Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
J Diabetes ; 16(3): e13504, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38035773

ABSTRACT

The skeleton is traditionally known for its structural support, organ protection, movement, and maintenance of mineral homeostasis. Over the last 10 years, bone has emerged as an endocrine organ with diverse physiological functions. The two key molecules in this context are fibroblast growth factor 23 (FGF23), secreted by osteocytes, and osteocalcin, a hormone produced by osteoblasts. FGF23 affects mineral homeostasis through its actions on the kidneys, and osteocalcin has beneficial effects in improving glucose homeostasis, muscle function, brain development, cognition, and male fertility. In addition, another osteoblast-derived hormone, lipocalin 2 (LCN2) has emerged into the researchers' field of vision. In this review, we mainly focus on LCN2's role in appetite regulation and glucose metabolism and also briefly introduce its effects in other pathophysiological conditions, such as nonalcoholic fatty liver disease, sarcopenic obesity, and cancer-induced cachexia.


Subject(s)
Bone and Bones , Hormones , Humans , Male , Animals , Mice , Lipocalin-2/metabolism , Osteocalcin , Bone and Bones/metabolism , Minerals
2.
Blood ; 142(5): 460-476, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37267505

ABSTRACT

The chromosome 9p21 locus comprises several tumor suppressor genes including MTAP, CDKN2A, and CDKN2B, and its homo- or heterozygous deletion is associated with reduced survival in multiple cancer types. We report that mice with germ line monoallelic deletion or induced biallelic deletion of the 9p21-syntenic locus (9p21s) developed a fatal myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN)-like disease associated with aberrant trabecular bone formation and/or fibrosis in the bone marrow (BM). Reciprocal BM transfers and conditional targeting of 9p21s suggested that the disease originates in the BM stroma. Single-cell analysis of 9p21s-deficient BM stroma revealed the expansion of chondrocyte and osteogenic precursors, reflected in increased osteogenic differentiation in vitro. It also showed reduced expression of factors maintaining hematopoietic stem/progenitor cells, including Cxcl12. Accordingly, 9p21s-deficient mice showed reduced levels of circulating Cxcl12 and concomitant upregulation of the profibrotic chemokine Cxcl13 and the osteogenesis- and fibrosis-related multifunctional glycoprotein osteopontin/Spp1. Our study highlights the potential of mutations in the BM microenvironment to drive MDS/MPN-like disease.


Subject(s)
Bone Marrow , Osteogenesis , Mice , Animals , Bone Marrow/pathology , Hematopoietic Stem Cells/metabolism , Genes, Tumor Suppressor , Cell Differentiation
3.
J Clin Invest ; 132(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36194488

ABSTRACT

The various functions of the skeleton are influenced by extracellular cues, hormones, and neurotransmitters. One type of neuronal regulation favors bone mass accrual by inhibiting sympathetic nervous system (SNS) activity. This observation raises questions about the transcriptional mechanisms regulating catecholamine synthesis. Using a combination of genetic and pharmacological studies, we found that the histone deacetylase sirtuin 1 (SIRT1) is a transcriptional modulator of the neuronal control of bone mass. Neuronal SIRT1 reduced bone mass by increasing SNS signaling. SIRT1 did so by increasing expression of monoamine oxidase A (MAO-A), a SIRT1 target that reduces brain serotonin levels by inducing its catabolism and by suppressing tryptophan hydroxylase 2 (Tph2) expression and serotonin synthesis in the brain stem. SIRT1 upregulated brain catecholamine synthesis indirectly through serotonin, but did not directly affect dopamine ß hydroxylase (Dbh) expression in the locus coeruleus. These results help us to understand skeletal changes associated with selective serotonin reuptake inhibitors (SSRIs) and may have implications for treating skeletal and metabolic diseases.


Subject(s)
Aging , Serotonin , Sirtuin 1 , Animals , Mice , Aging/genetics , Catecholamines , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Serotonin/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
4.
Cancer Discov ; 12(4): 1106-1127, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35046097

ABSTRACT

Remodeling of the microenvironment by tumor cells can activate pathways that favor cancer growth. Molecular delineation and targeting of such malignant-cell nonautonomous pathways may help overcome resistance to targeted therapies. Herein we leverage genetic mouse models, patient-derived xenografts, and patient samples to show that acute myeloid leukemia (AML) exploits peripheral serotonin signaling to remodel the endosteal niche to its advantage. AML progression requires the presence of serotonin receptor 1B (HTR1B) in osteoblasts and is driven by AML-secreted kynurenine, which acts as an oncometabolite and HTR1B ligand. AML cells utilize kynurenine to induce a proinflammatory state in osteoblasts that, through the acute-phase protein serum amyloid A (SAA), acts in a positive feedback loop on leukemia cells by increasing expression of IDO1-the rate-limiting enzyme for kynurenine synthesis-thereby enabling AML progression. This leukemia-osteoblast cross-talk, conferred by the kynurenine-HTR1B-SAA-IDO1 axis, could be exploited as a niche-focused therapeutic approach against AML, opening new avenues for cancer treatment. SIGNIFICANCE: AML remains recalcitrant to treatments due to the emergence of resistant clones. We show a leukemia-cell nonautonomous progression mechanism that involves activation of a kynurenine-HTR1B-SAA-IDO1 axis between AML cells and osteoblasts. Targeting the niche by interrupting this axis can be pharmacologically harnessed to hamper AML progression and overcome therapy resistance. This article is highlighted in the In This Issue feature, p. 873.


Subject(s)
Kynurenine , Leukemia, Myeloid, Acute , Animals , Humans , Kynurenine/metabolism , Kynurenine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Mice , Osteoblasts/metabolism , Signal Transduction , Tumor Microenvironment
5.
Elife ; 92020 11 24.
Article in English | MEDLINE | ID: mdl-33231171

ABSTRACT

In the mouse, the osteoblast-derived hormone Lipocalin-2 (LCN2) suppresses food intake and acts as a satiety signal. We show here that meal challenges increase serum LCN2 levels in persons with normal or overweight, but not in individuals with obesity. Postprandial LCN2 serum levels correlate inversely with hunger sensation in challenged subjects. We further show through brain PET scans of monkeys injected with radiolabeled recombinant human LCN2 (rh-LCN2) and autoradiography in baboon, macaque, and human brain sections, that LCN2 crosses the blood-brain barrier and localizes to the hypothalamus in primates. In addition, daily treatment of lean monkeys with rh-LCN2 decreases food intake by 21%, without overt side effects. These studies demonstrate the biology of LCN2 as a satiety factor and indicator and anorexigenic signal in primates. Failure to stimulate postprandial LCN2 in individuals with obesity may contribute to metabolic dysregulation, suggesting that LCN2 may be a novel target for obesity treatment.


Obesity has reached epidemic proportions worldwide and affects more than 40% of adults in the United States. People with obesity have a greater likelihood of developing type 2 diabetes, cardiovascular disease or chronic kidney disease. Changes in diet and exercise can be difficult to follow and result in minimal weight loss that is rarely sustained overtime. In fact, in people with obesity, weight loss can lower the metabolism leading to increased weight gain. New drugs may help some individuals achieve 5 to 10% weight loss but have side effects that prevent long-term use. Previous studies in mice show that a hormone called Lipocalin-2 (LCN2) suppresses appetite. It also reduces body weight and improves sugar metabolism in the animals. But whether this hormone has the same effects in humans or other primates is unclear. If it does, LCN2 might be a potential obesity treatment. Now, Petropoulou et al. show that LCN2 suppressed appetite in humans and monkeys. In human studies, LCN2 levels increased after a meal in individuals with normal weight or overweight, but not in individuals with obesity. Higher levels of LCN2 in a person's blood were also associated with a feeling of reduced hunger. Using brain scans, Petropoulou et al. showed that LCN2 crossed the blood-brain barrier in monkeys and bound to the hypothalamus, the brain center regulating appetite and energy balance. LCN2 also bound to human and monkey hypothalamus tissue in laboratory experiments. When injected into monkeys, the hormone suppressed food intake and lowered body weight without toxic effects in short-term studies. The experiments lay the initial groundwork for testing whether LCN2 might be a useful treatment for obesity. More studies in animals will help scientists understand how LCN2 works, which patients might benefit, how it would be given to patients and for how long. Clinical trials would also be needed to verify whether it is an effective and safe treatment for obesity.


