longitudinal studies of large numbers of patients are needed to determine the prognostic significance of somatic mutations and to guide optimal surveillance and treatment approaches to prevent long-term clonal complications.


Anemia, Aplastic/diagnosis , Anemia, Aplastic/therapy , Bone Marrow Diseases/diagnosis , Bone Marrow Diseases/therapy , Anemia, Aplastic/pathology , Bone Marrow Diseases/pathology , Bone Marrow Transplantation , Cyclosporine/therapeutic use , Hematopoietic Stem Cell Transplantation , Humans , Immunosuppression Therapy/methods , Siblings , Tissue Donors
17.
Blood ; 120(12): 2501-11, 2012 Sep 20.
Article En | MEDLINE | ID: mdl-22889760

Erythropoiesis is a robust process of cellular expansion and maturation occurring in murine bone marrow and spleen. We previously determined that sublethal irradiation, unlike bleeding or hemolysis, depletes almost all marrow and splenic erythroblasts but leaves peripheral erythrocytes intact. To better understand the erythroid stress response, we analyzed progenitor, precursor, and peripheral blood compartments of mice post-4 Gy total body irradiation. Erythroid recovery initiates with rapid expansion of late-stage erythroid progenitors-day 3 burst-forming units and colony-forming units, associated with markedly increased plasma erythropoietin (EPO). Although initial expansion of late-stage erythroid progenitors is dependent on EPO, this cellular compartment becomes sharply down-regulated despite elevated EPO levels. Loss of EPO-responsive progenitors is associated temporally with a wave of maturing erythroid precursors in marrow and with emergence of circulating erythroid progenitors and subsequent reestablishment of splenic erythropoiesis. These circulating progenitors selectively engraft and mature in irradiated spleen after short-term transplantation, supporting the concept that bone marrow erythroid progenitors migrate to spleen. We conclude that sublethal radiation is a unique model of endogenous stress erythropoiesis, with specific injury to the extravascular erythron, expansion and maturation of EPO-responsive late-stage progenitors exclusively in marrow, and subsequent reseeding of extramedullary sites.


Bone Marrow/pathology , Erythroid Precursor Cells/cytology , Erythropoiesis/physiology , Erythropoietin/administration & dosage , Radiation Injuries, Experimental/prevention & control , Spleen/cytology , Animals , Bone Marrow/radiation effects , Cell Proliferation , Colony-Forming Units Assay , Erythrocyte Transfusion , Erythroid Precursor Cells/radiation effects , Erythropoiesis/radiation effects , Erythropoietin/blood , Female , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Spleen/radiation effects , Stem Cells , Whole-Body Irradiation
18.
Exp Hematol ; 39(4): 434-45, 2011 Apr.
Article En | MEDLINE | ID: mdl-21291953

OBJECTIVE: Clastogenic injury of the erythroid lineage results in anemia, reticulocytopenia, and transient appearance of micronucleated reticulocytes. However, the micronucleated reticulocyte dose-response in murine models is only linear to 2 Gy total body irradiation and paradoxically decreases at higher exposures, suggesting complex radiation effects on erythroid intermediates. To better understand this phenomenon, we investigated the kinetics and apoptotic response of the erythron to sublethal radiation injury. MATERIALS AND METHODS: We analyzed the response to 1 and 4 Gy total body irradiation of erythroid progenitors and precursors using colony assays and imaging flow cytometry, respectively. We also investigated cell cycling and apoptotic gene expression of the steady-state erythron. RESULTS: After 1 Gy total body irradiation, erythroid progenitors and precursors were partially depleted. In contrast, essentially all bone marrow erythroid progenitors and precursors were lost within 2 days after 4 Gy irradiation. Imaging flow cytometry analysis revealed preferential loss of phenotypic erythroid colony-forming units and proerythroblasts immediately after sublethal irradiation. Furthermore, these populations underwent radiation-induced apoptosis, without changes in steady-state cellular proliferation, at much higher frequencies than later-stage erythroid precursors. Primary erythroid precursor maturation is associated with marked Bcl-xL upregulation and Bax and Bid downregulation. CONCLUSIONS: Micronucleated reticulocyte loss after higher sublethal radiation exposures results from rapid depletion of erythroid progenitors and precursors. This injury reveals that erythroid colony-forming units and proerythroblasts constitute a particularly proapoptotic compartment within the erythron. We conclude that the functional transition of primary proerythroblasts to later-stage erythroid precursors is characterized by a shift from a proapoptotic to an antiapoptotic phenotype.


Cell Differentiation/radiation effects , Erythroid Cells/radiation effects , Radiation Injuries, Experimental/physiopathology , Animals , Apoptosis/radiation effects , BH3 Interacting Domain Death Agonist Protein/genetics , Cell Cycle/radiation effects , Dose-Response Relationship, Radiation , Erythroblasts/cytology , Erythroblasts/metabolism , Erythroblasts/radiation effects , Erythroid Cells/cytology , Erythroid Cells/metabolism , Flow Cytometry , Gene Expression Regulation/radiation effects , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Whole-Body Irradiation , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
19.
Cancer Immunol Immunother ; 58(7): 1071-83, 2009 Jul.
Article En | MEDLINE | ID: mdl-19018532

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a multifunctional transcription factor that regulates adipogenesis, immunity and inflammation. Our laboratory previously demonstrated that PPARgamma ligands induce apoptosis in malignant B cells. While malignant B lineage cells such as B cell lymphoma express PPARgamma, its physiological function remains unknown. Herein, we demonstrate that silencing PPARgamma expression by RNAi in human Burkitt's type B lymphoma cells increased basal and mitogen-induced proliferation and survival, which was accompanied by enhanced NF-kappaB activity and increased expression of Bcl-2. These cells also had increased survival upon exposure to PPARgamma ligands and exhibited a less differentiated phenotype. In contrast, PPARgamma overexpression in B lymphoma cells inhibited cell growth and decreased their proliferative response to mitogenic stimuli. These cells were also more sensitive to PPARgamma-ligand induced growth arrest and displayed a more differentiated phenotype. Collectively, these findings support a regulatory role for PPARgamma in the proliferation, survival and differentiation of malignant B cells. These findings further suggest the potential of PPARgamma as a therapeutic target for B cell malignancy.


Burkitt Lymphoma/pathology , NF-kappa B/metabolism , PAX5 Transcription Factor/metabolism , PPAR gamma/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Repressor Proteins/metabolism , Burkitt Lymphoma/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Knockdown Techniques , Genetic Vectors/metabolism , Humans , Ligands , PPAR gamma/genetics , Positive Regulatory Domain I-Binding Factor 1 , RNA Interference
...