Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(5)2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38473716

ABSTRACT

Despite the well-known relevance of polyamines to many forms of life, little is known about how polyamines regulate osteogenesis and skeletal homeostasis. Here, we report a series of in vitro studies conducted with human-bone-marrow-derived pluripotent stromal cells (MSCs). First, we show that during osteogenic differentiation, mRNA levels of most polyamine-associated enzymes are relatively constant, except for the catabolic enzyme spermidine/spermine N1-acetyltransferase 1 (SAT1), which is strongly increased at both mRNA and protein levels. As a result, the intracellular spermidine to spermine ratio is significantly reduced during the early stages of osteoblastogenesis. Supplementation of cells with exogenous spermidine or spermine decreases matrix mineralization in a dose-dependent manner. Employing N-cyclohexyl-1,3-propanediamine (CDAP) to chemically inhibit spermine synthase (SMS), the enzyme catalyzing conversion of spermidine into spermine, also suppresses mineralization. Intriguingly, this reduced mineralization is rescued with DFMO, an inhibitor of the upstream polyamine enzyme ornithine decarboxylase (ODC1). Similarly, high concentrations of CDAP cause cytoplasmic vacuolization and alter mitochondrial function, which are also reversible with the addition of DFMO. Altogether, these studies suggest that excess polyamines, especially spermidine, negatively affect hydroxyapatite synthesis of primary MSCs, whereas inhibition of polyamine synthesis with DFMO rescues most, but not all of these defects. These findings are relevant for patients with Snyder-Robinson syndrome (SRS), as the presenting skeletal defects-associated with SMS deficiency-could potentially be ameliorated by treatment with DFMO.


Subject(s)
Mesenchymal Stem Cells , Spermidine , Humans , Spermidine/metabolism , Spermine/metabolism , Spermine Synthase/genetics , Ornithine Decarboxylase/metabolism , Osteogenesis , Polyamines/metabolism , Mesenchymal Stem Cells/metabolism , RNA, Messenger
2.
Endocr Relat Cancer ; 30(9)2023 09 01.
Article in English | MEDLINE | ID: mdl-37226936

ABSTRACT

Prostate cancer (PCa) is an increasingly prevalent health problem in the developed world. Effective treatment options exist for localized PCa, but metastatic PCa has fewer treatment options and shorter patient survival. PCa and bone health are strongly entwined, as PCa commonly metastasizes to the skeleton. Since androgen receptor signaling drives PCa growth, androgen-deprivation therapy whose sequelae reduce bone strength constitutes the foundation of advanced PCa treatment. The homeostatic process of bone remodeling - produced by concerted actions of bone-building osteoblasts, bone-resorbing osteoclasts, and regulatory osteocytes - may also be subverted by PCa to promote metastatic growth. Mechanisms driving skeletal development and homeostasis, such as regional hypoxia or matrix-embedded growth factors, may be subjugated by bone metastatic PCa. In this way, the biology that sustains bone is integrated into adaptive mechanisms for the growth and survival of PCa in bone. Skeletally metastatic PCa is difficult to investigate due to the entwined nature of bone biology and cancer biology. Herein, we survey PCa from origin, presentation, and clinical treatment to bone composition and structure and molecular mediators of PCa metastasis to bone. Our intent is to quickly yet effectively reduce barriers to team science across multiple disciplines that focuses on PCa and metastatic bone disease. We also introduce concepts of tissue engineering as a novel perspective to model, capture, and study complex cancer-microenvironment interactions.


Subject(s)
Bone Neoplasms , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/pathology , Bone Neoplasms/secondary , Androgen Antagonists/therapeutic use , Bone and Bones/metabolism , Treatment Outcome , Tumor Microenvironment
3.
JBMR Plus ; 7(4): e10733, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37065626

ABSTRACT

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.

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

ABSTRACT

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.

5.
JBMR Plus ; 5(5): e10462, 2021 May.
Article in English | MEDLINE | ID: mdl-33977198

ABSTRACT

Sclerostin antibody (romosozumab) was recently approved for clinical use in the United States to treat osteoporosis. We and others have explored Wnt-based combination therapy to disproportionately improve the anabolic effects of sclerostin inhibition, including cotreatment with sclerostin antibody (Scl-mAb) and Dkk1 antibody (Dkk1-mAb). To determine the optimal ratio of Scl-mAb and Dkk1-mAb for producing maximal anabolic action, the proportion of Scl-mAb and Dkk1-mAb were systematically varied while holding the total antibody dose constant. A 3:1 mixture of Scl-mAb to Dkk1-mAb produced two to three times as much cancellous bone mass as an equivalent dose of Scl-mAb alone. Further, a 75% reduction in the dose of the 3:1 mixture was equally efficacious to a full dose of Scl-mAb in the distal femur metaphysis. The Scl-mAb/Dkk1-mAb combination approach was highly efficacious in the cancellous bone mass, but the cortical compartment was much more subtly affected. The osteoanabolic effects of Wnt pathway targeting can be made more efficient if multiple antagonists are simultaneously targeted. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.

6.
Article in English | MEDLINE | ID: mdl-33071963

ABSTRACT

The skeleton is well-innervated, but only recently have the functions of this complex network in bone started to become known. Although our knowledge of skeletal sensory and sympathetic innervation is incomplete, including the specific locations and subtypes of nerves in bone, we are now able to reconcile early studies utilizing denervation models with recent work dissecting the molecular signaling between bone and nerve. In total, sensory innervation functions in bone much as it does elsewhere in the body-to sense and respond to stimuli, including mechanical loading. Similarly, sympathetic nerves regulate autonomic functions related to bone, including homeostatic remodeling and vascular tone. However, more study is required to translate our current knowledge of bone-nerve crosstalk to novel therapeutic strategies that can be effectively utilized to combat skeletal diseases, disorders of low bone mass, and age-related decreases in bone quality.


Subject(s)
Adaptation, Physiological , Aging/physiology , Bone Development , Bone and Bones/innervation , Bone and Bones/physiology , Animals , Autonomic Nervous System/physiology , Humans
7.
J Orthop Res ; 37(10): 2122-2129, 2019 10.
Article in English | MEDLINE | ID: mdl-31228216

ABSTRACT

The gut microbiome (GM) contributes to host development, metabolism, and disease. Perturbations in GM composition, elicited through chronic administration of oral antibiotics (Abx) or studied using germ-free environments, alter bone mass, and microarchitecture. However, studies primarily involved chronic Abx exposure to adult mice prior to evaluating the skeletal phenotype. Children are more prone to infection with bacterial pathogens than adults and are thus treated more frequently with broad-spectrum Abx; consequently, Abx treatment disproportionately occurs during periods of greatest skeletal plasticity to anabolic cues. Because early-life exposures may exert long-lasting effects on adult health, we hypothesized that acute Abx administration during a developmentally sensitive period would elicit lasting effects on the skeletal phenotype. To test this hypothesis, neonatal mice were treated with Abx (P7-P23; oral gavage) or vehicle (water); GM composition, gut physiology, and bone structural and material properties were assessed in adulthood (8 weeks). We found sexually dimorphic effects of neonatal Abx administration on GM composition, gut barrier permeability, and the skeleton, indicating a negative role for neonatal Abx on bone mass and quality. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2122-2129, 2019.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Bone and Bones/drug effects , Gastrointestinal Microbiome/drug effects , Sex Characteristics , Ampicillin/administration & dosage , Animals , Animals, Newborn , Colon/drug effects , Colon/microbiology , Female , Male , Mice , Mice, Inbred C57BL , Neomycin/administration & dosage , Permeability , Phenotype , Vancomycin/administration & dosage
8.
Curr Osteoporos Rep ; 17(1): 26-35, 2019 02.
Article in English | MEDLINE | ID: mdl-30725321

ABSTRACT

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.


Subject(s)
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
9.
Bone ; 116: 307-314, 2018 11.
Article in English | MEDLINE | ID: mdl-30172741

ABSTRACT

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.


Subject(s)
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
10.
Nat Rev Rheumatol ; 14(1): 42-51, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29255213

ABSTRACT

Connexons form the basis of hemichannels and gap junctions. They are composed of six tetraspan proteins called connexins. Connexons can function as individual hemichannels, releasing cytosolic factors (such as ATP) into the pericellular environment. Alternatively, two hemichannel connexons from neighbouring cells can come together to form gap junctions, membrane-spanning channels that facilitate cell-cell communication by enabling signalling molecules of approximately 1 kDa to pass from one cell to an adjacent cell. Connexins are expressed in joint tissues including bone, cartilage, skeletal muscle and the synovium. Indicative of their importance as gap junction components, connexins are also known as gap junction proteins, but individual connexin proteins are gaining recognition for their channel-independent roles, which include scaffolding and signalling functions. Considerable evidence indicates that connexons contribute to the function of bone and muscle, but less is known about the function of connexons in other joint tissues. However, the implication that connexins and gap junctional channels might be involved in joint disease, including age-related bone loss, osteoarthritis and rheumatoid arthritis, emphasizes the need for further research into these areas and highlights the therapeutic potential of connexins.


