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










Publication year range
1.
Blood Adv ; 5(21): 4435-4446, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34581760

ABSTRACT

Osteoclasts are multinuclear cells of monocytic lineage, with the ability to resorb bone. Studies in mouse have identified bone marrow clonal progenitors able to generate mature osteoclast cells (OCs) in vitro and in vivo. These osteoclast progenitors (OCPs) can also generate macrophages and dendritic cells. Interestingly, cells with equivalent potential can be detected in periphery. In humans, cells with OCP activity have been identified in bone marrow and periphery; however, their characterization has not been as extensive. We have developed reproducible methods to derive, from human pluripotent stem cells, a population containing monocyte progenitors able to generate functional OCs. Within this population, we have identified cells with monocyte and osteoclast progenitor activity based on CD11b and CD14 expression. A population double positive for CD11b and CD14 contains cells with expected osteoclastic potential. However, the double negative (DN) population, containing most of the hematopoietic progenitor activity, also presents a very high osteoclastic potential. These progenitor cells can also be differentiated to macrophage and dendritic cells. Further dissection within the DN population identified cells bearing the phenotype CD15-CD115+ as the population with highest monocytic progenitor and osteoclastic potential. When similar methodology was used to identify OCPs from human peripheral blood, we confirmed a published OCP population with the phenotype CD11b+CD14+. In addition, we identified a second population (CD14-CD11bloCD115+) with high monocytic progenitor activity that was also able to form osteoclast like cells, similar to the 2 populations identified from pluripotent stem cells.


Subject(s)
Osteoclasts , Pluripotent Stem Cells , Animals , Bone Marrow Cells , Cell Differentiation , Humans , Mice , Monocytes
2.
J Immunol ; 204(4): 868-878, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31915261

ABSTRACT

Osteoclasts (OC) originate from either bone marrow (BM)-resident or circulating myeloid OC progenitors (OCP) expressing the receptor CX3CR1. Multiple lines of evidence argue that OCP in homeostasis and inflammation differ. We investigated the relative contributions of BM-resident and circulating OCP to osteoclastogenesis during homeostasis and fracture repair. Using CX3CR1-EGFP/TRAP tdTomato mice, we found CX3CR1 expression in mononuclear cells, but not in multinucleated TRAP+ OC. However, CX3CR1-expressing cells generated TRAP+ OC on bone within 5 d in CX3CR1CreERT2/Ai14 tdTomato reporter mice. To define the role that circulating cells play in osteoclastogenesis during homeostasis, we parabiosed TRAP tdTomato mice (CD45.2) on a C57BL/6 background with wild-type (WT) mice (CD45.1). Flow cytometry (CD45.1/45.2) demonstrated abundant blood cell mixing between parabionts after 2 wk. At 4 wk, there were numerous tdTomato+ OC in the femurs of TRAP tdTomato mice but almost none in WT mice. Similarly, cultured BM stimulated to form OC demonstrated multiple fluorescent OC in cell cultures from TRAP tdTomato mice, but not from WT mice. Finally, flow cytometry confirmed low-level engraftment of BM cells between parabionts but significant engraftment in the spleens. In contrast, during fracture repair, we found that circulating CX3CR1+ cells migrated to bone, lost expression of CX3CR1, and became OC. These data demonstrate that OCP, but not mature OC, express CX3CR1 during both homeostasis and fracture repair. We conclude that, in homeostasis mature OC derive predominantly from BM-resident OCP, whereas during fracture repair, circulating CX3CR1+ cells can become OC.

3.
J Biol Chem ; 291(8): 3882-94, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26703472

ABSTRACT

Continuous parathyroid hormone (PTH) blocks its own osteogenic actions in marrow stromal cell cultures by inducing Cox2 and receptor activator of nuclear factor κB ligand (RANKL) in the osteoblastic lineage cells, which then cause the hematopoietic lineage cells to secrete an inhibitor of PTH-stimulated osteoblast differentiation. To identify this inhibitor, we used bone marrow macrophages (BMMs) and primary osteoblasts (POBs) from WT and Cox2 knock-out (KO) mice. Conditioned medium (CM) from RANKL-treated WT, but not KO, BMMs blocked PTH-stimulated cAMP production in POBs. Inhibition was reversed by pertussis toxin (PTX), which blocks Gαi/o activation. Saa3 was the most highly differentially expressed gene in a microarray comparison of RANKL-treated WT versus Cox2 KO BMMs, and RANKL induced Saa3 protein secretion only from WT BMMs. CM from RANKL-stimulated BMMs with Saa3 knockdown did not inhibit PTH-stimulated responses in POBs. SAA added to POBs inhibited PTH-stimulated cAMP responses, which was reversed by PTX. Selective agonists and antagonists of formyl peptide receptor 2 (Fpr2) suggested that Fpr2 mediated the inhibitory actions of Saa3 on osteoblasts. In BMMs committed to become osteoclasts by RANKL treatment, Saa3 expression peaked prior to appearance of multinucleated cells. Flow sorting of WT marrow revealed that Saa3 was secreted only from the RANKL-stimulated B220(-) CD3(-)CD11b(-/low) CD115(+) preosteoclast population. We conclude that Saa3 secretion from preosteoclasts, induced by RANKL in a Cox2-dependent manner, inhibits PTH-stimulated cAMP signaling and osteoblast differentiation via Gαi/o signaling. The induction of Saa3 by PTH may explain the suppression of bone formation when PTH is applied continuously and may be a new therapeutic target for osteoporosis.


