Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Drug Metab Dispos ; 51(10): 1391-1402, 2023 10.
Article in English | MEDLINE | ID: mdl-37524541

ABSTRACT

Numerous biomedical applications have been described for liver-humanized mouse models, such as in drug metabolism or drug-drug interaction (DDI) studies. However, the strong enlargement of the bile acid (BA) pool due to lack of recognition of murine intestine-derived fibroblast growth factor-15 by human hepatocytes and a resulting upregulation in the rate-controlling enzyme for BA synthesis, cytochrome P450 (CYP) 7A1, may pose a challenge in interpreting the results obtained from such mice. To address this challenge, the human fibroblast growth factor-19 (FGF19) gene was inserted into the Fah-/- , Rag2-/- , Il2rg-/- NOD (FRGN) mouse model, allowing repopulation with human hepatocytes capable of responding to FGF19. While a decrease in CYP7A1 expression in human hepatocytes from humanized FRGN19 mice (huFRGN19) and a concomitant reduction in BA production was previously shown, a detailed analysis of the BA pool in these animals has not been elucidated. Furthermore, there are sparse data on the use of this model to assess potential clinical DDI. In the present work, the change in BA composition in huFRGN19 compared with huFRGN control animals was systematically evaluated, and the ability of the model to recapitulate a clinically described CYP3A4-mediated DDI was assessed. In addition to a massive reduction in the total amount of BA, FGF19 expression in huFRGN19 mice resulted in significant changes in the profile of various primary, secondary, and sulfated BAs in serum and feces. Moreover, as observed clinically, administration of the pregnane X receptor agonist rifampicin reduced the oral exposure of the CYP3A4 substrate triazolam. SIGNIFICANCE STATEMENT: Transgenic expression of FGF19 normalizes the unphysiologically high level of bile acids in a chimeric liver-humanized mouse model and leads to massive changes in bile acid composition. These adaptations could overcome one of the potential impediments in the use of these mouse models for drug-drug interaction studies.


Subject(s)
Bile Acids and Salts , Cytochrome P-450 CYP3A , Mice , Humans , Animals , Bile Acids and Salts/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Mice, Inbred NOD , Liver/metabolism , Disease Models, Animal , Fibroblast Growth Factors/metabolism , Drug Interactions
2.
Cell Stem Cell ; 28(1): 33-47.e8, 2021 01 07.
Article in English | MEDLINE | ID: mdl-32997960

ABSTRACT

Bone marrow failure (BMF) in Fanconi anemia (FA) patients results from dysfunctional hematopoietic stem and progenitor cells (HSPCs). To identify determinants of BMF, we performed single-cell transcriptome profiling of primary HSPCs from FA patients. In addition to overexpression of p53 and TGF-ß pathway genes, we identified high levels of MYC expression. We correspondingly observed coexistence of distinct HSPC subpopulations expressing high levels of TP53 or MYC in FA bone marrow (BM). Inhibiting MYC expression with the BET bromodomain inhibitor (+)-JQ1 reduced the clonogenic potential of FA patient HSPCs but rescued physiological and genotoxic stress in HSPCs from FA mice, showing that MYC promotes proliferation while increasing DNA damage. MYC-high HSPCs showed significant downregulation of cell adhesion genes, consistent with enhanced egress of FA HSPCs from bone marrow to peripheral blood. We speculate that MYC overexpression impairs HSPC function in FA patients and contributes to exhaustion in FA bone marrow.


Subject(s)
Fanconi Anemia , Animals , Bone Marrow , DNA Damage , Fanconi Anemia/genetics , Hematopoietic Stem Cells , Humans , Mice , Transforming Growth Factor beta
3.
J Biol Chem ; 295(12): 3906-3917, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32054685