Subject(s)
Lipocalin-2/metabolism , Macaca/metabolism , Obesity/metabolism , Papio/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Eating , Humans , Lipocalin-2/genetics , Obesity/diagnostic imaging , Obesity/genetics , Obesity/physiopathology , Positron-Emission Tomography , Protein Transport
6.
J Exp Med ; 217(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32639539

ABSTRACT

Regulation of food intake is a recently identified endocrine function of bone that is mediated by Lipocalin-2 (LCN2). Osteoblast-secreted LCN2 suppresses appetite and decreases fat mass while improving glucose metabolism. We now show that serum LCN2 levels correlate with insulin levels and ß-cell function, indices of healthy glucose metabolism, in obese mice and obese, prediabetic women. However, LCN2 serum levels also correlate with body mass index and insulin resistance in the same individuals and are increased in obese mice. To dissect this apparent discrepancy, we modulated LCN2 levels in mice. Silencing Lcn2 expression worsens metabolic dysfunction in genetic and diet-induced obese mice. Conversely, increasing circulating LCN2 levels improves metabolic parameters and promotes ß-cell function in mouse models of ß-cell failure acting as a growth factor necessary for ß-cell adaptation to higher metabolic load. These results indicate that LCN2 up-regulation is a protective mechanism to counteract obesity-induced glucose intolerance by decreasing food intake and promoting adaptive ß-cell proliferation.


Subject(s)
Lipocalin-2/physiology , Obesity/metabolism , Prediabetic State/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Female , Glucose/metabolism , Humans , Insulin Resistance , Insulin-Secreting Cells/metabolism , Lipocalin-2/blood , Lipocalin-2/metabolism , Mice , Mice, Obese/blood , Mice, Obese/metabolism , Mice, Obese/physiology , Middle Aged , Obesity/blood , Prediabetic State/blood
7.
J Exp Med ; 217(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31873722

ABSTRACT

Numerous studies support a role of the microenvironment in maintenance of the leukemic clone, as well as in treatment resistance. It is clear that disruption of the normal bone marrow microenvironment is sufficient to promote leukemic transformation and survival in both a cell autonomous and non-cell autonomous manner. In this review, we provide a snapshot of the various cell types shown to contribute to the leukemic microenvironment as well as treatment resistance. Several of these studies suggest that leukemic blasts occupy specific cellular and biochemical "niches." Effective dissection of critical leukemic niche components using single-cell approaches has allowed a more precise and extensive characterization of complexity that underpins both the healthy and malignant bone marrow microenvironment. Knowledge gained from these observations can have an important impact in the development of microenvironment-directed targeted approaches aimed at mitigating disease relapse.


Subject(s)
Bone Marrow/pathology , Leukemia/metabolism , Leukemia/pathology , Tumor Microenvironment , Adipocytes/metabolism , Animals , B-Lymphocytes/immunology , Bone Marrow/metabolism , Endothelium, Vascular/metabolism , Humans , Immunotherapy, Adoptive , Leukemia/drug therapy , Leukemia/immunology , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/metabolism , Receptors, Chimeric Antigen , Signal Transduction/drug effects , Stem Cell Niche , T-Lymphocytes/immunology
9.
Nature ; 569(7755): 222-228, 2019 05.
Article in English | MEDLINE | ID: mdl-30971824