Subject(s)
Cyclic AMP/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Parathyroid Hormone/pharmacology , Second Messenger Systems/drug effects , Serum Amyloid A Protein/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cyclic AMP/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoclasts/cytology , Osteogenesis/drug effects , Osteogenesis/genetics , Parathyroid Hormone/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Second Messenger Systems/genetics , Serum Amyloid A Protein/genetics
4.
J Neurosci ; 34(11): 3901-9, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24623768

ABSTRACT

Intracerebral hemorrhage (ICH) is a devastating type of stroke that lacks a specific treatment. An intense immune response develops after ICH, which contributes to neuronal injury, disability, and death. However, the specific mediators of inflammation-induced injury remain unclear. The objective of the present study was to determine whether blood-derived CCR2+ Ly6C(hi) inflammatory monocytes contribute to disability. ICH was induced in mice and the resulting inflammatory response was quantified using flow cytometry, confocal microscopy, and neurobehavioral testing. Importantly, blood-derived monocytes were distinguished from resident microglia by differential CD45 staining and by using bone marrow chimeras with fluorescent leukocytes. After ICH, blood-derived CCR2+ Ly6C(hi) inflammatory monocytes trafficked into the brain, outnumbered other leukocytes, and produced tumor necrosis factor. Ccr2(-/-) mice, which have few circulating inflammatory monocytes, exhibited better motor function following ICH than control mice. Chimeric mice with wild-type CNS cells and Ccr2(-/-) hematopoietic cells also exhibited early improvement in motor function, as did wild-type mice after inflammatory monocyte depletion. These findings suggest that blood-derived inflammatory monocytes contribute to acute neurological disability. To determine the translational relevance of our experimental findings, we examined CCL2, the principle ligand for the CCR2 receptor, in ICH patients. Serum samples from 85 patients were collected prospectively at two hospitals. In patients, higher CCL2 levels at 24 h were independently associated with poor functional outcome at day 7 after adjusting for potential confounding variables. Together, these findings suggest that inflammatory monocytes worsen early disability after murine ICH and may represent a therapeutic target for patients.


Subject(s)
Antigens, Ly/genetics , Cerebral Hemorrhage/immunology , Encephalitis/immunology , Monocytes/immunology , Receptors, CCR2/genetics , Acute Disease , Animals , Antigens, Ly/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Brain/immunology , Brain/pathology , Cell Movement/immunology , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Chemokine CCL2/blood , Disability Evaluation , Encephalitis/pathology , Encephalitis/physiopathology , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Movement Disorders/immunology , Movement Disorders/pathology , Movement Disorders/physiopathology , Prospective Studies , Receptors, CCR2/immunology , Stroke/immunology , Stroke/pathology , Stroke/physiopathology
5.
Channels (Austin) ; 8(2): 142-56, 2014.
Article in English | MEDLINE | ID: mdl-24590064

ABSTRACT

Death of murine T cells induced by extracellular ATP is mainly triggered by activation of purinergic P2X 7 receptors (P2X 7Rs). However, a link between P2X 7Rs and pannexin1 (Panx1) channels, which are non-selective, has been recently demonstrated in other cell types. In this work, we characterized the expression and cellular distribution of pannexin family members (Panxs 1, 2 and 3) in isolated T cells. Panx1 was the main pannexin family member clearly detected in both helper (CD4+) and cytotoxic (CD8+) T cells, whereas low levels of Panx2 were found in both T-cell subsets. Using pharmacological and genetic approaches, Panx1 channels were found to mediate most ATP-induced ethidium uptake since this was drastically reduced by Panx1 channel blockers (10Panx1, Probenecid and low carbenoxolone concentration) and absent in T cells derived from Panx1-/- mice. Moreover, electrophysiological measurements in wild-type CD4+ cells treated with ATP unitary current events and pharmacological sensitivity compatible with Panx1 channels were found. In addition, ATP release from T cells treated with 4Br-A23187, a calcium ionophore, was completely blocked with inhibitors of both connexin hemichannels and Panx1 channels. Panx1 channel blockers drastically reduced the ATP-induced T-cell mortality, indicating that Panx1 channels mediate the ATP-induced T-cell death. However, mortality was not reduced in T cells of Panx1-/- mice, in which levels of P2X 7Rs and ATP-induced intracellular free Ca2+ responses were enhanced suggesting that P2X 7Rs take over Panx1 channels lose-function in mediating the onset of cell death induced by extracellular ATP.


