Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Exp Cell Res ; 417(1): 113204, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35588795

ABSTRACT

Muscle stem cells (MuSCs) are involved in muscle maintenance and regeneration. Mechanically loaded MuSCs within their native niche undergo tensile and shear deformations, but how MuSCs sense mechanical stimuli and translate these into biochemical signals regulating function and fate is still poorly understood. We aimed to investigate whether the glycocalyx is involved in the MuSC mechanoresponse, and whether MuSC morphology affects mechanical loading-induced pressure, shear stress, and fluid velocity distribution. FSS-induced deformation of active proliferating MuSCs (myoblasts) with intact or degraded glycocalyx was assessed by live-cell imaging. Glycocalyx-degradation did not significantly affect nitric oxide production, but reduced FSS-induced myoblast deformation and modulated gene expression. Finite-element analysis revealed that the distribution of FSS-induced pressure, shear stress, and fluid velocity on myoblasts was non-uniform, and the magnitude depended on myoblast morphology and apex-height. In conclusion, our results suggest that the glycocalyx does not play a role in NO production in myoblasts but might impact mechanotransduction and gene expression, which needs further investigation. Future studies will unravel the underlying mechanism by which the glycocalyx affects FSS-induced myoblast deformation, which might be related to increased drag forces. Moreover, MuSCs with varying apex-height experience different levels of FSS-induced pressure, shear stress, and fluid velocity, suggesting differential responsiveness to fluid shear forces.


Subject(s)
Glycocalyx , Mechanotransduction, Cellular , Glycocalyx/metabolism , Mechanotransduction, Cellular/physiology , Myoblasts/metabolism , Nitric Oxide/metabolism , Stress, Mechanical
2.
Curr Osteoporos Rep ; 21(6): 731-742, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37792246

ABSTRACT

PURPOSE OF REVIEW: Orthodontic tooth movement is characterized by periodontal tissue responses to mechanical loading, leading to clinically relevant functional adaptation of jaw bone. Since osteocytes are significant in mechanotransduction and orchestrate osteoclast and osteoblast activity, they likely play a central role in orthodontic tooth movement. In this review, we attempt to shed light on the impact and role of osteocyte mechanotransduction during orthodontic tooth movement. RECENT FINDINGS: Mechanically loaded osteocytes produce signaling molecules, e.g., bone morphogenetic proteins, Wnts, prostaglandins, osteopontin, nitric oxide, sclerostin, and RANKL, which modulate the recruitment, differentiation, and activity of osteoblasts and osteoclasts. The major signaling pathways activated by mechanical loading in osteocytes are the wingless-related integration site (Wnt)/ß-catenin and RANKL pathways, which are key regulators of bone metabolism. Moreover, osteocytes are capable of orchestrating bone adaptation during orthodontic tooth movement. A better understanding of the role of osteocyte mechanotransduction is crucial to advance orthodontic treatment. The optimal force level on the periodontal tissues for orthodontic tooth movement producing an adequate biological response, is debated. This review emphasizes that both mechanoresponses and inflammation are essential for achieving tooth movement clinically. To fully comprehend the role of osteocyte mechanotransduction in orthodontic tooth movement, more knowledge is needed of the biological pathways involved. This will contribute to optimization of orthodontic treatment and enhance patient outcomes.


Subject(s)
Mechanotransduction, Cellular , Osteocytes , Humans , Osteocytes/physiology , Tooth Movement Techniques , Osteoclasts/metabolism , Osteoblasts/metabolism , Bone Remodeling/physiology
3.
Calcif Tissue Int ; 110(3): 367-379, 2022 03.
Article in English | MEDLINE | ID: mdl-34647170