ABSTRACT

The bone marrow microenvironment has a key role in regulating haematopoiesis, but its molecular complexity and response to stress are incompletely understood. Here we map the transcriptional landscape of mouse bone marrow vascular, perivascular and osteoblast cell populations at single-cell resolution, both at homeostasis and under conditions of stress-induced haematopoiesis. This analysis revealed previously unappreciated levels of cellular heterogeneity within the bone marrow niche and resolved cellular sources of pro-haematopoietic growth factors, chemokines and membrane-bound ligands. Our studies demonstrate a considerable transcriptional remodelling of niche elements under stress conditions, including an adipocytic skewing of perivascular cells. Among the stress-induced changes, we observed that vascular Notch delta-like ligands (encoded by Dll1 and Dll4) were downregulated. In the absence of vascular Dll4, haematopoietic stem cells prematurely induced a myeloid transcriptional program. These findings refine our understanding of the cellular architecture of the bone marrow niche, reveal a dynamic and heterogeneous molecular landscape that is highly sensitive to stress and illustrate the utility of single-cell transcriptomic data in evaluating the regulation of haematopoiesis by discrete niche populations.


Subject(s)
Bone Marrow/blood supply , Cellular Microenvironment , Hematopoiesis , Hematopoietic Stem Cells , Single-Cell Analysis , Stem Cell Niche , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Differentiation , Cell Lineage , Endothelium, Vascular/cytology , Female , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Male , Mice , Myeloid Cells/cytology , Myeloid Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , RNA-Seq , Receptors, Notch/metabolism , Stem Cell Niche/genetics , Stress, Physiological/genetics , Transcriptome/genetics
10.
Genes Dev ; 32(5-6): 324-326, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29593065

ABSTRACT

Hematopoietic stem cells (HSCs) reside and are maintained in specialized microenvironments within the bone marrow known as niches, which are comprised of various cell types. Among them, leptin receptor (LepR)-expressing CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells are known to create a niche for HSCs and at the same time to give rise to osteoblasts. These two functions of CAR/LepR+ cells appear to be tightly but inversely regulated to ensure adequate physical space for HSCs. However, how osteogenesis is prevented in CAR cells to maintain spaces available for HSCs and hematopoiesis remains unclear. In this issue of Genes & Development, Seike and colleagues (pp. 359-372) report that the transcription factor early B-cell factor (Ebf3) is preferentially expressed by CAR/LepR+ cells and inhibits CAR cell differentiation into osteoblasts while at the same time maintaining self-renewal of CAR/LepR+ cells. Using conditional knockout and retroviral systems, the investigators show that loss of Ebf3 in CAR cells impairs HSC numbers and leads to osteosclerosis. This study provides novel insights into transcriptional requirements for CAR cell bone formation by identifying Ebf3 as a niche factor secreted from CAR/Lepr+ cells that regulates the interplay between osteogenesis and hematopoiesis.


Subject(s)
Osteogenesis , Stem Cell Niche , Bone Marrow , Hematopoiesis , Hematopoietic Stem Cells
11.
Article in English | MEDLINE | ID: mdl-28963115

ABSTRACT

Hematopoietic stem cells (HSCs) interact dynamically with an intricate network of cells in the bone marrow (BM) microenvironment or niche. These interactions provide instructive cues that influence the production and lineage determination of different types of blood cells and maintenance of HSC quiescence. They also contribute to hematopoietic deregulation and hematological myeloid malignancies. Alterations in the BM niche are commonly observed in myeloid malignancies and contribute to the aberrant function of myelodysplastic and leukemia-initiating stem cells. In this work, we review how different components of the BM niche affect normal hematopoiesis, the molecular signals that govern this interaction, and how genetic changes in stromal cells or alterations in remodeled malignant BM niches contribute to myeloid malignancies. Understanding the intricacies between normal and malignant niches and their modulation may provide insights into developing novel therapeutics for blood disorders.