Subject(s)
Connexin 43/metabolism , Connexins/metabolism , Gap Junctions/metabolism , Joint Diseases/metabolism , Animals , Arthritis, Rheumatoid/metabolism , Bone and Bones/metabolism , Cartilage/metabolism , Cell Communication/physiology , Cell Differentiation/physiology , Connexins/physiology , Connexins/therapeutic use , Gap Junctions/physiology , Humans , Ion Channel Gating/physiology , Ion Channels/physiology , Mice , Mice, Knockout , Musculoskeletal System/metabolism , Musculoskeletal System/pathology , Osteoarthritis/metabolism , Osteoporosis/metabolism , Synovial Membrane/metabolism
11.
PLoS One ; 12(10): e0187354, 2017.
Article in English | MEDLINE | ID: mdl-29088267

ABSTRACT

Underlying mechanisms contributing to the imbalance in bone turnover during osteoporosis remain only partially explained. Reduced sensory nerve function may contribute to this imbalance, as sensory neuropeptides affect the activity of osteoblasts and osteoclasts in vivo, especially during bone adaptation. In this study, we investigated bone adaptation in mice following two weeks of tibial compression (peak magnitude 3 N or 7 N). To induce decreased sensory nerve function, mice were treated with capsaicin as neonates. We hypothesized that decreased sensory nerve function would diminish the adaptation of bone to mechanical loading, assessed with µCT and dynamic histomorphometry. We found that tibial compression induced significant changes in cortical microarchitecture that depended on compression magnitude and location along the length of the tibia; in contrast, there was no effect of loading on trabecular bone of the tibial metaphysis. Tibial compression significantly increased periosteal, and decreased endosteal, bone formation. Contrary to our initial hypothesis, capsaicin-treated mice generally displayed a similar, if not larger, adaptive response to mechanical loading, including greater increases in bone mineral content and mineral apposition rate. To integrate mechanical loading of bone with sensory nerve activation, we examined whether concentration of the neuropeptides calcitonin gene-related peptide (CGRP) and substance P (SP) in bone were affected following 1 or 5 days of 5 N tibial compression or hindlimb unloading. We found that 1 day of tibial compression significantly increased CGRP concentrations in bone, and hindlimb unloading also exhibited a trend toward increased CGRP in bone. These results may suggest a role of sensory nerves in the bone adaptation response to the mechanical environment, though this remains unclear.


Subject(s)
Adaptation, Physiological , Disease Models, Animal , Peripheral Nerves/physiopathology , Tibia/physiopathology , Animals , Enzyme-Linked Immunosorbent Assay , Female , Hindlimb Suspension , Mice , Mice, Inbred C57BL , X-Ray Microtomography
12.
J R Soc Interface ; 14(127)2017 02.
Article in English | MEDLINE | ID: mdl-28179546

ABSTRACT

Spheroids formed of mesenchymal stem cells (MSCs) exhibit increased cell survival and trophic factor secretion compared with dissociated MSCs, making them therapeutically advantageous for cell therapy. Presently, there is no consensus for the mechanism of action. Many hypothesize that spheroid formation potentiates cell function by generating a hypoxic core within spheroids of sufficiently large diameters. The purpose of this study was to experimentally determine whether a hypoxic core is generated in MSC spheroids by measuring oxygen tension in aggregates of increasing diameter and correlating oxygen tension values with cell function. MSC spheroids were formed with 15 000, 30 000 or 60 000 cells per spheroid, resulting in radii of 176 ± 8 µm, 251 ± 12 µm and 353 ± 18 µm, respectively. Oxygen tension values coupled with mathematical modelling revealed a gradient that varied less than 10% from the outer diameter within the largest spheroids. Despite the modest radial variance in oxygen tension, cellular metabolism from spheroids significantly decreased as the number of cells and resultant spheroid size increased. This may be due to adaptive reductions in matrix deposition and packing density with increases in spheroid diameter, enabling spheroids to avoid the formation of a hypoxic core. Overall, these data provide evidence that the enhanced function of MSC spheroids is not oxygen mediated.


Subject(s)
Mesenchymal Stem Cells/metabolism , Oxygen/metabolism , Spheroids, Cellular/metabolism , Humans , Mesenchymal Stem Cells/cytology , Spheroids, Cellular/cytology
13.
Bone ; 92: 180-188, 2016 11.
Article in English | MEDLINE | ID: mdl-27601226

ABSTRACT

Sclerostin (Sost) is a negative regulator of bone formation that acts upon the Wnt signaling pathway. Sost is mechanically regulated at both mRNA and protein level such that loading represses and unloading enhances Sost expression, in osteocytes and in circulation. The non-coding evolutionarily conserved enhancer ECR5 has been previously reported as a transcriptional regulatory element required for modulating Sost expression in osteocytes. Here we explored the mechanisms by which ECR5, or several other putative transcriptional enhancers regulate Sost expression, in response to mechanical stimulation. We found that in vivo ulna loading is equally osteoanabolic in wildtype and Sost-/- mice, although Sost is required for proper distribution of load-induced bone formation to regions of high strain. Using Luciferase reporters carrying the ECR5 non-coding enhancer and heterologous or homologous hSOST promoters, we found that ECR5 is mechanosensitive in vitro and that ECR5-driven Luciferase activity decreases in osteoblasts exposed to oscillatory fluid flow. Yet, ECR5-/- mice showed similar magnitude of load-induced bone formation and similar periosteal distribution of bone formation to high-strain regions compared to wildtype mice. Further, we found that in contrast to Sost-/- mice, which are resistant to disuse-induced bone loss, ECR5-/- mice lose bone upon unloading to a degree similar to wildtype control mice. ECR5 deletion did not abrogate positive effects of unloading on Sost, suggesting that additional transcriptional regulators and regulatory elements contribute to load-induced regulation of Sost.


Subject(s)
Adaptation, Physiological/physiology , Enhancer Elements, Genetic/physiology , Glycoproteins/deficiency , Osteocytes/physiology , Osteogenesis/physiology , Adaptor Proteins, Signal Transducing , Animals , Biomechanical Phenomena/physiology , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Female , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Mice, Transgenic , RNA, Untranslated/genetics
14.
Bone ; 81: 97-103, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26151122

ABSTRACT

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.


Subject(s)
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
15.
Arthritis Res Ther ; 17: 30, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25888819

ABSTRACT

INTRODUCTION: Previous studies in animal models of osteoarthritis suggest that alendronate (ALN) has antiresorptive and chondroprotective effects, and can reduce osteophyte formation. However, these studies used non-physiologic injury methods, and did not investigate early time points during which bone is rapidly remodeled prior to cartilage degeneration. The current study utilized a non-invasive model of knee injury in mice to investigate the effect of ALN treatment on subchondral bone changes, articular cartilage degeneration, and osteophyte formation following injury. METHODS: Non-invasive knee injury via tibial compression overload or sham injury was performed on a total of 90 mice. Mice were treated with twice weekly subcutaneous injections of low-dose ALN (40 µg/kg/dose), high-dose ALN (1,000 µg/kg/dose), or vehicle, starting immediately after injury until sacrifice at 7, 14 or 56 days. Trabecular bone of the femoral epiphysis, subchondral cortical bone, and osteophyte volume were quantified using micro-computed tomography (µCT). Whole-joint histology was performed at all time points to analyze articular cartilage and joint degeneration. Blood was collected at sacrifice, and serum was analyzed for biomarkers of bone formation and resorption. RESULTS: µCT analysis revealed significant loss of trabecular bone from the femoral epiphysis 7 and 14 days post-injury, which was effectively prevented by high-dose ALN treatment. High-dose ALN treatment was also able to reduce subchondral bone thickening 56 days post-injury, and was able to partially preserve articular cartilage 14 days post-injury. However, ALN treatment was not able to reduce osteophyte formation at 56 days post-injury, nor was it able to prevent articular cartilage and joint degeneration at this time point. Analysis of serum biomarkers revealed an increase in bone resorption at 7 and 14 days post-injury, with no change in bone formation at any time points. CONCLUSIONS: High-dose ALN treatment was able to prevent early trabecular bone loss and cartilage degeneration following non-invasive knee injury, but was not able to mitigate long-term joint degeneration. These data contribute to understanding the effect of bisphosphonates on the development of osteoarthritis, and may support the use of anti-resorptive drugs to prevent joint degeneration following injury, although further investigation is warranted.


Subject(s)
Alendronate/therapeutic use , Anterior Cruciate Ligament Injuries , Bone Density Conservation Agents/therapeutic use , Knee Injuries/drug therapy , Osteoarthritis, Knee/prevention & control , Animals , Bone Resorption/prevention & control , Cartilage Diseases/prevention & control , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Female , Injections, Subcutaneous , Mice , Mice, Inbred C57BL
16.
PLoS One ; 9(9): e107482, 2014.
Article in English | MEDLINE | ID: mdl-25222280

ABSTRACT

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.


Subject(s)
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
17.
Tissue Eng Part A ; 20(17-18): 2305-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24593020

ABSTRACT

The goal of this study was to assess the long-term biocompatibility of single-wall carbon nanotubes (SWNTs) for tissue engineering of articular cartilage. We hypothesized that SWNT nanocomposite scaffolds in cartilage tissue engineering can provide an improved molecular-sized substrate for stimulation of chondrocyte growth, as well as structural reinforcement of the scaffold's mechanical properties. The effect of SWNT surface functionalization (-COOH or -PEG) on chondrocyte viability and biochemical matrix deposition was examined in two-dimensional cultures, in three-dimensional (3D) pellet cultures, and in a 3D nanocomposite scaffold consisting of hydrogels+SWNTs. Outcome measures included cell viability, histological and SEM evaluation, GAG biochemical content, compressive and tensile biomechanical properties, and gene expression quantification, including extracellular matrix (ECM) markers aggrecan (Agc), collagen-1 (Col1a1), collagen-2 (Col2a1), collagen-10 (Col10a1), surface adhesion proteins fibronectin (Fn), CD44 antigen (CD44), and tumor marker (Tp53). Our findings indicate that chondrocytes tolerate functionalized SWNTs well, with minimal toxicity of cells in 3D culture systems (pellet and nanocomposite constructs). Both SWNT-PEG and SWNT-COOH groups increased the GAG content in nanocomposites relative to control. The compressive biomechanical properties of cell-laden SWNT-COOH nanocomposites were significantly elevated relative to control. Increases in the tensile modulus and ultimate stress were observed, indicative of a tensile reinforcement of the nanocomposite scaffolds. Surface coating of SWNTs with -COOH also resulted in increased Col2a1 and Fn gene expression throughout the culture in nanocomposite constructs, indicative of increased chondrocyte metabolic activity. In contrast, surface coating of SWNTs with a neutral -PEG moiety had no significant effect on Col2a1 or Fn gene expression, suggesting that the charged nature of the -COOH surface functionalization may promote ECM expression in this culture system. The results of this study indicate that SWNTs exhibit a unique potential for cartilage tissue engineering, where functionalization with bioactive molecules may provide an improved substrate for stimulation of cellular growth and repair.


Subject(s)
Cartilage/growth & development , Chondrocytes/physiology , Chondrogenesis/physiology , Nanocomposites/chemistry , Nanotubes, Carbon/chemistry , Tissue Engineering/instrumentation , Tissue Scaffolds , Biocompatible Materials/chemical synthesis , Cartilage/cytology , Cell Proliferation/physiology , Chondrocytes/cytology , Compressive Strength/physiology , Elastic Modulus , Equipment Design , Equipment Failure Analysis , Hardness/physiology , Humans , Materials Testing , Nanocomposites/ultrastructure , Nanotubes, Carbon/ultrastructure , Particle Size , Stress, Mechanical , Surface Properties , Tensile Strength/physiology
18.
Tissue Eng Part A ; 20(7-8): 1156-64, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24131310

ABSTRACT

The survival of transplanted cells and their resulting efficacy in cell-based therapies is markedly impaired due to serum deprivation and hypoxia (SD/H) resulting from poor vascularization within tissue defects. Lysophosphatidic acid (LPA) is a platelet-derived growth factor with pleiotropic effects on many cell types. Mesenchymal stromal cells (MSC) exhibit unique secretory and stimulatory characteristics depending on their differentiation state. In light of the potential of MSC in cell-based therapies, we examined the ability of LPA to abrogate SD/H-induced apoptosis in human MSC at increasing stages of osteogenic differentiation in vitro and assessed MSC survival in vivo. Undifferentiated MSC were rescued from SD/H-induced apoptosis by treatment with both 25 and 100 µM LPA. However, MSC conditioned with osteogenic supplements responded to 25 µM LPA, and cells conditioned with dexamethasone-containing osteogenic media required 100 µM LPA. This rescue was mediated through LPA1 in all cases. The addition of 25 µM LPA enhanced vascular endothelial growth factor (VEGF) secretion by MSC in all conditions, but VEGF availability was not responsible for protection against apoptosis. We also showed that codelivery of 25 µM LPA with MSC in alginate hydrogels significantly improved the persistence of undifferentiated MSC in vivo over 4 weeks as measured by bioluminescence imaging. Osteogenic differentiation alone was protective of SD/H-induced apoptosis in vitro, and the synergistic delivery of LPA did not enhance persistence of osteogenically induced MSC in vivo. These data demonstrate that the capacity of LPA to inhibit SD/H-induced apoptosis in MSC is dependent on both the differentiation state and dosage. This information will be valuable for optimizing osteogenic conditioning regimens for MSC before in vivo implementation.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Cytoprotection/drug effects , Lysophospholipids/pharmacology , Mesenchymal Stem Cells/cytology , Animals , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Serum-Free/chemistry , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Osteogenesis/drug effects , Receptors, Lysophosphatidic Acid/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
19.
Brief Funct Genomics ; 12(5): 391-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23788797

ABSTRACT

Microarray technologies provide high-throughput analysis of genes that are differentially expressed in humans and other species, and thereby provide a means to measure how biological systems are altered during development or disease states. Within, we review how high-throughput genomic technologies have increased our understanding about the molecular complexity of breast cancer, identified distinct molecular phenotypes and how they can be used to increase the accuracy of predicted clinical outcome.


Subject(s)
Biomedical Research , Breast Neoplasms/genetics , Genomics/methods , Breast Neoplasms/classification , Breast Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Neoplasm Grading , Signal Transduction/genetics
20.
Proc Natl Acad Sci U S A ; 109(35): 14092-7, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22886088

ABSTRACT

The Wnt antagonist Sost has emerged as a key regulator of bone homeostasis through the modulation of Lrp4/5/6 Wnt coreceptors. In humans, lack of Sclerostin causes sclerosteosis and van Buchem (VB) disease, two generalized skeletal hyperostosis disorders that result from hyperactive Wnt signaling. Unlike sclerosteosis, VB patients lack SOST coding mutations but carry a homozygous 52 kb noncoding deletion that is essential for the transcriptional activation of SOST in bone. We recently identified a putative bone enhancer, ECR5, in the VB deletion region, and showed that the transcriptional activity of ECR5 is controlled by Mef2C transcription factor in vitro. Here we report that mice lacking ECR5 or Mef2C through Col1-Cre osteoblast/osteocyte-specific ablation result in high bone mass (HBM) due to elevated bone formation rates. We conclude that the absence of the Sost-specific long-range regulatory element ECR5 causes VB disease in rodents, and that Mef2C is the main transcription factor responsible for ECR5-dependent Sost transcriptional activation in the adult skeleton.


Subject(s)
Bone Remodeling/genetics , Enhancer Elements, Genetic/genetics , Glycoproteins/genetics , Hyperostosis/genetics , Myogenic Regulatory Factors/genetics , Osteocytes/physiology , Syndactyly/genetics , Adaptor Proteins, Signal Transducing , Age Factors , Animals , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Female , Femur/cytology , Femur/physiology , Gene Deletion , Glycoproteins/metabolism , Hyperostosis/metabolism , Intercellular Signaling Peptides and Proteins , Lac Operon , MEF2 Transcription Factors , Male , Mandible/abnormalities , Mandible/metabolism , Mice , Mice, Transgenic , Myogenic Regulatory Factors/metabolism , Osteochondrodysplasias , Osteosclerosis/genetics , Osteosclerosis/metabolism , Signal Transduction/genetics , Skull/abnormalities , Skull/metabolism , Syndactyly/metabolism , Transcriptional Activation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...