Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 41
1.
J Orthop Res ; 42(5): 1033-1044, 2024 May.
Article En | MEDLINE | ID: mdl-38044472

High fat diets overwhelm the physiological mechanisms for absorption, storage, and utilization of triglycerides (TG); consequently TG, TG-rich lipoproteins (TGRL), and TGRL remnants accumulate, circulate systemically, producing dyslipidemia. This associates with, or is causative for increased atherosclerotic cardiovascular risk, ischemic stroke, fatty liver disease, and pancreatitis. TGRL hydrolysis by endothelial surface-bound lipoprotein lipase (LPL) generates metabolites like free fatty acids which have proinflammatory properties. While osteoblasts utilize fatty acids as an energy source, dyslipidemia is associated with negative effects on the skeleton. In this study we investigated the effects of TGRL lipolysis products (TGRL-LP) on expression of a stress responsive transcription factor, termed activating transcription factor 3 (ATF3), reactive oxygen species (ROS), ATF3 target genes, and angiopoietin-like 4 (Angptl4) in osteoblasts. As ATF3 negatively associates with osteoblast differentiation, we also investigated the skeletal effects of global ATF3 deletion in mice. TGRL-LP increased expression of Atf3, proinflammatory proteins Ptgs2 and IL-6, and induced ROS in MC3T3-E1 osteoblastic cells. Angptl4 is an endogenous inhibitor of LPL which was transcriptionally induced by TGRL-LP, while recombinant Angptl4 prevented TG-driven Atf3 induction. Atf3 global knockout male mice demonstrated increased trabecular and cortical microarchitectural parameters. In summary, we find that TGRL-LP induce osteoblastic cell stress as evidenced by expression of ATF3, which may contribute to the negative impact of dyslipidemia in the skeleton. Further, concomitant induction of Angptl4 in osteoblasts might play a protective role by reducing local lipolysis.


Dyslipidemias , Lipolysis , Male , Animals , Mice , Lipolysis/physiology , Reactive Oxygen Species/metabolism , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Heat-Shock Proteins/metabolism , Triglycerides/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Osteoblasts/metabolism
2.
JBMR Plus ; 7(4): e10733, 2023 Apr.
Article En | MEDLINE | ID: mdl-37065626

Hypoxia-inducible factors (HIFs) are oxygen-dependent heterodimeric transcription factors that mediate molecular responses to reductions in cellular oxygen (hypoxia). HIF signaling involves stable HIF-ß subunits and labile, oxygen-sensitive HIF-α subunits. Under hypoxic conditions, the HIF-α subunit is stabilized, complexes with nucleus-confined HIF-ß subunit, and transcriptionally regulates hypoxia-adaptive genes. Transcriptional responses to hypoxia include altered energy metabolism, angiogenesis, erythropoiesis, and cell fate. Three isoforms of HIF-α-HIF-1α, HIF-2α, and HIF-3α-are found in diverse cell types. HIF-1α and HIF-2α serve as transcriptional activators, whereas HIF-3α restricts HIF-1α and HIF-2α. The structure and isoform-specific functions of HIF-1α in mediating molecular responses to hypoxia are well established across a wide range of cell and tissue types. The contributions of HIF-2α to hypoxic adaptation are often unconsidered if not outrightly attributed to HIF-1α. This review establishes what is currently known about the diverse roles of HIF-2α in mediating the hypoxic response in skeletal tissues, with specific focus on development and maintenance of skeletal fitness. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

