Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
2.
Bone ; 184: 117092, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38575048

ABSTRACT

PURPOSE: Bone is susceptible to fluctuations in iron homeostasis, as both iron deficiency and overload are linked to poor bone strength in humans. In mice, however, inconsistent results have been reported, likely due to different diet setups or genetic backgrounds. Here, we assessed the effect of different high and low iron diets on bone in six inbred mouse strains (C57BL/6J, A/J, BALB/cJ, AKR/J, C3H/HeJ, and DBA/2J). METHODS: Mice received a high (20,000 ppm) or low-iron diet (∼10 ppm) after weaning for 6-8 weeks. For C57BL/6J males, we used two dietary setups with similar amounts of iron, yet different nutritional compositions that were either richer ("TUD study") or poorer ("UCLA study") in minerals and vitamins. After sacrifice, liver, blood and bone parameters as well as bone turnover markers in the serum were analyzed. RESULTS: Almost all mice on the UCLA study high iron diet had a significant decrease of cortical and trabecular bone mass accompanied by high bone resorption. Iron deficiency did not change bone microarchitecture or turnover in C57BL/6J, A/J, and DBA/2J mice, but increased trabecular bone mass in BALB/cJ, C3H/HeJ and AKR/J mice. In contrast to the UCLA study, male C57BL/6J mice in the TUD study did not display any changes in trabecular bone mass or turnover on high or low iron diet. However, cortical bone parameters were also decreased in TUD mice on the high iron diet. CONCLUSION: Thus, these data show that cortical bone is more susceptible to iron overload than trabecular bone and highlight the importance of a nutrient-rich diet to potentially mitigate the negative effects of iron overload on bone.


Subject(s)
Bone and Bones , Iron Overload , Animals , Male , Bone and Bones/metabolism , Bone and Bones/drug effects , Iron Overload/metabolism , Mice , Iron Deficiencies , Diet , Mice, Inbred C57BL , Iron, Dietary/administration & dosage , Liver/metabolism
3.
JBMR Plus ; 8(3): ziad016, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38505222

ABSTRACT

Bone development and remodeling are controlled by the phosphoinositide-3-kinase (Pi3k) signaling pathway. We investigated the effects of downregulation of phosphatase and tensin homolog (Pten), a negative regulator of Pi3k signaling, in a mouse model of Pten deficiency in preosteoblasts. We aimed to identify mechanisms that are involved in the regulation of bone turnover and are linked to bone disorders. Femora, tibiae, and bone marrow stromal cells (BMSCs) isolated from mice with a conditional deletion of Pten (Pten cKO) in Osterix/Sp7-expressing osteoprogenitor cells were compared to Cre-negative controls. Bone phenotyping was performed by µCT measurements, bone histomorphometry, quantification of bone turnover markers CTX and procollagen type 1 N propeptide (P1NP), and three-point bending test. Proliferation of BMSCs was measured by counting nuclei and Ki-67-stained cells. In vitro, osteogenic differentiation capacity was determined by ALP staining, as well as by detecting gene expression of osteogenic markers. BMSCs from Pten cKO mice were functionally different from control BMSCs. Osteogenic markers were increased in BMSCs derived from Pten cKO mice, while Pten protein expression was lower and Akt phosphorylation was increased. We detected a higher trabecular bone volume and an altered cortical bone morphology in Pten cKO bones with a progressive decrease in bone and tissue mineral density. Pten cKO bones displayed fewer osteoclasts and more osteoblasts (P = .00095) per trabecular bone surface and a higher trabecular bone formation rate. Biomechanical analysis revealed a significantly higher bone strength (P = .00012 for males) and elasticity of Pten cKO femora. On the cellular level, both proliferation and osteogenic differentiation capacity of Pten cKO BMSCs were significantly increased compared to controls. Our findings suggest that Pten knockout in osteoprogenitor cells increases bone stability and elasticity by increasing trabecular bone mass and leads to increased proliferation and osteogenic differentiation of BMSCs.