Subject(s)
Adenosine Triphosphate/pharmacology , Apoptosis/drug effects , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Purinergic P2X7/metabolism , T-Lymphocytes/drug effects , Adenosine Triphosphate/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/physiology , Calcimycin/pharmacology , Calcium Signaling/drug effects , Cell Membrane Permeability/drug effects , Cells, Cultured , Connexins/antagonists & inhibitors , Connexins/genetics , Humans , Jurkat Cells , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Time-Lapse Imaging
6.
J Bone Miner Res ; 27(5): 1030-42, 2012 May.
Article in English | MEDLINE | ID: mdl-22258693

ABSTRACT

Interleukin-7 is a critical cytokine for lymphoid development and a direct inhibitor of in vitro osteoclastogenesis in murine bone marrow cultures. To explore the role of IL-7 in bone, we generated transgenic mouse lines bearing the 2.3-kb rat collagen 1α1 promoter driving the expression of human IL-7 specifically in osteoblasts. In addition, we crossed these mice with IL-7-deficient mice to determine if the alterations in lymphopoiesis, bone mass, and osteoclast formation observed in the IL-7 knockout (KO) mice could be rescued by osteoblast-specific overexpression of IL-7. Here, we show that mice overexpressing human IL-7 in the osteoblast lineage showed increased trabecular bone volume in vivo by µCT and decreased osteoclast formation in vitro. Furthermore, targeted overexpression of IL-7 in osteoblasts rescued the osteopenic bone phenotype and B-cell development of IL-7 KO mice but did not have an effect on T lymphopoiesis, which occurs in the periphery. The bone phenotypes in IL-7 KO mice and targeted IL-7-overexpressing mouse models were observed only in females. These results likely reflect both direct inhibitory effects of IL-7 on osteoclastogenesis in vivo and sex-specific differences in responses to IL-7.


Subject(s)
Gene Expression Regulation , Interleukin-7/deficiency , Interleukin-7/genetics , Osteoblasts/metabolism , Animals , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Lymphopoiesis , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Polymerase Chain Reaction , Rats , Sex Factors
7.
Stem Cells ; 30(2): 187-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22083974

ABSTRACT

Adult mesenchymal progenitor cells have enormous potential for use in regenerative medicine. However, the true identity of the progenitors in vivo and their progeny has not been precisely defined. We hypothesize that cells expressing a smooth muscle α-actin promoter (αSMA)-directed Cre transgene represent mesenchymal progenitors of adult bone tissue. By combining complementary colors in combination with transgenes activating at mature stages of the lineage, we characterized the phenotype and confirmed the ability of isolated αSMA(+) cells to progress from a progenitor to fully mature state. In vivo lineage tracing experiments using a new bone formation model confirmed the osteogenic phenotype of αSMA(+) cells. In vitro analysis of the in vivo-labeled SMA9(+) cells supported their differentiation potential into mesenchymal lineages. Using a fracture-healing model, αSMA9(+) cells served as a pool of fibrocartilage and skeletal progenitors. Confirmation of the transition of αSMA9(+) progenitor cells to mature osteoblasts during fracture healing was assessed by activation of bone-specific Col2.3emd transgene. Our findings provide a novel in vivo identification of defined population of mesenchymal progenitor cells with active role in bone remodeling and regeneration.


Subject(s)
Cell Lineage , Mesenchymal Stem Cells/metabolism , Actins/genetics , Actins/metabolism , Animals , Antigens, Differentiation/metabolism , Bone Marrow Cells/metabolism , Bone Regeneration , Bone Remodeling , Cell Differentiation , Female , Fracture Healing , Gene Expression Regulation , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Transgenic , Phenotype , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Tibia/pathology
8.
Am J Pathol ; 180(2): 811-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22155108

ABSTRACT

Bone marrow (BM) fibrosis is a feature of severe hyperparathyroidism. Consistent with this observation, mice expressing constitutively active parathyroid hormone (PTH)/PTH-related peptide receptors (PPR) in osteoblasts (PPR*Tg) display BM fibrosis. To obtain insight into the nature of BM fibrosis in such a model, a double-mutant mouse expressing constitutively active PPR and green fluorescent protein (GFP) under the control of the type I collagen promoter (PPR*Tg/GFP) was generated. Confocal microscopy and flow cytometry revealed the presence of a cell population expressing GFP (GFP(+)) that was also positive for the hematopoietic marker CD45 in the BM of both PPR*Tg/GFP and control animals. This cell population was expanded in PPR*Tg/GFP. The existence of cells expressing both type I collagen and CD45 in the adult BM was confirmed by IHC and fluorescence-activated cell sorting. An analysis of total RNA extracted from sorted GFP(+)CD45(+) cells showed that these cells produced type I collagen and PTH/PTH-related peptide receptor and receptor activator for NF-κB mRNAs, further supporting their features of being both mesenchymal and hematopoietic lineages. Similar cells, known as fibrocytes, are also present in pathological fibroses. Our findings, thus, indicate that the BM is a permissive microenvironment for the differentiation of fibrocyte-like cells and raise the possibility that these cells could contribute to the pathogenesis of BM fibrosis.


Subject(s)
Biomarkers/metabolism , Hematopoietic Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Primary Myelofibrosis/pathology , Animals , Bone Marrow/metabolism , Cell Differentiation , Collagen Type I , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Parathyroid Hormone-Related Protein/metabolism , Primary Myelofibrosis/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism
9.
J Leukoc Biol ; 88(2): 347-59, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20430777

ABSTRACT

The robust and consistent expression of the CD13 cell surface marker on very early as well as differentiated myeloid hematopoietic cells has prompted numerous investigations seeking to define roles for CD13 in myeloid cells. To address the function of myeloid CD13 directly, we created a CD13 null mouse and assessed the responses of purified primary macrophages or DCs from WT and CD13 null animals in cell assays and inflammatory disease models, where CD13 has been implicated previously. We find that mice lacking CD13 develop normally with normal hematopoietic profiles except for an increase in thymic but not peripheral T cell numbers. Moreover, in in vitro assays, CD13 appears to be largely dispensable for the aspects of phagocytosis, proliferation, and antigen presentation that we tested, although we observed a slight decrease in actin-independent erythrocyte uptake. However, in agreement with our published studies, we show that lack of monocytic CD13 completely ablates anti-CD13-dependent monocyte adhesion to WT endothelial cells. In vivo assessment of four inflammatory disease models showed that lack of CD13 has little effect on disease onset or progression. Nominal alterations in gene expression levels between CD13 WT and null macrophages argue against compensatory mechanisms. Therefore, although CD13 is highly expressed on myeloid cells and is a reliable marker of the myeloid lineage of normal and leukemic cells, it is not a critical regulator of hematopoietic development, hemostasis, or myeloid cell function.


Subject(s)
CD13 Antigens/physiology , Hematopoiesis/genetics , Myeloid Cells/physiology , Animals , CD13 Antigens/analysis , CD13 Antigens/genetics , Dendritic Cells , Gene Expression Regulation , Hematopoietic Stem Cells , Inflammation/etiology , Macrophages , Mice , Mice, Knockout , Myeloid Cells/chemistry
10.
Blood ; 115(12): 2380-90, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-19965672

ABSTRACT

Integrins contribute to lymphopoiesis, whereas Toll-like receptors (TLRs) facilitate the myeloid replenishment during inflammation. The combined role of TLRs and integrin on hematopoiesis remains unclear. gp96 (grp94, HSP90b1) is an endoplasmic reticulum master chaperone for multiple TLRs. We report herein that gp96 is also essential for expression of 14 hematopoietic system-specific integrins. Genetic deletion of gp96 thus enables us to determine the collective roles of gp96, integrins, and TLRs in hematopoiesis. We found that gp96-null hematopoietic stem cells could support long-term myelopoiesis. B- and T-cell development, however, was severely compromised with transitional block from pro-B to pre-B cells and the inability of thymocytes to develop beyond the CD4(-)CD8(-) stage. These defects were cell-intrinsic and could be recapitulated on bone marrow stromal cell culture. Furthermore, defective lymphopoiesis correlated strongly with failure of hematopoietic progenitors to form close contact with stromal cell niche and was not the result of the defect in the assembly of antigen receptor or interleukin-7 signaling. These findings define gp96 as the only known molecular chaperone to specifically regulate T- and B-cell development.


Subject(s)
B-Lymphocytes/physiology , Lymphopoiesis/physiology , Membrane Glycoproteins/metabolism , Molecular Chaperones/metabolism , T-Lymphocytes/physiology , Animals , B-Lymphocytes/cytology , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Cell Line , Cell Lineage/immunology , Endoplasmic Reticulum/metabolism , Female , Integrins/metabolism , Interleukin-7/metabolism , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Chaperones/genetics , Myelopoiesis/physiology , Precursor Cells, B-Lymphoid/cytology , Signal Transduction/immunology , Stromal Cells/cytology , Stromal Cells/physiology , T-Lymphocytes/cytology , Thymus Gland/cytology , Thymus Gland/physiology , Toll-Like Receptors/metabolism
11.
Bone ; 45(4): 640-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19591967

