Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Mol Syst Biol ; 18(8): e10961, 2022 08.
Article in English | MEDLINE | ID: mdl-35975552

ABSTRACT

Cell-intrinsic responses mounted in PBMCs during mild and severe COVID-19 differ quantitatively and qualitatively. Whether they are triggered by signals emitted by productively infected cells of the respiratory tract or result from physical interaction with virus particles remains unclear. Here, we analyzed susceptibility and expression profiles of PBMCs from healthy donors upon ex vivo exposure to SARS-CoV and SARS-CoV-2. In line with the absence of detectable ACE2 receptor expression, human PBMCs were refractory to productive infection. RT-PCR experiments and single-cell RNA sequencing revealed JAK/STAT-dependent induction of interferon-stimulated genes (ISGs) but not proinflammatory cytokines. This SARS-CoV-2-specific response was most pronounced in monocytes. SARS-CoV-2-RNA-positive monocytes displayed a lower ISG signature as compared to bystander cells of the identical culture. This suggests a preferential invasion of cells with a low ISG baseline profile or delivery of a SARS-CoV-2-specific sensing antagonist upon efficient particle internalization. Together, nonproductive physical interaction of PBMCs with SARS-CoV-2- and, to a much lesser extent, SARS-CoV particles stimulate JAK/STAT-dependent, monocyte-accentuated innate immune responses that resemble those detected in vivo in patients with mild COVID-19.


Subject(s)
COVID-19 , Severe acute respiratory syndrome-related coronavirus , Humans , Immunity, Innate , Interferons , SARS-CoV-2
2.
PLoS Genet ; 8(5): e1002726, 2012.
Article in English | MEDLINE | ID: mdl-22615584

ABSTRACT

Maintenance of normal mineral ion homeostasis is crucial for many biological activities, including proper mineralization of the skeleton. Parathyroid hormone (PTH), Klotho, and FGF23 have been shown to act as key regulators of serum calcium and phosphate homeostasis through a complex feedback mechanism. The phenotypes of Fgf23(-/-) and Klotho(-/-) (Kl(-/-)) mice are very similar and include hypercalcemia, hyperphosphatemia, hypervitaminosis D, suppressed PTH levels, and severe osteomalacia/osteoidosis. We recently reported that complete ablation of PTH from Fgf23(-/-) mice ameliorated the phenotype in Fgf23(-/-)/PTH(-/-) mice by suppressing serum vitamin D and calcium levels. The severe osteomalacia in Fgf23(-/-) mice, however, persisted, suggesting that a different mechanism is responsible for this mineralization defect. In the current study, we demonstrate that deletion of PTH from Kl(-/-) (Kl(-/-)/PTH(-/-) or DKO) mice corrects the abnormal skeletal phenotype. Bone turnover markers are restored to wild-type levels; and, more importantly, the skeletal mineralization defect is completely rescued in Kl(-/-)/PTH(-/-) mice. Interestingly, the correction of the osteomalacia is accompanied by a reduction in the high levels of osteopontin (Opn) in bone and serum. Such a reduction in Opn levels could not be observed in Fgf23(-/-)/PTH(-/-) mice, and these mice showed sustained osteomalacia. This significant in vivo finding is corroborated by in vitro studies using calvarial osteoblast cultures that show normalized Opn expression and rescued mineralization in Kl(-/-)/PTH(-/-) mice. Moreover, continuous PTH infusion of Kl(-/-) mice significantly increased Opn levels and osteoid volume, and decreased trabecular bone volume. In summary, our results demonstrate for the first time that PTH directly impacts the mineralization disorders and skeletal deformities of Kl(-/-), but not of Fgf23(-/-) mice, possibly by regulating Opn expression. These are significant new perceptions into the role of PTH in skeletal and disease processes and suggest FGF23-independent interactions of PTH with Klotho.


Subject(s)
Bone and Bones/metabolism , Calcification, Physiologic , Fibroblast Growth Factors , Glucuronidase , Osteopontin , Parathyroid Hormone , Animals , Calcification, Physiologic/genetics , Calcium/blood , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Glucuronidase/genetics , Glucuronidase/metabolism , Klotho Proteins , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , Osteomalacia/metabolism , Osteopontin/blood , Osteopontin/genetics , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Phosphates/blood , Vitamin D/blood
3.
PLoS Genet ; 4(8): e1000154, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18688277