3.
JBMR Plus ; 7(4): e10724, 2023 Apr.
Article En | MEDLINE | ID: mdl-37065633

Molecular oxygen levels vary during development and disease. Adaptations to decreased oxygen bioavailability (hypoxia) are mediated by hypoxia-inducible factor (HIF) transcription factors. HIFs are composed of an oxygen-dependent α subunit (HIF-α), of which there are two transcriptionally active isoforms (HIF-1α and HIF-2α), and a constitutively expressed ß subunit (HIFß). Under normoxic conditions, HIF-α is hydroxylated via prolyl hydroxylase domain (PHD) proteins and targeted for degradation via Von Hippel-Lindau (VHL). Under hypoxic conditions, hydroxylation via PHD is inhibited, allowing for HIF-α stabilization and induction of target transcriptional changes. Our previous studies showed that Vhl deletion in osteocytes (Dmp1-cre; Vhl f/f ) resulted in HIF-α stabilization and generation of a high bone mass (HBM) phenotype. The skeletal impact of HIF-1α accumulation has been well characterized; however, the unique skeletal impacts of HIF-2α remain understudied. Because osteocytes orchestrate skeletal development and homeostasis, we investigated the role of osteocytic HIF-α isoforms in driving HBM phenotypes via osteocyte-specific loss-of-function and gain-of-function HIF-1α and HIF-2α mutations in C57BL/6 female mice. Deletion of Hif1a or Hif2a in osteocytes showed no effect on skeletal microarchitecture. Constitutively stable, degradation-resistant HIF-2α (HIF-2α cDR), but not HIF-1α cDR, generated dramatic increases in bone mass, enhanced osteoclast activity, and expansion of metaphyseal marrow stromal tissue at the expense of hematopoietic tissue. Our studies reveal a novel influence of osteocytic HIF-2α in driving HBM phenotypes that can potentially be harnessed pharmacologically to improve bone mass and reduce fracture risk. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

4.
Vet Sci ; 9(10)2022 Sep 25.
Article En | MEDLINE | ID: mdl-36288137

Sex steroids regulate bone metabolism directly and indirectly through receptors on bone. Estrogen receptors (ER-∝, ER-ß), progesterone receptor (PR), and androgen receptor (AR), have been previously identified on human osteosarcoma (OSA) cells, and are considered to influence tumor growth, but their expression and role in canine OSA is unknown. The aim of this study was to characterize sex hormone receptor expression levels in naturally occurring OSA tissue and in three canine OSA cell lines. The expression of ER-α, ER-ß, PR, and AR was investigated using RT-PCR. PR expression levels were also quantified in OSA cells cultured under hypoxic conditions or in the presence of estradiol. The effects of progesterone on cell proliferation were quantified. Results demonstrated varying expression levels of these receptors in five OSA subtypes. OSA cell lines demonstrated high gene expression levels of PR and low gene expression levels of ER-α and ER-ß and no gene expression of AR. PR expression was increased in OSA cells cultured under hypoxic conditions in a HIF-∝ independent manner. Interestingly, one cell line expressed very high levels of PR, expression of which decreased in response to estradiol. In addition, progesterone decreased OSA cell proliferation in this particular cell line. Further investigation of the role of sex steroids, particularly PR and its ligands, in regulation of canine OSA is recommended.

5.
PLoS One ; 14(9): e0222462, 2019.
Article En | MEDLINE | ID: mdl-31513647

Migration of cells from both local and systemic sources is essential for the inflammatory and regenerative processes that occur during normal wound healing. CXCL12 is considered a critical regulator of CXCR4-positive cell migration during tissue regeneration. In this study, we investigated the expression of Cxcl12 and Cxcr4 during healing of a murine full thickness ear wound. We also investigated the expression of angiopoietin-like 4, which has been shown to participate in wound angiogenesis and reepithelialization. At time points up to 48hrs, complete blood counts were performed using automated hematology analysis, and the numbers of circulating stem and progenitor cells quantified using flow cytometry. Expression of both Cxcr4 and Angptl4 was significantly elevated within 3 days of wounding, and both were strongly expressed in cells of the epidermis. ANGPTL4 protein expression remained elevated in the epithelium through day 14. Cxcl12 expression was increased significantly at day 3, and remained elevated through day 21. Faint Cxcl12 staining was detectable in the epithelium at day 1, and thereafter staining was faint and more generalized. There were significantly fewer circulating total white blood cells and lymphocytes 1hr following ear punching. Similarly, there was a significant early (1hr) reduction in the number of circulating endothelial progenitor cells. Further studies are warranted to investigate whether ANGPTL4 and CXCL12/CXCR4 interact or synergize to facilitate cell recruitment and migration, and to potentiate reepithelialization and wound healing.