ABSTRACT

Osteocytes are mechanosensory cells which are embedded in calcified collagenous matrix. The specific native matrix of osteocytes affects their regulatory activity, i.e., transmission of signaling molecules to osteoclasts and/or osteoblasts, in the mechanical adaptation of bone. Unfortunately, no existing in vitro model of cortical bone is currently available to study the mechanosensory function of human osteocytes in their native matrix. Therefore, we aimed to develop an in vitro three-dimensional mechanical loading model of human osteocytes in their native matrix. Human cortical bone explants containing osteocytes in their three-dimensional native matrix were cultured and mechanically loaded by three-point bending using a custom-made loading apparatus generating sinusoidal displacement. Osteocyte viability and sclerostin expression were measured 1-2 days before 5 min loading and 1 day after loading. Bone microdamage was visualized and quantified by micro-CT analysis and histology using BaSO4 staining. A linear relationship was found between loading magnitude (2302-13,811 µÉ›) and force (1.6-4.9 N) exerted on the bone explants. At 24 h post-loading, osteocyte viability was not affected by 1600 µÉ› loading. Sclerostin expression and bone microdamage were unaffected by loading up to 8000 µÉ›. In conclusion, we developed an in vitro 3D mechanical loading model to study mechanoresponsiveness of viable osteocytes residing in their native matrix. This model is suitable to study the effect of changed bone matrix composition in metabolic bone disease on osteocyte mechanoresponsiveness.


Subject(s)
Osteoclasts , Osteocytes , Bone Matrix , Bone and Bones , Humans , Osteoblasts , Osteocytes/metabolism , Stress, Mechanical
4.
Biophys J ; 120(13): 2665-2678, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34087215

ABSTRACT

Muscle stem cells (MuSCs) are requisite for skeletal muscle regeneration and homeostasis. Proper functioning of MuSCs, including activation, proliferation, and fate decision, is determined by an orchestrated series of events and communication between MuSCs and their niche. A multitude of biochemical stimuli are known to regulate MuSC fate and function. However, in addition to biochemical factors, it is conceivable that MuSCs are subjected to mechanical forces during muscle stretch-shortening cycles because of myofascial connections between MuSCs and myofibers. MuSCs respond to mechanical forces in vitro, but it remains to be proven whether physical forces are also exerted on MuSCs in their native niche and whether they contribute to the functioning and fate of MuSCs. MuSC deformation in their native niche resulting from mechanical loading of ex vivo myofiber bundles was visualized utilizing mT/mG double-fluorescent Cre-reporter mouse and multiphoton microscopy. MuSCs were subjected to 1 h pulsating fluid shear stress (PFSS) with a peak shear stress rate of 6.5 Pa/s. After PFSS treatment, nitric oxide, messenger RNA (mRNA) expression levels of genes involved in regulation of MuSC proliferation and differentiation, ERK 1/2, p38, and AKT activation were determined. Ex vivo stretching of extensor digitorum longus and soleus myofiber bundles caused compression as well as tensile and shear deformation of MuSCs in their niche. MuSCs responded to PFSS in vitro with increased nitric oxide production and an upward trend in iNOS mRNA levels. PFSS enhanced gene expression of c-Fos, Cdk4, and IL-6, whereas expression of Wnt1, MyoD, Myog, Wnt5a, COX2, Rspo1, Vangl2, Wnt10b, and MGF remained unchanged. ERK 1/2 and p38 MAPK signaling were also upregulated after PFSS treatment. We conclude that MuSCs in their native niche are subjected to force-induced deformations due to myofiber stretch-shortening. Moreover, MuSCs are mechanoresponsive, as evidenced by PFSS-mediated expression of factors by MuSCs known to promote proliferation.


Subject(s)
Muscle, Skeletal , Myoblasts , Animals , Cell Differentiation , Gene Expression , Mice , Stress, Mechanical
5.
Biofouling ; 37(2): 184-193, 2021 02.
Article in English | MEDLINE | ID: mdl-33615928

ABSTRACT

In this in vitro study the effect of XZ.700, a new endolysin, on methicillin resistant Staphylococcus aureus (MRSA) biofilms grown on titanium was evaluated. Biofilms of S. aureus USA300 were grown statically and under flow, and treatment with XZ.700 was compared with povidone-iodine (PVP-I) and gentamicin. To evaluate the cytotoxic effects of XZ.700 and derived biofilm lysates, human osteocyte-like cells were exposed to biofilm supernatants, and metabolism and proliferation were quantified. XZ.700 showed a significant, concentration dependent reduction in biofilm viability, compared with carrier controls. Metabolism and proliferation of human osteocyte-like cells were not affected by XZ.700 or lysates, unlike PVP-I and gentamicin lysates which significantly inhibited proliferation. Using time-lapse microscopy, rapid biofilm killing and removal was observed for XZ.700. In comparison, PVP-I and gentamicin showed slower biofilm killing, with no apparent biofilm removal. In conclusion, XZ.700 reduced MRSA biofilms, especially under flow condition, without toxicity for surrounding bone cells.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Anti-Bacterial Agents/toxicity , Biofilms , Endopeptidases , Humans , Osteocytes , Staphylococcus aureus
6.
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
7.
Curr Osteoporos Rep ; 18(5): 541-550, 2020 10.
Article in English | MEDLINE | ID: mdl-32676786

