Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 17129, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34429458

ABSTRACT

Production of red blood cells relies on proper mitochondrial function, both for their increased energy demands during differentiation and for proper heme and iron homeostasis. Mutations in genes regulating mitochondrial function have been reported in patients with anemia, yet their pathophysiological role often remains unclear. PGC1ß is a critical coactivator of mitochondrial biogenesis, with increased expression during terminal erythroid differentiation. The role of PGC1ß has however mainly been studied in skeletal muscle, adipose and hepatic tissues, and its function in erythropoiesis remains largely unknown. Here we show that perturbed PGC1ß expression in human hematopoietic stem/progenitor cells from both bone marrow and cord blood results in impaired formation of early erythroid progenitors and delayed terminal erythroid differentiation in vitro, with accumulations of polychromatic erythroblasts, similar to MDS-related refractory anemia. Reduced levels of PGC1ß resulted in deregulated expression of iron, heme and globin related genes in polychromatic erythroblasts, and reduced hemoglobin content in the more mature bone marrow derived reticulocytes. Furthermore, PGC1ß knock-down resulted in disturbed cell cycle exit with accumulation of erythroblasts in S-phase and enhanced expression of G1-S regulating genes, with smaller reticulocytes as a result. Taken together, we demonstrate that PGC1ß is directly involved in production of hemoglobin and regulation of G1-S transition and is ultimately required for proper terminal erythroid differentiation.


Subject(s)
Erythroid Cells/metabolism , Erythropoiesis , RNA-Binding Proteins/metabolism , Cell Cycle , Cells, Cultured , Erythroid Cells/cytology , Hemoglobins/metabolism , Humans , RNA-Binding Proteins/genetics
2.
Sci Rep ; 11(1): 15898, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34354145

ABSTRACT

The YPEL family genes are highly conserved across a diverse range of eukaryotic organisms and thus potentially involved in essential cellular processes. Ypel4, one of five YPEL family gene orthologs in mouse and human, is highly and specifically expressed in late terminal erythroid differentiation (TED). In this study, we investigated the role of Ypel4 in murine erythropoiesis, providing for the first time an in-depth description of a Ypel4-null phenotype in vivo. We demonstrated that the Ypel4-null mice displayed a secondary polycythemia with macro- and reticulocytosis. While lack of Ypel4 did not affect steady-state TED in the bone marrow or spleen, the anemia-recovering capacity of Ypel4-null cells was diminished. Furthermore, Ypel4-null red blood cells (RBC) were cleared from the circulation at an increased rate, demonstrating an intrinsic defect of RBCs. Scanning electron micrographs revealed an ovalocytic morphology of Ypel4-null RBCs and functional testing confirmed reduced deformability. Even though Band 3 protein levels were shown to be reduced in Ypel4-null RBC membranes, we could not find support for a physical interaction between YPEL4 and the Band 3 protein. In conclusion, our findings provide crucial insights into the role of Ypel4 in preserving normal red cell membrane integrity.


Subject(s)
Carrier Proteins/genetics , Erythrocyte Membrane/physiology , Erythropoiesis/genetics , Anemia/metabolism , Animals , Anion Exchange Protein 1, Erythrocyte/metabolism , Carrier Proteins/metabolism , Erythrocyte Membrane/genetics , Erythrocytes/metabolism , Erythrocytes, Abnormal/metabolism , Erythropoiesis/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycythemia/genetics , Spleen
3.
Haematologica ; 105(11): 2561-2571, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33131245