4.
Sci Rep ; 14(1): 4887, 2024 02 28.
Article in English | MEDLINE | ID: mdl-38418857

ABSTRACT

Iron is an essential nutrient for all living organisms. Both iron deficiency and excess can be harmful. Bone, a highly metabolic active organ, is particularly sensitive to fluctuations in iron levels. In this study, we investigated the effects of dietary iron overload on bone homeostasis with a specific focus on two frequently utilized mouse strains: 129/Sv and C57BL/6J. Our findings revealed that after 6 weeks on an iron-rich diet, 129/Sv mice exhibited a decrease in trabecular and cortical bone density in both vertebral and femoral bones, which was linked to reduced bone turnover. In contrast, there was no evidence of bone changes associated with iron overload in age-matched C57BL/6J mice. Interestingly, 129/Sv mice exposed to an iron-rich diet during their prenatal development were protected from iron-induced bone loss, suggesting the presence of potential adaptive mechanisms. Overall, our study underscores the critical role of genetic background in modulating the effects of iron overload on bone health. This should be considered when studying effects of iron on bone.


Subject(s)
Iron Overload , Iron, Dietary , Pregnancy , Female , Mice , Animals , Mice, Inbred C57BL , Bone and Bones , Mice, Inbred Strains , Iron Overload/complications , Iron
5.
Immunity ; 57(2): 364-378.e9, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38301651

ABSTRACT

Mutations of the CBP/p300 histone acetyltransferase (HAT) domain can be linked to leukemic transformation in humans, suggestive of a checkpoint of leukocyte compartment sizes. Here, we examined the impact of reversible inhibition of this domain by the small-molecule A485. We found that A485 triggered acute and transient mobilization of leukocytes from the bone marrow into the blood. Leukocyte mobilization by A485 was equally potent as, but mechanistically distinct from, granulocyte colony-stimulating factor (G-CSF), which allowed for additive neutrophil mobilization when both compounds were combined. These effects were maintained in models of leukopenia and conferred augmented host defenses. Mechanistically, activation of the hypothalamus-pituitary-adrenal gland (HPA) axis by A485 relayed shifts in leukocyte distribution through corticotropin-releasing hormone receptor 1 (CRHR1) and adrenocorticotropic hormone (ACTH), but independently of glucocorticoids. Our findings identify a strategy for rapid expansion of the blood leukocyte compartment via a neuroendocrine loop, with implications for the treatment of human pathologies.


Subject(s)
Bone Marrow , Histone Acetyltransferases , Humans , Histone Acetyltransferases/metabolism , Bone Marrow/metabolism , Histones/metabolism , Neutrophils/metabolism , Hypothalamo-Hypophyseal System/metabolism
6.
J Cell Physiol ; 239(2): e31172, 2024 02.
Article in English | MEDLINE | ID: mdl-38214117

ABSTRACT

Periodontitis is associated with significant alveolar bone loss. Patients with iron overload suffer more frequently from periodontitis, however, the underlying mechanisms remain largely elusive. Here, we investigated the role of transferrin receptor 2 (Tfr2), one of the main regulators of iron homeostasis, in the pathogenesis of periodontitis and the dental phenotype under basal conditions in mice. As Tfr2 suppresses osteoclastogenesis, we hypothesized that deficiency of Tfr2 may exacerbate periodontitis-induced bone loss. Mice lacking Tfr2 (Tfr2-/- ) and wild-type (Tfr2+/+ ) littermates were challenged with experimental periodontitis. Mandibles and maxillae were collected for microcomputed tomography and histology analyses. Osteoclast cultures from Tfr2+/+ and Tfr2-/- mice were established and analyzed for differentiation efficiency, by performing messenger RNA expression and protein signaling pathways. After 8 days, Tfr2-deficient mice revealed a more severe course of periodontitis paralleled by higher immune cell infiltration and a higher histological inflammation index than Tfr2+/+ mice. Moreover, Tfr2-deficient mice lost more alveolar bone compared to Tfr2+/+ littermates, an effect that was only partially iron-dependent. Histological analysis revealed a higher number of osteoclasts in the alveolar bone of Tfr2-deficient mice. In line, Tfr2-deficient osteoclastic differentiation ex vivo was faster and more efficient as reflected by a higher number of osteoclasts, a higher expression of osteoclast markers, and an increased resorptive activity. Mechanistically, Tfr2-deficient osteoclasts showed a higher p38-MAPK signaling and inhibition of p38-MAPK signaling in Tfr2-deficient cells reverted osteoclast formation to Tfr2+/+ levels. Taken together, our data indicate that Tfr2 modulates the inflammatory response in periodontitis thereby mitigating effects on alveolar bone loss.