Subject(s)
Bone Marrow Neoplasms/pathology , Bone Marrow/physiology , Cellular Microenvironment/physiology , Chemokine CXCL12/metabolism , Endothelial Cells/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Nestin/metabolism , Neuroglia/physiology , Neurons/physiology , Osteoblasts/physiology , Osteocytes/physiology , Receptors, Leptin/metabolism , Stem Cell Niche/physiology , Sympathetic Nervous System/physiology
12.
Curr Mol Biol Rep ; 3(2): 53-62, 2017 Jun.
Article in English | MEDLINE | ID: mdl-29098141

ABSTRACT

PURPOSE OF REVIEW: This review focuses on evidence highlighting the bidirectional crosstalk between the hematopoietic stem cell (HSC) and their surrounding stromal cells, with a particular emphasis on cells of the osteoblast lineage. The role and molecular functions of osteoblasts in normal hematopoiesis and in myeloid hematological malignancies is discussed. RECENT FINDINGS: Cells of the osteoblast lineage have emerged as potent regulators of HSC expansion that regulate their recruitment and, depending on their stage of differentiation, their activity, proliferation and differentiation along the lymphoid, myeloid and erythroid lineages. In addition, mutations in mature osteoblasts or their progenitors induce myeloid malignancies. Conversely, signals from myelodysplastic cells can remodel the osteoblastic niche to favor self-perpetuation. SUMMARY: Understanding cellular crosstalk between osteoblastic cells and HSCs in the bone marrow microenvironment is of fundamental importance for developing therapies against benign and malignant hematological diseases.

14.
Nature ; 543(7645): 385-390, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28273060

ABSTRACT

Bone has recently emerged as a pleiotropic endocrine organ that secretes at least two hormones, FGF23 and osteocalcin, which regulate kidney function and glucose homeostasis, respectively. These findings have raised the question of whether other bone-derived hormones exist and what their potential functions are. Here we identify, through molecular and genetic analyses in mice, lipocalin 2 (LCN2) as an osteoblast-enriched, secreted protein. Loss- and gain-of-function experiments in mice demonstrate that osteoblast-derived LCN2 maintains glucose homeostasis by inducing insulin secretion and improves glucose tolerance and insulin sensitivity. In addition, osteoblast-derived LCN2 inhibits food intake. LCN2 crosses the blood-brain barrier, binds to the melanocortin 4 receptor (MC4R) in the paraventricular and ventromedial neurons of the hypothalamus and activates an MC4R-dependent anorexigenic (appetite-suppressing) pathway. These results identify LCN2 as a bone-derived hormone with metabolic regulatory effects, which suppresses appetite in a MC4R-dependent manner, and show that the control of appetite is an endocrine function of bone.


Subject(s)
Appetite Regulation/physiology , Bone and Bones/metabolism , Lipocalin-2/metabolism , Receptor, Melanocortin, Type 4/metabolism , Animals , Blood-Brain Barrier/metabolism , Bone and Bones/cytology , Cyclic AMP/metabolism , Eating/physiology , Female , Fibroblast Growth Factor-23 , Glucose/metabolism , Homeostasis , Hypothalamus/cytology , Hypothalamus/metabolism , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Male , Mice , Neurons/metabolism , Obesity/metabolism , Osteoblasts/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Thinness/metabolism
15.
J Bone Miner Res ; 32(6): 1267-1273, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28218468

ABSTRACT

We have previously reported that premenopausal women with idiopathic osteoporosis (IOP) have profound microarchitectural deficiencies and heterogeneous bone remodeling. Those with the lowest bone formation rate have higher baseline serum insulin-like growth factor-1 (IGF-1) levels and less robust response to teriparatide. Because IGF-1 stimulates bone formation and is critical for teriparatide action on osteoblasts, these findings suggest a state of IGF-1 resistance in some IOP women. To further investigate the hypothesis that osteoblast and IGF-1-related mechanisms mediate differential responsiveness to teriparatide in IOP, we studied circulating osteoblast progenitor (COP) cells and their IGF-1 receptor (IGF-1R) expression. In premenopausal women with IOP, peripheral blood mononuclear cells (PBMCs) were obtained at baseline (n = 25) and over 24 months of teriparatide treatment (n = 11). Flow cytometry was used to identify and quantify COPs (non-hematopoetic lineage cells expressing osteocalcin and RUNX2) and to quantify IGF-1R expression levels. At baseline, both the percent of PBMCs that were COPs (%COP) and COP cell-surface IGF-1R expression correlated directly with several histomorphometric indices of bone formation in tetracycline-labeled transiliac biopsies. In treated subjects, both %COP and IGF-1R expression increased promptly after teriparatide, returning toward baseline by 18 months. Although neither baseline %COP nor increase in %COP after 3 months predicted the bone mineral density (BMD) response to teriparatide, the percent increase in IGF-1R expression on COPs at 3 months correlated directly with the BMD response to teriparatide. Additionally, lower IGF-1R expression after teriparatide was associated with higher body fat, suggesting links between teriparatide resistance, body composition, and the GH/IGF-1 axis. In conclusion, these assays may be useful to characterize bone remodeling noninvasively and may serve to predict early response to teriparatide and possibly other bone formation-stimulating medications. These new tools may also have utility in the mechanistic investigation of teriparatide resistance in premenopausal IOP and perhaps in other populations. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Osteoblasts/metabolism , Osteogenesis , Osteoporosis/drug therapy , Osteoporosis/physiopathology , Premenopause/physiology , Receptor, IGF Type 1/metabolism , Stem Cells/metabolism , Teriparatide/therapeutic use , Adipose Tissue/drug effects , Adolescent , Adult , Biopsy , Body Composition/drug effects , Bone Density/drug effects , Cohort Studies , Female , Humans , Middle Aged , Osteoblasts/drug effects , Osteogenesis/drug effects , Premenopause/drug effects , Stem Cells/drug effects , Teriparatide/pharmacology , Young Adult
17.
J Bone Miner Res ; 31(11): 1988-1996, 2016 11.
Article in English | MEDLINE | ID: mdl-27283956

ABSTRACT

Chronic immune activation associated with human immunodeficiency virus (HIV) infection may have negative consequences on bone acquisition in individuals infected with HIV early in life. Bone mineral density (BMD) and microarchitecture were characterized in 38 HIV-infected men on antiretroviral therapy (18 perinatally-infected, 20 adolescence-infected) and 20 uninfected men age 20 to 25 years by dual-energy X-ray absorptiometry (DXA), high-resolution peripheral quantitative computed tomography (HRpQCT). Flow cytometry was utilized to measure CD4+/CD8+ activation (HLADR+CD38+) and senescence (CD28-CD57+) and to quantify circulating osteogenic precursor (COP) cells in peripheral blood mononuclear cells using antibodies to RUNX2 and osteocalcin (OCN). Telomere lengths were measured in sorted COP cells using qPCR. DXA-derived areal BMD Z-scores and HRpQCT-derived volumetric BMD (vBMD) measures were lower in HIV-infected than uninfected men. Proportion of activated and senescent CD4+ and CD8+ T cells were higher in HIV-infected than uninfected men. The percentage of COP cells (mean ± SE) was lower in HIV-infected than uninfected (0.19% ± 0.02% versus 0.43% ± 0.06%; p < 0.0001) men, and also lower in perinatally-infected than adolescence-infected men (0.15% ± 0.02% versus 0.22% ± 0.03%; p < 0.04). A higher proportion of COP cells correlated with higher bone stiffness, a measure of bone strength, whereas a higher proportion of activated CD4+ T cells correlated with lower BMD and stiffness and lower proportion of COP cells. T cell activation with HIV-infection was associated with decreased numbers of osteogenic precursors as well as lower peak bone mass and bone strength. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone and Bones/abnormalities , Cellular Senescence , HIV Infections/immunology , HIV Infections/pathology , Osteogenesis , Stem Cells/pathology , Absorptiometry, Photon , Biomarkers/metabolism , Bone Density , Bone Remodeling , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Cell Movement , Humans , Lymphocyte Activation/immunology , Male , Radius/diagnostic imaging , Radius/pathology , T-Lymphocytes/immunology , Tibia/diagnostic imaging , Tibia/pathology , Tomography, X-Ray Computed , Young Adult
18.
Biochim Biophys Acta ; 1863(3): 490-498, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26681532