ABSTRACT

MIF is an important regulator of innate and adaptive immunity, which is produced by a variety of cell types including activated T cells and macrophages. We examined the effects of MIF on osteoclastogenesis in bone marrow (BM) cultures from WT and MIF-deficient (KO) mice as well as the bone mass of MIF KO mice. Exogenous MIF inhibited osteoclast formation in BM cultures by decreasing fusion in cells that were treated with M-CSF and RANKL. However, inhibition of OCL formation by MIF treatment was not mediated by fusion-related molecules in heterogeneous bone marrow cultures. BM cultures from MIF KO mice that were treated with M-CSF and RANKL, PTH or vitamin D had significantly increased OCL number compared to cells from WT mice. MIF also significantly inhibited OCL formation in cultures of RAW 264.7 cells that were treated with RANKL. In addition, the number of CFU-GM and Mac-1(+) cells in the BM of MIF KO mice was greater than in WT controls. Trabecular bone volume (TBV) in the femurs and vertebrae of MIF KO mice was decreased compared to WT mice. In addition, serum bone resorption and formation markers were decreased in MIF KO mice compared to WT mice. These results demonstrate that MIF has inhibitory effects on OCL formation in vitro. We also found that BM cell cultures from MIF KO mice had an increased capacity to form osteoclasts. Furthermore, MIF KO animals had significantly decreased TBV with low turnover. We conclude that MIF is an inhibitor of osteoclastogenesis in vitro, which may regulate bone turnover via indirect mechanism in vivo.


Subject(s)
Macrophage Migration-Inhibitory Factors/metabolism , Osteoclasts/cytology , Osteogenesis , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteocalcin/blood , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RANK Ligand/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
12.
J Immunol ; 181(11): 7728-37, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017962

ABSTRACT

We compared how CD4 vs CD8 cells attain the capacity to express the effector cytokine IFN-gamma under both immunogenic and tolerogenic conditions. Although the Ifng gene locus was epigenetically repressed in naive Ag-inexperienced CD4 cells, it had already undergone partial remodeling toward a transcriptionally competent configuration in naive CD8 cells. After TCR stimulation, CD8 cells fully remodeled the Ifng locus and gained the capacity to express high levels of IFN-gamma more rapidly than CD4 cells. Enforced dual costimulation through OX40 and 4-1BB redirected CD8 cells encountering soluble exogenous peptide to expand and differentiate into IFN-gamma and TNF-alpha double-producing effectors rather than becoming tolerant. Despite this and the stronger tendency of CD8 compared with CD4 cells to differentiate into IFN-gamma-expressing effectors, when parenchymal self-Ag was the source of tolerizing Ag, enforced dual costimulation selectively boosted expansion but did not push effector differentiation in CD8 cells while both expansion and effector differentiation were dramatically boosted in CD4 cells. Notably, enforced dual costimulation was able to push effector differentiation in CD8 cells encountering cognate parenchymal self-Ag when CD4 cells were simultaneously engaged. Thus, the ability of enforced OX40 plus 4-1BB dual costimulation to redirect CD8 cells to undergo effector differentiation was unexpectedly influenced by the source of tolerizing Ag and help was selectively required to facilitate CD8 cell effector differentiation when the tolerizing Ag derived from self.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immune Tolerance , Receptors, OX40/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , Autoantigens/genetics , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Immune Tolerance/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Transgenic , Peptides/genetics , Peptides/immunology , Quantitative Trait Loci/immunology , Receptors, OX40/agonists , Receptors, OX40/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
13.
Bone ; 43(3): 501-10, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18571490

ABSTRACT

Identification of a reliable marker of skeletal precursor cells within calcified and soft tissues remains a major challenge for the field. To address this, we used a transgenic model in which osteoblasts can be eliminated by pharmacological treatment. Following osteoblast ablation a dramatic increase in a population of alpha-smooth muscle actin (alpha-SMA) positive cells was observed. During early recovery phase from ablation we have detected cells with the simultaneous expression of alpha-SMA and a preosteoblastic 3.6GFP marker, indicating the potential for transition of alpha-SMA+ cells towards osteoprogenitor lineage. Utilizing alpha-SMAGFP transgene, alpha-SMAGFP+ positive cells were detected in the microvasculature and in the osteoprogenitor population within bone marrow stromal cells. Osteogenic and adipogenic induction stimulated expression of bone and fat markers in the alpha-SMAGFP+ population derived from bone marrow or adipose tissue. In adipose tissue, alpha-SMA+ cells were localized within the smooth muscle cell layer and in pericytes. After in vitro expansion, alpha-SMA+/CD45-/Sca1+ progenitors were highly enriched. Following cell sorting and transplantation of expanded pericyte/myofibroblast populations, donor-derived differentiated osteoblasts and new bone formation was detected. Our results show that cells with a pericyte/myofibroblast phenotype have the potential to differentiate into functional osteoblasts.