Subject(s)
Alveolar Bone Loss , Periodontitis , Animals , Humans , Mice , Alveolar Bone Loss/genetics , Alveolar Bone Loss/metabolism , Iron , Osteoclasts , Periodontitis/genetics , Periodontitis/metabolism , Receptors, Transferrin/genetics , X-Ray Microtomography , Mice, Inbred C57BL , Cells, Cultured
7.
Am J Physiol Endocrinol Metab ; 324(6): E514-E530, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37126848

ABSTRACT

Elevated serum concentrations of glucocorticoids (GCs) result in excessive lipid accumulation in white adipose tissue (WAT) as well as dysfunction of thermogenic brown adipose tissue (BAT), ultimately leading to the development of obesity and metabolic disease. Here, we hypothesized that activation of the sympathetic nervous system either via cold exposure or the use of a selective ß3-adrenergic receptor (ß3-AR) agonist alleviates the adverse metabolic effects of chronic GC exposure in rodents. To this end, male 10-wk-old C57BL/6NRj mice were treated with corticosterone via drinking water or placebo for 4 wk while being maintained at 29°C (thermoneutrality), 22°C (room temperature), or 13°C (cold temperature); in a follow-up study mice received a selective ß3-AR agonist or placebo with and without corticosterone while being maintained at room temperature. Body weight and food intake were monitored throughout the study. Histological and molecular analyses were performed on white and brown adipose depots. Cold exposure not only preserved the thermogenic function of brown adipose tissue but also reversed GC-induced lipid accumulation in white adipose tissue and corrected GC-driven obesity, hyperinsulinemia, and hyperglycemia. The metabolic benefits of cold exposure were associated with enhanced sympathetic activity in adipose tissue, thus potentially linking an increase in sympathetic signaling to the observed metabolic benefits. In line with this concept, chronic administration of a selective ß3-AR agonist reproduced the beneficial metabolic effects of cold adaption during exposure to exogenous GCs. This preclinical study demonstrates the potential of ß3-AR as a therapeutic target in the management and prevention of GC-induced metabolic disease.NEW & NOTEWORTHY This preclinical study in mice shows that the ß3-adrenergic receptor can be a potential therapeutic approach to counteracting glucocorticoid (GC)-induced obesity and metabolic dysfunction. Both cold acclimation and ß3-adrenergic receptor stimulation in a mouse model of excess glucocorticoids were adequate in not only preventing obesity, adiposity, and adipose tissue dysfunction but also correcting hyperinsulinemia, hyperleptinemia, and dyslipidemia.


Subject(s)
Glucocorticoids , Receptors, Adrenergic, beta , Male , Animals , Mice , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , Receptors, Adrenergic, beta/metabolism , Corticosterone/metabolism , Follow-Up Studies , Mice, Inbred C57BL , Adipose Tissue/metabolism , Obesity/chemically induced , Obesity/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Receptors, Adrenergic, beta-3/metabolism , Lipids , Thermogenesis
8.
Redox Biol ; 60: 102616, 2023 04.
Article in English | MEDLINE | ID: mdl-36746004