ABSTRACT

Massive expansion of erythroid progenitor cells is essential for surviving anemic stress. Research towards understanding this critical process, referred to as stress-erythropoiesis, has been hampered due to lack of specific marker-combinations enabling analysis of the distinct stress-progenitor cells capable of providing radioprotection and enhanced red blood cell production. Here we present a method for precise identification and in vivo validation of progenitor cells contributing to both steady-state and stress-erythropoiesis, enabling for the first time in-depth molecular characterization of these cells. Differential expression of surface markers CD150, CD9 and Sca1 defines a hierarchy of splenic stress-progenitors during irradiation-induced stress recovery in mice, and provides high-purity isolation of the functional stress-BFU-Es with a 100-fold improved enrichment compared to state-of-the-art. By transplanting purified stress-progenitors expressing the fluorescent protein Kusabira Orange, we determined their kinetics in vivo and demonstrated that CD150+CD9+Sca1- stress-BFU-Es provide a massive but transient radioprotective erythroid wave, followed by multi-lineage reconstitution from CD150+CD9+Sca1+ multi-potent stem/progenitor cells. Whole genome transcriptional analysis revealed that stress-BFU-Es express gene signatures more associated with erythropoiesis and proliferation compared to steady-state BFU-Es, and are BMP-responsive. Evaluation of chromatin accessibility through ATAC sequencing reveals enhanced and differential accessibility to binding sites of the chromatin-looping transcription factor CTCF in stress-BFU-Es compared to steady-state BFU-Es. Our findings offer molecular insight to the unique capacity of stress-BFU-Es to rapidly form erythroid cells in response to anemia and constitute an important step towards identifying novel erythropoiesis stimulating agents.


Subject(s)
Erythropoietin , Transcriptome , Animals , Epigenesis, Genetic , Erythroid Cells , Erythroid Precursor Cells , Erythropoiesis/genetics , Mice
4.
Exp Hematol ; 88: 28-41, 2020 08.
Article in English | MEDLINE | ID: mdl-32629063

ABSTRACT

Erythropoiesis is intimately coupled to cell division, and deletion of the cell cycle regulator retinoblastoma protein (pRb) causes anemia in mice. Erythroid-specific deletion of pRb has been found to result in inefficient erythropoiesis because of deregulated coordination of cell cycle exit and mitochondrial biogenesis. However, the pathophysiology remains to be fully described, and further characterization of the link between cell cycle regulation and mitochondrial function is needed. To this end we further assessed conditional erythroid-specific deletion of pRb. This resulted in macrocytic anemia, despite elevated levels of erythropoietin (Epo), and an accumulation of erythroid progenitors in the bone marrow, a phenotype strongly resembling refractory anemia associated with myelodysplastic syndromes (MDS). Using high-fractionation fluorescence-activated cell sorting analysis for improved phenotypic characterization, we illustrate that erythroid differentiation was disrupted at the orthochromatic stage. Transcriptional profiling of sequential purified populations revealed failure to upregulate genes critical for mitochondrial function such as Pgc1ß, Alas2, and Abcb7 specifically at the block, together with disturbed heme production and iron transport. Notably, deregulated ABCB7 causes ring sideroblastic anemia in MDS patients, and the mitochondrial co-activator PGC1ß is heterozygously lost in del5q MDS. Importantly, the anemia could be rescued through enhanced PPAR signaling in vivo via either overexpression of Pgc1ß or bezafibrate administration. In conclusion, lack of pRb results in MDS-like anemia with disrupted differentiation and impaired mitochondrial function at the orthochromatic erythroblast stage. Our findings reveal for the first time a role for pRb in heme and iron regulation, and indicate that pRb-induced anemia can be rescued in vivo through therapeutic enhancement of PPAR signaling.


Subject(s)
Anemia/metabolism , Erythroblasts/metabolism , Erythropoiesis , Mitochondria/metabolism , Myelodysplastic Syndromes/metabolism , Retinoblastoma Protein/deficiency , Anemia/genetics , Anemia/pathology , Animals , Erythroblasts/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Retinoblastoma Protein/metabolism
5.
Exp Hematol ; 54: 4-11, 2017 10.
Article in English | MEDLINE | ID: mdl-28757433