Chemokine CXCL12/genetics , Receptors, CXCR4/genetics , Wound Healing/physiology , Angiopoietin-Like Protein 4/genetics , Angiopoietin-Like Protein 4/metabolism , Animals , Cell Movement , Chemokine CXCL12/metabolism , Ear/pathology , Endothelial Progenitor Cells/metabolism , Epidermal Cells/metabolism , Epidermis/metabolism , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Stem Cells/metabolism , Transcriptome/genetics
6.
Curr Osteoporos Rep ; 17(1): 26-35, 2019 02.
Article En | MEDLINE | ID: mdl-30725321

PURPOSE OF REVIEW: We reviewed recent literature on oxygen sensing in osteogenic cells and its contribution to development of a skeletal phenotype, the coupling of osteogenesis with angiogenesis and integration of hypoxia into canonical Wnt signaling, and opportunities to manipulate oxygen sensing to promote skeletal repair. RECENT FINDINGS: Oxygen sensing in osteocytes can confer a high bone mass phenotype in murine models; common and unique targets of HIF-1α and HIF-2α and lineage-specific deletion of oxygen sensing machinery suggest differentia utilization and requirement of HIF-α proteins in the differentiation from mesenchymal stem cell to osteoblast to osteocyte; oxygen-dependent but HIF-α-independent signaling may contribute to observed skeletal phenotypes. Manipulating oxygen sensing machinery in osteogenic cells influences skeletal phenotype through angiogenesis-dependent and angiogenesis-independent pathways and involves HIF-1α, HIF-2α, or both proteins. Clinically, an FDA-approved iron chelator promotes angiogenesis and osteogenesis, thereby enhancing the rate of fracture repair.


Bone and Bones/metabolism , Hypoxia/metabolism , Osteocytes/metabolism , Osteogenesis/physiology , Oxygen/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Density/physiology , Bone Regeneration/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Neovascularization, Physiologic/physiology , Wnt Signaling Pathway/physiology
7.
J Bone Miner Res ; 34(1): 157-170, 2019 01.
Article En | MEDLINE | ID: mdl-30189111

The most reliable predictor of future fracture risk is a previous fracture of any kind. The etiology of this increased fracture risk is not fully known, but it is possible that fracture initiates systemic bone loss, leading to greater fracture risk at all skeletal sites. In this study, we investigated systemic bone loss and recovery after femoral fracture in young (3-month-old) and middle-aged (12-month-old) mice. Transverse femur fractures were created using a controlled impact, and whole-body bone mineral density (BMD), trabecular and cortical microstructure, bone mechanical properties, bone formation and resorption rates, mouse voluntary movement, and systemic inflammation were quantified at multiple time points post-fracture. We found that fracture led to decreased whole-body BMD in both young and middle-aged mice 2 weeks post-fracture; this bone loss was recovered by 6 weeks in young but not middle-aged mice. Similarly, trabecular bone volume fraction (BV/TV) of the L5 vertebral body was significantly reduced in fractured mice relative to control mice 2 weeks post-fracture (-11% for young mice, -18% for middle-aged mice); no significant differences were observed 6 weeks post-fracture. At 3 days post-fracture, we observed significant increases in serum levels of interleukin-6 and significant decreases in voluntary movement in fractured mice compared with control mice, with considerably greater changes in middle-aged mice than in young mice. At this time point, we also observed increased osteoclast number on L5 vertebral body trabecular bone of fractured mice compared with control mice. These data show that systemic bone loss occurs after fracture in both young and middle-aged mice, and recovery from this bone loss may vary with age. This systemic response could contribute to increased future fracture risk after fracture; these data may inform clinical treatment of fractures with respect to improving long-term skeletal health. © 2018 American Society for Bone and Mineral Research.


Aging , Bone Density , Bone Diseases, Metabolic , Femoral Fractures , Osteogenesis , Aging/metabolism , Aging/pathology , Animals , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone Diseases, Metabolic/physiopathology , Female , Femoral Fractures/metabolism , Femoral Fractures/pathology , Femoral Fractures/physiopathology , Mice
8.
Bone ; 116: 307-314, 2018 11.
Article En | MEDLINE | ID: mdl-30172741

Tissue oxygen (O2) levels vary during development and disease; adaptations to decreased O2 (hypoxia) are mediated by hypoxia-inducible factor (HIF) transcription factors. HIFs are active in the skeleton, and stabilizing HIF-α isoforms cause high bone mass (HBM) phenotypes. A fundamental limitation of previous studies examining the obligate role for HIF-α isoforms in the skeleton involves the persistence of gene deletion as osteolineage cells differentiate into osteocytes. Because osteocytes orchestrate skeletal development and homeostasis, we evaluated the influence of Vhl or Hif1a disruption in osteocytes. Osteocytic Vhl deletion caused HBM phenotype, but Hif1a was dispensable in osteocytes. Vhl cKO mice revealed enhanced canonical Wnt signaling. B cell development was reduced while myelopoiesis increased in osteocytic Vhl cKO, revealing a novel influence of Vhl/HIF-α function in osteocytes on maintenance of bone microarchitecture via canonical Wnt signaling and effects on hematopoiesis.


Bone and Bones/metabolism , Hematopoiesis , Osteocytes/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Cancellous Bone/pathology , Cortical Bone/pathology , Gene Deletion , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphopoiesis , Mice, Inbred C57BL , Organ Size , Wnt Signaling Pathway
9.
Bone ; 81: 97-103, 2015 Dec.
Article En | MEDLINE | ID: mdl-26151122

Osteoblasts perceive and respond to changes in their pericellular environment, including biophysical signals and oxygen availability, to elicit an anabolic or catabolic response. Parathyroid hormone (PTH) affects each arm of skeletal remodeling, with net anabolic or catabolic effects dependent upon duration of exposure. Similarly, the capacity of osteoblastic cells to perceive pericellular oxygen has a profound effect on skeletal mass and architecture, as mice expressing stable hypoxia-inducible factor (HIF)-1α and -2α demonstrate age-dependent increases in bone volume per tissue volume and osteoblast number. Further, HIF levels and signaling can be influenced in an oxygen-independent manner. Because the cellular mechanisms involved in PTH regulation of the skeleton remain vague, we sought whether PTH could influence HIF-1α expression and HIF-α-driven luciferase activity independently of altered oxygen availability. Using UMR106.01 mature osteoblasts, we observed that 100nM hPTH(1-34) decreased HIF-1α and HIF-responsive luciferase activity in a process involving heat shock protein 90 (Hsp90) and cyclic AMP but not intracellular calcium. Altering activity of the small GTPase RhoA and its effector kinase ROCK altered HIF-α-driven luciferase activity in the absence and presence of PTH. Taken together, these data introduce PTH as a regulator of oxygen-independent HIF-1α levels through a mechanism involving cyclic AMP, Hsp90, and the cytoskeleton.


Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Osteoblasts/metabolism , Osteogenesis/physiology , Parathyroid Hormone/metabolism , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Blotting, Western , Cell Line , Humans , Mutagenesis, Site-Directed , Rats , Transfection
10.
J Orthop Res ; 33(9): 1364-73, 2015 Sep.
Article En | MEDLINE | ID: mdl-25864912

Vascular disruption that occurs as a consequence of bone fracture, leads to hypoxia at the site of damage. Hypoxia regulates the expression of a number of genes that can modulate energy conservation, cell survival, tissue regeneration and angiogenesis. In this study we investigated the expression of Angiopoietin-like 4, an adipocytokine that has additional roles in angiogenesis, at the fracture site. We demonstrate that Angptl4 mRNA expression increased early during fracture healing (day 3) returning close to baseline at day14. In the callus, Angptl4 mRNA was visualized in areas of condensing mesenchymal cells, callus cartilage and was especially high in mineralizing osteoblasts located in areas of new bone formation. In vitro, Angptl4 mRNA expression in osteoblasts increased under hypoxic conditions and in cells treated with the hypoxia mimetic desferrioxamine. Angptl4 levels were strongly induced at day 14 in differentiating MC3T3-E1 osteoblastic cells. Exogenous ANGPTL4 increased expression of Runx2, Spp1, vegfa, and Alp mRNA in differentiating osteoblasts. We suggest that the distribution of Angptl4 in the callus may be driven by hypoxia and that Angptl4 may play a role in osteoblastic differentiation, and possibly angiogenesis via regulation of VEGF. Further studies could reveal a dual role for Angptl4 in angiogenesis and osteogenesis.


Angiopoietins/metabolism , Fracture Healing , Fractures, Bone/pathology , Hypoxia/pathology , Neovascularization, Physiologic , Osteoblasts/pathology , 3T3 Cells , Angiopoietin-Like Protein 4 , Animals , Bone and Bones/pathology , Cell Differentiation , Gene Expression Profiling , Gene Expression Regulation , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism
11.
PLoS One ; 9(9): e107482, 2014.
Article En | MEDLINE | ID: mdl-25222280

Annexins are a class of calcium-binding proteins with diverse functions in the regulation of lipid rafts, inflammation, fibrinolysis, transcriptional programming and ion transport. Within bone, they are well-characterized as components of mineralizing matrix vesicles, although little else is known as to their function during osteogenesis. We employed shRNA to generate annexin A2 (AnxA2)- or annexin A5 (AnxA5)-knockdown pre-osteoblasts, and determined whether proliferation or osteogenic differentiation was altered in knockdown cells, compared to pSiren (Si) controls. We report that DNA content, a marker of proliferation, was significantly reduced in both AnxA2 and AnxA5 knockdown cells. Alkaline phosphatase expression and activity were also suppressed in AnxA2- or AnxA5-knockdown after 14 days of culture. The pattern of osteogenic gene expression was altered in knockdown cells, with Col1a1 expressed more rapidly in knock-down cells, compared to pSiren. In contrast, Runx2, Ibsp, and Bglap all revealed decreased expression after 14 days of culture. In both AnxA2- and AnxA5-knockdown, interleukin-induced STAT6 signaling was markedly attenuated compared to pSiren controls. These data suggest that AnxA2 and AnxA5 can influence bone formation via regulation of osteoprogenitor proliferation, differentiation, and responsiveness to cytokines in addition to their well-studied function in matrix vesicles.


Annexin A2/genetics , Annexin A5/genetics , Cell Differentiation/genetics , Osteogenesis/genetics , Animals , Cell Proliferation/genetics , Collagen Type I/biosynthesis , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/biosynthesis , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Integrin-Binding Sialoprotein/biosynthesis , Mice , Osteoblasts/metabolism , RNA, Small Interfering , STAT6 Transcription Factor/biosynthesis , STAT6 Transcription Factor/genetics , Signal Transduction/genetics
12.
BMC Vet Res ; 10: 173, 2014 Aug 05.
Article En | MEDLINE | ID: mdl-25088159

BACKGROUND: The intermediate filament protein nestin is used as a marker for neural stem cells, and its expression is inversely correlated with cellular differentiation. More recently, nestin expression has also been described in other cell types including multipotential mesenchymal stromal cells (MSCs). In this study, we examined the expression of nestin in equine, canine and human bone marrow-derived MSCs undergoing osteogenic differentiation, to determine whether nestin levels were attenuated as the cells acquired a more mature phenotype. In addition, the expression of nestin may be under the influence of cellular hypoxia, as nestin expression is known to increase in areas of ischemic tissue damage. Therefore, we also examined the effects of hypoxia on expression of nestin in human MSCs and examined a role for hypoxia inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF) in the response. Additionally, we quantified the temporal expression of nestin in the fracture callus during bone regeneration, a site that has been characterized as hypoxic. RESULTS: There were no significant changes in nestin expression in MSCs during osteogenic differentiation. There was a significant increase in expression of nestin mRNA and protein in human MSCs in response to hypoxia (1% O2) or the chemical hypoxia mimetic desferroxamine. This may be due to upregulation of VEGF under hypoxia, as treatment of cells with the VEGF receptor antagonist CPO-P11 attenuated hypoxia-induced nestin expression. A significant increase in nestin mRNA expression was observed in the fracture callus of mice three and seven days post fracture. CONCLUSIONS: Nestin was not a selective marker for MSCs, as its expression was maintained during osteogenic differentiation, in all species examined. Furthermore our data suggest that nestin expression can be induced by hypoxia, and that this increase in nestin is partially regulated by HIF-1α and VEGF. Interestingly, nestin levels were significantly upregulated at the fracture site. Further studies are required to understand the role of nestin in bone cell biology and ultimately bone regeneration.


Bone Marrow Cells/metabolism , Gene Expression Regulation/physiology , Mesenchymal Stem Cells/metabolism , Nestin/metabolism , Osteogenesis/physiology , Oxygen/pharmacology , Animals , Cells, Cultured , Fracture Healing/physiology , Fractures, Bone/metabolism , Humans , Nestin/genetics , Species Specificity
13.
J Biomed Mater Res A ; 102(12): 4480-90, 2014 Dec.
Article En | MEDLINE | ID: mdl-24616346

There is a need for improved biomaterials for use in treating non-healing bone defects. A number of natural and synthetic biomaterials have been used for the regeneration of bone tissue with mixed results. One approach is to modify native tissue via decellularization or other treatment for use as natural scaffolding for tissue repair. In this study, our goal was to improve on our previously published alternating solution immersion (ASI) method to fabricate a robust, biocompatible, and mechanically competent biomaterial from natural demineralized bone matrix (DBM). The improved method includes an antigen removal (AR) treatment step which improves mineralization and stiffness while removing unwanted proteins. The chemistry of the mineral in the remineralized bone matrix (RBM) was consistent with dicalcium phosphate dihydrate (brushite), a material used clinically in bone healing applications. Mass spectrometry identified proteins removed from the matrix with AR treatment to include α-2 HS-glycoprotein and osteopontin, noncollagenous proteins (NCPs) and known inhibitors of biomineralization. Additionally, the RBM supported the survival, proliferation, and differentiation of human mesenchymal stromal cells (MSCs) in vitro as well or better than other widely used biomaterials including DBM and PLG scaffolds. DNA content increased more than 10-fold on RBM compared to DBM and PLG; likewise, osteogenic gene expression was significantly increased after 1 and 2 weeks. We demonstrated that ASI remineralization has the capacity to fabricate mechanically stiff and biocompatible RBM, a suitable biomaterial for cell culture applications.


Cell Differentiation , Extracellular Matrix/chemistry , Mesenchymal Stem Cells/metabolism , Metacarpal Bones/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Antigens, Differentiation/biosynthesis , Cells, Cultured , Gene Expression Regulation , Horses , Humans , Mesenchymal Stem Cells/cytology , Osteogenesis
14.
Vet Surg ; 42(7): 814-8, 2013 Oct.
Article En | MEDLINE | ID: mdl-24033641

OBJECTIVE: To determine presence of bacterial DNA in canine stifles with cranial cruciate ligament rupture (CCLR) and medial patellar luxation (MPL) compared to normal canine stifles (control). STUDY DESIGN: Prospective clinical study. ANIMALS: Dogs (n = 44). METHODS: Dogs of varying age, breed, sex, and weight residing in California were assessed for stifle pathology (CCLR, MPL, or normal control). Synovial fluid of all stifles was assessed for the presence of bacterial DNA using broad-ranging 16S rRNA primers and PCR. RESULTS: Bacterial DNA was detected in normal control stifles and those with CCLR and MPL. There were no statistical differences in the copy numbers of bacterial DNA in the stifle synovial fluid among groups (P > .05); however, synovial fluid specimens from dogs with stifle pathology (CCLR and MPL combined) tended to have higher copy numbers of bacterial DNA than those from controls (P = .06). There was no significant difference in the number of bacterial DNA between the CCLR and MPL groups (P = .57). The copy numbers of bacterial DNA had a weak positive significant correlation with the duration of lameness in CCLR group (P < .05). CONCLUSIONS: Increased detection of bacterial DNA in the stifle synovial fluid may indicate joint pathology but not be directly linked to a specific joint disease.


Bacterial Infections/diagnosis , DNA, Bacterial/isolation & purification , Dog Diseases/diagnosis , Joint Diseases/veterinary , Polymerase Chain Reaction/veterinary , Stifle/pathology , Synovial Fluid/microbiology , Animals , Anterior Cruciate Ligament/pathology , Dog Diseases/microbiology , Dogs , Female , Joint Diseases/diagnosis , Joint Diseases/microbiology , Male , Patellar Dislocation/pathology , Patellar Dislocation/veterinary , Polymerase Chain Reaction/methods
15.
Cytotherapy ; 15(9): 1136-47, 2013 Sep.
Article En | MEDLINE | ID: mdl-23831362

BACKGROUND AIMS: Delivery of bone marrow-derived stem and progenitor cells to the site of injury is an effective strategy to enhance bone healing. An alternate approach is to mobilize endogenous, heterogeneous stem cells that will home to the site of injury. AMD3100 is an antagonist of the chemokine receptor 4 (CXCR4) that rapidly mobilizes stem cell populations into peripheral blood. Our hypothesis was that increasing circulating numbers of stem and progenitor cells using AMD3100 will improve bone fracture healing. METHODS: A transverse femoral fracture was induced in C57BL/6 mice, after which they were subcutaneously injected for 3 d with AMD3100 or saline control. Mesenchymal stromal cells, hematopoietic stem and progenitor cells and endothelial progenitor cells in the peripheral blood and bone marrow were evaluated by means of flow cytometry, automated hematology analysis and cell culture 24 h after injection and/or fracture. Healing was assessed up to 84 d after fracture by histomorphometry and micro-computed tomography. RESULTS: AMD3100 injection resulted in higher numbers of circulating mesenchymal stromal cells, hematopoietic stem cells and endothelial progenitor cells. Micro-computed tomography data demonstrated that the fracture callus was significantly larger compared with the saline controls at day 21 and significantly smaller (remodeled) at day 84. AMD3100-treated mice have a significantly higher bone mineral density than do saline-treated counterparts at day 84. CONCLUSIONS: Our data demonstrate that early cell mobilization had significant positive effects on healing throughout the regenerative process. Rapid mobilization of endogenous stem cells could provide an effective alternative strategy to cell transplantation for enhancing tissue regeneration.


Cell Culture Techniques/methods , Femoral Fractures/therapy , Fracture Healing/physiology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Stem Cells/physiology , Animals , Benzylamines , Bone Density/drug effects , Bone Density/physiology , Bone Marrow/drug effects , Bone Marrow/physiology , Cells, Cultured , Cyclams , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/physiology , Femoral Fractures/drug therapy , Femoral Fractures/physiopathology , Fracture Healing/drug effects , Hematopoietic Stem Cells/drug effects , Heterocyclic Compounds/therapeutic use , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Stem Cells/drug effects
16.
J Orthop Res ; 30(11): 1853-9, 2012 Nov.
Article En | MEDLINE | ID: mdl-22592891

Fracture healing involves rapid stem and progenitor cell migration, homing, and differentiation. SDF-1 (CXCL12) is considered a master regulator of CXCR4-positive stem and progenitor cell trafficking to sites of ischemic (hypoxic) injury and regulates their subsequent differentiation into mature reparative cells. In this study, we investigated the role of SDF-1/CXCR4 signaling in fracture healing where vascular disruption results in hypoxia and SDF-1 expression. Mice were injected with AMD3100, a CXCR4 antagonist, or vehicle twice daily until euthanasia with the intent to impair stem cell homing to the fracture site and/or their differentiation. Fracture healing was evaluated using micro-computed tomography, histology, quantitative PCR, and mechanical testing. AMD3100 administration resulted in a significantly reduced hyaline cartilage volume (day 14), callus volume (day 42) and mineralized bone volume (day 42) and reduced expression of genes associated with endochondral ossification including collagen Type 1 alpha 1, collagen Type 2 alpha 1, vascular endothelial growth factor, Annexin A5, nitric oxide synthase 2, and mechanistic target of rapamycin. Our data suggest that the SDF-1/CXCR4 signaling plays a central role in bone healing possibly by regulating the recruitment and/or differentiation of stem and progenitor cells.


Chemokine CXCL12/metabolism , Fracture Healing , Heterocyclic Compounds/pharmacology , Receptors, CXCR4/metabolism , Animals , Benzylamines , Biomechanical Phenomena , Cyclams , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Osteogenesis , Polymerase Chain Reaction , Receptors, CXCR4/antagonists & inhibitors , X-Ray Microtomography
17.
Res Vet Sci ; 92(1): 66-75, 2012 Feb.
Article En | MEDLINE | ID: mdl-21075407

The aim of this study was to compare the osteogenic and proliferative potential of canine mesenchymal stromal cells (cMSCs) derived from bone marrow (BM-cMSCs) and adipose tissue (AT-cMSCs). Proliferation potential was determined under varying oxygen tensions (1%, 5%, and 21% O(2)). Effects of reduced oxygen levels on the osteogenic differentiation of AT-cMSCs were also investigated. AT-cMSCs proliferated at a significantly faster rate than BM-cMSCs, although both cell types showed robust osteogenic differentiation. Culture in 5% and 1% O(2) impaired proliferation in cMSC from both sources and osteogenic differentiation in AT-cMSCs. Our data suggests that AT-cMSCs might be more suitable for use in a clinical situation, where large cell numbers are required for bone repair, due to their rapid proliferation combined with robust osteogenic potential. Our data also suggests that the inhibitory effects of hypoxia on both cell proliferation and differentiation should be considered when using MSCs in a potentially hypoxic environment such as a fracture site.


Bone Marrow Cells/cytology , Cell Differentiation , Cell Proliferation , Hypoxia , Mesenchymal Stem Cells/cytology , Osteogenesis , Subcutaneous Fat/cytology , Animals , Cells, Cultured , Dogs , In Vitro Techniques
18.
J Cell Biochem ; 112(11): 3354-63, 2011 Nov.
Article En | MEDLINE | ID: mdl-21751239

Tenascins regulate cell interaction with the surrounding pericellular matrix. Within bone, tenascins C and W influence osteoblast adhesion and differentiation, although little is known about the regulation of tenascin expression. In this study we examined the effect of osteogenic differentiation, bone morphogenetic protein (BMP) and Wnt growth factors, and mechanical loading on tenascin expression in osteogenic cells. Osteogenic differentiation increased tenascin C (TnC), and decreased tenascin W (TnW), expression. Both growth factors and mechanical loading increased both TnC and TnW expression, albeit via distinct signaling mechanisms. Both BMP-2 and Wnt5a induction of tenascin expression were mediated by MAP kinases. These data establish a role for BMP, Wnts, and mechanical loading in the regulation of tenascin expression in osteoblasts.


Bone and Bones/metabolism , Tenascin/metabolism , Animals , Blotting, Western , Bone Morphogenetic Proteins/physiology , Bone and Bones/cytology , Cell Differentiation , Cell Line , Mechanotransduction, Cellular , Mice , Mitogen-Activated Protein Kinases/metabolism , NFATC Transcription Factors/metabolism , Polymerase Chain Reaction , RNA, Messenger/genetics , Signal Transduction , Tenascin/genetics , Type C Phospholipases/metabolism , Wnt Proteins/physiology
19.
Cell Biol Int ; 35(10): 981-9, 2011 Oct.
Article En | MEDLINE | ID: mdl-21574962

A variety of pathologies such as skeletal fracture, neoplasia and inflammation compromise tissue perfusion and thereby decrease tissue oxygen tension. We and others have demonstrated that hypoxia is a potent stimulant for MSC (mesenchymal stem cell) recruitment and differentiation, yet to date little research has focused on the effects of oxygen tension on MSC migration. In the present study, we examined the effects of hypoxia and the potential role of the GTPase RhoA and HIF-1α (hypoxia-inducible factor 1α) on MSC migration. Our results demonstrate that hypoxia decreases MSC migration through an HIF-1α and RhoA-mediated pathway. The active GTP-bound form of RhoA was reduced in 1% oxygen, whereas activation of RhoA under hypoxic conditions rescued migration. Furthermore, stabilization of HIF-1α under normoxic conditions attenuated cell migration similar to that of hypoxia. These results suggest that hypoxia negatively affects MSC migration by regulating activation of GTPases. These results highlight the importance of oxygen in regulating the recruitment of progenitor cells to areas of ischaemic tissue damage.


Cell Hypoxia , Cell Movement , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mesenchymal Stem Cells/cytology , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/metabolism , Adolescent , Bone Marrow Cells/cytology , Humans , Male , Signal Transduction
20.
Biorheology ; 48(1): 65-74, 2011.
Article En | MEDLINE | ID: mdl-21515937

The ability of bone cells to detect and transduce mechanical signals is central to the mechanism whereby bone adapts to mechanical load and maintains healthy bone mass. Src, a non-receptor tyrosine kinase, is located in focal adhesions, highly specialized and localized sites of attachment, that are thought to be a primary site of mechanotransduction. While Src is activated by mechanical loads in other cell types, its role in osteoblast mechanotransduction is unclear. In this study we examined whether oscillatory fluid flow influenced Src phosphorylation, and Src's role in the flow-induced osteopontin response. Additionally, we investigated the effect of constitutively active Src on osteopontin expression. Oscillatory fluid flow induced a statistically significant increase in phosphorylation of Src at tyrosine residue 416 after a 15 min exposure. Transfection with constitutively-active Src resulted in an increase in Src-Y416 phosphorylation and an increase in osteopontin mRNA transcript under static conditions. However, inhibition of Src activity had no effect on oscillatory fluid flow-stimulated osteopontin expression or ERK1/2 phosphorylation. These data suggest that although Src activity regulates osteopontin expression under static conditions, and is induced under conditions of shear stress, it is not required for load-induced osteopontin expression.


Osteoblasts/metabolism , Osteopontin/genetics , Transcriptional Activation , src-Family Kinases/metabolism , Animals , Cell Line , Hydrodynamics , Mechanotransduction, Cellular/genetics , Mice , Osteoblasts/cytology , Osteopontin/biosynthesis , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tyrosine/metabolism , src-Family Kinases/chemistry
...