ABSTRACT

OBJECTIVE: Rheumatoid arthritis is an inflammatory joint disease in which synovial iron deposition has been described. Transferrin receptor 2 (Tfr2) represents a critical regulator of systemic iron levels. Loss of Tfr2 function in humans and mice results in iron overload. As iron contributes to inflammatory processes, we investigated whether Tfr2-deletion affects the pathogenesis of inflammatory arthritis in an iron-dependent manner. METHODS: Using a global and conditional genetic disruption of Tfr2, we assessed the relevance of Tfr2 in K/BxN serum-transfer arthritis (STA) and macrophage polarization. RESULTS: Male Tfr2-/- mice subjected to STA developed pronounced joint swelling, and bone erosion as compared to Tfr2+/+ littermate-controls (P < 0.01). Furthermore, an increase of neutrophils and macrophages/monocytes was observed in the inflammatory infiltrate within the paws of Tfr2-/- mice. To elucidate whether Tfr2 in myeloid cells has a direct role in the pathogenesis of arthritis or whether the effects were mediated via the systemic iron overload, we induced STA in Tfr2fl/fl-LysMCre + mice, which showed normal iron-loading. Cre + female mice displayed increased disease development compared to Cre-controls. As macrophages regulate iron availability and innate immunity, we hypothesized that Tfr2-deficiency would polarize macrophages toward a pro-inflammatory state (M1) that contributes to arthritis progression. In response to IFN-γ stimulation, Tfr2-/- macrophages showed increased expression of M1-like cytokines, IFN-γ-target genes, nitric-oxide production, and prolonged STAT1 activation compared to Tfr2+/+ macrophages (P < 0.01), while pre-treatment with ruxolitinib abolished Tfr2-driven M1-like polarization. CONCLUSION: Taken together, these findings suggest a protective role of Tfr2 in macrophages on the progression of arthritis via suppression of M1-like polarization.


Subject(s)
Arthritis , Iron Overload , Humans , Mice , Male , Female , Animals , Mice, Knockout , Iron/metabolism , Iron Overload/pathology , Macrophages/metabolism , Arthritis/metabolism , Receptors, Transferrin/genetics
10.
Trends Endocrinol Metab ; 33(9): 652-663, 2022 09.
Article in English | MEDLINE | ID: mdl-35871125

ABSTRACT

Hereditary hemochromatosis (HH) is a genetic disorder in which mutations affect systemic iron homeostasis. Most subtypes of HH result in low hepcidin levels and iron overload. Accumulation of iron in various tissues can lead to widespread organ damage and to various complications, including liver cirrhosis, arthritis, and diabetes. Osteoporosis is another frequent complication of HH, and the underlying mechanisms are poorly understood. Currently, it is unknown whether iron overload in HH directly damages bone or whether complications associated with HH, such as liver cirrhosis or hypogonadism, affect bone secondarily. This review summarizes current knowledge of bone metabolism in HH and highlights possible implications of metabolic dysfunction in HH-driven bone loss. We further discuss therapeutic considerations managing osteoporosis in HH.


Subject(s)
Hemochromatosis , Iron Overload , Osteoporosis , Hemochromatosis/complications , Hemochromatosis/genetics , Hemochromatosis/metabolism , Humans , Iron/metabolism , Iron Overload/complications , Iron Overload/genetics , Liver Cirrhosis/complications , Osteoporosis/genetics
11.
12.
Front Endocrinol (Lausanne) ; 13: 835720, 2022.
Article in English | MEDLINE | ID: mdl-35432217

ABSTRACT

Despite the continued development of specialized immunosuppressive therapies in the form of monoclonal antibodies, glucocorticoids remain a mainstay in the treatment of rheumatological and auto-inflammatory disorders. Therapeutic glucocorticoids are unmatched in the breadth of their immunosuppressive properties and deliver their anti-inflammatory effects at unparalleled speed. However, long-term exposure to therapeutic doses of glucocorticoids decreases bone mass and increases the risk of fractures - particularly in the spine - thus limiting their clinical use. Due to the abundant expression of glucocorticoid receptors across all skeletal cell populations and their respective progenitors, therapeutic glucocorticoids affect skeletal quality through a plethora of cellular targets and molecular mechanisms. However, recent evidence from rodent studies, supported by clinical data, highlights the considerable role of cells of the osteoblast lineage in the pathogenesis of glucocorticoid-induced osteoporosis: it is now appreciated that cells of the osteoblast lineage are key targets of therapeutic glucocorticoids and have an outsized role in mediating their undesirable skeletal effects. As part of this article, we review the molecular mechanisms underpinning the detrimental effects of supraphysiological levels of glucocorticoids on cells of the osteoblast lineage including osteocytes and highlight the clinical implications of recent discoveries in the field.