Subject(s)
Actins/metabolism , Adipocytes/cytology , Genes, Reporter/genetics , Green Fluorescent Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Osteogenesis/physiology , Stem Cells/cytology , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Ataxin-1 , Ataxins , Bone Marrow Cells/cytology , Bone and Bones/metabolism , Leukocyte Common Antigens/biosynthesis , Mice , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Osteoblasts/metabolism , Osteogenesis/genetics
14.
Blood ; 111(6): 3229-35, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18056838

ABSTRACT

Polycythemia is often associated with erythropoietin (EPO) overexpression and defective oxygen sensing. In normal cells, intracellular oxygen concentrations are directly sensed by prolyl hydroxylase domain (PHD)-containing proteins, which tag hypoxia-inducible factor (HIF) alpha subunits for polyubiquitination and proteasomal degradation by oxygen-dependent prolyl hydroxylation. Here we show that different PHD isoforms differentially regulate HIF-alpha stability in the adult liver and kidney and suppress Epo expression and erythropoiesis through distinct mechanisms. Although Phd1(-/-) or Phd3(-/-) mice had no apparent defects, double knockout of Phd1 and Phd3 led to moderate erythrocytosis. HIF-2alpha, which is known to activate Epo expression, accumulated in the liver. In adult mice deficient for PHD2, the prototypic Epo transcriptional activator HIF-1alpha accumulated in both the kidney and liver. Elevated HIF-1alpha levels were associated with dramatically increased concentrations of both Epo mRNA in the kidney and Epo protein in the serum, which led to severe erythrocytosis. In contrast, heterozygous mutation of Phd2 had no detectable effects on blood homeostasis. These findings suggest that PHD1/3 double deficiency leads to erythrocytosis partly by activating the hepatic HIF-2alpha/Epo pathway, whereas PHD2 deficiency leads to erythrocytosis by activating the renal Epo pathway.


Subject(s)
Aging/physiology , DNA-Binding Proteins/metabolism , Erythropoiesis , Immediate-Early Proteins/metabolism , Procollagen-Proline Dioxygenase/metabolism , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hypoxia-Inducible Factor-Proline Dioxygenases , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/genetics , Mice , Mice, Knockout , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics
15.
J Bone Miner Res ; 21(11): 1704-12, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17002560

ABSTRACT

UNLABELLED: We examined OVX-induced bone loss in three TLD mouse models. In TLD mice, OVX caused trabecular bone loss equivalent to that of WT. In contrast, cortical bone loss with OVX was variable. We conclude that T lymphocytes do not influence OVX-induced trabecular bone loss. INTRODUCTION: We examined ovariectomy (OVX)-induced bone loss in three T lymphocyte-deficient (TLD) mouse models: nude mice, recombination activating gene 2-deficient (RAG2 KO) mice, and T cell receptor alpha chain-deficient (TCRalpha KO) mice. MATERIALS AND METHODS: Bone mass was examined by DXA, microCT, and histomorphometry. We also examined the effect of OVX on T lymphocytes in the bone marrow and spleens of wildtype (WT) mice and on in vitro osteoclastogenesis and colony forming unit-granulocyte macrophage (CFU-GM) activity in the bone marrow of WT and nude mice. RESULTS: In WT mice, OVX did not alter T lymphocyte number in the bone marrow but did increase T lymphocytes in the spleen. Comparison of bone mass in nude, RAG2 KO, and TCRalpha KO mice with WT as measured by DXA showed decreased femoral bone mass in nude mice and increased vertebral bone mass in RAG2 KO mice. In TCRalpha KO mice, femoral, tibial, and vertebral bone mass were decreased. In vertebrae and long bones, bone loss with OVX was consistently present in WT mice but variably present in TLD mice as measured by DXA. In contrast, microCT and histomorphometry showed similar trabecular bone loss after OVX in all mice. However, femoral cortical bone loss occurred only in WT and RAG2 KO mice. OVX produced similar trabecular bone loss in WT and TCRalpha KO mice and also induced cortical bone loss in both. Histomorphometry showed that TRACP(+) area in bones was increased by OVX in femurs from both WT and nude mice as was in vitro osteoclast-like cell formation and CFU-GM activity. CONCLUSIONS: These results show that OVX caused similar trabecular bone loss in both WT and TLD mice. The ability of DXA and measurement of cortical bone loss to show OVX-induced effects on bone mass was variable. It seems that T lymphocytes are not critical for OVX-induced trabecular bone loss in these mouse models.


Subject(s)
Bone and Bones/physiology , Ovariectomy/methods , T-Lymphocytes/physiology , Animals , Bone Marrow Cells/metabolism , Bone and Bones/metabolism , Female , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Osteoclasts/metabolism , Recombination, Genetic , Stem Cells/metabolism , T-Lymphocytes/metabolism
16.
Bone ; 39(6): 1302-12, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16938497

ABSTRACT

The type I collagen promoter has been used to develop transgenic constructs that are able to mark different stages of osteoblastic differentiation. The pOBCol3.6 promoter is active in early mesenchymal progenitors, including preosteoblasts and osteoblasts, while the pOBCol2.3 promoter is more restricted, showing expression in mature osteoblasts and osteocytes. Transgenic mouse lines have been created that express various GFP reporters under the control of both promoters. These transgenic mice permit the tracking of osteoblastic lineage progression in vitro. They also represent a system to test lineage progression in vivo after the transplantation of progenitors. A parabiosis system was used in which pOBCol3.6GFP transgenic mice were surgically joined with mice bearing a Col2.3DeltaTK transgene. The Col2.3DeltaTK transgenic mouse bears a herpes thymidine kinase gene driven by the pOBCol2.3 promoter, and upon treatment with gancyclovir (GCV) displays extensive destruction of the bone lining cells. After a common circulation was established, parabiotic pairs were treated with GCV for 15 days. Histological analysis of their bones showed the clear presence of GFP positive cells in the Col2.3DeltaTK parabionts, around trabecular bone and on the endosteal and periosteal surfaces. Stromal cell cultures from these Col2.3DeltaTK parabionts did not display mineralized colonies coexpressing GFP. In contrast, scattered GFP positive clusters that contained large cells with morphology similar to osteoclast like cells (OCLs) were observed. These cells were also TRAP positive. They were readily detected in Col2.3DeltaTK mice treated with GCV and transplanted with purified hematopoietic stem cells (HSCs) isolated from pOBCol3.6GFP mice. OCLs were also generated in vitro from osteoclast progenitor cells obtained from pOBCol3.6GFP mice that were defined by the B220- CD3- CD11b- c-fms+ phenotype. Molecular analysis showed that OCLs did not express type I collagen indicating that the Col3.6 promoter contains elements that are active during osteoclastogenesis and are not strictly related to collagen transcription. In summary, we demonstrate that pOBCol3.6 unexpectedly directs the expression of transgenes in the osteoclast lineage and this effect must be considered when utilizing this promoter to study of mesenchymal progenitor cells.


Subject(s)
Collagen Type I/genetics , Osteoblasts/metabolism , Osteoclasts/metabolism , Animals , Base Sequence , Bone Marrow Transplantation , Collagen Type I, alpha 1 Chain , DNA/genetics , Female , Ganciclovir/pharmacology , Gene Expression , Green Fluorescent Proteins/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Parabiosis , Promoter Regions, Genetic , Rats , Recombinant Proteins/genetics , Transplantation Chimera
17.
J Immunol ; 177(1): 234-45, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785519

ABSTRACT

IL-18 induces inflammation resulting in either enhanced protection from pathogens or exacerbation of autoimmunity, and T cells are profoundly activated during these responses. How IL-18 influences T cell activation is unknown, but this study in mice shows that IL-18 boosted Ag-specific T cell clonal expansion of effector T cells and induced a subpopulation of IFN-gamma superproducing T cells. Commitment to IFN-gamma production through IL-18 was independent of NK cells and IL-12 but dependent on host-derived IFN-gamma. To determine how expansion of these effectors occurred, IL-18 was shown to induce OX40L on dendritic cells, whereas peptide stimulation induced CD134 (OX40) on specific T cells. CD134 blockade inhibited T cell effector expansion thereby reducing the number of IFN-gamma superproducers by 12-fold. Thus, independent of IL-12, IL-18 impacts T cell immunity throughout lymphoid and nonlymphoid tissue by bridging the innate and adaptive arms of the immune system through IFN-gamma and the CD134 costimulatory pathway.


Subject(s)
Adjuvants, Immunologic/physiology , Interferon-gamma/physiology , Interleukin-18/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/immunology , Adjuvants, Immunologic/administration & dosage , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Immunity, Cellular/genetics , Immunity, Innate/genetics , Interferon-gamma/biosynthesis , Interleukin-12/physiology , Interleukin-18/administration & dosage , Interleukin-18 Receptor alpha Subunit , Killer Cells, Natural/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Receptors, Interleukin-18 , Receptors, OX40 , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/genetics , Spleen/cytology , Spleen/immunology , Spleen/transplantation
18.
J Bone Miner Res ; 21(1): 67-77, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16355275

ABSTRACT

UNLABELLED: Murine BM was fractionated using a series of hematopoietic markers to characterize its osteoclast progenitor populations. We found that the early osteoclastogenic activity in total BM was recapitulated by a population of cells contained within the CD11b(-/low) CD45R- CD3- CD115high fraction. INTRODUCTION: Osteoclasts are of hematopoietic origin and they have been shown to share the same lineage as macrophages. We further characterized the phenotype of osteoclast progenitor populations in murine bone marrow (BM) by analyzing their cell surface markers. MATERIALS AND METHODS: We used fluorescence-activated cell sorting (FACS) to identify the subsets of BM cells that contained osteoclast progenitors. We fractionated BM according to several markers and cultured the sorted populations for a period of 2-6 days with macrophage-colony stimulating factor (M-CSF) and RANKL. The numbers of multinucleated osteoclast-like cells (OCLs) that formed in the cultures were counted. RESULTS: We found that the CD45R- CD11b(-/low) population recapitulated the early osteoclastogenic activity of total BM. In addition, although previous experiments indicated that osteoclastogenic activity was enriched within the CD45R+ population, we found that highly purified CD45R+ BM was incapable of differentiating into osteoclasts in vitro. We also found that CD45R- CD11b(high) BM cells were an inefficient source of osteoclast progenitors. However, CD11b was transiently upregulated by cells of the CD45R- CD11b(-/low) fraction early (within 24 h) during culture with M-CSF. Finally, further fractionation of BM using CD115 and CD117 showed that, as osteoclast precursor cells matured, they downregulate CD117 but remain CD115+. Curiously, pure populations of CD117- (CD115high) cells isolated fresh from BM have low osteoclastogenic activity in vitro. CONCLUSIONS: We provided a refined analysis of the precise subpopulations of murine BM that are capable of differentiating into OCLs in vitro when treated with M-CSF and RANKL.