ABSTRACT

Osteoblasts are emerging regulators of myeloid malignancies since genetic alterations in them, such as constitutive activation of ß-catenin, instigate their appearance. The LDL receptor-related protein 5 (LRP5), initially proposed to be a co-receptor for Wnt proteins, in fact favors bone formation by suppressing gut-serotonin synthesis. This function of Lrp5 occurring in the gut is independent of ß-catenin activation in osteoblasts. However, it is unknown whether Lrp5 can act directly in osteoblast to influence other functions that require ß-catenin signaling, particularly, the deregulation of hematopoiesis and leukemogenic properties of ß-catenin activation in osteoblasts, that lead to development of acute myeloid leukemia (AML). Using mice with gain-of-function (GOF) Lrp5 alleles (Lrp5(A214V)) that recapitulate the human high bone mass (HBM) phenotype, as well as patients with the T253I HBM Lrp5 mutation, we show here that Lrp5 GOF mutations in both humans and mice do not activate ß-catenin signaling in osteoblasts. Consistent with a lack of ß-catenin activation in their osteoblasts, Lrp5(A214V) mice have normal trilinear hematopoiesis. In contrast to leukemic mice with constitutive activation of ß-catenin in osteoblasts (Ctnnb1(CAosb)), accumulation of early myeloid progenitors, a characteristic of AML, myeloid-blasts in blood, and segmented neutrophils or dysplastic megakaryocytes in the bone marrow, are not observed in Lrp5(A214V) mice. Likewise, peripheral blood count analysis in HBM patients showed normal hematopoiesis, normal percentage of myeloid cells, and lack of anemia. We conclude that Lrp5 GOF mutations do not activate ß-catenin signaling in osteoblasts. As a result, myeloid lineage differentiation is normal in HBM patients and mice. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.


Subject(s)
Hematopoiesis , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Mutation , Osteoblasts/metabolism , beta Catenin/metabolism , Adult , Aged , Animals , Bone Density/genetics , Cell Line , Cell Line, Tumor , Female , Flow Cytometry , Humans , Male , Mice, Knockout , Middle Aged , Osteogenesis/genetics , Signal Transduction/genetics , Young Adult
20.
Blood ; 124(18): 2834-46, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25139351

ABSTRACT

The bone marrow niche is thought to act as a permissive microenvironment required for emergence or progression of hematologic cancers. We hypothesized that osteoblasts, components of the niche involved in hematopoietic stem cell (HSC) function, influence the fate of leukemic blasts. We show that osteoblast numbers decrease by 55% in myelodysplasia and acute myeloid leukemia patients. Further, genetic depletion of osteoblasts in mouse models of acute leukemia increased circulating blasts and tumor engraftment in the marrow and spleen leading to higher tumor burden and shorter survival. Myelopoiesis increased and was coupled with a reduction in B lymphopoiesis and compromised erythropoiesis, suggesting that hematopoietic lineage/progression was altered. Treatment of mice with acute myeloid or lymphoblastic leukemia with a pharmacologic inhibitor of the synthesis of duodenal serotonin, a hormone suppressing osteoblast numbers, inhibited loss of osteoblasts. Maintenance of the osteoblast pool restored normal marrow function, reduced tumor burden, and prolonged survival. Leukemia prevention was attributable to maintenance of osteoblast numbers because inhibition of serotonin receptors alone in leukemic blasts did not affect leukemia progression. These results suggest that osteoblasts play a fundamental role in propagating leukemia in the marrow and may be a therapeutic target to induce hostility of the niche to leukemia blasts.


Subject(s)
Disease Progression , Leukemia/pathology , Osteoblasts/pathology , Animals , Cell Count , Cell Lineage/drug effects , Cell Proliferation/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Leukemia/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Osteoblasts/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...