Subject(s)
Osteocytes , Osteoporosis , Bone and Bones/metabolism , Glucocorticoids/metabolism , Humans , Osteoblasts/metabolism , Osteocytes/metabolism , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Osteoporosis/metabolism
13.
Semin Hematol ; 58(3): 188-200, 2021 07.
Article in English | MEDLINE | ID: mdl-34389111

ABSTRACT

Well-controlled iron levels are indispensable for health. Iron deficiency is the most common cause of anemia, whereas iron overload, either hereditary or secondary due to disorders of ineffective erythropoiesis, causes widespread organ failure. Bone is particularly sensitive to fluctuations in systemic iron levels as both iron deficiency and overload are associated with low bone mineral density and fragility. Recent studies have shown that not only iron itself, but also iron-regulatory proteins that are mutated in hereditary hemochromatosis can control bone mass. This review will summarize the current knowledge on the effects of iron on bone homeostasis and bone cell activities, and on the role of proteins that regulate iron homeostasis, i.e. hemochromatosis proteins and proteins of the bone morphogenetic protein pathway, on bone remodeling. As disorders of iron homeostasis are closely linked to bone fragility, deeper insights into common regulatory mechanisms may provide new opportunities to concurrently treat disorders affecting iron homeostasis and bone.


Subject(s)
Hemochromatosis , Iron Deficiencies , Iron Overload , Hemochromatosis/complications , Hemochromatosis/genetics , Hemochromatosis/therapy , Homeostasis , Humans , Iron/metabolism , Iron Overload/complications
14.
Front Immunol ; 12: 665208, 2021.
Article in English | MEDLINE | ID: mdl-34149700

ABSTRACT

Bone marrow adipose tissue (BMAT) has recently been recognized as a distinct fat depot with endocrine functions. However, if and how it is regulated by chronic inflammation remains unknown. Here, we investigate the amount of white fat and BMAT in HLA-B27 transgenic rats and curdlan-challenged SKG mice, two well-established models of chronic inflammatory spondyloarthritis (SpA). Subcutaneous and gonadal white adipose tissue and BMAT was reduced by 65-70% and by up to 90% in both experimental models. Consistently, B27 rats had a 2-3-fold decrease in the serum concentrations of the adipocyte-derived cytokines adiponectin and leptin as well as a 2-fold lower concentration of triglycerides. The bone marrow of B27 rats was further characterized by higher numbers of neutrophils, lower numbers of erythroblast precursors, and higher numbers of IL-17 producing CD4+ T cells. IL-17 concentration was also increased in the serum of B27 rats. Using a cell culture model, we show that high levels of IL-17 in the serum of B27 rats negatively impacted adipogenesis (-76%), an effect that was reversed in the presence of neutralizing anti-IL-17 antibody. In summary, these findings show BMAT is severely reduced in two experimental models of chronic inflammatory SpA and suggest that IL-17 is involved in this process.


Subject(s)
Adipose Tissue/pathology , Bone Marrow/pathology , HLA-B27 Antigen/genetics , Interleukin-17/blood , Spondylarthritis/pathology , Animals , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Mice, Transgenic , Rats , Rats, Inbred F344 , Rats, Transgenic , Spondylarthritis/genetics , Spondylarthritis/immunology
15.
iScience ; 24(3): 102224, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33748710

ABSTRACT

The Wnt inhibitor Dickkopf-1 (DKK1) is a negative regulator of bone formation and bone mass and is dysregulated in various bone diseases. How DKK1 contributes to postmenopausal osteoporosis, however, remains poorly understood. Here, we show that mice lacking DKK1 in T cells are protected from ovariectomy-induced bone loss. Ovariectomy activated CD4+ and CD8+ T cells and increased their production of DKK1. Co-culture of activated T cells with osteoblasts inhibited Wnt signaling in osteoblasts, leading to impaired differentiation. Importantly, DKK1 expression in T cells also controlled physiological bone remodeling. T-cell-deficient Dkk1 knock-out mice had a higher bone mass with an increased bone formation rate and decreased numbers of osteoclasts compared with controls, a phenotype that was rescued by adoptive transfer of wild-type T cells. Thus, these findings highlight that T cells control bone remodeling in health and disease via their expression of DKK1.

16.
Front Endocrinol (Lausanne) ; 12: 782118, 2021.
Article in English | MEDLINE | ID: mdl-34975756

ABSTRACT

Glucocorticoid-induced osteoporosis (GIOP) leads to fractures in up to 40% of patients with chronic glucocorticoid (GC) therapy when left untreated. GCs rapidly increase fracture risk, and thus many patients with anticipated chronic GC exposures should start anti-osteoporosis pharmacotherapy to prevent fractures. In addition to low awareness of the need for anti-osteoporosis therapy among clinicians treating patients with GCs, a major barrier to prevention of fractures from GIOP is a lack of clear guideline recommendations on when to start and stop anti-osteoporosis treatment in patients with GC use. The aim of this narrative review is to summarize current evidence and provide considerations for the duration of anti-osteoporosis treatment in patients taking GCs based on pre-clinical, clinical, epidemiologic, and pharmacologic evidence. We review the pathophysiology of GIOP, outline current guideline recommendations on initiating and stopping anti-osteoporosis therapy for GIOP, and present considerations for the duration of anti-osteoporosis treatment based on existing evidence. In each section, we illustrate major points through a patient case example. Finally, we conclude with proposed areas for future research and emerging areas of interest related to GIOP clinical management.


Subject(s)
Bone Density Conservation Agents/administration & dosage , Bone Density/drug effects , Glucocorticoids/adverse effects , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Withholding Treatment , Bone Density/physiology , Female , Fractures, Bone/diagnostic imaging , Fractures, Bone/prevention & control , Glucocorticoids/administration & dosage , Humans , Lupus Erythematosus, Systemic/diagnostic imaging , Lupus Erythematosus, Systemic/drug therapy , Middle Aged , Osteoporosis/diagnostic imaging , Time-to-Treatment
17.
J Clin Endocrinol Metab ; 106(1): e288-e299, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33034626

ABSTRACT

CONTEXT: Osteoporosis and anemia are among the most common diseases in the aging population with an increasing prevalence worldwide. OBJECTIVE: As the bone-derived hormone fibroblast growth factor 23 (FGF-23) was recently reported to regulate erythropoiesis, we examined age-related associations between hemoglobin levels and bone quality, bone turnover, and FGF-23 concentrations. DESIGN: We used data from more than 5000 adult subjects who participated in the population-based cohorts of the Study of Health in Pomerania (SHIP and SHIP-Trend). Bone quality was assessed by quantitative ultrasound at the heel, bone turnover by measurement of carboxy-terminal telopeptide of type I collagen (CTX), and intact amino-terminal propeptide of type I procollagen (P1NP) serum concentrations, respectively. Anemia was defined as hemoglobin <13 g/dL in men and <12 g/dL in women. Carboxy-terminal FGF-23 levels were measured in plasma in a subset of 852 subjects. RESULTS: Anemic subjects had poorer bone quality, higher fracture risk, and lower serum levels of P1NP than nonanemic individuals. Linear regression models revealed positive associations between hemoglobin and bone quality in subjects aged 40 or above and inverse associations with CTX in subjects aged 60 or above. Hemoglobin and FGF-23 concentrations were inversely associated, while FGF-23 was not related to bone quality or turnover. CONCLUSION: Our data corroborate a close link between FGF-23 and anemia, which is related to poor bone quality in elderly people. We observed no direct association of FGF-23 with bone parameters. Further studies are needed clarifying the role of FGF-23 on bone and red blood cell production.


Subject(s)
Anemia , Bone and Bones/physiology , Fibroblast Growth Factors/blood , Adult , Aged , Anemia/blood , Anemia/complications , Anemia/epidemiology , Bone Density/physiology , Cohort Studies , Female , Fibroblast Growth Factor-23 , Germany/epidemiology , Healthy Volunteers , Humans , Male , Middle Aged , Osteoporosis/blood , Osteoporosis/epidemiology , Osteoporosis/etiology , Osteoporotic Fractures/blood , Osteoporotic Fractures/epidemiology , Osteoporotic Fractures/etiology , Risk Factors , Young Adult
18.
JCI Insight ; 5(15)2020 08 06.
Article in English | MEDLINE | ID: mdl-32759495

ABSTRACT

Myelodysplastic syndromes (MDS) are clonal malignant hematopoietic disorders in the elderly characterized by ineffective hematopoiesis. This is accompanied by an altered bone microenvironment, which contributes to MDS progression and higher bone fragility. The underlying mechanisms remain largely unexplored. Here, we show that myelodysplastic NUP98­HOXD13 (NHD13) transgenic mice display an abnormally high number of osteoblasts, yet a higher fraction of nonmineralized bone, indicating delayed bone mineralization. This was accompanied by high fibroblast growth factor-23 (FGF-23) serum levels, a phosphaturic hormone that inhibits bone mineralization and erythropoiesis. While Fgf23 mRNA expression was low in bone, brain, and kidney of NHD13 mice, its expression was increased in erythroid precursors. Coculturing these precursors with WT osteoblasts induced osteoblast marker gene expression, which was inhibited by blocking FGF-23. Finally, antibody-based neutralization of FGF-23 in myelodysplastic NHD13 mice improved bone mineralization and bone microarchitecture, and it ameliorated anemia. Importantly, higher serum levels of FGF­23 and an elevated amount of nonmineralized bone in patients with MDS validated the findings. C­terminal FGF­23 correlated negatively with hemoglobin levels and positively with the amount of nonmineralized bone. Thus, our study identifies FGF-23 as a link between altered bone structure and ineffective erythropoiesis in MDS with the prospects of a targeted therapeutic intervention.


Subject(s)
Bone Diseases/diagnosis , Calcification, Physiologic , Erythropoiesis , Fibroblast Growth Factors/blood , Homeodomain Proteins/physiology , Myelodysplastic Syndromes/complications , Nuclear Pore Complex Proteins/physiology , Oncogene Proteins, Fusion/physiology , Osteoblasts/pathology , Aged , Animals , Bone Diseases/blood , Bone Diseases/etiology , Bone Remodeling , Case-Control Studies , Female , Fibroblast Growth Factor-23 , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Osteoblasts/metabolism
19.
Sci Rep ; 10(1): 9344, 2020 Jun 04.
Article in English | MEDLINE | ID: mdl-32494045

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

20.
Bone ; 137: 115400, 2020 08.
Article in English | MEDLINE | ID: mdl-32380257

ABSTRACT

Ferroportin (FPN) is the only known iron exporter. Mutations conferring resistance of FPN to hepcidin-mediated degradation cause the iron overload disorder hereditary hemochromatosis type 4. While iron overload is associated with low bone mass, the mechanisms involved are not completely understood. Here, we aimed to investigate whether the disruption in the hepcidin/FPN axis in FpnC326S mice and subsequent systemic iron accumulation impacts on bone tissue to a similar extent as in Hfe-/- mice, which are hallmarked by a milder iron overload phenotype. Hfe-/- and FpnC326S mice show increased plasma iron levels and liver iron content, whereas iron overload was more pronounced in FpnC326S compared to Hfe-/- mice. Bone volume fraction and trabecular thickness at the femur were not different between 10 and 14-week-old male wild-type (WT), Hfe-/- and FpnC326S mice. By contrast, both Hfe-/- and FpnC326S mice exhibited a lower bone volume fraction [Hfe-/-, 24%; FpnC326S, 33%; p < 0.05] and trabecular thickness [Hfe-/-, 10%; FpnC326S, 15%; p < 0.05] in the fourth lumbar vertebra compared to WT mice. Analysis of the bone formation rate at the tibia showed no difference in both genotypes, but it was reduced in the vertebral bone of FpnC326S [36%, p < 0.05] compared to WT mice. Serum levels of the bone formation marker, P1NP, were significantly reduced in both, Hfe-/- and FpnC326S compared with WT mice [Hfe-/-, 35%; FpnC326S, 40%; p < 0.05]. Also, the intrinsic differentiation capacity of FpnC326S osteoblasts was impaired. Osteoclast parameters were not grossly affected. Interestingly, the liver iron content and plasma iron levels negatively correlated with the bone formation rate and serum levels of P1NP. Thus, disruption of the hepcidin/ferroportin regulatory axis in FpnC326S mice results in axial bone loss due to suppressed bone formation.


Subject(s)
Cation Transport Proteins , Hemochromatosis , Iron Overload , Animals , Cation Transport Proteins/genetics , Hepcidins/genetics , Hepcidins/metabolism , Male , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL