Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Bioeng Biotechnol ; 10: 1011853, 2022.
Article in English | MEDLINE | ID: mdl-36338134

ABSTRACT

Bioactive coatings are promising for improving osseointegration and the long-term success of titanium dental or orthopaedic implants. Biomimetic octacalcium phosphate (OCP) coating can be used as a carrier for osteoinductive agents. κ-Carrageenan, a highly hydrophilic and biocompatible seaweed-derived sulfated-polysaccharide, promotes pre-osteoblast activity required for bone regeneration. Whether κ-carrageenan can functionalize OCP-coating to enhance osseointegration of titanium implants is unclear. This study aimed to analyze carrageenan-functionalized biomimetic OCP-coated titanium structure, and effects of carrageenan functionalization on pre-osteoblast behavior and osteogenic differentiation. Titanium discs were coated with OCP/κ-carrageenan at 0.125-2 mg/ml OCP solution, and physicochemical and biological properties were investigated. κ-Carrageenan (2 mg/ml) in the OCP coating of titanium discs decreased the pore size in the sheet-like OCP crystal by 41.32%. None of the κ-carrageenan concentrations tested in the OCP-coating did affect hydrophilicity. However, κ-carrageenan (2 mg/ml) increased (1.26-fold) MC3T3-E1 pre-osteoblast spreading at 1 h i.e., κ-Carrageenan in the OCP-coating increased pre-osteoblast proliferation (max. 1.92-fold at 2 mg/ml, day 1), metabolic activity (max. 1.50-fold at 2 mg/ml, day 3), and alkaline phosphatase protein (max. 4.21-fold at 2 mg/ml, day 3), as well as matrix mineralization (max. 5.45-fold at 2 mg/ml, day 21). κ-Carrageenan (2 mg/ml) in the OCP-coating increased gene expression of Mepe (4.93-fold) at day 14, and Runx2 (2.94-fold), Opn (3.59-fold), Fgf2 (3.47-fold), Ocn (3.88-fold), and Dmp1 (4.59-fold) at day 21 in pre-osteoblasts. In conclusion, κ-carrageenan modified the morphology and microstructure of OCP-coating on titanium discs, and enhanced pre-osteoblast metabolic activity, proliferation, and osteogenic differentiation. This suggests that κ-carrageenan-functionalized OCP coating may be promising for in vivo improvement of titanium implant osseointegration.

2.
Molecules ; 26(20)2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34684714

ABSTRACT

Current cell-based bone tissue regeneration strategies cannot cover large bone defects. K-carrageenan is a highly hydrophilic and biocompatible seaweed-derived sulfated polysaccharide, that has been proposed as a promising candidate for tissue engineering applications. Whether κ-carrageenan can be used to enhance bone regeneration is still unclear. In this study, we aimed to investigate whether κ-carrageenan has osteogenic potential by testing its effect on pre-osteoblast proliferation and osteogenic differentiation in vitro. Treatment with κ-carrageenan (0.5 and 2 mg/mL) increased both MC3T3-E1 pre-osteoblast adhesion and spreading at 1 h. K-carrageenan (0.125-2 mg/mL) dose-dependently increased pre-osteoblast proliferation and metabolic activity, with a maximum effect at 2 mg/mL at day three. K-carrageenan (0.5 and 2 mg/mL) increased osteogenic differentiation, as shown by enhanced alkaline phosphatase activity (1.8-fold increase at 2 mg/mL) at day four, and matrix mineralization (6.2-fold increase at 2 mg/mL) at day 21. K-carrageenan enhanced osteogenic gene expression (Opn, Dmp1, and Mepe) at day 14 and 21. In conclusion, κ-carrageenan promoted MC3T3-E1 pre-osteoblast adhesion and spreading, metabolic activity, proliferation, and osteogenic differentiation, suggesting that κ-carrageenan is a potential osteogenic inductive factor for clinical application to enhance bone regeneration.


Subject(s)
Bone Regeneration/physiology , Carrageenan/pharmacology , Osteogenesis/drug effects , Animals , Bone Regeneration/drug effects , Carrageenan/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/physiology , Tissue Engineering/methods
3.
Biophys J ; 117(6): 1029-1036, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31495446

ABSTRACT

From observations of colloidal tracer particles in fibrin undergoing gelation, we introduce an analytical framework that allows the determination of the probability density function for a stochastic process beyond fractional Brownian motion. Using passive microrheology via videomicroscopy, mean square displacements of tracer particles suspended in fibrin at different ageing times are obtained. The anomalous diffusion is then described by a damped white noise process with memory, with analytical results closely matching experimental plots of mean square displacements and probability density function. We further show that the white noise functional stochastic approach applied to passive microrheology reveals the existence of a gelation parameter µ which elucidates the dynamics of constrained tracer particles embedded in a time-dependent soft material. In addition, we found that microstructural heterogeneity of particle environments decreases as the ageing time increases. This study offers experimental insights on the ageing of fibrin gels while presenting a white noise functional stochastic approach that could be applied to other systems exhibiting non-Markovian diffusive behavior.


Subject(s)
Fibrin/chemistry , Gels/chemistry , Diffusion , Humans , Probability , Time Factors
4.
Int J Oral Sci ; 10(1): 2, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483534

ABSTRACT

Bone mass is important for dental implant success and is regulated by mechanoresponsive osteocytes. We aimed to investigate the relationship between the levels and orientation of tensile strain and morphology and orientation of osteocytes at different dental implant positions in the maxillary bone. Bone biopsies were retrieved from eight patients who underwent maxillary sinus-floor elevation with ß-tricalcium phosphate prior to implant placement. Gap versus free-ending locations were compared using 1) a three-dimensional finite-element model of the maxilla to predict the tensile strain magnitude and direction and 2) histology and histomorphometric analyses. The finite-element model predicted larger, differently directed tensile strains in the gap versus free-ending locations. The mean percentage of mineralised residual native-tissue volume, osteocyte number (mean ± standard deviations: 97 ± 40/region-of-interest), and osteocyte shape (~90% elongated, ~10% round) were similar for both locations. However, the osteocyte surface area was 1.5-times larger in the gap than in the free-ending locations, and the elongated osteocytes in these locations were more cranially caudally oriented. In conclusion, significant differences in the osteocyte surface area and orientation seem to exist locally in the maxillary bone, which may be related to the tensile strain magnitude and orientation. This might reflect local differences in the osteocyte mechanosensitivity and bone quality, suggesting differences in dental implant success based on the location in the maxilla.


Subject(s)
Bone-Implant Interface , Dental Implants , Osteocytes/physiology , Biopsy , Calcium Phosphates/pharmacology , Finite Element Analysis , Humans , Maxilla/surgery , Radiography, Panoramic , Sinus Floor Augmentation , Tensile Strength
5.
Curr Osteoporos Rep ; 13(2): 61-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25663071

ABSTRACT

There is considerable variation in the shape of osteocyte lacunae, which is likely to influence the function of osteocytes as the professional mechanosensors of bone. In this review, we first discussed how mechanical loading could affect the shape of osteocyte lacunae. Recent studies show that osteocyte lacunae are aligned to collagen. Since collagen fiber orientation is affected by loading mode, this alignment may help to understand how mechanical loading shapes the osteocyte lacuna. Secondly, we discussed how the shape of osteocytes could influence their mechanosensation. In vitro, round osteocytes are more mechanosensitive than flat osteocytes. Altered lacunar morphology has been associated with bone pathology. It is important to know whether osteocyte shape is part of the etiology.


Subject(s)
Cell Shape/physiology , Osteocytes/cytology , Stress, Mechanical , Animals , Collagen/physiology , Humans , In Vitro Techniques , Mechanotransduction, Cellular/physiology , Osteocytes/physiology , Weight-Bearing/physiology
6.
J Biomech ; 48(5): 855-65, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25582356

ABSTRACT

Adaptation of bone to mechanical stresses normally produces a bone architecture that combines a proper resistance against failure with a minimal use of material. This adaptive process is governed by mechanosensitive osteocytes that transduce the mechanical signals into chemical responses, i.e. the osteocytes release signaling molecules, which orchestrate the recruitment and activity of bone forming osteoblasts and/or bone resorbing osteoclasts. Computer models have shown that the maintenance of a mechanically-efficient bone architecture depends on the intensity and spatial distribution of the mechanical stimulus as well as on the osteocyte response. Osteoporosis is a condition characterized by a reduced bone mass and a compromized resistance of bone against mechanical loads, which has led us to hypothesize that mechanotransduction by osteocytes is altered in osteoporosis. One of the major causal factors for osteoporosis is the loss of estrogen, the major hormonal regulator of bone metabolism. Loss of estrogen may increase osteocyte-mediated activation of bone remodeling, resulting in impaired bone mass and architecture. In this review we highlight current insights on how osteocytes perceive mechanical stimuli placed on whole bones. Particular emphasis is placed on the role of estrogen in signaling pathway activation by mechanical stimuli, and on computer simulation in combination with cell biology to unravel biological processes contributing to bone strength.


Subject(s)
Estrogens/deficiency , Osteoblasts/physiology , Osteoporosis/physiopathology , Animals , Bone and Bones/physiology , Estrogens/physiology , Humans , Mechanotransduction, Cellular/physiology , Osteoporosis/metabolism , Stress, Mechanical
7.
Biophys J ; 105(10): 2240-51, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24268136

ABSTRACT

During wound healing and angiogenesis, fibrin serves as a provisional extracellular matrix. We use a model system of fibroblasts embedded in fibrin gels to study how cell-mediated contraction may influence the macroscopic mechanical properties of their extracellular matrix during such processes. We demonstrate by macroscopic shear rheology that the cells increase the elastic modulus of the fibrin gels. Microscopy observations show that this stiffening sets in when the cells spread and apply traction forces on the fibrin fibers. We further show that the stiffening response mimics the effect of an external stress applied by mechanical shear. We propose that stiffening is a consequence of active myosin-driven cell contraction, which provokes a nonlinear elastic response of the fibrin matrix. Cell-induced stiffening is limited to a factor 3 even though fibrin gels can in principle stiffen much more before breaking. We discuss this observation in light of recent models of fibrin gel elasticity, and conclude that the fibroblasts pull out floppy modes, such as thermal bending undulations, from the fibrin network, but do not axially stretch the fibers. Our findings are relevant for understanding the role of matrix contraction by cells during wound healing and cancer development, and may provide design parameters for materials to guide morphogenesis in tissue engineering.


Subject(s)
Fibrin/metabolism , Stress, Mechanical , Biomechanical Phenomena , Extracellular Matrix/metabolism , Fibrin/chemistry , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Rheology
8.
Bone ; 54(2): 182-90, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23085083

ABSTRACT

The human skeleton is a miracle of engineering, combining both toughness and light weight. It does so because bones possess cellular mechanisms wherein external mechanical loads are sensed. These mechanical loads are transformed into biological signals, which ultimately direct bone formation and/or bone resorption. Osteocytes, since they are ubiquitous in the mineralized matrix, are the cells that sense mechanical loads and transduce the mechanical signals into a chemical response. The osteocytes then release signaling molecules, which orchestrate the recruitment and activity of osteoblasts or osteoclasts, resulting in the adaptation of bone mass and structure. In this review, we highlight current insights in bone adaptation to external mechanical loading, with an emphasis on how a mechanical load placed on whole bones is translated and amplified into a mechanical signal that is subsequently sensed by the osteocytes.


Subject(s)
Mechanotransduction, Cellular , Osteocytes/metabolism , Animals , Humans , Stress, Mechanical
9.
Integr Biol (Camb) ; 4(10): 1198-206, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22858651

ABSTRACT

Osteocytes play a pivotal role in the regulation of skeletal mass. Osteocyte processes are thought to sense the flow of interstitial fluid that is driven through the osteocyte canaliculi by mechanical stimuli placed upon bone, but how this flow elicits a cellular response is virtually unknown. Modern theoretical models assume that osteocyte canaliculi contain ultrastructural features that amplify the fluid flow-derived mechanical signal. Unfortunately the calcified bone matrix has considerably hampered studies on the osteocyte process within its canaliculus. Using one of the few ultra high voltage electron microscopes (UHVEM) available worldwide, we applied UHVEM tomography at 2 MeV to reconstruct unique three-dimensional images of osteocyte canaliculi in 1 µm sections of human bone. A realistic three-dimensional image-based model of a single canaliculus was constructed, and the fluid dynamics of a Newtonian fluid flow within the canaliculus was analyzed. We created virtual 2.2 nm thick sections through a canaliculus and found that traditional TEM techniques create a false impression that osteocyte processes are directly attached to the canalicular wall. The canalicular wall had a highly irregular surface and contained protruding axisymmetric structures similar in size and shape to collagen fibrils. We also found that the microscopic surface roughness of the canalicular wall strongly influenced the fluid flow profiles, whereby highly inhomogeneous flow patterns emerged. These inhomogeneous flow patterns may induce deformation of cytoskeletal elements in the osteocyte process, thereby amplifying mechanical signals. Based on these observations, new and realistic models can be developed that will significantly enhance our understanding of the process of mechanotransduction in bone.


Subject(s)
Osteocytes/cytology , Biomechanical Phenomena , Bone Matrix/physiology , Bone Matrix/ultrastructure , Bone and Bones/physiology , Bone and Bones/ultrastructure , Collagen/chemistry , Computer Simulation , Extracellular Matrix/metabolism , Humans , Imaging, Three-Dimensional , Male , Mechanotransduction, Cellular/physiology , Microscopy, Electron, Transmission/methods , Middle Aged , Models, Biological , Models, Statistical , Osteocytes/ultrastructure , Tomography, X-Ray Computed/methods
10.
Biophys J ; 98(10): 2281-9, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20483337

ABSTRACT

Fibrin gels are responsible for the mechanical strength of blood clots, which are among the most resilient protein materials in nature. Here we investigate the physical origin of this mechanical behavior by performing rheology measurements on reconstituted fibrin gels. We find that increasing levels of shear strain induce a succession of distinct elastic responses that reflect stretching processes on different length scales. We present a theoretical model that explains these observations in terms of the unique hierarchical architecture of the fibers. The fibers are bundles of semiflexible protofibrils that are loosely connected by flexible linker chains. This architecture makes the fibers 100-fold more flexible to bending than anticipated based on their large diameter. Moreover, in contrast with other biopolymers, fibrin fibers intrinsically stiffen when stretched. The resulting hierarchy of elastic regimes explains the incredible resilience of fibrin clots against large deformations.


Subject(s)
Elasticity/physiology , Fibrin/chemistry , Gels/chemistry , Models, Molecular , Shear Strength/physiology , Biomechanical Phenomena , Materials Testing , Mechanics , Rheology , Tensile Strength/physiology
11.
Technol Health Care ; 17(1): 57-65, 2009.
Article in English | MEDLINE | ID: mdl-19478406

ABSTRACT

Stochastic resonance is exhibited by many biological systems, where the response to a small stimulus is enhanced with the aid of noise. This intriguing possibility provides a novel paradigm for understanding previously reported osteogenic benefits of low amplitude dynamic loading. However, it is unknown whether bone cell mechanosensitivity is enhanced by noise as an alternative mechanism for an amplified response to small stresses. We studied whether noise of varying intensities enhanced the mechanosensitivity of MC3T3-E1 cells. Nitric oxide (NO) production was measured as the parameter for bone cell activation. Dynamic fluid shear stress stimulated bone cells provided an initial-stress kick was implemented. Without the initial stress-kick bone cells did not release a significant amount of NO demonstrating an essential non-linearity to bone cell responses to stress and the possibility of stochastic resonance in bone cell mechanosensitivity. The rapid NO response of MC3T3-E1 cells to a small periodic fluid shear stress was increased with the addition of noise compared to the response to stress with only noise. This confirms the possibility of stochastic resonance enhancement of NO production by bone cells. Since NO regulate bone formation as well as resorption, our results suggest that noise enhances the activity of bone cells in driving the mechanical adaptation of bone.


Subject(s)
Mechanotransduction, Cellular/physiology , Nitric Oxide/biosynthesis , Noise , Osteocytes/metabolism , Animals , Bone Resorption/metabolism , Bone Resorption/physiopathology , Bone and Bones/cytology , Cells, Cultured , Osteocytes/cytology , Osteogenesis/physiology , Stress, Mechanical
12.
J Biomech ; 41(7): 1590-8, 2008.
Article in English | MEDLINE | ID: mdl-18402963

ABSTRACT

There is increasing evidence that cell function and mechanical properties are closely related to morphology. However, most in vitro studies investigate flat adherent cells, which might not reflect physiological geometries in vivo. Osteocytes, the mechanosensors in bone, reside within ellipsoid containment, while osteoblasts adhere to flatter bone surfaces. It is unknown whether morphology difference, dictated by the geometry of attachment is important for cell rheology and mechanosensing. We developed a novel methodology for investigating the rheology and mechanosensitivity of bone cells under different morphologies using atomic force microscopy and our two-particle assay for optical tweezers. We found that the elastic constant of MLO-Y4 osteocytes when flat and adherent (>1 kPa) largely differed when round but partially adherent (<1 kPa). The elastic constant of round suspended MLO-Y4 osteocytes, MC3T3-E1 osteoblasts, and primary osteoblasts were similarly <1 kPa. The mechanosensitivity of round suspended MLO-Y4 osteocytes was investigated by monitoring nitric oxide (NO) release, an essential signaling molecule in bone. A preliminary observation of high NO release from round suspended MLO-Y4 osteocytes in response to approximately 5 pN force is reported here, in contrast with previous studies where flat cells routinely release lesser NO while being stimulated with higher force. Our results suggest that a round cellular morphology supports a less stiff cytoskeleton configuration compared with flat cellular morphology. This implies that osteocytes take advantage of their ellipsoid morphology in vivo to sense small strains benefiting bone health. Our assay provides novel opportunities for in vitro studies under a controlled suspended morphology versus commonly studied adherent morphologies.


Subject(s)
Cell Shape/physiology , Mechanotransduction, Cellular/physiology , Osteocytes/physiology , Animals , Cell Line , Elasticity , Humans , Microscopy, Atomic Force/methods , Nitric Oxide/metabolism , Optical Tweezers , Osteocytes/ultrastructure , Rheology
13.
FASEB J ; 20(7): 858-64, 2006 May.
Article in English | MEDLINE | ID: mdl-16675843

ABSTRACT

Mechanosensing by cells directs changes in bone mass and structure in response to the challenges of mechanical loading. Low-amplitude, high-frequency loading stimulates bone growth by enhancing bone formation and inhibiting disuse osteoporosis. However, how bone cells sense vibration stress is unknown. Hence, we investigated bone cell responses to vibration stress at a wide frequency range (5-100 Hz). We used NO and prostaglandin E2 (PGE2) release, and COX-2 mRNA expression as parameters for bone cell response since these molecules regulate bone adaptation to mechanical loading. NO release positively correlated whereas PGE2 release negatively correlated to the maximum acceleration rate of the vibration stress. COX-2 mRNA expression increased in a frequency-dependent manner, which relates to increased NO release at high frequencies, confirming our previous results. The negatively correlated release of NO and PGE2 suggests that these signaling molecules play different roles in bone adaptation to high-frequency loading. The maximum acceleration rate is proportional to omega3 (frequency=omega/2pi), which is commensurate with the Stokes-Einstein relation for modeling cell nucleus motion within the cytoplasm due to vibration stress. Correlations of NO and PGE2 with the maximum acceleration rate then relate to nucleus oscillations, providing a physical basis for cellular mechanosensing of high-frequency loading.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Osteoblasts/metabolism , Vibration , 3T3 Cells , Animals , Biomechanical Phenomena , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic , Mice , Nitric Oxide/metabolism
14.
J Orthop Res ; 24(6): 1170-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16705700

ABSTRACT

Loading frequency is an important parameter for the stimulation of bone formation in vivo. It is still unclear how the information of external loading characteristics is conveyed to osteoblasts and osteoclasts. Osteocytes are thought to detect mechanical loads by sensing fluid flow through the lacuno-canalicular network within bone and to translate this information into chemical signals. The signaling molecules nitric oxide (NO) and prostaglandin E2 (PGE2) are known to play important roles in the adaptive response of bone to mechanical loads. We have investigated the effects of fluid flow frequency on the production of PGE2 and NO in bone cells in vitro. Pulsatile fluid flow with different frequencies stimulated the release of NO by MC3T3-E1 osteoblasts in a dose-dependent manner. In contrast, PGE2 production was enhanced consistently by all fluid flow regimes, independent of flow frequency. This implies that the NO response may play a role in mediating the differential effects of the various loading patterns on bone.


Subject(s)
Dinoprostone/metabolism , Nitric Oxide/metabolism , Osteocytes/metabolism , Pulsatile Flow/physiology , 3T3 Cells , Animals , Cell Line, Transformed , Mice , Osteocytes/cytology , Signal Transduction , Stress, Mechanical
15.
Ann Biomed Eng ; 33(1): 104-10, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15709711

ABSTRACT

The shear stress induced by the loading-mediated flow of interstitial fluid through the lacuno-canalicular network is a likely stimulus for bone cell adaptive responses. Furthermore, the magnitude of the cellular response is related to the rate of mechanical loading rather than its magnitude. Thus, bone cells might be very sensitive to sudden stress-kicks, as occuring e.g., during impact loading. There is evidence that cells change stiffness under stress, which might make them more sensitive to subsequent loading. We studied the influence of a stress-kick on the mechanosensitivity of MC3T3-E1 osteoblast-like cells under different peak shear rate conditions, as measured by nitric oxide production. MC3T3-E1 bone cells were treated with steady or pulsating fluid shear stress (PFSS) for 5 min with different peak rates (9.70, 17.5, and 22.0 Pa Hz) using varying frequencies (5 and 9 Hz), and amplitudes (0.70 and 0.31 Pa). PFSS treatment was done with or without fluid flow pretreatment phase, which removed the initial stress-kick by first applying a slow fluid flow increase. Nitric oxide production in response to fluid shear stress was rate dependent, but necessitated an initial stress-kick to occur. This suggests that high-rate stimuli condition bone cells to be more sensitive for high-frequency, low-amplitude loads.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Mechanotransduction, Cellular/physiology , Nitric Oxide/biosynthesis , Osteoblasts/physiology , Cell Line , Humans , Stress, Physiological , Vibration
16.
J Biomech ; 38(1): 159-67, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15519352

ABSTRACT

An in vitro model using a parallel-plate fluid flow chamber is supposed to simulate in vivo fluid shear stresses on various cell types exposed to dynamic fluid flow in their physiological environment. The metabolic response of cells in vitro is associated with the wall shear stress. However, parallel-plate flow chambers have not been characterized for dynamic fluid flow experiments. We use a dimensionless ratio h / lambda(v), in determining the exact magnitude of the dynamic wall shear stress, with its oscillating components scaled by a shear factor T. It is shown that, in order to expose cells to predictable levels of dynamic fluid shear stress, two conditions have to be met: (1) h / lambda(v) < 2, where h is the distance between the plates and lambda(v) is the viscous penetration depth; and (2) f(0) < f(c) / m, where the critical frequency f(c) is the upper threshold for this flow regime, m is the highest harmonic mode of the flow, and f(0) is the fundamental frequency of fluid flow.


Subject(s)
Body Fluids/physiology , Cell Physiological Phenomena , Models, Biological , Physiology/instrumentation , Animals , Computer Simulation , Differential Threshold , Equipment Design , Humans , Stress, Mechanical , Viscosity
17.
Biochem Biophys Res Commun ; 315(4): 823-9, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14985086

ABSTRACT

Shear stress due to mechanical loading-induced flow of interstitial fluid through the lacuno-canalicular network is a likely signal for bone cell adaptive responses. Moreover, the rate (determined by frequency and magnitude) of mechanical loading determines the amount of bone formation. Whether the bone cells' response to fluid shear stress is rate dependent is unknown. Here we investigated whether bone cell activation by fluid shear stress is rate dependent. MC3T3-E1 osteoblastic cells were subjected for 15 min to fluid shear stress of varying frequencies and amplitudes, resulting in peak fluid shear stress rates ranging from 0 to 39.6 Pa-Hz. Nitric oxide production, a parameter for bone cell activation, was found to be linearly dependent on the fluid shear stress rate; the slope was steepest at 5 min (0.11 Pa-Hz(-1)) and decreased to 0.03 Pa-Hz(-1) at 15 min. We conclude that the fluid shear stress rate is an important parameter for bone cell activation.


Subject(s)
Bone and Bones/metabolism , Nitric Oxide/biosynthesis , Animals , Biomechanical Phenomena , Biosensing Techniques/methods , Bone and Bones/cytology , Cells, Cultured , Hip Joint/physiology , Humans , Male , Mice , Pulsatile Flow , Regression Analysis , Stress, Mechanical , Walking
SELECTION OF CITATIONS
SEARCH DETAIL
...