ABSTRACT

Transferrin receptor 2 (TFR2) is a transmembrane protein expressed mainly in hepatocytes and in developing erythroid cells and is an important focal point in systemic iron regulation. Loss of TFR2 function results in a rare form of the iron-overload disease hereditary hemochromatosis. Although TFR2 in the liver has been shown to be important for regulating iron homeostasis in the body, TFR2's function in erythroid progenitors remains controversial. In this report, we analyzed TFR2-deficient mice in the presence or absence of iron overload to distinguish between the effects caused by a high iron load and those caused by loss of TFR2 function. Analysis of bone marrow from TFR2-deficient mice revealed a reduction in the early burst-forming unit-erythroid and an expansion of late-stage erythroblasts that was independent of iron overload. Spleens of TFR2-deficient mice displayed an increase in colony-forming unit-erythroid progenitors and in all erythroblast populations regardless of iron overload. This expansion of the erythroid compartment coincided with increased erythroferrone (ERFE) expression and serum erythropoietin (EPO) levels. Rescue of hepatic TFR2 expression normalized hepcidin expression and the total cell count of the bone marrow and spleen, but it had no effect on erythroid progenitor frequency. On the basis of these results, we propose a model of TFR2's function in murine erythropoiesis, indicating that deficiency in this receptor is associated with increased erythroid development and expression of EPO and ERFE in extrahepatic tissues independent of TFR's role in the liver.


Subject(s)
Erythropoiesis , Iron Overload/pathology , Receptors, Transferrin/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cytokines/metabolism , Erythropoietin/blood , Hepcidins/metabolism , Iron Overload/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/metabolism , Receptors, Transferrin/deficiency , Spleen/pathology , Stem Cells/cytology , Stem Cells/metabolism
4.
Stem Cells ; 33(11): 3304-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26119982

ABSTRACT

Based on its physical interactions with histone-modifying enzymes, the transcriptional corepressor Rcor1 has been implicated in the epigenetic regulation blood cell development. Previously, we have demonstrated that Rcor1 is essential for the maturation of definitive erythroid cells and fetal survival. To determine the functional role of Rcor1 in steady-state hematopoiesis in the adult, we used a conditional knockout approach. Here, we show that the loss of Rcor1 expression results in the rapid onset of severe anemia due to a complete, cell autonomous block in the maturation of committed erythroid progenitors. By contrast, both the frequency of megakaryocyte progenitors and their capacity to produce platelets were normal. Although the frequency of common lymphoid progenitors and T cells was not altered, B cells were significantly reduced and showed increased apoptosis. However, Rcor1-deficient bone marrow sustained normal levels of B-cells following transplantation, indicating a non-cell autonomous requirement for Rcor1 in B-cell survival. Evaluation of the myelomonocytic lineage revealed an absence of mature neutrophils and a significant increase in the absolute number of monocytic cells. Rcor1-deficient monocytes were less apoptotic and showed ∼100-fold more colony-forming activity than their normal counterparts, but did not give rise to leukemia. Moreover, Rcor1(-/-) monocytes exhibited extensive, cytokine-dependent self-renewal and overexpressed genes associated with hematopoietic stem/progenitor cell expansion including Gata2, Meis1, and Hoxa9. Taken together, these data demonstrate that Rcor1 is essential for the normal differentiation of myeloerythroid progenitors and for appropriately regulating self-renewal activity in the monocyte lineage.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Co-Repressor Proteins/metabolism , Erythroid Precursor Cells/metabolism , Neutrophils/metabolism , Animals , Erythroid Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/metabolism
5.
Radiat Res ; 183(3): 338-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25738896

ABSTRACT

Bone marrow suppression due to exposure to ionizing radiation is a significant clinical problem associated with radiation therapy as well as with nonmedical radiation exposure. Currently, there are no small molecule agents available that can enhance hematopoietic regeneration after radiation exposure. Here, we report on the effective mitigation of acute hematopoietic radiation syndrome in mice by the synthetic triterpenoid, RTA 408. The administration of a brief course of RTA 408 treatment, beginning 24 h after lethal doses of radiation to bone marrow, significantly increased overall survival. Importantly, treatment with RTA 408 led to the full recovery of steady state hematopoiesis with normalization of the frequency of hematopoietic stem and progenitor cells. Moreover, hematopoietic stem cells from RTA 408-mitigated mice showed lineage-balanced, long-term, multilineage potential in serial transplantation assays, indicative of their normal self-renewal activity. The potency of RTA 408 in mitigating radiation-induced bone marrow suppression makes it an attractive candidate for potential clinical use in treating both therapy-related and unanticipated radiation exposure.


Subject(s)
Acute Radiation Syndrome/drug therapy , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Triterpenes/administration & dosage , Acute Radiation Syndrome/pathology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Dose-Response Relationship, Radiation , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/radiation effects , Mice , Radiation, Ionizing , Radiotherapy/adverse effects , Whole-Body Irradiation
6.
Blood ; 123(20): 3175-84, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24652990

ABSTRACT

The corepressor Rcor1 has been linked biochemically to hematopoiesis, but its function in vivo remains unknown. We show that mice deleted for Rcor1 are profoundly anemic and die in late gestation. Definitive erythroid cells from mutant mice arrest at the transition from proerythroblast to basophilic erythroblast. Remarkably, Rcor1 null erythroid progenitors cultured in vitro form myeloid colonies instead of erythroid colonies. The mutant proerythroblasts also aberrantly express genes of the myeloid lineage as well as genes typical of hematopoietic stem cells (HSCs) and/or progenitor cells. The colony-stimulating factor 2 receptor ß subunit (Csf2rb), which codes for a receptor implicated in myeloid cytokine signaling, is a direct target for both Rcor1 and the transcription repressor Gfi1b in erythroid cells. In the absence of Rcor1, the Csf2rb gene is highly induced, and Rcor1(-/-) progenitors exhibit CSF2-dependent phospho-Stat5 hypersensitivity. Blocking this pathway can partially reduce myeloid colony formation by Rcor1-deficient erythroid progenitors. Thus, Rcor1 promotes erythropoiesis by repressing HSC and/or progenitor genes, as well as the genes and signaling pathways that lead to myeloid cell fate.


Subject(s)
Co-Repressor Proteins/metabolism , Erythropoiesis , Animals , Cells, Cultured , Co-Repressor Proteins/genetics , Cytokine Receptor Common beta Subunit/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Erythroblasts/cytology , Erythroblasts/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Gene Deletion , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Myeloid Cells/cytology , Receptors, Interleukin-3/metabolism , Signal Transduction
7.
Leukemia ; 28(10): 1978-1987, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24637335

ABSTRACT

Vascular endothelial cells are a critical component of the hematopoietic microenvironment that regulates blood cell production. Recent studies suggest the existence of functional cross-talk between hematologic malignancies and vascular endothelium. Here we show that human acute myeloid leukemia (AML) localizes to the vasculature in both patients and in a xenograft model. A significant number of vascular tissue-associated AML cells (V-AML) integrate into vasculature in vivo and can fuse with endothelial cells. V-AML cells acquire several endothelial cell-like characteristics, including the upregulation of CD105, a receptor associated with activated endothelium. Remarkably, endothelial-integrated V-AML shows an almost fourfold reduction in proliferative activity compared with non-vascular-associated AML. Primary AML cells can be induced to downregulate the expression of their hematopoietic markers in vitro and differentiate into phenotypically and functionally defined endothelial-like cells. After transplantation, these leukemia-derived endothelial cells are capable of giving rise to AML. These novel functional interactions between AML cells and normal endothelium along with the reversible endothelial cell potential of AML suggest that vascular endothelium may serve as a previously unrecognized reservoir for AML.


Subject(s)
Endothelium, Vascular/metabolism , Leukemia, Myeloid, Acute/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Cell Differentiation , Cell Line , Cell Survival , Cells, Cultured , Endoglin , Female , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Mice, Inbred NOD , Middle Aged , Neoplasm Transplantation , Phenotype , Receptors, Cell Surface/metabolism , Recurrence , Young Adult
8.
Biol Blood Marrow Transplant ; 20(1): 132-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24161922

ABSTRACT

Human cytomegalovirus (HCMV) infection, including primary infection resulting from transmission from a seropositive donor to a seronegative recipient (D(+)/R(-)), remains a significant problem in the setting of peripheral blood stem cell transplantation (PBSCT). The lack of a suitable animal model for studying HCMV transmission after PBSCT is a major barrier to understanding this process and, consequently, developing novel interventions to prevent HCMV infection. Our previous work demonstrated that human CD34(+) progenitor cell-engrafted NOD-scid IL2Rγc(null) (NSG) mice support latent HCMV infection after direct inoculation and reactivation after treatment with granulocyte colony-stimulating factor. To more accurately recapitulate HCMV infection in the D(+)/R(-) PBSCT setting, granulocyte colony-stimulating factor-mobilized peripheral blood stem cells from seropositive donors were used to engraft NSG mice. All recipient mice demonstrated evidence of HCMV infection in liver, spleen, and bone marrow. These findings validate the NSG mouse model for studying HCMV transmission during PBSCT.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Peripheral Blood Stem Cell Transplantation , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Bone Marrow/virology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Humans , Liver/immunology , Liver/pathology , Liver/virology , Mice , Mice, Transgenic , Spleen/immunology , Spleen/pathology , Spleen/virology , Transplantation, Heterologous , Viral Load , Virus Activation , Virus Replication
9.
Stem Cell Res ; 11(3): 1013-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23939266

ABSTRACT

Endothelial cells (ECs) are an essential component of the hematopoietic microenvironment, which maintains and regulates hematopoietic stem cells (HSCs). Although ECs can support the regeneration of otherwise lethally-irradiated HSCs, the mechanisms are not well understood. To further understand this phenomenon, we studied HSC regeneration from irradiated bone marrow using co-culture with human aortic ECs (HAECs). Co-culture with HAECs induced a 24-fold expansion of long-term HSCs (CD150(+), lineage(lo), Sca-1(+), c-Kit(+); CD150(+)LSK cells) in vitro. These cells gave rise to functional hematopoietic stem and progenitor cells (HSPCs) with colony-forming activity, multilineage reconstitution and serial transplantation potential. Furthermore, HAECs significantly reduced DNA damage in irradiated LSK cells within 24h. Remarkably, we were able to delay the exposure of irradiated bone marrow to the regenerative, HAEC-derived signals for up to 48h and still rescue functional HSCs. G-CSF is the gold standard for promoting hematopoietic regeneration in vivo. However, when compared to HAECs, in vitro G-CSF treatment promoted lineage differentiation and regenerated 5-fold fewer CD150(+)LSK cells. Together, our results show that HAECs are powerful, direct mitigators of HSC injury and DNA damage. Identification of the HAEC-derived factors that rescue HSCs may lead to improved therapies for hematopoietic regeneration after radiation injury.


Subject(s)
DNA Damage/radiation effects , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/radiation effects , Whole-Body Irradiation , Animals , Aorta/cytology , Bone Marrow Cells/cytology , Bone Marrow Cells/radiation effects , Bone Marrow Transplantation , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis , Humans , Male , Mice
10.
PLoS Pathog ; 7(12): e1002444, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22241980

ABSTRACT

Clinical strains of HCMV encode 20 putative ORFs within a region of the genome termed ULb' that are postulated to encode functions related to persistence or immune evasion. We have previously identified ULb'-encoded pUL138 as necessary, but not sufficient, for HCMV latency in CD34+ hematopoietic progenitor cells (HPCs) infected in vitro. pUL138 is encoded on polycistronic transcripts that also encode 3 additional proteins, pUL133, pUL135, and pUL136, collectively comprising the UL133-UL138 locus. This work represents the first characterization of these proteins and identifies a role for this locus in infection. Similar to pUL138, pUL133, pUL135, and pUL136 are integral membrane proteins that partially co-localized with pUL138 in the Golgi during productive infection in fibroblasts. As expected of ULb' sequences, the UL133-UL138 locus was dispensable for replication in cultured fibroblasts. In CD34+ HPCs, this locus suppressed viral replication in HPCs, an activity attributable to both pUL133 and pUL138. Strikingly, the UL133-UL138 locus was required for efficient replication in endothelial cells. The association of this locus with three context-dependent phenotypes suggests an exciting role for the UL133-UL138 locus in modulating the outcome of viral infection in different contexts of infection. Differential profiles of protein expression from the UL133-UL138 locus correlated with the cell-type dependent phenotypes associated with this locus. We extended our in vitro findings to analyze viral replication and dissemination in a NOD-scid IL2Rγ(c) (null)-humanized mouse model. The UL133-UL138(NULL) virus exhibited an increased capacity for replication and/or dissemination following stem cell mobilization relative to the wild-type virus, suggesting an important role in viral persistence and spread in the host. As pUL133, pUL135, pUL136, and pUL138 are conserved in virus strains infecting higher order primates, but not lower order mammals, the functions encoded likely represent host-specific viral adaptations.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Genetic Loci , Hematopoietic Stem Cells/virology , Host-Pathogen Interactions/physiology , Viral Tropism/physiology , Virus Replication/physiology , Animals , Cell Line , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/pathology , Disease Models, Animal , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID
11.
Cell Host Microbe ; 8(3): 284-91, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20833379

ABSTRACT

Human cytomegalovirus (HCMV) is a significant cause of morbidity and mortality in organ transplant recipients. The use of granulocyte-colony stimulating factor (G-CSF)-mobilized stem cells from HCMV seropositive donors is suggested to double the risk of late-onset HCMV disease and chronic graft-versus-host disease in recipients when compared to conventional bone marrow transplantation with HCMV seropositive donors, although the etiology of the increased risk is unknown. To understand mechanisms of HCMV transmission in patients receiving G-CSF-mobilized blood products, we generated a NOD-scid IL2Rγ(c)(null)-humanized mouse model in which HCMV establishes latent infection in human hematopoietic cells. In this model, G-CSF induces the reactivation of latent HCMV in monocytes/macrophages that have migrated into organ tissues. In addition to establishing a humanized mouse model for systemic and latent HCMV infection, these results suggest that the use of G-CSF mobilized blood products from seropositive donors pose an elevated risk for HCMV transmission to recipients.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Macrophages/virology , Virus Activation , Virus Latency , Animals , Antigens, CD34/analysis , Bone Marrow Cells/immunology , Bone Marrow Cells/virology , Cytokines/blood , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus/isolation & purification , Disease Models, Animal , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/virology , Humans , Mice , Mice, SCID , Monocytes/virology
12.
Blood ; 116(24): 5140-8, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20826722

ABSTRACT

Progressive bone marrow failure is a major cause of morbidity and mortality in human Fanconi Anemia patients. In an effort to develop a Fanconi Anemia murine model to study bone marrow failure, we found that Fancd2(-/-) mice have readily measurable hematopoietic defects. Fancd2 deficiency was associated with a significant decline in the size of the c-Kit(+)Sca-1(+)Lineage(-) (KSL) pool and reduced stem cell repopulation and spleen colony-forming capacity. Fancd2(-/-) KSL cells showed an abnormal cell cycle status and loss of quiescence. In addition, the supportive function of the marrow microenvironment was compromised in Fancd2(-/-) mice. Treatment with Sirt1-mimetic and the antioxidant drug, resveratrol, maintained Fancd2(-/-) KSL cells in quiescence, improved the marrow microenvironment, partially corrected the abnormal cell cycle status, and significantly improved the spleen colony-forming capacity of Fancd2(-/-) bone marrow cells. We conclude that Fancd2(-/-) mice have readily quantifiable hematopoietic defects, and that this model is well suited for pharmacologic screening studies.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia/drug therapy , Hematopoietic System/drug effects , Stilbenes/pharmacology , Animals , Antioxidants , Bone Marrow/drug effects , Cell Cycle , Cell Lineage , Colony-Forming Units Assay , Fanconi Anemia Complementation Group D2 Protein/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Resveratrol , Spleen/cytology , Stilbenes/therapeutic use , Treatment Outcome
13.
Stem Cell Res ; 4(1): 17-24, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19720572

ABSTRACT

Recent studies suggest that endothelial cells are a critical component of the normal hematopoietic microenvironment. Therefore, we sought to determine whether primary endothelial cells have the capacity to repair damaged hematopoietic stem cells. Highly purified populations of primary CD31(+) microvascular endothelial cells isolated from the brain or lung did not express the pan hematopoietic marker CD45, most hematopoietic lineage markers, or the progenitor marker c-kit and did not give rise to hematopoietic cells in vitro or in vivo. Remarkably, the transplantation of small numbers of these microvascular endothelial cells consistently restored hematopoiesis following bone marrow lethal doses of irradiation. Analysis of the peripheral blood of rescued recipients demonstrated that both short-term and long-term multilineage hematopoietic reconstitution was exclusively of host origin. Secondary transplantation studies revealed that microvascular endothelial cell-mediated hematopoietic regeneration also occurs at the level of the hematopoietic stem cell. These findings suggest a potential therapeutic role for microvascular endothelial cells in the self-renewal and repair of adult hematopoietic stem cells.


Subject(s)
Cell Differentiation , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Animals , Biomarkers , Cell Lineage , Cell Separation , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Hematopoietic Stem Cells/metabolism , Leukocyte Common Antigens/metabolism , Mice , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
14.
Blood ; 114(20): 4393-401, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19759357

ABSTRACT

Bone morphogenetic protein 4 (BMP4) is required for mesoderm commitment to the hematopoietic lineage during early embryogenesis. However, deletion of BMP4 is early embryonically lethal and its functional role in definitive hematopoiesis is unknown. Consequently, we used a BMP4 hypomorph to investigate the role of BMP4 in regulating hematopoietic stem cell (HSC) function and maintaining steady-state hematopoiesis in the adult. Reporter gene expression shows that Bmp4 is expressed in cells associated with the hematopoietic microenvironment including osteoblasts, endothelial cells, and megakaryocytes. Although resting hematopoiesis is normal in a BMP4-deficient background, the number of c-Kit+, Sca-1+, Lineage- cells is significantly reduced. Serial transplantation studies reveal that BMP4-deficient recipients have a microenvironmental defect that reduces the repopulating activity of wild-type HSCs. This defect is even more pronounced in a parabiosis model that demonstrates a profound reduction in wild-type hematopoietic cells within the bone marrow of BMP4-deficient recipients. Furthermore, wild-type HSCs that successfully engraft into the BMP4-deficient bone marrow show a marked decrease in functional stem cell activity when tested in a competitive repopulation assay. Taken together, these findings indicate BMP4 is a critical component of the hematopoietic microenvironment that regulates both HSC number and function.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Stem Cell Niche/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Flow Cytometry , Gene Expression , Mice , Parabiosis
15.
Mech Dev ; 126(3-4): 117-27, 2009.
Article in English | MEDLINE | ID: mdl-19116164

ABSTRACT

Vertebrate Bmp2 and Bmp4 diverged from a common ancestral gene and encode closely related proteins. Mice homozygous for null mutations in either gene show early embryonic lethality, thereby precluding analysis of shared functions. In the current studies, we present phenotypic analysis of compound mutant mice heterozygous for a null allele of Bmp2 in combination with null or hypomorphic alleles of Bmp4. Whereas mice lacking a single copy of Bmp2 or Bmp4 are viable and have subtle developmental defects, compound mutants show embryonic and postnatal lethality due to defects in multiple organ systems including the allantois, placental vasculature, ventral body wall, skeleton, eye and heart. Within the heart, BMP2 and BMP4 function coordinately to direct normal lengthening of the outflow tract, proper positioning of the outflow vessels, and septation of the atria, ventricle and atrioventricular canal. Our results identify numerous BMP4-dependent developmental processes that are also very sensitive to BMP2 dosage, thus revealing novel functions of Bmp2.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 4/genetics , Organogenesis , Animals , Bone and Bones/abnormalities , Chorioallantoic Membrane/embryology , Chorioallantoic Membrane/metabolism , Crosses, Genetic , Embryo Loss/genetics , Embryonic Development , Extremities/embryology , Eye/embryology , Female , Fetus/blood supply , Fetus/metabolism , Gene Deletion , Genotype , Heart Atria/abnormalities , Heart Atria/embryology , Heart Defects, Congenital , Heart Ventricles/abnormalities , Heart Ventricles/embryology , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
16.
PLoS One ; 3(11): e3812, 2008.
Article in English | MEDLINE | ID: mdl-19043576

ABSTRACT

BACKGROUND: Although the lymphatic system arises as an extension of venous vessels in the embryo, little is known about the role of circulating progenitors in the maintenance or development of lymphatic endothelium. Here, we investigated whether hematopoietic stem cells (HSCs) have the potential to give rise to lymphatic endothelial cells (LEC). METHODOLOGY/PRINCIPAL FINDINGS: Following the transfer of marked HSCs into irradiated recipients, donor-derived LEC that co-express the lymphatic endothelial markers Lyve-1 and VEGFR-3 were identified in several tissues. HSC-derived LEC persisted for more than 12 months and contributed to approximately 3-4% of lymphatic vessels. Donor-derived LECs were not detected in mice transplanted with common myeloid progenitors and granulocyte/macrophage progenitors, suggesting that myeloid lineage commitment is not a requisite step in HSC contribution to lymphatic endothelium. Analysis of parabiotic mice revealed direct evidence for the existence of functional, circulating lymphatic progenitors in the absence of acute injury. Furthermore, the transplantation of HSCs into Apc(Min/+) mice resulted in the incorporation of donor-derived LEC into the lymphatic vessels of spontaneously arising intestinal tumors. CONCLUSIONS/SIGNIFICANCE: Our results indicate that HSCs can contribute to normal and tumor associated lymphatic endothelium. These findings suggest that the modification of HSCs may be a novel approach for targeting tumor metastasis and attenuating diseases of the lymphatic system.


Subject(s)
Endothelium, Lymphatic/cytology , Hematopoietic Stem Cells/physiology , Stem Cell Transplantation , Animals , Biomarkers/analysis , Granulocytes , Hematopoietic Stem Cells/cytology , Intestinal Neoplasms/pathology , Macrophages , Mice , Myeloid Cells
17.
J Exp Med ; 205(8): 1843-58, 2008 Aug 04.
Article in English | MEDLINE | ID: mdl-18625744

ABSTRACT

In mantle cell lymphoma (MCL) and some cases of multiple myeloma (MM), cyclin D1 expression is deregulated by chromosome translocations involving the immunoglobulin heavy chain (IgH) locus. To evaluate the mechanisms responsible, gene targeting was used to study long-distance gene regulation. Remarkably, these targeted cell lines lost the translocated chromosome (t(11;14)). In these MCL and MM cells, the nonrearranged cyclin D1 (CCND1) locus reverts from CpG hypomethylated to hypermethylated. Reintroduction of the translocated chromosome induced a loss of methylation at the unrearranged CCND1 locus, providing evidence of a transallelic regulatory effect. In these cell lines and primary MCL patient samples, the CCND1 loci are packaged in chromatin-containing CCCTC binding factor (CTCF) and nucleophosmin (NPM) at the nucleolus. We show that CTCF and NPM are bound at the IgH 3' regulatory elements only in the t(11;14) MCL cell lines. Furthermore, NPM short hairpin RNA produces a specific growth arrest in these cells. Our data demonstrate transvection in human cancer and suggest a functional role for CTCF and NPM.


Subject(s)
Genes, bcl-1 , Lymphoma, Mantle-Cell/genetics , Translocation, Genetic , CCCTC-Binding Factor , Cell Line, Tumor , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , CpG Islands , Cyclin D , Cyclins/genetics , Cyclins/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Targeting , Genes, Immunoglobulin , Humans , Hybrid Cells , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Promoter Regions, Genetic , Repressor Proteins/metabolism
18.
Dev Biol ; 310(2): 454-69, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17850784

ABSTRACT

In Xenopus, primitive blood originates from the mesoderm, but extrinsic signals from the ectoderm are required during gastrulation to enable these cells to differentiate as erythrocytes. The nature of these signals, and how they are transcriptionally regulated, is not well understood. We have previously shown that bone morphogenetic proteins (BMPs) are required to signal to ectodermal cells to generate secondary non-cell-autonomous signal(s) necessary for primitive erythropoiesis, and that calmodulin-dependent protein kinase IV (CaM KIV) antagonizes BMP signaling. The current studies demonstrate that Gata-2 functions downstream of BMP receptor activation in these same cells, and is a direct target for antagonism by CaM KIV. We show, using loss of function analysis in whole embryos and in explants, that ectodermal Gata-2 is required for primitive erythropoiesis, and that BMP signals cannot rescue blood defects caused by ectoderm removal or loss of ectodermal GATA-2. Furthermore, we provide evidence that acetylation of GATA-2 is required for its function in primitive blood formation in vivo. Our data support a model in which Gata-2 is a transcriptional target downstream of BMPs within ectodermal cells, while activation of the CaM KIV signaling pathway alters GATA-2 function posttranslationally, by inhibiting its acetylation.