ABSTRACT

The hematopoietic system is responsible for transporting oxygen and nutrients, fighting infections, and repairing tissue damage. Hematopoietic system dysfunction therefore causes a range of serious health consequences. Lifelong hematopoiesis is maintained by repopulating multipotent hematopoietic stem cells (HSCs) that replenish shorter-lived, mature blood cell types. A prokaryotic mechanism of immunity, the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 nuclease system, has been recently "repurposed" to mutate mammalian genomes efficiently and in a sequence-specific manner. The application of this genome-editing technology to hematology has afforded new approaches for functional genomics and even the prospect of "correcting" dysfunctional HSCs in the treatment of serious genetic hematological diseases. In this Perspective, we provide an overview of three recent CRISPR/Cas9 methods in hematology: gene disruption, gene targeting, and saturating mutagenesis. We also summarize the technical considerations and advice provided during the May 2017 International Society of Experimental Hematology New Investigator Committee webinar on the same topic.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Gene Targeting/methods , Genome , Hematology/methods , Mutagenesis , Animals , Computational Biology , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
6.
Exp Hematol ; 50: 22-26, 2017 06.
Article in English | MEDLINE | ID: mdl-28189651

ABSTRACT

Hematopoietic stem cells (HSCs) reside in specialized microenvironments known as niches. The niche is essential to support HSC function and to maintain a correct balance between self-renewal and differentiation. Recent advances in defining different mesenchymal and endothelial bone marrow cell populations, as well as hematopoietic stem and progenitor cells, greatly enhanced our understanding of these niches and of the molecular mechanisms by which they regulate HSC function. In addition to the role in maintaining HSC homeostasis, the niche has also been implicated in the pathogenesis of blood disorders including hematological malignancies. Characterizing the extrinsic regulators and the cellular context in which the niches interact with HSCs will be crucial to define new strategies to enhance blood regeneration. Furthermore, a better understanding of the role of the niche in leukemia development will open new possibilities for the treatment of these disorders by using therapies aiming to target the leukemic niche specifically. To update on recent findings on this topic, the International Society for Experimental Hematology (ISEH) organized a webinar, presented by Prof. Sean J. Morrison and Dr. Simón Méndez-Ferrer and moderated by Dr. Cristina Lo Celso, entitled "The evolving view of the hematopoietic stem cell niche," which we summarize here.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Stem Cell Niche/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation , Disease Susceptibility , Hematopoiesis , Homeostasis , Humans , Osteogenesis
7.
Cell Rep ; 15(11): 2550-62, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27264182

ABSTRACT

Erythroid cell commitment and differentiation proceed through activation of a lineage-restricted transcriptional network orchestrated by a group of well characterized genes. However, the minimal set of factors necessary for instructing red blood cell (RBC) development remains undefined. We employed a screen for transcription factors allowing direct lineage reprograming from fibroblasts to induced erythroid progenitors/precursors (iEPs). We show that Gata1, Tal1, Lmo2, and c-Myc (GTLM) can rapidly convert murine and human fibroblasts directly to iEPs. The transcriptional signature of murine iEPs resembled mainly that of primitive erythroid progenitors in the yolk sac, whereas addition of Klf1 or Myb to the GTLM cocktail resulted in iEPs with a more adult-type globin expression pattern. Our results demonstrate that direct lineage conversion is a suitable platform for defining and studying the core factors inducing the different waves of erythroid development.


Subject(s)
Cell Lineage , Erythropoiesis , Transcription Factors/metabolism , Aging , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cellular Reprogramming/genetics , Colony-Forming Units Assay , Erythroblasts/cytology , Erythroblasts/metabolism , Erythropoiesis/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation , Globins/genetics , Globins/metabolism , Humans , Mice, Inbred C57BL
8.
Exp Hematol ; 43(9): 756-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26143581

ABSTRACT

Transplantation of hematopoietic stem cells (HSCs) to treat hematologic disorders is routinely used in the clinic. However, HSC therapy is hindered by the requirements of finding human leukocyte antigen (HLA)-matched donors and attaining sufficient numbers of long-term HSCs in the graft. Therefore, ex vivo expansion of transplantable HSCs remains one of the "holy grails" of hematology. Without the ability to maintain and expand human HSCs in vitro, two complementary approaches involving cellular reprogramming to generate transplantable HSCs have emerged. Reprogrammed HSCs represent a potentially inexhaustible supply of autologous tissue. On March 18th, 2015, Dr. George Q. Daley and Dr. Derrick J. Rossi, two pioneers in the field, presented and discussed their most recent research on these topics in a webinar organized by the International Society for Experimental Hematology (ISEH). Here, we summarize these seminars and discuss the possibilities and challenges in the field of hematopoietic specification.


Subject(s)
Hematologic Diseases , Hematopoietic Stem Cells , Pluripotent Stem Cells , Animals , Cellular Reprogramming , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Hematologic Diseases/therapy , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology
9.
Cytokine ; 68(2): 101-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24767864

ABSTRACT

OBJECTIVE: To identify how the gp130-signaling cytokine oncostatin M (OSM), acting alone or in concert with IL-1ß or TNFα, affects synovial fibroblast expression of genes relevant to inflammation and bone erosion in inflammatory arthritis. METHODS: Synovial fibroblasts (SFs) were isolated from non-arthritic wild type (WT) or OSM receptor deficient (OSMR(-/-)) mice and stimulated with OSM, IL-1ß or TNFα and their combinations. Cytokine gene expression was assessed by quantitative RT-PCR. ELISA, flow cytometry and immunohistochemistry identified protein expression. Gene expression patterns were confirmed in SFs isolated from patients with osteoarthritis (OASFs) and rheumatoid arthritis (RASFs). RESULTS: Expression of OSM and its receptors, gp130, OSMR and LIFR, was increased in synovial tissue from the mouse antigen-induced arthritis model. In isolated WT mouse synovial fibroblasts OSM alone, or in synergy with IL-1ß, or together with TNFα, potently induced expression of the pro-inflammatory cytokine IL-6. OSM also induced a sustained increase in mRNA levels of the pro-osteoclastic cytokine RANKL. Combining OSM with IL-1ß, but not with TNFα, further increased RANKL expression. Importantly these effects of OSM were all dependent on the expression of OSMR. Furthermore, OSM also increased expression of its own receptors, gp130 and OSMR and the IL-1 receptor, IL1-R1; the latter effects were also observed in both human OASFs and RASFs. CONCLUSION: Together our data suggests that OSM signaling via OSMR in SFs has the potential to contribute significantly to joint destruction in inflammatory arthritis. It not only induces expression of pro-inflammatory and pro-osteoclastic cytokines but can also augment its own actions and that of IL-1 by inducing expression of OSMR and IL-1R1.


Subject(s)
Fibroblasts/metabolism , Interleukin-1beta/metabolism , Oncostatin M/metabolism , Receptors, Oncostatin M/metabolism , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Gene Expression Regulation , Humans , Interleukin-1beta/genetics , Mice, Inbred C57BL , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-1/metabolism , Receptors, Oncostatin M/deficiency
10.
Haematologica ; 99(4): 647-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24415629

ABSTRACT

The proto-oncogene SKI is highly expressed in human myeloid leukemia and also in murine hematopoietic stem cells. However, its operative relevance in these cells remains elusive. We have over-expressed SKI to define its intrinsic role in hematopoiesis and myeloid neoplasms, which resulted in a robust competitive advantage upon transplantation, a complete dominance of the stem and progenitor compartments, and a marked enhancement of myeloid differentiation at the expense of other lineages. Accordingly, enforced expression of SKI induced a gene signature associated with hematopoietic stem cells and myeloid differentiation, as well as hepatocyte growth factor signaling. Here we demonstrate that, in contrast to what has generally been assumed, the significant impact of SKI on hematopoiesis is independent of its ability to inhibit TGF-beta signaling. Instead, myeloid progenitors expressing SKI are partially dependent on functional hepatocyte growth factor signaling. Collectively our results demonstrate that SKI is an important regulator of hematopoietic stem cell activity and its overexpression leads to myeloproliferative disease.


Subject(s)
DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/metabolism , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Differentiation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Erythropoiesis/genetics , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hepatocyte Growth Factor/metabolism , Humans , Lymphopoiesis/genetics , Mice , Myelopoiesis/genetics , Myeloproliferative Disorders/metabolism , Phenotype , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcriptional Activation , Transforming Growth Factor beta/metabolism
11.
Genom Data ; 2: 189-91, 2014 Dec.
Article in English | MEDLINE | ID: mdl-26484093

ABSTRACT

The proto-oncogene SKI is highly expressed in human myeloid leukemia and also in murine hematopoietic stem cells. However, its operative relevance in these cells remains elusive. We have over-expressed SKI to define its intrinsic role in hematopoiesis and myeloid neoplasms, which resulted in a robust competitive advantage upon transplantation, a complete dominance of the stem and progenitor compartments, and a marked enhancement of myeloid differentiation at the expense of other lineages. Accordingly, enforced expression of SKI induced gene signatures associated with hematopoietic stem cells and myeloid differentiation. Here we provide detailed experimental methods and analysis for the gene expression profiling described in our recently published study of Singbrant et al. (2014) in Haematologica. Our data sets (available at http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE39457) provide a resource for exploring the underlying molecular mechanisms of the involvement of the proto-oncogene SKI in hematopoietic stem cell function and development of myeloid neoplasms.

12.
J Cell Biochem ; 112(6): 1486-90, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21503954

ABSTRACT

Understanding the in vivo regulation of hematopoietic stem cells (HSCs) will be critical to identifying key factors involved in the regulation of HSC self-renewal and differentiation. The niche (microenvironment) in which HSCs reside has recently regained attention accompanied by a dramatic increase in the understanding of the cellular constituents of the bone marrow HSC niche. The use of sophisticated genetic models allowing modulation of specific lineages has demonstrated roles for mesenchymal-derived elements such as osteoblasts and adipocytes, vasculature, nerves, and a range of hematopoietic progeny of the HSC as being participants in the regulation of the bone marrow microenvironment. Whilst providing significant insight into the cellular composition of the niche, is it possible to manipulate any given cell lineage in vivo without impacting, knowingly or unknowingly, on those that remain?


Subject(s)
Hematopoietic Stem Cells/cytology , Stem Cell Niche/cytology , Animals , Bone Marrow Cells/cytology , Humans , Mice
13.
Blood ; 117(21): 5631-42, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21421837

ABSTRACT

Erythropoietin (Epo) has been used in the treatment of anemia resulting from numerous etiologies, including renal disease and cancer. However, its effects are controversial and the expression pattern of the Epo receptor (Epo-R) is debated. Using in vivo lineage tracing, we document that within the hematopoietic and mesenchymal lineage, expression of Epo-R is essentially restricted to erythroid lineage cells. As expected, adult mice treated with a clinically relevant dose of Epo had expanded erythropoiesis because of amplification of committed erythroid precursors. Surprisingly, we also found that Epo induced a rapid 26% loss of the trabecular bone volume and impaired B-lymphopoiesis within the bone marrow microenvironment. Despite the loss of trabecular bone, hematopoietic stem cell populations were unaffected. Inhibition of the osteoclast activity with bisphosphonate therapy blocked the Epo-induced bone loss. Intriguingly, bisphosphonate treatment also reduced the magnitude of the erythroid response to Epo. These data demonstrate a previously unrecognized in vivo regulatory network coordinating erythropoiesis, B-lymphopoiesis, and skeletal homeostasis. Importantly, these findings may be relevant to the clinical application of Epo.


Subject(s)
B-Lymphocytes/metabolism , Bone Marrow/drug effects , Bone and Bones/metabolism , Erythropoiesis/physiology , Erythropoietin/pharmacology , Homeostasis , Lymphopoiesis/physiology , Animals , Bone Marrow/metabolism , Bone Remodeling/physiology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Erythroblasts/metabolism , Flow Cytometry , Gene Expression , Humans , Male , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Receptors, Erythropoietin/metabolism , Recombinant Proteins , Spleen/cytology , Spleen/metabolism
14.
Blood ; 115(23): 4689-98, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20371744