ABSTRACT

Maintenance of physiologic phosphate balance is of crucial biological importance, as it is fundamental to cellular function, energy metabolism, and skeletal mineralization. Fibroblast growth factor-23 (FGF-23) is a master regulator of phosphate homeostasis, but the molecular mechanism of such regulation is not yet completely understood. Targeted disruption of the Fgf-23 gene in mice (Fgf-23-/-) elicits hyperphosphatemia, and an increase in renal sodium/phosphate co-transporter 2a (NaPi2a) protein abundance. To elucidate the pathophysiological role of augmented renal proximal tubular expression of NaPi2a in Fgf-23-/- mice and to examine serum phosphate-independent functions of Fgf23 in bone, we generated a new mouse line deficient in both Fgf-23 and NaPi2a genes, and determined the effect of genomic ablation of NaPi2a from Fgf-23-/- mice on phosphate homeostasis and skeletal mineralization. Fgf-23-/-/NaPi2a-/- double mutant mice are viable and exhibit normal physical activities when compared to Fgf-23-/- animals. Biochemical analyses show that ablation of NaPi2a from Fgf-23-/- mice reversed hyperphosphatemia to hypophosphatemia by 6 weeks of age. Surprisingly, despite the complete reversal of serum phosphate levels in Fgf-23-/-/NaPi2a-/-, their skeletal phenotype still resembles the one of Fgf23-/- animals. The results of this study provide the first genetic evidence of an in vivo pathologic role of NaPi2a in regulating abnormal phosphate homeostasis in Fgf-23-/- mice by deletion of both NaPi2a and Fgf-23 genes in the same animal. The persistence of the skeletal anomalies in double mutants suggests that Fgf-23 affects bone mineralization independently of systemic phosphate homeostasis. Finally, our data support (1) that regulation of phosphate homeostasis is a systemic effect of Fgf-23, while (2) skeletal mineralization and chondrocyte differentiation appear to be effects of Fgf-23 that are independent of phosphate homeostasis.


Subject(s)
Bone and Bones/metabolism , Fibroblast Growth Factors/metabolism , Hypophosphatemia/metabolism , Phosphates/metabolism , Animals , Bone Density , Bone and Bones/physiopathology , Calcification, Physiologic , Cells, Cultured , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Gene Expression , Hypophosphatemia/genetics , Hypophosphatemia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Osteoblasts/metabolism , Phenotype , Serum/chemistry , Skull/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Urine/chemistry
4.
Life Sci Alliance ; 4(7)2021 07.
Article in English | MEDLINE | ID: mdl-34078739

ABSTRACT

Interferon-induced transmembrane (IFITM) proteins restrict membrane fusion and virion internalization of several enveloped viruses. The role of IFITM proteins during alphaviral infection of human cells and viral counteraction strategies are insufficiently understood. Here, we characterized the impact of human IFITMs on the entry and spread of chikungunya virus and Mayaro virus and provide first evidence for a CHIKV-mediated antagonism of IFITMs. IFITM1, 2, and 3 restricted infection at the level of alphavirus glycoprotein-mediated entry, both in the context of direct infection and cell-to-cell transmission. Relocalization of normally endosomal IFITM3 to the plasma membrane resulted in loss of antiviral activity. rs12252-C, a naturally occurring variant of IFITM3 that may associate with severe influenza in humans, restricted CHIKV, MAYV, and influenza A virus infection as efficiently as wild-type IFITM3 Antivirally active IFITM variants displayed reduced cell surface levels in CHIKV-infected cells involving a posttranscriptional process mediated by one or several nonstructural protein(s) of CHIKV. Finally, IFITM3-imposed reduction of specific infectivity of nascent particles provides a rationale for the necessity of a virus-encoded counteraction strategy against this restriction factor.


Subject(s)
Alphavirus Infections/prevention & control , Chikungunya Fever/prevention & control , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Alphavirus/pathogenicity , Alphavirus Infections/metabolism , Alphavirus Infections/virology , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Chikungunya Fever/metabolism , Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Endosomes/metabolism , Humans , Membrane Proteins/physiology , RNA-Binding Proteins/physiology , Virus Internalization
5.
J Nutr ; 139(4): 703-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19211826

ABSTRACT

In this experiment, we investigated the long-term effects of a marginal zinc (Zn) supply on bone metabolism in aged rats. Nine-mo-old female Fischer-344 rats were divided into 8 weight-matched groups of 8 rats each. All rats were adapted for 1 mo to restrictive feeding (7.5 g/d) of a purified diet containing 8 g/kg sodium phytate and 64 mg/kg Zn. Control rats were pair-fed throughout the experiment. During the 1-mo depletion phase, controls received the Zn-replete diet with 64 mg/kg Zn, whereas Zn-deficient rats were fed the same diet with 2.2 mg/kg Zn. The depletion phase was followed by a 3-mo marginal phase in which the rats fed the diet with 2.2 mg/kg Zn received an additional daily Zn supplement of 75 microg Zn/rat by gavage. In the following 2-mo repletion phase, a marginal group was switched to the Zn-replete diet, while the other groups were maintained on marginal Zn supply or on the Zn-replete diet. Zn depletion and marginal Zn reduced serum and bone Zn and serum alkaline phosphatase activity. Zn repletion normalized serum Zn. However, apart from subtle changes in bone mineralization density distribution, Zn deficiency was not associated with detrimental effects on bone mineral density, turnover, architecture, or biomechanics relative to control rats at any time point. Our data suggest that Zn does not play an essential role in bone metabolism in aged rats and cast doubt on the hypthosis that Zn deficiency is a risk factor for osteoporosis.


Subject(s)
Aging/drug effects , Zinc/pharmacology , Animals , Body Weight/drug effects , Bone Density/drug effects , Bone and Bones/drug effects , Feeding Behavior/drug effects , Female , Rats , Rats, Inbred F344 , Weight-Bearing
6.
Sci Rep ; 9(1): 10153, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31300685

ABSTRACT

Intra-articular (IA) injection of mesenchymal stem cells (MSCs) promotes articular cartilage repair. However, cell fate and action after transplantation remain unclear. This study aimed at evaluating the biodistribution and efficacy of MSCs after IA injection. We used an immunocompetent, dual transgenic rat model, which is based on donor rats ubiquitously expressing heat stable human placental alkaline phosphatase (ALPP), and recipient rats expressing a heat sensitive ALPP form. A focal cartilage defect was created in the patellofemoral groove of recipient rats. Bone marrow-derived MSCs isolated from donor rats were injected into the synovial cavity of recipients, and cell tracking was performed in distant organs and knees over 6 months post-injection. A few donor MSCs were observed in the lung of one of the recipients, 1 day post-injection. We failed to detect donor MSCs in any of the studied tissues at all later time points. IA-injected MSCs remained in the synovial cavity, engrafted within the cartilage lesion, and were detectable up to 1 month post-injection. Although the number of MSCs decreased over time, MSCs injection promoted cartilage regeneration as evidenced by histology and immunofluorescent collagen staining. Our study supports the safety and efficacy of using MSCs for cartilage repair via IA delivery.


Subject(s)
Cartilage, Articular/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Regeneration/physiology , Alkaline Phosphatase/genetics , Animals , Cartilage, Articular/cytology , Cell Survival , GPI-Linked Proteins/genetics , Injections, Intra-Articular , Isoenzymes/genetics , Knee Joint/cytology , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/physiology , Rats, Inbred F344 , Rats, Transgenic , Tissue Distribution
7.
J Vis Exp ; (151)2019 09 16.
Article in English | MEDLINE | ID: mdl-31566615

ABSTRACT

RNA yield and integrity are decisive for RNA analysis. However, it is often technically challenging to maintain RNA integrity throughout the entire laser capture microdissection (LCM) procedure. Since LCM studies work with low amounts of material, concerns about limited RNA yields are also important. Therefore, an LCM protocol was developed to obtain sufficient quantity of high-quality RNA for gene expression analysis in bone cells. The effect of staining protocol, thickness of cryosections, microdissected tissue quantity, RNA extraction kit, and LCM system used on RNA yield and integrity obtained from microdissected bone cells was evaluated. Eight-µm-thick frozen bone sections were made using an adhesive film and stained using a rapid protocol for a commercial LCM stain. The sample was sandwiched between a polyethylene terephthalate (PET) membrane and the adhesive film. An LCM system that uses gravity for sample collection and a column-based RNA extraction method were used to obtain high quality RNAs of sufficient yield. The current study focusses on mouse femur sections. However, the LCM protocol reported here can be used to study in situ gene expression in cells of any hard tissue in both physiological conditions and disease processes.


Subject(s)
Femur/physiology , Frozen Sections/methods , Laser Capture Microdissection/methods , RNA/physiology , Animals , Femur/chemistry , Gene Expression Profiling/methods , Male , Mice , Mice, Inbred C57BL , RNA/analysis , Staining and Labeling/methods
8.
Article in English | MEDLINE | ID: mdl-29942284

ABSTRACT

Chronic kidney disease-mineral and bone disorder (CKD-MBD) is a systemic disorder of mineral and bone metabolism caused by CKD. Impaired bone mineralization together with increased bony secretion of fibroblast growth factor-23 (FGF23) are hallmarks of CKD-MBD. We recently showed that FGF23 suppresses the expression of tissue nonspecific alkaline phosphatase (TNAP) in bone cells by a Klotho-independent, FGF receptor-3-mediated signaling axis, leading to the accumulation of the mineralization inhibitor pyrophosphate. Therefore, we hypothesized that excessive FGF23 secretion may locally impair bone mineralization in CKD-MBD. To test this hypothesis, we induced CKD by 5/6 nephrectomy in 3-month-old wild-type (WT) mice and Fgf23-/-/VDRΔ/Δ (Fgf23/VDR) compound mutant mice maintained on a diet enriched with calcium, phosphate, and lactose. Eight weeks postsurgery, WT CKD mice were characterized by reduced bone mineral density at the axial and appendicular skeleton, hyperphosphatemia, secondary hyperparathyroidism, increased serum intact Fgf23, and impaired bone mineralization as evidenced by bone histomorphometry. Laser capture microdissection in bone cryosections showed that both osteoblasts and osteocytes contributed to the CKD-induced increase in Fgf23 mRNA abundance. In line with our hypothesis, osteoblastic and osteocytic activity of alkaline phosphatase was reduced, and bone pyrophosphate concentration was ~2.5-fold higher in CKD mice, relative to Sham controls. In Fgf23/VDR compound mice lacking Fgf23, 5/6-Nx induced secondary hyperparathyroidism and bone loss. However, 5/6-Nx failed to suppress TNAP activity, and bone pyrophosphate concentrations remained unchanged in Fgf23/VDR CKD mice. Collectively, our data suggest that elevated Fgf23 production in bone contributes to the mineralization defect in CKD-MBD by auto-/paracrine suppression of TNAP and subsequent accumulation of pyrophosphate in bone. Hence, our study has identified a novel mechanism involved in the pathogenesis of CKD-MBD.

9.
J Bone Miner Res ; 32(10): 2049-2061, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28600880

ABSTRACT

Fibroblast growth factor-23 (FGF23) is a bone-derived hormone regulating vitamin D hormone production and renal handling of minerals by signaling through an FGF receptor/αKlotho (Klotho) receptor complex. Whether Klotho has FGF23-independent effects on mineral homeostasis is a controversial issue. Here, we aimed to shed more light on this controversy by comparing male and female triple knockout mice with simultaneous deficiency in Fgf23 and Klotho and a nonfunctioning vitamin D receptor (VDR) (Fgf23/Klotho/VDR) with double (Fgf23/VDR, Klotho/VDR, and Fgf23/Klotho) and single Fgf23, Klotho, and VDR mutants. As expected, 4-week-old Fgf23, Klotho, and Fgf23/Klotho knockout mice were hypercalcemic and hyperphosphatemic, whereas VDR, Fgf23/VDR, and Klotho/VDR mice on rescue diet were normocalcemic and normophosphatemic. Serum levels of calcium, phosphate, and sodium did not differ between 4-week-old triple Fgf23/Klotho/VDR and double Fgf23/VDR or Klotho/VDR knockout mice. Notably, 3-month-old Fgf23/Klotho/VDR triple knockout mice were indistinguishable from double Fgf23/VDR and Klotho/VDR compound mutants in terms of serum calcium, serum phosphate, serum sodium, and serum PTH, as well as urinary calcium and sodium excretion. Protein expression analysis revealed increased membrane abundance of sodium-phosphate co-transporter 2a (NaPi-2a), and decreased expression of sodium-chloride co-transporter (NCC) and transient receptor potential cation channel subfamily V member 5 (TRPV5) in Fgf23/Klotho/VDR, Fgf23/VDR, and Klotho/VDR mice, relative to wild-type and VDR mice, but no differences between triple and double knockouts. Further, ex vivo treatment of live kidney slices isolated from wild-type and Klotho/VDR mice with soluble Klotho did not induce changes in intracellular phosphate, calcium or sodium accumulation assessed by two-photon microscopy. In conclusion, our data suggest that the main physiological function of Klotho for mineral homeostasis in vivo is its role as co-receptor mediating Fgf23 action. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucuronidase/metabolism , Homeostasis , Minerals/metabolism , Animals , Biological Transport , Bone and Bones/pathology , Calcium/metabolism , Fibroblast Growth Factor-23 , Kidney/metabolism , Klotho Proteins , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Phenotype , Phosphates/metabolism , Receptors, Calcitriol/metabolism , Recombinant Proteins/pharmacology , Sodium/metabolism , Solubility
10.
Sci Rep ; 7(1): 6460, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28744019

ABSTRACT

Estrogen is critical for skeletal homeostasis and regulates bone remodeling, in part, by modulating the expression of receptor activator of NF-κB ligand (RANKL), an essential cytokine for bone resorption by osteoclasts. RANKL can be produced by a variety of hematopoietic (e.g. T and B-cell) and mesenchymal (osteoblast lineage, chondrocyte) cell types. The cellular mechanisms by which estrogen acts on bone are still a matter of controversy. By using murine reconstitution models that allow for selective deletion of estrogen receptor-alpha (ERα) or selective inhibition of RANKL in hematopoietic vs. mesenchymal cells, in conjunction with in situ expression profiling in bone cells, we identified bone lining cells as important gatekeepers of estrogen-controlled bone resorption. Our data indicate that the increase in bone resorption observed in states of estrogen deficiency in mice is mainly caused by lack of ERα-mediated suppression of RANKL expression in bone lining cells.


Subject(s)
Bone Remodeling/physiology , Bone and Bones/cytology , Estrogens/metabolism , RANK Ligand/genetics , Alkaline Phosphatase/genetics , Animals , Bone Density , Bone Marrow Transplantation/methods , Bone Remodeling/genetics , Bone and Bones/physiology , Estrogen Receptor alpha/genetics , Estrogens/genetics , Female , GPI-Linked Proteins/genetics , Gene Expression Regulation , Humans , Isoenzymes/genetics , Mesenchymal Stem Cells/radiation effects , Mice, Knockout , Mice, Transgenic , RANK Ligand/metabolism , Rats, Inbred F344
11.
Endocrinology ; 157(10): 3924-3934, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27526032

ABSTRACT

The musculoskeletal effects of exercise are attenuated by estrogen deficiency. The peroxisome proliferator-activated receptor-α agonist fenofibrate exerts beneficial effects in bone and muscle. We therefore examined whether fenofibrate could enhance the musculoskeletal training response during estrogen deficiency. We investigated the combined effects of 8 weeks of fenofibrate and jumping exercise in ovariectomized (OVX) Sprague Dawley rats. Female rats were allocated to a sham-operated group and four OVX groups; fenofibrate (OVX-Fen), exercise (OVX-Ex), combined fenofibrate and exercise (OVX-FenEx), and a control group (OVX-Ctr) (n = 12/group). Fenofibrate (90 mg/kg/d) or methylcellulose was given by gavage. The combination of exercise and fenofibrate resulted in enhanced femoral bone mineral density (BMD) and improved bone microarchitecture compared with fenofibrate alone as well as increased trabecular BMD compared with OVX-Ctr. These effects were not seen in the OVX-Ex group. Femoral BMD was normalized in both exercise groups relative to sham and increased more in all intervention groups compared with OVX-Ctr. A higher plasma level of the bone formation marker type 1 collagen amino propeptide was observed in the OVX-Fen and OVX-FenEx groups compared with controls. Lean mass and soleus muscle weight were higher in the OVX-FenEx group than in the OVX-Ctr group, which coincided with lower mRNA levels of Atrogin1. These results suggest that peroxisome proliferator-activated receptor-α activation improves the musculoskeletal effects of exercise during estrogen deficiency.


Subject(s)
Bone and Bones/drug effects , Estrogens/deficiency , Fenofibrate/therapeutic use , PPAR alpha/agonists , Physical Conditioning, Animal , Animals , Body Composition/drug effects , Bone Density/drug effects , Bone and Bones/metabolism , Drug Evaluation, Preclinical , Female , Fenofibrate/pharmacology , Humans , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Osteogenesis/drug effects , Ovariectomy , Random Allocation , Rats, Sprague-Dawley , Tibia/diagnostic imaging , Tibia/drug effects , X-Ray Microtomography
12.
J Control Release ; 239: 137-48, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27586186

ABSTRACT

Transcript therapies using chemically modified messenger RNAs (cmRNAs) are emerging as safe and promising alternatives for gene and recombinant protein therapies. However, their applications have been limited due to transient translation and relatively low stability of cmRNAs compared to DNA. Here we show that vacuum-dried cmRNA-loaded collagen sponges, termed transcript activated matrices (TAMs), can serve as depots for sustained delivery of cmRNA. TAMs provide steady state protein production for up to six days, and substantial residual expression until 11days post transfection. Another advantage of this technology was nearly 100% transfection efficiency as well as low toxicity in vitro. TAMs were stable for at least 6months at room temperature. Human BMP-2-encoding TAMs induced osteogenic differentiation of MC3T3-E1 cells in vitro and bone regeneration in a non-critical rat femoral bone defect model in vivo. In summary, TAMs are a promising tool for bone regeneration and potentially also for other applications in regenerative medicine and tissue engineering.


Subject(s)
Bone Regeneration/genetics , Collagen/administration & dosage , Gene Transfer Techniques , Genetic Therapy/methods , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , A549 Cells , Animals , Bone Regeneration/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Collagen/chemistry , Collagen/metabolism , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/metabolism , Femur/diagnostic imaging , Femur/drug effects , Femur/metabolism , Hep G2 Cells , Humans , Male , Mice , NIH 3T3 Cells , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
13.
J Endocrinol ; 230(2): 251-62, 2016 08.
Article in English | MEDLINE | ID: mdl-27325243

ABSTRACT

Epidemiological studies suggest an increased fracture risk in patients taking proton pump inhibitors (PPIs) for long term. The underlying mechanism, however, has been disputed. By binding to the gastric proton pump, PPIs inhibit gastric acid secretion. We have previously shown that proton pump (H(+)/K(+)ATPase beta subunit) KO mice exhibit reduced bone mineral density (BMD) and inferior bone strength compared with WT mice. Patients using PPIs as well as these KO mice exhibit gastric hypoacidity, and subsequently increased serum concentrations of the hormone gastrin. In this study, we wanted to examine whether inhibition of the gastrin/CCK2 receptor influences bone quality in these mice. KO and WT mice were given either the gastrin/CCK2 receptor antagonist netazepide dissolved in polyethylene glycol (PEG) or only PEG for 1year. We found significantly lower bone mineral content and BMD, as well as inferior bone microarchitecture in KO mice compared with WT. Biomechanical properties by three-point bending test also proved inferior in KO mice. KO mice receiving netazepide exhibited significantly higher cortical thickness, cortical area fraction, trabecular thickness and trabecular BMD by micro-CT compared with the control group. Three-point bending test also showed higher Young's modulus of elasticity in the netazepide KO group compared with control mice. In conclusion, we observed that the gastrin receptor antagonist netazepide slightly improved bone quality in this mouse model, suggesting that hypergastrinemia may contribute to deteriorated bone quality during acid inhibition.


Subject(s)
Benzodiazepinones/therapeutic use , Bone and Bones/drug effects , H(+)-K(+)-Exchanging ATPase/deficiency , Osteoporosis/prevention & control , Phenylurea Compounds/therapeutic use , Receptor, Cholecystokinin B/antagonists & inhibitors , Absorptiometry, Photon , Adaptor Proteins, Signal Transducing , Animals , Benzodiazepinones/pharmacology , Bone Density/drug effects , Bone and Bones/diagnostic imaging , Drug Evaluation, Preclinical , Female , Gastrins/blood , Glycoproteins/blood , H(+)-K(+)-Exchanging ATPase/genetics , Intercellular Signaling Peptides and Proteins , Leptin/blood , Mice, Inbred BALB C , Mice, Knockout , Osteocalcin/blood , Osteoporosis/chemically induced , Phenylurea Compounds/pharmacology , Proton Pump Inhibitors/adverse effects , RANK Ligand/blood , Stomach/drug effects , X-Ray Microtomography
14.
J Bone Miner Res ; 29(3): 693-704, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24038141

ABSTRACT

Excessive FGF23 has been identified as a pivotal phosphaturic factor leading to renal phosphate-wasting and the subsequent development of rickets and osteomalacia. In contrast, loss of FGF23 in mice (Fgf23(-/-) ) leads to high serum phosphate, calcium, and 1,25-vitamin D levels, resulting in early lethality attributable to severe ectopic soft-tissue calcifications and organ failure. Paradoxically, Fgf23(-/-) mice exhibit a severe defect in skeletal mineralization despite high levels of systemic mineral ions and abundant ectopic mineralization, an abnormality that remains largely unexplained. Through use of in situ hybridization, immunohistochemistry, and immunogold labeling coupled with electron microscopy of bone samples, we discovered that expression and accumulation of osteopontin (Opn/OPN) was markedly increased in Fgf23(-/-) mice. These results were confirmed by qPCR analyses of Fgf23(-/-) bones and ELISA measurements of serum OPN. To investigate whether elevated OPN levels were contributing to the bone mineralization defect in Fgf23(-/-) mice, we generated Fgf23(-/-) /Opn(-/-) double-knockout mice (DKO). Biochemical analyses showed that the hypercalcemia and hyperphosphatemia observed in Fgf23(-/-) mice remained unchanged in DKO mice; however, micro-computed tomography (µCT) and histomorphometric analyses showed a significant improvement in total mineralized bone volume. The severe osteoidosis was markedly reduced and a normal mineral apposition rate was present in DKO mice, indicating that increased OPN levels in Fgf23(-/-) mice are at least in part responsible for the osteomalacia. Moreover, the increased OPN levels were significantly decreased upon lowering serum phosphate by feeding a low-phosphate diet or after deletion of NaPi2a, indicating that phosphate levels contribute in part to the high OPN levels in Fgf23(-/-) mice. In summary, our results suggest that increased OPN is an important pathogenic factor mediating the mineralization defect and the alterations in bone metabolism observed in Fgf23(-/-) bones.


Subject(s)
Calcification, Physiologic , Fibroblast Growth Factors/physiology , Osteopontin/blood , Animals , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Hyperphosphatemia/blood , In Situ Hybridization , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction
15.
Bone ; 50(1): 140-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22019458

ABSTRACT

Previous data have shown gender-related differences in the skeletal effects of the immunosuppressive drug cyclosporin A (CsA) in rats. To test the hypothesis that the gender-related skeletal effects of CsA are caused by gender-specific metabolism of this drug, we treated aged male and female sham-operated, gonadectomized (GX) as well as sex hormone-supplemented GX rats with 5 mg/kg CsA three times per week for 2 months, and analyzed the bone phenotype as well as the concentrations of CsA and its major metabolites AM1, AM1c, AM9, and AM4N in blood, urine, and liver tissue. CsA treatment induced high turnover osteopenia in males, but not females. Male rats showed several-fold higher CsA and CsA metabolite blood levels compared with females. Renal clearance data revealed that CsA undergoes selective tubular reabsorption in male, but not female rats. However, a mathematical modeling approach demonstrated that the higher CsA blood levels in males were almost exclusively caused by a 6-fold lower hepatic clearance rate compared with females. In addition, we subcutaneously treated female rats with up to 6-fold higher doses of CsA. Similar to males, high dose CsA induced high turnover osteopenia in female rats. Our data show that the gender-related differences in the skeletal effects of CsA are caused by a higher hepatic clearance rate for CsA in female compared to male rats, and not by a differential skeletal response to CsA. Moreover, our study indicates that CsA blood levels of ≤200 ng/ml measured by HPLC do not induce high turnover osteopenia in aged rats.


Subject(s)
Bone and Bones/drug effects , Bone and Bones/metabolism , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Animals , Bone Density , Bone Diseases, Metabolic/chemically induced , Bone and Bones/anatomy & histology , Cyclosporine/metabolism , Dose-Response Relationship, Drug , Female , Immunosuppressive Agents/metabolism , Liver/drug effects , Liver/metabolism , Male , Models, Theoretical , Rats , Rats, Inbred F344 , Sex Factors
16.
Endocrinology ; 153(4): 1725-33, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22334713

ABSTRACT

Estrogen replacement is an effective therapy of postmenopausal symptoms such as hot flushes, bone loss, and vaginal dryness. Undesired estrogen effects are the stimulation of uterine and mammary gland epithelial cell proliferation as well as hepatic estrogenicity. In this study, we examined the influence of different estradiol release kinetics on tissue responsivity in ovariectomized (OVX) rats. Pulsed release kinetics was achieved by ip or sc administration of estradiol dissolved in physiological saline containing 10% ethanol (EtOH/NaCl) whereas continuous release kinetics was achieved by sc injection of estradiol dissolved in benzylbenzoate/ricinus oil (1+4, vol/vol). Initial 3-d experiments in OVX rats showed that pulsed ip estradiol administration had profoundly reduced stimulatory effects on the uterus and the liver compared with continuous release kinetics. On the other hand, both administration forms prevented severe vaginal atrophy. Based on these results, we compared the effects of pulsed (sc in EtOH/NaCl) vs. continuous (sc in benzylbenzoate/ricinus oil) estradiol release kinetics on bone, uterus, mammary gland, and liver in a 4-month study in OVX rats. Ovariectomy-induced bone loss was prevented by both administration regimes. However, pulsed estradiol resulted in lower uterine weight, reduced induction of hepatic gene expression, and reduced mammary epithelial hyperplasia relative to continuous estradiol exposure. We conclude that organ responsivity is influenced by different hormone release kinetics, a fact that might be exploited to reduce undesired estradiol effects in postmenopausal women.


Subject(s)
Bone and Bones/drug effects , Estradiol/pharmacology , Estradiol/pharmacokinetics , Liver/drug effects , Ovariectomy , Uterus/drug effects , Animals , Atrophy/chemically induced , Bone Density/drug effects , Bone and Bones/pathology , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Estrogen Replacement Therapy/adverse effects , Female , Hyperplasia/chemically induced , Injections , Liver/pathology , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Models, Animal , Rats , Rats, Wistar , Time Factors , Uterus/pathology , Vagina/drug effects , Vagina/pathology
17.
Bone ; 50(1): 237-44, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22056328

ABSTRACT

Although the bone anabolic properties of intermittent parathyroid hormone (PTH) have long been employed in the treatment of osteoporosis, the molecular mechanisms behind this action remain largely unknown. Previous studies showed that PTH increases the expression and the activity of epidermal growth factor receptor (EGFR) in osteoblasts, and activation of ERK1/2 by PTH in osteoblasts was demonstrated to induce the proteolytical release of EGFR ligands and EGFR transactivation. However, conclusive evidence for an important role of the EGFR system in mediating the anabolic actions of intermittent PTH on bone in vivo is lacking. Here, we evaluated the effects of intermittent PTH on bone in Waved-5 (Wa5) mice which carry an antimorphic Egfr allele whose product acts as a dominant negative receptor. Heterozygous Wa5 females and control littermates received a subcutaneous injection of PTH (80 µg/kg) or buffer on 5 days per week for 4 weeks. Wa5 mice had slightly lower total bone mineral density (BMD), but normal cancellous bone volume and turnover in the distal femoral metaphysis. The presence of the antimorphic Egfr allele neither influenced the PTH-induced increase in serum osteocalcin nor the increases in distal femoral BMD, cortical thickness, cancellous bone volume, and cancellous bone formation rate. Similarly, the PTH-induced rise in lumbar vertebral BMD was unchanged in Wa5 relative to wild-type mice. Wa5-derived osteoblasts showed considerably lower basal extracellular signal-regulated kinase 1/2 (ERK1/2) activation as compared to control osteoblasts. Whereas activation of ERK1/2 by the EGFR ligand amphiregulin was largely blocked in Wa5 osteoblasts, treatment with PTH induced ERK1/2 activation comparable to that observed in control osteoblasts, relative to baseline levels. Our data indicate that impairment of EGFR signaling does not affect the anabolic action of intermittent PTH on cancellous and cortical bone.


Subject(s)
Anabolic Agents/pharmacology , Bone and Bones/drug effects , Bone and Bones/metabolism , ErbB Receptors/metabolism , Parathyroid Hormone/pharmacology , Signal Transduction/physiology , Animals , Bone Density/drug effects , Bone and Bones/ultrastructure , ErbB Receptors/genetics , Female , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/blood , X-Ray Microtomography
18.
J Bone Miner Res ; 27(10): 2085-96, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22648978

ABSTRACT

Bone remodeling involves tightly regulated bone-resorbing osteoclasts and bone-forming osteoblasts. Determining osteoclast function is central to understanding bone diseases such as osteoporosis and osteopetrosis. Here, we report a novel function of the F-actin binding and regulatory protein SWAP-70 in osteoclast biology. F-actin ring formation, cell morphology, and bone resorption are impaired in Swap-70(-/-) osteoclasts, whereas the expression of osteoclast differentiation markers induced in vitro by macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) remains unaffected. Swap-70(-/-) mice develop osteopetrosis with increased bone mass, abnormally dense bone, and impaired osteoclast function. Ectopic expression of SWAP-70 in Swap-70(-/-) osteoclasts in vitro rescues their deficiencies in bone resorption and F-actin ring formation. Rescue requires a functional pleckstrin homology (PH) domain, known to support membrane localization of SWAP-70, and the F-actin binding domain. Transplantation of SWAP-70-proficient bone marrow into Swap-70(-/-) mice restores osteoclast resorption capacity in vivo. The identification of the role of SWAP-70 in promoting osteoclast function through modulating membrane-proximal F-actin rearrangements reveals a new pathway to control osteoclasts and bone homeostasis.


Subject(s)
Actins/metabolism , Bone and Bones/pathology , DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Nuclear Proteins/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Animals , Bone Resorption/complications , Bone Resorption/pathology , Cell Line , Cell Movement , DNA-Binding Proteins/deficiency , Guanine Nucleotide Exchange Factors/deficiency , Humans , Mice , Minor Histocompatibility Antigens , Nuclear Proteins/deficiency , Organ Size , Osteoblasts/pathology , Osteopetrosis/complications , Osteopetrosis/pathology
19.
J Bone Miner Res ; 26(9): 2026-35, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21590742

ABSTRACT

Parathyroid hormone (PTH) is widely recognized as a key regulator of mineral ion homeostasis. Daily intermittent administration of PTH is the only currently available anabolic therapy for bone disorders such as osteoporosis. Recent studies have shown that PTH increases transcription and secretion of fibroblast growth factor 23 (FGF-23), another important regulator of phosphate homeostasis and skeletal metabolism. However, the full relationship between PTH and FGF-23 is largely unknown. This study evaluated the effect of FGF-23/Klotho signaling on the phosphaturic and anabolic functions of PTH. Eight-day-old wild-type (WT) Fgf23(-/-) and Kl(-/-) mice were injected with 100 µg/kg PTH(1-34) or vehicle daily for a 2-week-period and then euthanized. Intermittent injection of PTH successfully reduced the serum phosphate levels and reversed the hyperphosphatemia of Fgf23(-/-) and Kl(-/-) mice. Bone changes were analyzed in the distal femur metaphysis by peripheral quantitative computed tomography (pQCT), micro-computed tomography (µCT), and histomorphometry. PTH treatment induced substantial increases in bone mineral density (BMD) and trabecular bone volume in each mouse genotype. Expression of osteoblastic marker genes, including Runx2, Col1, Alp, Ocn, and Sost, was similarly altered. In addition, primary osteoblasts were isolated and treated with 100 nM PTH in vitro. PTH treatment similarly induced cAMP accumulation and phosphorylation of ERK1/2 and CREB in the osteoblasts from each genotype. Taken together, our results demonstrate that FGF-23/Klotho signaling is not essential for the phosphaturic and anabolic functions of PTH, suggesting that PTH can function as a therapeutic agent to improve the skeletal quality of patients even in the presence of abnormal serum FGF-23 levels.


Subject(s)
Anabolic Agents/pharmacology , Fibroblast Growth Factors/metabolism , Glucuronidase/metabolism , Hypophosphatemia, Familial/metabolism , Parathyroid Hormone/pharmacology , Signal Transduction/drug effects , Anabolic Agents/administration & dosage , Animals , Bone Density/drug effects , Bone and Bones/drug effects , Bone and Bones/pathology , Calcitriol/blood , Calcium/blood , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/deficiency , Gene Expression Regulation/drug effects , Glucuronidase/deficiency , Humans , Hypophosphatemia, Familial/pathology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Klotho Proteins , Mice , Organ Size/drug effects , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoblasts/pathology , Parathyroid Hormone/administration & dosage , Phenotype , Phosphates/blood
20.
J Bone Miner Res ; 24(1): 22-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18767932

ABSTRACT

Cell proliferation and PTH secretion in the parathyroid gland are known to be regulated by vitamin D and extracellular calcium. Here, we examined the vitamin D-independent effects of correction of extracellular calcium in an adult-onset secondary hyperparathyroidism (sHPT) model, using mice with a nonfunctioning vitamin D receptor (VDR). Wildtype and homozygous VDR mutant mice were kept on a rescue diet (RD) containing 2% calcium (Ca), 1.25% phosphorus (P), and 20% lactose until they were 4 mo or 1 yr of age. Subsequently, 4-mo-old mice were switched to a challenge diet (CD) containing the following: 0.5% Ca, 0.4% P, and 0% lactose. After 2 mo on the CD, groups of VDR mutant mice were either fed CD, a normal mouse chow with 0.9% Ca, 0.7% P, and 0% lactose, or the RD for another 3 mo. Feeding the RD protected VDR mutants against sHPT over 1 yr, showing that vitamin D is not essential for long-term control of the function and proliferation of parathyroid cells. When 4-mo-old VDR mutants were switched from the RD to the CD for 2 mo, they developed severe sHPT associated with hypertrophy and hyperplasia of parathyroid glands and profound bone loss. Subsequent feeding of the RD during a 3-mo therapy phase fully corrected sHPT, reduced chief cell proliferation, and reduced maximum parathyroid gland area by 25% by cell atrophy. There was no evidence of RD-induced chief cell apoptosis. We conclude that signaling by the calcium-sensing receptor regulates chief cell function and size in the absence of signaling through the VDR.


Subject(s)
Calcium/metabolism , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/metabolism , Vitamin D/metabolism , Animals , Apoptosis , Calcium/therapeutic use , Cell Proliferation , Disease Models, Animal , Female , Hypertrophy , Male , Mice , Mice, Inbred C57BL , Models, Biological , Parathyroid Glands/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL