Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502232

ABSTRACT

Cellular senescence and its senescence-associated secretory phenotype (SASP) are widely regarded as promising therapeutic targets for aging-related diseases, such as osteoporosis. However, the expression pattern of cellular senescence and multiple SASP secretion remains unclear, thus leaving a large gap in the knowledge for a desirable intervention targeting cellular senescence. Therefore, there is a critical need to understand the molecular mechanism of SASP secretion in the bone microenvironment that can ameliorate aging-related degenerative pathologies including osteoporosis. In this study, osteocyte-like cells (MLO-Y4) were induced to cellular senescence by 2 Gy γ-rays; then, senescence phenotype changes and adverse effects of SASP on bone marrow mesenchymal stem cell (BMSC) differentiation potential were investigated. The results revealed that 2 Gy irradiation could hinder cell viability, shorten cell dendrites, and induce cellular senescence, as evidenced by the higher expression of senescence markers p16 and p21 and the elevated formation of senescence-associated heterochromatin foci (SAHF), which was accompanied by the enhanced secretion of SASP markers such as IL-1α, IL-6, MMP-3, IGFBP-6, resistin, and adiponectin. When 0.8 µM JAK1 inhibitors were added to block SASP secretion, the higher expression of SASP was blunted, but the inhibition in osteogenic and adipogenic differentiation potential of BMSCs co-cultured with irradiated MLO-Y4 cell conditioned medium (CM- 2 Gy) was alleviated. These results suggest that senescent osteocytes can perturb BMSCs' differential potential via the paracrine signaling of SASP, which was also demonstrated by in vivo experiments. In conclusion, we identified the SASP factor partially responsible for the degenerative differentiation of BMSCs, which allowed us to hypothesize that senescent osteocytes and their SASPs may contribute to radiation-induced bone loss.


Subject(s)
Bone Resorption/pathology , Cell Differentiation , Cellular Senescence , Gamma Rays/adverse effects , Mesenchymal Stem Cells/pathology , Osteocytes/pathology , Paracrine Communication , Animals , Bone Resorption/etiology , Bone Resorption/metabolism , Male , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Inbred BALB C , Osteocytes/radiation effects , Osteogenesis
2.
Arch Biochem Biophys ; 685: 108333, 2020 05 30.
Article in English | MEDLINE | ID: mdl-32194044

ABSTRACT

This study summarizes the available evidence from systematic reviews on the in vitro effects of photobiomodulation on the proliferation and differentiation of human bone and stromal cells by appraising their methodological quality. Improvements for future studies are also highlighted, with particular emphasis on in vitro protocols and cell-related characteristics. Six reviews using explicit eligibility criteria and methods selected in order to minimize bias were included. There was no compelling evidence on the cellular mechanisms of action or treatment parameters of photobiomodulation; compliance with quality assessment was poor. A rigorous description of laser parameters (wavelength, power, beam spot size, power density, energy density, repetition rate, pulse duration or duty cycle, exposure duration, frequency of treatments, and total radiant energy), exposure conditions (methods to ensure a uniform irradiation and to avoid cross-irradiation, laser-cell culture surface distance, lid presence during irradiation) and cell-related characteristics (cell type or line, isolation and culture conditions, donor-related factors where applicable, tissue source, cell phenotype, cell density, number of cell passages in culture) should be included among eligibility criteria for study inclusion. These methodological improvements will maximize the contribution of in vitro studies on the effects of photobiomodulation on human bone and stromal cells to evidence-based translational research.


Subject(s)
Low-Level Light Therapy , Osteocytes/metabolism , Stromal Cells/metabolism , Animals , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Humans , Osteocytes/radiation effects , Stromal Cells/radiation effects , Systematic Reviews as Topic
3.
J Cell Physiol ; 234(7): 10588-10601, 2019 07.
Article in English | MEDLINE | ID: mdl-30422320

ABSTRACT

Growing evidence has shown that pulsed electromagnetic fields (PEMF) can modulate bone metabolism in vivo and regulate the activities of osteoblasts and osteoclasts in vitro. Osteocytes, accounting for 95% of bone cells, act as the major mechanosensors in bone for transducing external mechanical signals and producing cytokines to regulate osteoblastic and osteoclastic activities. Targeting osteocytic signaling pathways is becoming an emerging therapeutic strategy for bone diseases. We herein systematically investigated the changes of osteocyte behaviors, functions, and its regulation on osteoclastogenesis in response to PEMF. The osteocyte-like MLO-Y4 cells were exposed to 15 Hz PEMF stimulation with different intensities (0, 5, and 30 Gauss [G]) for 2 hr. We found that the cell apoptosis and cytoskeleton organization of osteocytes were regulated by PEMF with an intensity-dependent manner. Moreover, PEMF exposure with 5 G significantly inhibited apoptosis-related gene expression and also suppressed the gene and protein expression of the receptor activator of nuclear factor κB ligand/osteoprotegerin (RANKL/OPG) ratio in MLO-Y4 cells. The formation, maturation, and osteoclastic bone-resorption capability of in vitro osteoclasts were significantly suppressed after treated with the conditioned medium from PEMF-exposed (5 G) osteocytes. Our results also revealed that the inhibition of osteoclastic formation, maturation, and bone-resorption capability induced by the conditioned medium from 5 G PEMF-exposed osteocytes was significantly attenuated after abrogating primary cilia in osteocytes using the polaris siRNA transfection. Together, our findings highlight that PEMF with 5 G can inhibit cellular apoptosis, modulate cytoskeletal distribution, and decrease RANKL/OPG expression in osteocytes, and also inhibit osteocyte-mediated osteoclastogenesis, which requires the existence of primary cilia in osteocytes. This study enriches our basic knowledge for further understanding the biological behaviors of osteocytes and is also helpful for providing a more comprehensive mechanistic understanding of the effect of electromagnetic stimulation on bone and relevant skeletal diseases (e.g., bone fracture and osteoporosis).


Subject(s)
Bone Resorption/genetics , Osteogenesis/genetics , Osteoprotegerin/genetics , RANK Ligand/genetics , Animals , Apoptosis/genetics , Bone Resorption/pathology , Bone Resorption/therapy , Cells, Cultured , Cilia/genetics , Cilia/radiation effects , Cytoskeleton/genetics , Electromagnetic Fields , Gene Expression Regulation/radiation effects , Humans , Mice , Osteoclasts/radiation effects , Osteocytes/radiation effects , Osteogenesis/radiation effects , Signal Transduction/genetics
4.
Arch Orthop Trauma Surg ; 139(11): 1599-1605, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31289845

ABSTRACT

OBJECTIVES: The Gekkou-drill® is an industrial drill that is highly efficient due to reduced cutting resistance resulting from its characteristic drill point shape. In this experiment, we compared the degree of thermal damage to bone tissue caused by conventional medical drills and these same drills with Gekkou modifications. METHODS: Holes were created in the tibias of living pigs using two different 3.2-mm diameter drills and their modified versions. Regarding the drilling parameters, the thrust force was 10 N and the drilling speeds were 800 revolutions per minute (rpm) and 1500 rpm. We compared the original and modified drills in terms of the bone temperature around the drill bit and the total time necessary to create each hole, the latter calculated using imaging data captured during drilling. In histopathological examination, the percentages of empty lacunae in osteocytes of the cortical bone beneath the periosteum were evaluated at 400 × magnification with an optical microscope. RESULTS: Compared to the original drills, the modified drills required significantly less time to create each hole and caused a significantly lower temperature rise during bone drilling. With the modified drills, the percentages of empty lacunae around the drilling holes were about 1/2-1/3 of those with the original drills, and were significantly lower for both drilling speeds. CONCLUSIONS: Gekkou-modified medical drills shortened drilling times despite low thrust force, and histopathological assessment demonstrated a significant reduction in osteocyte damage.


Subject(s)
Hot Temperature/adverse effects , Orthopedic Procedures/instrumentation , Osteocytes/radiation effects , Tibia , Animals , Swine , Tibia/pathology , Tibia/radiation effects
5.
Ultrastruct Pathol ; 42(2): 124-132, 2018.
Article in English | MEDLINE | ID: mdl-29424622

ABSTRACT

OBJECTIVES: The aim of the present study is to investigate the morphological and cellular changes in dental extraction socket that has been irradiated after the tooth extraction and to describe morphological characteristics of the osteocytes and osteocyte-lacunar-canalicular network (LCN) by scanning electron microscopy (SEM). MATERIAL AND METHODS: Five beagle dogs aged 1-2 years were used in this study. One side of each mandible was irradiated in two sessions and the other side of mandible (non-irradiated) served as a control. The mandible bone blocks were processed by bulk staining en bloc in basic fuchsin and the specimens were embedded routinely in polymethyl methacrylate resin without preliminary decalcification. All blocks were subjected to micro-CT imaging, after that the specimens were prepared for light microscopy and SEM. RESULTS: Alterations in bone macrostructure are minimal in irradiated bone, but the changes in LCN are clear. In the area of the tooth extraction socket, the connections of osteocytes to the vessels and to neighboring osteocytes were not observed both in irradiated and nonirradiated bone. However, osteoclasts were located in the bone surface entering inside to the bone between osteons. In the lamellar bone of lateral sides, a decrease in canalicular connections between osteocytes and periosteum was found in irradiated bone as compared to the non-irradiated side. CONCLUSIONS: The novelty of the present study is that radiation disrupts osteocytes and their dendrites.


Subject(s)
Alveolar Process/radiation effects , Bone Remodeling/radiation effects , Mandible/radiation effects , Osteocytes/radiation effects , Tooth Extraction/adverse effects , Alveolar Process/pathology , Alveolar Process/ultrastructure , Animals , Disease Models, Animal , Dogs , Mandible/pathology , Mandible/ultrastructure , Microscopy, Electron, Scanning , Osteocytes/pathology , Osteocytes/ultrastructure
6.
Lasers Med Sci ; 32(7): 1545-1560, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28725994

ABSTRACT

The aim of this study was to evaluate the effects of photobiomodulation (PBM) on cancellous bone in rat models of ovariectomized induced osteoporosis (OVX-D) and glucocorticoid-induced osteoporosis (GIOP). The experiment comprised of nine groups. A group of healthy rats was used for baseline evaluations. The OVX-D rats were further divided into groups as follows: control rats with osteoporosis, OVX-D rats that received alendronate (1 mg/kg 60 days), OVX-D rats treated with pulsed wave laser (890 nm, 80 Hz, 900 s, 0.0061 W/cm2, 5.5 J/cm2, three times a week, 60 days), and OVX-D rats treated with alendronate + pulsed laser. Dexamethasone was administered to the remaining rats that were split into four groups: control, alendronate-treated rats, laser-treated rats, and GIOP rats treated with alendronate + laser. T12, L1, L2, and L3 vertebrae were subjected to laser. Results of the current study demonstrated that OVX-D and GIOP significantly decreased some stereological parameters, and type 1 collagen gene expression compared to the healthy group. There was a significant increase in osteoclast number in both OVX-D and glucocorticoid administration compared to the healthy group. However, the detrimental effect of the OVX-D procedure on bone was more serious than glucocorticoid administration. Results showed that laser alone had a detrimental effect on trabecular bone volume, and cortical bone volume in groups GIOP and OVX-D compared to those in the healthy group. Alendronate significantly improved total vertebral bone volume, trabecular bone volume, and cortical bone volume, in GIOP and OVX-D groups compared to the laser-treated groups. Furthermore, the alendronate + laser in OVX-D rats and GIOP rats produced significantly increased osteoblast number and type 1 collagen gene expression and caused a significant decrease in osteoclast number compared to the controls.


Subject(s)
Low-Level Light Therapy , Lumbar Vertebrae/pathology , Lumbar Vertebrae/radiation effects , Osteoporosis/radiotherapy , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Count , Collagen Type I/metabolism , Disease Models, Animal , Female , Insulin-Like Growth Factor I/metabolism , Male , Osteoblasts/pathology , Osteoblasts/radiation effects , Osteocytes/pathology , Osteocytes/radiation effects , Osteoporosis/pathology , Ovariectomy , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism
7.
J Biol Chem ; 290(1): 157-67, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25336648

ABSTRACT

Focal radiotherapy for cancer patients has detrimental effects on bones within the radiation field and the primary clinical signs of bone damage include the loss of functional osteoblasts. We reported previously that daily injection of parathyroid hormone (PTH, 1-34) alleviates radiation-induced osteopenia in a preclinical radiotherapy model by improving osteoblast survival. To elucidate the molecular mechanisms, we irradiated osteoblastic UMR 106-01 cells and calvarial organ culture and demonstrated an anti-apoptosis effect of PTH1-34 on these cultures. Inhibitor assay indicated that PTH exerts its radioprotective action mainly through protein kinase A/ß-catenin pathway. γ-H2AX foci staining and comet assay revealed that PTH efficiently promotes the repair of DNA double strand breaks (DSBs) in irradiated osteoblasts via activating the ß-catenin pathway. Interestingly, Wnt3a alone also blocked cell death and accelerated DNA repair in primary osteoprogenitors, osteoblastic and osteocytic cells after radiation through the canonical signaling. Further investigations revealed that both Wnt3a and PTH increase the amount of Ku70, a core protein for initiating the assembly of DSB repair machinery, in osteoblasts after radiation. Moreover, down-regulation of Ku70 by siRNA abrogated the prosurvival effect of PTH and Wnt3a on irradiated osteoblasts. In summary, our results identify a novel role of PTH and canonical Wnt signaling in regulating DSB repair machinery and apoptosis in osteoblasts and shed light on using PTH1-34 or Wnt agonist as possible therapy for radiation-induced osteoporosis.


Subject(s)
Apoptosis/drug effects , DNA Repair/drug effects , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Radiation-Protective Agents/pharmacology , Animals , Animals, Newborn , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Apoptosis/radiation effects , Cell Differentiation , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Ku Autoantigen , Osteoblasts/cytology , Osteoblasts/radiation effects , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/radiation effects , Rats , Recombinant Proteins/pharmacology , Signal Transduction , Skull/cytology , Skull/drug effects , Skull/radiation effects , Tissue Culture Techniques , Wnt3A Protein/metabolism , Wnt3A Protein/pharmacology , X-Rays , beta Catenin/genetics , beta Catenin/metabolism
8.
J Oral Maxillofac Surg ; 73(12): 2419-28, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26341682

ABSTRACT

PURPOSE: The devastation radiation therapy (XRT) causes to endogenous tissue in patients with head and neck cancer can be a prohibitive obstacle in reconstruction of the mandible, demanding a better understanding of XRT-induced damage and options for reconstruction. This study investigated the cellular damage caused by radiation in an isogenic murine model of mandibular distraction osteogenesis (DO). The authors posited that radiation would result in fewer osteocytes, with increased empty lacunae and immature osteoid. MATERIALS AND METHODS: Twenty Lewis rats were randomly assigned to a DO group (n = 10) or a XRT/DO group (n = 10). These groups underwent an osteotomy and mandibular DO across a 5.1-mm gap. XRT was administered to the XRT/DO group at a fractionated human equivalent dose of 35 Gy before surgery. Animals were sacrificed on postoperative day 40 and mandibles were harvested and sectioned for histologic analysis. RESULTS: Bone that underwent radiation showed a significantly decreased osteocyte count and complementary increase in empty lacunae compared with non-XRT bone (P = .019 and P = .000). In addition, XRT bone exhibited increased immature osteoid and decreased mature woven bone compared with nonradiated bone (P = .001 and P = .003, respectively). Furthermore, analysis of the ratio of immature osteoid to woven bone volume exhibited a significant increase in the XRT bone, further showing the devastating damage from XRT (P = .001). CONCLUSION: These results clearly show the cellular diminution that occurs as a result of radiation. This foundational study provides the groundwork on which to investigate cellular therapies in an immuno-privileged model of mandibular DO.


Subject(s)
Mandible/surgery , Osteogenesis, Distraction , Radiation Injuries/pathology , Animals , Cell Count , Disease Models, Animal , Male , Mandible/pathology , Mandible/radiation effects , Osteocytes/pathology , Osteocytes/radiation effects , Radiation Injuries/surgery , Rats , Rats, Inbred Lew
9.
J Oral Maxillofac Surg ; 72(3): 559-66, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24342580

ABSTRACT

PURPOSE: Radiation is known to decrease osteocyte count and function, leading to bone weakening. A treatment strategy to mitigate these consequences could have immense therapeutic ramifications. The authors previously reported significantly decreased osteocyte count and mineralization capacity in a rat model of fracture healing after radiotherapy. They hypothesized that amifostine (AMF) would preserve osteocyte number and function in this model. MATERIALS AND METHODS: Thirty-six rats were divided into 3 groups: fracture, radiated fracture, and radiated fracture with AMF. Radiated groups underwent human-equivalent radiotherapy to the mandible before fixator placement and mandibular osteotomy. The AMF group received a subcutaneous injection before each dose of radiotherapy. After 40 days, mandibles were harvested for histologic processing. Quantification of osteocyte count (Oc), empty lacunae (EL), and osteoid ratio (osteoid volume [OV] to tissue volume [TV]) was performed and the results were compared using analysis of variance (P < .05). RESULTS: Radiated fractures showed significantly decreased Oc, increased EL, and a decreased capacity to produce new osteoid at the fracture site as measured with OV/TV compared with nonradiated fractures. In mandibles treated with AMF, these metrics were not statistically different than the control, indicating a preservation of osteocyte number and function. CONCLUSIONS: These results support the hypothesis that AMF preserves osteocyte number and function, thereby preventing the pernicious effects of radiotherapy on the cellular environment of fracture healing. Based on these findings, the authors encourage future investigation of this promising therapy for use in the prevention of pathologic fractures and osteoradionecrosis.


Subject(s)
Amifostine/therapeutic use , Fracture Healing/drug effects , Fracture Healing/radiation effects , Osteocytes/drug effects , Osteocytes/radiation effects , Radiation-Protective Agents/therapeutic use , Radiotherapy/adverse effects , Animals , Bone Regeneration/drug effects , Bone Regeneration/radiation effects , Calcification, Physiologic/drug effects , Calcification, Physiologic/radiation effects , Cell Count , Male , Mandibular Osteotomy , Rats , Rats, Sprague-Dawley
10.
Lasers Med Sci ; 28(6): 1495-502, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23324954

ABSTRACT

This study was aimed to evaluate the effects of low-level laser therapy (LLLT) in the treatment of trabecular bone loss induced by skeletal unloading. Twelve mice have taken denervation operation. At 2 weeks after denervation, LLLT (wavelength, 660 nm; energy, 3 J) was applied to the right tibiae of 6 mice (LASER) for 5 days/week over 2 weeks by using a minimally invasive laser needle system (MILNS) which consists of a 100 µm optical fiber in a fine needle (diameter, 130 µm) [corrected]. Structural parameters and histograms of bone mineralization density distribution (BMDD) were obtained before LLLT and at 2 weeks after LLLT. In addition, osteocyte, osteoblast, and osteoclast populations were counted. Two weeks after LLLT, bone volume fraction, trabeculae number, and trabeculae thickness were significantly increased and trabecular separations, trabecular bone pattern factor, and structure model index were significantly decreased in LASER than SHAM (p < 0.05). BMDD in LASER was maintained while that in SHAM was shifted to lower mineralization. Osteocyte and osteoblast populations were significantly increased but osteoclast population was significantly decreased in LASER when compared with those in SHAM (p < 0.05). The results indicate that LLLT with the MILNS may enhance bone quality and bone homeostasis associated with enhancement of bone formation and suppression of bone resorption.


Subject(s)
Bone Resorption/radiotherapy , Low-Level Light Therapy/instrumentation , Animals , Bone Density/radiation effects , Bone Resorption/etiology , Bone Resorption/pathology , Female , Hindlimb Suspension/adverse effects , Mice , Mice, Inbred ICR , Needles , Optical Fibers , Osteoblasts/pathology , Osteoblasts/radiation effects , Osteoclasts/pathology , Osteoclasts/radiation effects , Osteocytes/pathology , Osteocytes/radiation effects
11.
Electromagn Biol Med ; 30(4): 261-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22047464

ABSTRACT

Studies of animals and humans subjected to spaceflight demonstrate that weightlessness negatively affects the mass and mechanical properties of bone tissue. Bone cells could sense and respond to the gravity unloading, and genes sensitive to gravity change were considered to play a critical role in the mechanotransduction of bone cells. To evaluate the fold-change of gene expression, appropriate reference genes should be identified because there is no housekeeping gene having stable expression in all experimental conditions. Consequently, expression stability of ten candidate housekeeping genes were examined in osteoblast-like MC3T3-E1, osteocyte-like MLO-Y4, and preosteoclast-like FLG29.1 cells under different apparent gravities (µg, 1 g, and 2 g) in the high-intensity gradient magnetic field produced by a superconducting magnet. The results showed that the relative expression of these ten candidate housekeeping genes was different in different bone cells; Moreover, the most suitable reference genes of the same cells in altered gravity conditions were also different from that in strong magnetic field. It demonstrated the importance of selecting suitable reference genes in experimental set-ups. Furthermore, it provides an alternative choice to the traditionally accepted housekeeping genes used so far about studies of gravitational biology and magneto biology.


Subject(s)
Gene Expression Profiling/methods , Magnetic Fields , Osteocytes/radiation effects , Real-Time Polymerase Chain Reaction/methods , 3T3 Cells , Algorithms , Animals , Mechanotransduction, Cellular/radiation effects , Mice , Osteocytes/cytology , Osteocytes/metabolism
12.
Mol Cancer Res ; 19(10): 1739-1750, 2021 10.
Article in English | MEDLINE | ID: mdl-34039648

ABSTRACT

Radiation-induced bystander effects have been implicated in contributing to the growth delay of disseminated tumor cells (DTC) caused by 223RaCl2, an alpha particle-emitting radiopharmaceutical. To understand how 223RaCl2 affects the growth, we have quantified biological changes caused by direct effects of radiation and bystander effects caused by the emitted radiations on DTC and osteocytes. Characterizing these effects contribute to understanding the efficacy of alpha particle-emitting radiopharmaceuticals and guide expansion of their use clinically. MDA-MB-231 or MCF-7 human breast cancer cells were inoculated intratibially into nude mice that were previously injected intravenously with 50 or 600 kBq/kg 223RaCl2. At 1-day and 3-days postinoculation, tibiae were harvested and examined for DNA damage (γ-H2AX foci) and apoptosis in osteocytes and cancer cells located within and beyond the range (70 µm) of alpha particles emitted from the bone surface. Irradiated and bystander MDA-MB-231 and MCF-7 cells harbored DNA damage. Bystander MDA-MB-231 cells expressed DNA damage at both treatment levels while bystander MCF-7 cells required the higher administered activity. Osteocytes also had DNA damage regardless of inoculated cancer cell line. The extent of DNA damage was quantified by increases in low (1-2 foci), medium (3-5 foci), and high (5+ foci) damage. MDA-MB-231 but not MCF-7 bystander cells showed increases in apoptosis in 223RaCl2-treated animals, as did irradiated osteocytes. In summary, radiation-induced bystander effects contribute to DTC cytotoxicity caused by 223RaCl2. IMPLICATIONS: This observation supports clinical investigation of the efficacy of 223RaCl2 to prevent breast cancer DTC from progressing to oligometastases.


Subject(s)
Apoptosis/radiation effects , Bone Marrow/radiation effects , Bystander Effect/radiation effects , DNA Damage/radiation effects , Radium/pharmacology , Alpha Particles/therapeutic use , Animals , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Cell Proliferation/radiation effects , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Osteocytes/radiation effects
13.
Cells ; 10(12)2021 12 13.
Article in English | MEDLINE | ID: mdl-34944025

ABSTRACT

There are numerous studies that investigate the effects of static magnetic fields (SMFs) on osteoblasts and osteoclasts. However, although osteocytes are the most abundant cell type in bone tissue, there are few studies on the biological effects of osteocytes under magnetic fields. Iron is a necessary microelement that is involved in numerous life activities in cells. Studies have shown that high static magnetic fields (HiSMF) can regulate cellular iron metabolism. To illustrate the effect of HiSMF on activities of osteocytes, and whether iron is involved in this process, HiSMF of 16 tesla (T) was used, and the changes in cellular morphology, cytoskeleton, function-related protein expression, secretion of various cytokines, and iron metabolism in osteocytes under HiSMF were studied. In addition, the biological effects of HiSMF combined with iron preparation and iron chelator on osteocytes were also investigated. The results showed that HiSMF promoted cellular viability, decreased apoptosis, increased the fractal dimension of the cytoskeleton, altered the secretion of cytokines, and increased iron levels in osteocytes. Moreover, it was found that the biological effects of osteocytes under HiSMF are attenuated or enhanced by treatment with a certain concentration of iron. These data suggest that HiSMF-regulated cellular iron metabolism may be involved in altering the biological effects of osteocytes under HiSMF exposure.


Subject(s)
Apoptosis/genetics , Cell Survival/genetics , Iron/metabolism , Osteocytes/radiation effects , Animals , Apoptosis/radiation effects , Cell Survival/radiation effects , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/radiation effects , Gene Expression Regulation/radiation effects , Iron/radiation effects , Magnetic Fields/adverse effects , Mice , Microtubules/genetics , Microtubules/radiation effects , Osteoblasts/metabolism , Osteoblasts/radiation effects , Osteoclasts/metabolism , Osteoclasts/radiation effects , Osteocytes/metabolism , RAW 264.7 Cells
14.
Braz Oral Res ; 32: e61, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30379208

ABSTRACT

To evaluate the impact of the GaAlAs diode laser with energy densities of 160 J/cm2, 320 J/cm2, and 640 J/cm2 on the periodontal tissues under continuous orthodontic force application and on the rate of orthodontic tooth movement in rats with type-2 diabetes mellitus. The intensity of primary alveolar bone formation was also investigated through the immune-positive osteocytes for OPN antibody. Forty adult male Wistar rats were divided into eight groups of 5 rats: normoglycemic (N), 160 J-laser-normoglycemic (160 J-LN), 320 J-laser-normoglycemic (320 J-LN), 640 J-laser-normoglycemic (640 J-LN), diabetic (D), 160 J-laser-diabetic (160 J-LD), 320 J-laser-diabetic (320 J-LD), and 640 J-laser-diabetic (640 J-LD) rats. Diabetes mellitus was induced by a single intravenous injection of 40 mg/kg monohydrated-alloxan. An orthodontic force magnitude of 20cN was applied. The laser parameters were continuous emission of 780-nm wavelength, output power of 20mW, and fiber probe with a spot size of 0.04 cm in diameter. Radiographic, histomorphological, and immunohistochemical analysis were performed after a period of 21 days. The photobiomodulation using the energy density of 640 J/cm2 strongly stimulated the alveolar bone formation and contributed the reorganization of the soft periodontal tissues, followed by the 320 J/cm2. Extensive alveolar bone loss, intense infiltration of inflammatory cells, and degradation of the PDJ tissue were mainly found in the D and 160 J-LD groups. The rate of orthodontic tooth movement was represented by the interdental distance between the cementoenamel junctions of the right mandibular first and second molars . This distance was larger in the diabetic groups (D: 39.98±1.97, 160 J-LD: 34.84±6.01, 320 J-LD: 29.82±1.73, and 640 J-LD: 35.47±4.56) than in the normoglycemic groups (N: 21.13±1.19; 160 J-LN: 22.69±0.72, 320 J-LN: 22.28±0.78, and 640 J-LN: 24.56±2.11). The number of osteopontin-positive osteocytes was significantly greater in the 640 J-LD (14.72 ± 0.82; p < 0.01) and 640 J-LN (13.62 ± 1.33; p < 0.05) groups than with D (9.82 ± 1.17) and 160 J-LD (9.77 ± 1.10) groups. Therefore, the energy density of 640 J/cm2 provided the best maintenance and integrity of the periodontal tissue microarchitecture under continuous orthodontic force when compared with the other dosages, mainly in the uncontrolled diabetic rats. The interdental distance was greater in the D and 160 J-LD groups due to presence of severe periodontitis caused by diabetes plus the mechanical stress generated by continuous orthodontic forces, implying, thus, an insufficient biostimulatory effect for the dosage of 160 J/cm2.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Low-Level Light Therapy/methods , Periodontium/radiation effects , Tooth Movement Techniques/methods , Alveolar Bone Loss/pathology , Animals , Diabetes Mellitus, Experimental , Immunohistochemistry , Lasers, Semiconductor/therapeutic use , Male , Orthodontic Appliances , Osteoclasts/radiation effects , Osteocytes/radiation effects , Osteogenesis/radiation effects , Osteopontin/analysis , Periodontium/diagnostic imaging , Periodontium/pathology , Radiation Dosage , Radiography , Random Allocation , Rats, Wistar , Reference Values , Reproducibility of Results
15.
Photomed Laser Surg ; 25(3): 197-204, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17603861

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the effect of laser therapy (lambda = 780 nm) on bone tissue submitted to ionizing radiation. BACKGROUND DATA: The biostimulation effect of laser in normal bone tissue has already been demonstrated successfully; however its effect on bone tissue submitted to radiotherapy has not yet been studied. METHODS: Twenty-two Wistar rats were randomly divided into four groups: group I, control (n = 4), submitted only to radiotherapy; group II, laser starting 1 day prior to radiotherapy (n = 6); group III, laser started immediately after radiotherapy (n = 6); group IV, laser 4 weeks after radiotherapy (n = 6). The source of ionizing radiation used was Cobalt 60, which was applied in a single dose of 3000 cGy on the femur. The laser groups received seven applications with a 48-h interval in four points per session of DE = 4 J/cm(2), P = 40 mW, t = 100 sec, and beam diameter of 0.04 cm(2). All animals were killed 6 weeks after radiotherapy. RESULTS: Clinical examination revealed cutaneous erosions on experimental groups (II, III, and IV) starting at the 6th week after radiotherapy. The radiographic findings showed higher bone density in groups II and IV (p < 0.05) compared to the control group. The results further showed an increase of bone marrow cells, and number of osteocytes and Haversian canals in experimental groups II and IV (p < 0.05). It was also found an increase of osteoblastic activity, in groups II, III, and IV (p < 0.05). CONCLUSION: Laser therapy on bone tissue in rats presented a positive biostimulative effect, especially when applied before or 4 weeks after radiotherapy. However, the use of laser in the parameters above should be used with caution due to epithelial erosions.


Subject(s)
Bone and Bones/radiation effects , Low-Level Light Therapy , Animals , Body Weight/radiation effects , Bone Marrow Cells/radiation effects , Bone and Bones/pathology , Cell Count , Haversian System/radiation effects , Male , Osteocytes/radiation effects , Rats , Rats, Wistar
16.
J Orthop Res ; 35(12): 2799-2807, 2017 12.
Article in English | MEDLINE | ID: mdl-28485534

ABSTRACT

Polymethylmethacrylate bone cement has been widely used for the anchorage of artificial implants in various orthopedic surgeries. Although it is one of the most successful biomaterials in use, excess heat generation intrinsically causes thermal damage to bone cells adjacent to the bone cement. To estimate a risk of thermal injury, a response of bone cells to cement polymerization must be elucidated because of the occurrence of thermal damage. Thermal damage is affected not only by maximal temperature but also by exposure time, temperature history, and cell type. This study aimed at quantifying the thermal tolerance of bone cells for the development of a thermal injury model, and applying this model for the estimation of thermal damage during cement polymerization in total knee arthroplasty. Osteocytes, osteoblasts, and fibroblasts were respectively subjected to steady supraphysiological temperatures ranging from 45 to 50°C. Survival curves of each cell and temperatures were used to formulate the Arrhenius model. A three-dimensional heat conduction analysis for total knee arthroplasty was conducted using the finite element model based on serial CT images of human knee. A maximal temperature rise of 50°C was observed at the interface between the 3-mm thick cement and the tissue immediately beneath the tibial tray of the prosthesis. The probability of thermal damage to the osteocyte, which was calculated using the Arrhenius model, was negligible at a distance of at least 1 mm away from the cement-bone interface. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2799-2807, 2017.


Subject(s)
Arthroplasty, Replacement, Knee/adverse effects , Fibroblasts/radiation effects , Hot Temperature/adverse effects , Infrared Rays/adverse effects , Osteocytes/radiation effects , Animals , Cell Line , Mice
17.
Eur Rev Med Pharmacol Sci ; 21(24): 5562-5570, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29271987

ABSTRACT

OBJECTIVE: Kienböck's disease is a commonly seen posttraumatic avascular necrosis characterized by avascular necrosis of the lunate bone of the wrist which involves the dominant hand. In our study, we aimed to present midterm outcomes of 12 cases treated with radial metaphyseal core decompression. PATIENTS AND METHODS: In our clinic, 12 patients who applied to our outpatient clinic with intractable pain despite at least six weeks of conservative treatment were previously diagnosed and evaluated as Kienböck's disease between the years 2006 and 2014. Patients at early stage received radial metaphyseal core decompression. RESULTS: The patients were evaluated as postoperative grip strength, flexion-extension gap, ulnar-radial deviation gap, VAS, Quick DASH and MAYO wrist scoring and patient satisfaction. CONCLUSIONS: We determined that interventions performed for Kienböck's disease cannot halt radiological progression. We are of the opinion that radial metaphyseal core decompression, aiming at increasing blood perfusion, improve early diagnosis and treatment of Kienböck's disease, increasing the patient satisfaction.


Subject(s)
Adipose Tissue/cytology , Apoptosis/drug effects , Exosomes/physiology , Low-Level Light Therapy , Osteocytes/radiation effects , Stem Cells/physiology , Animals , Cell Hypoxia , Male , Mice , Mice, Inbred C57BL , Osteocytes/pathology
18.
In Vivo ; 20(3): 325-31, 2006.
Article in English | MEDLINE | ID: mdl-16724665

ABSTRACT

BACKGROUND: The alpha-emitter 223Ra, which localizes in osteoblastic active zones, including on skeletal surfaces and in osteoblastic metastases, has recently been introduced as a potential therapeutic agent against skeletal metastases. Here, the adverse effects of high dosages in animals were investigated. MATERIALS AND METHODS: Balb/c mice received intravenously (i.v.) either 1250, 2500, or 3750 kBq/kg of dissolved 223RaCl2 and were followed in the initial toxicity phase. At the 4-week end-point, the animals were sacrificed and blood samples were collected to study the effects on clinical chemistry and hematological parameters. Selected organs were weighed and tissue samples examined by microscopy. RESULTS: Treatment with 223Ra caused a dose-related minimal to moderate depletion of osteocytes and osteoblasts in the bones. Furthermore, a dose-related minimal to marked depletion of the hematopoietic cells in the bone marrow, and a minimal to slight extramedullary hematopoiesis in the spleen and in the mandibular and mesenteric lymph nodes were observed. The LD50 for acute toxicity, defined as death within 4 weeks of receiving the substance, was not reached. CONCLUSION: This study demonstrated that high doses of the bone-seeker 223Ra did not completely inactivate the blood-producing cells. The relatively high tolerance to skeletal alpha doses was probably caused by the surviving pockets of red bone marrow cells beyond the range of alpha particles from the bone surfaces, and the recruitment of peripheral stems cells.


Subject(s)
Alpha Particles/adverse effects , Hematopoiesis/radiation effects , Osteoclasts/radiation effects , Osteocytes/radiation effects , Radium/toxicity , Animals , Clinical Chemistry Tests , Dose-Response Relationship, Radiation , Female , Femur/diagnostic imaging , Femur/metabolism , Hematologic Tests , Histological Techniques , Injections, Intravenous , Lymph Nodes/diagnostic imaging , Lymph Nodes/metabolism , Male , Mice , Mice, Inbred BALB C , Radionuclide Imaging , Radium/administration & dosage , Spleen/diagnostic imaging , Spleen/metabolism , Sternum/diagnostic imaging , Sternum/metabolism , Time Factors
19.
Radiat Res ; 186(3): 254-63, 2016 09.
Article in English | MEDLINE | ID: mdl-27538114

ABSTRACT

Exploration missions to the Moon or Mars will expose astronauts to galactic cosmic radiation and low gravitational fields. Exposure to reduced weightbearing and radiation independently result in bone loss. However, no data exist regarding the skeletal consequences of combining low-dose, high-linear energy transfer (LET) radiation and partial weightbearing. We hypothesized that simulated galactic cosmic radiation would exacerbate bone loss in animals held at one-sixth body weight (G/6) without radiation exposure. Female BALB/cByJ four-month-old mice were randomly assigned to one of the following treatment groups: 1 gravity (1G) control; 1G with radiation; G/6 control; and G/6 with radiation. Mice were exposed to either silicon-28 or X-ray radiation. (28)Si radiation (300 MeV/nucleon) was administered at acute doses of 0 (sham), 0.17 and 0.5 Gy, or in three fractionated doses of 0.17 Gy each over seven days. X radiation (250 kV) was administered at acute doses of 0 (sham), 0.17, 0.5 and 1 Gy, or in three fractionated doses of 0.33 Gy each over 14 days. Bones were harvested 21 days after the first exposure. Acute 1 Gy X-ray irradiation during G/6, and acute or fractionated 0.5 Gy (28)Si irradiation during 1G resulted in significantly lower cancellous mass [percentage bone volume/total volume (%BV/TV), by microcomputed tomography]. In addition, G/6 significantly reduced %BV/TV compared to 1G controls. When acute X-ray irradiation was combined with G/6, distal femur %BV/TV was significantly lower compared to G/6 control. Fractionated X-ray irradiation during G/6 protected against radiation-induced losses in %BV/TV and trabecular number, while fractionated (28)Si irradiation during 1G exacerbated the effects compared to single-dose exposure. Impaired bone formation capacity, measured by percentage mineralizing surface, can partially explain the lower cortical bone thickness. Moreover, both partial weightbearing and (28)Si-ion exposure contribute to a higher proportion of sclerostin-positive osteocytes in cortical bone. Taken together, these data suggest that partial weightbearing and low-dose, high-LET radiation negatively impact maintenance of bone mass by lowering bone formation and increasing bone resorption. The impaired bone formation response is associated with sclerostin-induced suppression of Wnt signaling. Therefore, exposure to low-dose, high-LET radiation during long-duration spaceflight missions may reduce bone formation capacity, decrease cancellous bone mass and increase bone resorption. Future countermeasure strategies should aim to restore mechanical loads on bone to those experienced in one gravity. Moreover, low-doses of high-LET radiation during long-duration spaceflight should be limited or countermeasure strategies employed to mitigate bone loss.


Subject(s)
Bone Resorption/physiopathology , Glycoproteins/metabolism , Linear Energy Transfer , Moon , Osteocytes/radiation effects , Weight-Bearing , Weightlessness Simulation , Adaptor Proteins, Signal Transducing , Animals , Biomarkers/metabolism , Body Weight/radiation effects , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Resorption/pathology , Cosmic Radiation/adverse effects , Dose-Response Relationship, Radiation , Female , Femur/pathology , Femur/physiopathology , Femur/radiation effects , Intercellular Signaling Peptides and Proteins , Mice , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoclasts/radiation effects , Osteocytes/metabolism , Osteocytes/pathology
20.
J Craniomaxillofac Surg ; 43(2): 214-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25523397

ABSTRACT

BACKGROUND: Radiation therapy (RT) as part head and neck cancer treatment often leads to irradiation of surrounding normal tissue, such as mandibular bone. A reduced reparative capacity of the bone can lead to osteoradionecrosis (ORN). Hyperbaric oxygen therapy (HBOT) is used to treat ORN, based on its potential to raise the oxygen tension in tissues. However, prevention of radiation-induced damage is of great interest. Our purpose was to investigate whether HBOT could prevent radiation-induced damage to murine mandibles. METHODS: Twenty-eight mice were irradiated in the head and neck region with a single dose (15 Gy) and half of them were subsequently subjected to HBOT. Another 14 mice did not receive any treatment and served as controls. Ten and 24 weeks after RT, mandibles were harvested and analysed histologically and by microcomputed tomography (micro-CT). RESULTS: Micro-CT analysis showed a reduction in relative bone volume by RT, which was partly recovered by HBOT. Trabecular thickness and separation were also positively influenced by HBOT. Morphologically, HBOT suppressed the osteoclast number, indicating decreased resorption, and decreased the amount of lacunae devoid of osteocytes, indicating increased bone viability. CONCLUSIONS: HBOT was able to partly reduce radiation-induced effects on microarchitectural parameters, resorption, and bone viability in mouse mandibles. HBOT could therefore potentially play a role in the prevention of radiation-induced damage to human mandibular bone.


Subject(s)
Hyperbaric Oxygenation/methods , Mandibular Diseases/prevention & control , Osteoradionecrosis/prevention & control , Radiation Injuries, Experimental/prevention & control , Animals , Bone Resorption/prevention & control , Cell Count , Female , Mandible/pathology , Mandible/radiation effects , Mice , Mice, Inbred C3H , Organ Size/radiation effects , Osteoclasts/pathology , Osteoclasts/radiation effects , Osteocytes/pathology , Osteocytes/radiation effects , Osteogenesis/radiation effects , Radiotherapy Dosage , Time Factors , Tissue Survival/radiation effects , X-Ray Microtomography/methods
SELECTION OF CITATIONS
SEARCH DETAIL