ABSTRACT

Numerous publications have described the importance of bone morphogenetic protein (BMP) signaling in the specification of hematopoietic tissue in developing embryos. Here we investigate the full role of canonical BMP signaling in both adult and fetal liver hematopoiesis using conditional knockout strategies because conventional disruption of components of the BMP signaling pathway result in early death of the embryo. By targeting both Smad1 and Smad5, we have generated a double-knockout mouse with complete disruption of canonical BMP signaling. Interestingly, concurrent deletion of Smad1 and Smad5 results in death because of extrahematopoietic pathologic changes in the colon. However, Smad1/Smad5-deficient bone marrow cells can compete normally with wild-type cells and display unaffected self-renewal and differentiation capacity when transplanted into lethally irradiated recipients. Moreover, although BMP receptor expression is increased in fetal liver, fetal liver cells deficient in both Smad1 and Smad5 remain competent to long-term reconstitute lethally irradiated recipients in a multilineage manner. In conclusion, canonical BMP signaling is not required to maintain either adult or fetal liver hematopoiesis, despite its crucial role in the initial patterning of hematopoiesis in early embryonic development.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Fetus/embryology , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells/metabolism , Liver/embryology , Signal Transduction/physiology , Animals , Bone Morphogenetic Protein Receptors/biosynthesis , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Proteins/genetics , Cell Differentiation/physiology , Colon/embryology , Colon/metabolism , Embryo Loss/genetics , Embryo Loss/metabolism , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cell Transplantation , Liver/metabolism , Mice , Mice, Knockout , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Transplantation, Homologous
15.
Stem Cells ; 25(11): 2809-19, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17673527

ABSTRACT

Endoglin is a transforming growth factor-beta (TGF-beta) accessory receptor recently identified as being highly expressed on long-term repopulating hematopoietic stem cells (HSC). However, little is known regarding its function in these cells. We have used two complementary approaches toward understanding endoglin's role in HSC biology: one that efficiently knocks down expression via lentiviral-driven short hairpin RNA and another that uses retroviral-mediated overexpression. Altering endoglin expression had functional consequences for hematopoietic progenitors in vitro such that endoglin-suppressed myeloid progenitors (colony-forming unit-granulocyte macrophage) displayed a higher degree of sensitivity to TGF-beta-mediated growth inhibition, whereas endoglin-overexpressing cells were partially resistant. However, transplantation of transduced bone marrow enriched in primitive hematopoietic stem and progenitor cells revealed that neither endoglin suppression nor endoglin overexpression affected the ability of stem cells to short-term or long-term repopulate recipient marrow. Furthermore, transplantation of cells altered in endoglin expression yielded normal white blood cell proportions and peripheral blood platelets. Interestingly, decreasing endoglin expression increased the clonogenic capacity of early blast-forming unit-erythroid progenitors, whereas overexpression compromised erythroid differentiation at the basophilic erythroblast phase, suggesting a pivotal role for endoglin at key stages of adult erythropoietic development.


Subject(s)
Aging/physiology , Erythropoiesis/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Intracellular Signaling Peptides and Proteins/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Endoglin , HeLa Cells , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells
16.
J Exp Med ; 204(3): 467-74, 2007 Mar 19.
Article in English | MEDLINE | ID: mdl-17353364

ABSTRACT

Members of the transforming growth factor beta (TGF-beta) superfamily of growth factors have been shown to regulate the in vitro proliferation and maintenance of hematopoietic stem cells (HSCs). Working at a common level of convergence for all TGF-beta superfamily signals, Smad4 is key in orchestrating these effects. The role of Smad4 in HSC function has remained elusive because of the early embryonic lethality of the conventional knockout. We clarify its role by using an inducible model of Smad4 deletion coupled with transplantation experiments. Remarkably, systemic induction of Smad4 deletion through activation of MxCre was incompatible with survival 4 wk after induction because of anemia and histopathological changes in the colonic mucosa. Isolation of Smad4 deletion to the hematopoietic system via several transplantation approaches demonstrated a role for Smad4 in the maintenance of HSC self-renewal and reconstituting capacity, leaving homing potential, viability, and differentiation intact. Furthermore, the observed down-regulation of notch1 and c-myc in Smad4(-/-) primitive cells places Smad4 within a network of genes involved in the regulation HSC renewal.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Smad4 Protein/physiology , Animals , Cell Differentiation/genetics , Down-Regulation/genetics , Genes, Lethal , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Immunophenotyping , Inflammation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/biosynthesis , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/biosynthesis , Signal Transduction/genetics , Smad4 Protein/deficiency , Smad4 Protein/genetics , Transforming Growth Factor beta/deficiency , Transforming Growth Factor beta/genetics
17.
Blood ; 108(13): 4246-54, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16917010