ABSTRACT

PURPOSE OF REVIEW: Bone regeneration plays an important role in contemporary clinical treatment. Bone tissue engineering should result in successful bone regeneration to restore congenital or acquired bone defects in the human skeleton. Osteocytes are thought to have a governing role in bone remodeling by regulating osteoclast and osteoblast activity, and thus bone loss and formation. In this review, we address the so far largely unknown role osteocytes may play in bone tissue regeneration. RECENT FINDINGS: Osteocytes release biochemical signaling molecules involved in bone remodeling such as prostaglandins, nitric oxide, Wnts, and insulin-like growth factor-1 (IGF-1). Treatment of mesenchymal stem cells in bone tissue engineering with prostaglandins (e.g., PGE2, PGI2, PGF2α), nitric oxide, IGF-1, or Wnts (e.g., Wnt3a) improves osteogenesis. This review provides an overview of the functions of osteocytes in bone tissue, their interaction with other bone cells, and their role in bone remodeling. We postulate that osteocytes may have a pivotal role in bone regeneration as well, and consequently that the bone regeneration process may be improved effectively and rapidly if osteocytes are optimally used and stimulated.


Subject(s)
Bone Regeneration/physiology , Bone Remodeling/physiology , Osteocytes/physiology , Bone Resorption , Guided Tissue Regeneration , Humans , Insulin-Like Growth Factor I , Nitric Oxide , Osteoblasts/physiology , Osteoclasts/physiology , Osteocytes/metabolism , Osteogenesis , Prostaglandins , Signal Transduction , Tissue Engineering , Wnt Proteins/metabolism
8.
Int J Mol Sci ; 21(21)2020 Nov 07.
Article in English | MEDLINE | ID: mdl-33171812

ABSTRACT

Mechanical loading preserves bone mass and function-yet, little is known about the cell biological basis behind this preservation. For example, cell and nucleus morphology are critically important for cell function, but how these morphological characteristics are affected by the physiological mechanical loading of bone cells is under-investigated. This study aims to determine the effects of fluid shear stress on cell and nucleus morphology and volume of osteoblasts, and how these effects relate to changes in actin cytoskeleton and focal adhesion formation. Mouse calvaria 3T3-E1 (MC3T3-E1) osteoblasts were treated with or without 1 h pulsating fluid flow (PFF). Live-cell imaging was performed every 10 min during PFF and immediately after PFF. Cytoskeletal organization and focal adhesions were visualized, and gene and protein expression quantified. Two-dimensional (2D) and three-dimensional (3D) morphometric analyses were made using MeasureStack and medical imaging interaction toolkit (MITK) software. 2D-images revealed that 1 h PFF changed cell morphology from polygonal to triangular, and nucleus morphology from round to ellipsoid. PFF also reduced cell surface area (0.3-fold), cell volume (0.3-fold), and nucleus volume (0.2-fold). During PFF, the live-cell volume gradually decreased from 6000 to 3000 µm3. After PFF, α-tubulin orientation was more disorganized, but F-actin fluorescence intensity was enhanced, particularly around the nucleus. 3D-images obtained from Z-stacks indicated that PFF increased F-actin fluorescence signal distribution around the nucleus in the XZ and YZ direction (2.3-fold). PFF increased protein expression of phospho-paxillin (2.0-fold) and integrin-α5 (2.8-fold), but did not increase mRNA expression of paxillin-a (PXNA), paxillin-b (PXNB), integrin-α5 (ITGA51), or α-tubulin protein expression. In conclusion, PFF induced substantial changes in osteoblast cytoskeleton, as well as cell and nucleus morphology and volume, which was accompanied by elevated gene and protein expression of adhesion and structural proteins. More insights into the mechanisms whereby mechanical cues drive morphological changes in bone cells, and thereby, possibly in bone cell behavior, will aid the guidance of clinical treatment, particularly in the field of orthodontics, (oral) implantology, and orthopedics.