Subject(s)
Bone Marrow Cells/physiology , Cell Differentiation/physiology , Osteoclasts/physiology , Stem Cells/physiology , Animals , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Bone Marrow Cells/cytology , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Flow Cytometry/methods , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Male , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Mice , Osteoclasts/cytology , Osteogenesis , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Stem Cells/cytology
19.
Proc Natl Acad Sci U S A ; 101(15): 5616-21, 2004 Apr 13.
Article in English | MEDLINE | ID: mdl-15060278

ABSTRACT

IL-15 and the IL-15 receptor (IL-15R)alpha chain are essential for normal development of naive CD8 T cells, intestinal intraepithelial lymphocytes (IEL), and natural killer (NK)/NK/T cells. However, whether IL-15R alpha expression by these subsets is necessary for their production and which cell type needs to produce IL-15 to drive development are unknown. We analyzed the requirements for IL-15 and IL-15R alpha expression by bone marrow-derived or parenchymal cells for mediating lymphocyte subset development. Naive CD8 T cell development required IL-15R alpha expression by both bone marrow-derived and parenchymal cells, whereas memory-phenotype CD8 T cells required IL-15R alpha expression only by hematopoietic cells. In contrast and surprisingly, the development of IEL subsets, particularly CD8 alpha alpha Thy1(-)V gamma 5(+) T cell antigen receptor gamma delta and the CD8 alpha alpha Thy1(-) T cell antigen receptor alpha beta IEL populations, depended completely on parenchymal cell expression of IL-15R alpha and IL-15 but not IL-15R beta. In the case of NK and NK/T cell generation and maturation, expression of IL-15 and IL-15R alpha by both parenchymal and hematopoietic cells was important, although the latter played the greatest role. These results demonstrated dichotomous mechanisms by which IL-15 regulated lymphoid development, interacting with distinct cell types depending on the developmental pathway.


Subject(s)
Interleukin-15/physiology , Lymphocytes/immunology , Receptors, Interleukin-2/physiology , Animals , Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epithelial Cells/immunology , Immunologic Memory , Interleukin-15/biosynthesis , Intestinal Mucosa/cytology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver/cytology , Lymphocytes/cytology , Lymphocytes/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Interleukin-15 , Receptors, Interleukin-2/biosynthesis , Receptors, Interleukin-2/deficiency , Spleen/cytology , T-Lymphocyte Subsets/metabolism , Transplantation Chimera
20.
Blood ; 103(9): 3258-64, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14726388

ABSTRACT

We previously reported a transgenic mouse model expressing herpesvirus thymidine kinase (TK) gene under the control of a 2.3-kilobase fragment of the rat collagen alpha1 type I promoter (Col2.3 Delta TK). This construct confers lineage-specific expression in developing osteoblasts, allowing the conditional ablation of osteoblast lineage after treatment with ganciclovir (GCV). After GCV treatment these mice have profound alterations on bone formation leading to a progressive bone loss. In addition, treated animals also lose bone marrow cellularity. In this report we characterized hematopoietic parameters in GCV-treated Col2.3 Delta TK mice, and we show that after treatment transgenic animals lose lymphoid, erythroid, and myeloid progenitors in the bone marrow, followed by decreases in the number of hematopoietic stem cells (HSCs). Together with the decrease in bone marrow hematopoiesis, active extramedullary hematopoiesis was observed in the spleen and liver, as measured by an increase in peripheral HSCs and active primary in vitro hematopoiesis. After withdrawal of GCV, osteoblasts reappeared in the bone compartment together with a recovery of medullary and decrease in extramedullary hematopoiesis. These observations directly demonstrate the role of osteoblasts in hematopoiesis and provide a model to study the interactions between the mesenchymal and hematopoietic compartments in the marrow.


Subject(s)
Genetic Vectors/pharmacology , Hematopoiesis , Osteoblasts/physiology , Animals , Bone Marrow Cells , Cell Lineage , Collagen Type I/genetics , Ganciclovir/pharmacology , Hematopoiesis, Extramedullary , Hematopoietic Stem Cells , Mice , Mice, Transgenic , Models, Animal , Osteoblasts/metabolism , Promoter Regions, Genetic , Rats , Thymidine Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...