ABSTRACT

The Smad-signaling pathway downstream of the transforming growth factor-beta superfamily of ligands is an evolutionarily conserved signaling circuitry with critical functions in a wide variety of biologic processes. To investigate the role of this pathway in the regulation of hematopoietic stem cells (HSCs), we have blocked Smad signaling by retroviral gene transfer of the inhibitory Smad7 to murine HSCs. We report here that the self-renewal capacity of HSCs is promoted in vivo upon blocking of the entire Smad pathway, as shown by both primary and secondary bone marrow (BM) transplantations. Importantly, HSCs overexpressing Smad7 have an unperturbed differentiation capacity as evidenced by normal contribution to both lymphoid and myeloid cell lineages, suggesting that the Smad pathway regulates self-renewal independently of differentiation. Moreover, phosphorylation of Smads was inhibited in response to ligand stimulation in BM cells, thus verifying impairment of the Smad-signaling cascade in Smad7-overexpressing cells. Taken together, these data reveal an important and previously unappreciated role for the Smad-signaling pathway in the regulation of self-renewal of HSCs in vivo.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Myeloid Progenitor Cells/physiology , Signal Transduction/physiology , Smad7 Protein/metabolism , Animals , Cells, Cultured , Gene Expression , Ligands , Mice , Myeloid Progenitor Cells/cytology , Phosphorylation , Protein Processing, Post-Translational/physiology , Smad7 Protein/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
18.
Blood ; 108(12): 3707-12, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16896158

ABSTRACT

Smad5 is known to transduce intracellular signals from bone morphogenetic proteins (BMPs), which belong to the transforming growth factor-beta (TGF-beta) superfamily and are involved in the regulation of hematopoiesis. Recent findings suggest that BMP4 stimulates proliferation of human primitive hematopoietic progenitors in vitro, while early progenitors from mice deficient in Smad5 display increased self-renewal capacity in murine embryonic hematopoiesis. Here, we evaluate the role of Smad5 in the regulation of hematopoietic stem cell (HSC) fate decisions in adult mice by using an inducible MxCre-mediated conditional knockout model. Surprisingly, analysis of induced animals revealed unperturbed cell numbers and lineage distribution in peripheral blood (PB), bone marrow (BM), and the spleen. Furthermore, phenotypic characterization of the stem cell compartment revealed normal numbers of primitive lin(-)Sca-1(+)c-Kit(+) (LSK) cells in Smad5(-)(/)(-) BM. When transplanted in a competitive fashion into lethally irradiated primary and secondary recipients, Smad5-deficient BM cells competed normally with wild-type (wt) cells, were able to provide long-term reconstitution for the hosts, and displayed normal lineage distribution. Taken together, Smad5-deficient HSCs from adult mice show unaltered differentiation, proliferation, and repopulating capacity. Therefore, in contrast to its role in embryonic hematopoiesis, Smad5 is dispensable for hematopoiesis in the adult mouse.


Subject(s)
Cell Differentiation , Cell Proliferation , Hematopoiesis , Hematopoietic Stem Cells/physiology , Smad5 Protein/metabolism , Animals , Antigens, Differentiation/metabolism , Bone Marrow/physiology , Cell Differentiation/genetics , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Hematopoiesis/genetics , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/cytology , Leukocyte Count , Mice , Mice, Knockout , Smad5 Protein/deficiency , Spleen/cytology , Spleen/physiology , Stem Cell Transplantation/methods , Transplantation Chimera/physiology , Whole-Body Irradiation/methods
SELECTION OF CITATIONS
SEARCH DETAIL