Subject(s)
Cell Nucleus/physiology , Mechanotransduction, Cellular/physiology , Osteoblasts/metabolism , 3T3 Cells , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Mice , Osteoblasts/physiology , Osteocytes/metabolism , RNA, Messenger/genetics , Shear Strength/physiology , Signal Transduction/physiology , Stress, Mechanical
9.
J Cell Physiol ; 234(11): 20520-20532, 2019 11.
Article in English | MEDLINE | ID: mdl-31016754

ABSTRACT

Fracture repair is characterized by cytokine production and hypoxia. To better predict cytokine modulation of mesenchymal stem cell (MSC)-aided bone healing, we investigated whether interleukin 4 (IL-4), IL-6, and their combination, affect osteogenic differentiation, vascular endothelial growth factor (VEGF) production, and/or mammalian target of rapamycin complex 1 (mTORC1) activation by MSCs under normoxia or hypoxia. Human adipose stem cells (hASCs) were cultured with IL-4, IL-6, or their combination for 3 days under normoxia (20% O 2 ) or hypoxia (1% O 2 ), followed by 11 days without cytokines under normoxia or hypoxia. Hypoxia did not alter IL-4 or IL-6-modulated gene or protein expression by hASCs. IL-4 alone decreased runt-related transcription factor 2 (RUNX2) and collagen type 1 (COL1) gene expression, alkaline phosphatase (ALP) activity, and VEGF protein production by hASCs under normoxia and hypoxia, and decreased mineralization of hASCs under hypoxia. In contrast, IL-6 increased mineralization of hASCs under normoxia, and enhanced RUNX2 gene expression under normoxia and hypoxia. Neither IL-4 nor IL-6 affected phosphorylation of the mTORC1 effector protein P70S6K. IL-4 combined with IL-6 diminished the inhibitory effect of IL-4 on ALP activity, bone nodule formation, and VEGF production, and decreased RUNX2 and COL1 expression, similar to IL-4 alone, under normoxia and hypoxia. In conclusion, IL-4 alone, but not in combination with IL-6, inhibits osteogenic differentiation and angiogenic stimulation potential of hASCs under normoxia and hypoxia, likely through pathways other than mTORC1. These results indicate that cytokines may differentially affect bone healing and regeneration when applied in isolation or in combination.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/drug effects , Interleukin-4/pharmacology , Interleukin-6/pharmacology , Osteogenesis/drug effects , Stem Cells/drug effects , Stem Cells/physiology , Adult , Bone Development/drug effects , Cell Differentiation/physiology , Cell Proliferation , Female , Gene Expression Regulation/drug effects , Humans , Middle Aged , Osteogenesis/physiology , Oxygen , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
10.
Curr Osteoporos Rep ; 17(4): 207-216, 2019 08.
Article in English | MEDLINE | ID: mdl-31240566

ABSTRACT

PURPOSE OF REVIEW: Osteocytes are responsible for mechanosensing and mechanotransduction in bone and play a crucial role in bone homeostasis. They are embedded in a calcified collagenous matrix and connected with each other through the lacuno-canalicular network. Due to this specific native environment, it is a challenge to isolate primary osteocytes without losing their specific characteristics in vitro. This review summarizes the commonly used and recently established models to study the function of osteocytes in vitro. RECENT FINDINGS: Osteocytes are mostly studied in monolayer culture, but recently, 3D models of osteocyte-like cells and primary osteocytes in vitro have been established as well. These models mimic the native environment of osteocytes and show superior osteocyte morphology and behavior, enabling the development of human disease models. Osteocyte-like cell lines as well as primary osteocytes isolated from bone are widely used to study the role of osteocytes in bone homeostasis. Both cells lines and primary cells are cultured in 2D-monolayer and 3D-models. The use of these models and their advantages and shortcomings are discussed in this review.