Subject(s)
Bone Morphogenetic Proteins/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 4/physiology , GATA2 Transcription Factor/physiology , Hematopoiesis , Xenopus Proteins/physiology , Xenopus/physiology , Animals , Ectoderm/physiology , Erythropoiesis , Female , Xenopus/embryology
19.
Dev Biol ; 298(2): 415-29, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16887115

ABSTRACT

Gata4, Gata5, and Gata6 represent a subfamily of zinc-finger transcriptional regulators that are important in the development and differentiation of numerous tissues, including many endodermally-derived organs. We demonstrate that Gata4 and Gata6 have overlapping expression patterns in the early pancreatic epithelium. Later, Gata4 becomes restricted to exocrine tissue and Gata6 becomes restricted to a subset of endocrine cells. In addition, we show Gata6, but not Gata4, physically interacts with Nkx2.2, an essential islet transcription factor. To begin determining the roles that Gata4 and Gata6 play during pancreatic development, we expressed Gata4-Engrailed and Gata6-Engrailed dominant repressor fusion proteins in the pancreatic epithelium and in the islet. At e17.5, transgenic Gata6-Engrailed embryos exhibit two distinct phenotypes: a complete absence of pancreas or a reduction in pancreatic tissue. In the embryos that do form pancreas, there is a significant reduction of all pancreatic cell types, with the few differentiated endocrine cells clustered within, or in close proximity to, enlarged ductal structures. Conversely, the majority of transgenic Gata4-Engrailed embryos do not have a pancreatic phenotype. This study suggests that Gata6 is an important regulator of pancreas specification.


Subject(s)
GATA4 Transcription Factor/metabolism , GATA6 Transcription Factor/physiology , Pancreas/embryology , Animals , Cell Differentiation , Cell Line , Cell Lineage , Drosophila Proteins , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Organogenesis , Pancreas/metabolism , Promoter Regions, Genetic , Rats , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins
20.
Development ; 133(10): 1933-42, 2006 May.
Article in English | MEDLINE | ID: mdl-16624858

ABSTRACT

ProBMP4 is initially cleaved at a site adjacent to the mature ligand (the S1 site) allowing for subsequent cleavage at an upstream (S2) site. Mature BMP4 synthesized from a precursor in which the S2 site cannot be cleaved remains in a complex with the prodomain that is targeted for lysosomal degradation, and is thus less active when overexpressed in Xenopus. Here we report that mice carrying a point mutation that prevents S2 processing show severe loss of BMP4 activity in some tissues, such as testes and germ cells, whereas other tissues that are sensitive to Bmp4 dosage, such as the limb, dorsal vertebrae and kidney, develop normally. In a haploinsufficient background, inability to cleave the S2 site leads to embryonic and postnatal lethality due to defects in multiple organ systems including the allantois, placental vasculature, ventral body wall, eye and heart. These data demonstrate that cleavage of the S2 site is essential for normal development and, more importantly, suggest that this site might be selectively cleaved in a tissue-specific fashion. In addition, these studies provide the first genetic evidence that BMP4 is required for dorsal vertebral fusion and closure of the ventral body wall.


Subject(s)
Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/metabolism , Germ Cells/metabolism , Point Mutation , Testis/metabolism , Animals , Bone Morphogenetic Protein 4 , Embryonic Development/genetics , Gene Dosage , Gene Targeting , Germ Cells/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Organ Specificity , Protein Structure, Tertiary , Testis/cytology , Xenopus Proteins
SELECTION OF CITATIONS
SEARCH DETAIL