Subject(s)
Bone Matrix , In Vitro Techniques , Osteocytes , Cell Line , Culture Techniques , Humans , Primary Cell Culture
11.
Curr Osteoporos Rep ; 17(5): 235-249, 2019 10.
Article in English | MEDLINE | ID: mdl-31428977

ABSTRACT

PURPOSE OF REVIEW: Bone and muscle mass increase in response to mechanical loading and biochemical cues. Bone-forming osteoblasts differentiate into early osteocytes which ultimately mature into late osteocytes encapsulated in stiff calcified matrix. Increased muscle mass originates from muscle stem cells (MuSCs) enclosed between their plasma membrane and basal lamina. Stem cell fate and function are strongly determined by physical and chemical properties of their microenvironment, i.e., the cell niche. RECENT FINDINGS: The cellular niche is a three-dimensional structure consisting of extracellular matrix components, signaling molecules, and/or other cells. Via mechanical interaction with their niche, osteocytes and MuSCs are subjected to mechanical loads causing deformations of membrane, cytoskeleton, and/or nucleus, which elicit biochemical responses and secretion of signaling molecules into the niche. The latter may modulate metabolism, morphology, and mechanosensitivity of the secreting cells, or signal to neighboring cells and cells at a distance. Little is known about how mechanical loading of bone and muscle tissue affects osteocytes and MuSCs within their niches. This review provides an overview of physicochemical niche conditions of (early) osteocytes and MuSCs and how these are sensed and determine cell fate and function. Moreover, we discuss how state-of-the-art imaging techniques may enhance our understanding of these conditions and mechanisms.


Subject(s)
Mechanotransduction, Cellular/physiology , Muscle Cells/physiology , Osteocytes/physiology , Animals , Cell Differentiation , Extracellular Matrix , Humans , Stress, Mechanical
12.
Calcif Tissue Int ; 103(6): 675-685, 2018 12.
Article in English | MEDLINE | ID: mdl-30109376

ABSTRACT

Hormonal changes during lactation are associated with profound changes in bone cell biology, such as osteocytic osteolysis, resulting in larger lacunae. Larger lacuna shape theoretically enhances the transmission of mechanical signals to osteocytes. We aimed to provide experimental evidence supporting this theory by comparing the mechanoresponse of osteocytes in the bone of lactating mice, which have enlarged lacunae due to osteocytic osteolysis, with the response of osteocytes in bone from age-matched virgin mice. The osteocyte mechanoresponse was measured in excised fibulae that were cultured in hormone-free medium for 24 h and cyclically loaded for 10 min (sinusoidal compressive load, 3000 µÎµ, 5 Hz) by quantifying loading-related changes in Sost mRNA expression (qPCR) and sclerostin and ß-catenin protein expression (immunohistochemistry). Loading decreased Sost expression by ~ threefold in fibulae of lactating mice. The loading-induced decrease in sclerostin protein expression by osteocytes was larger in lactating mice (55% decrease ± 14 (± SD), n = 8) than virgin mice (33% decrease ± 15, n = 7). Mechanical loading upregulated ß-catenin expression in osteocytes in lactating mice by 3.5-fold (± 0.2, n = 6) which is significantly (p < 0.01) higher than the 1.6-fold increase in ß-catenin expression by osteocytes in fibulae from virgin mice (± 0.12, n = 4). These results suggest that osteocytes in fibulae from lactating mice with large lacunae may respond stronger to mechanical loading than those from virgin mice. This could indicate that osteocytes residing in larger lacuna show a stronger response to mechanical loading.


Subject(s)
Bone Remodeling/physiology , Fibula/physiology , Lactation/physiology , Mechanotransduction, Cellular/physiology , Osteocytes/physiology , Animals , Female , Fibula/cytology , Mice , Mice, Inbred C57BL , Osteocytes/cytology , Stress, Mechanical
13.
Clin Oral Implants Res ; 29(5): 488-498, 2018 May.
Article in English | MEDLINE | ID: mdl-29638012

ABSTRACT

OBJECTIVES: Synthetic biphasic calcium phosphate (BCP) with a hydroxyapatite/ß-tricalcium phosphate (HA/ß-TCP) ratio of 60/40 (BCP60/40) is successfully used as alternative for autologous bone in patients undergoing maxillary sinus floor elevation (MSFE) for dental implant placement. A high percentage of HA in BCP60/40 may hamper efficient scaffold remodeling. Osteogenesis and neovascularization are pivotal in effective bone regeneration. We aimed to investigate whether differences exist in osteogenic and/or vasculogenic potential of BCP60/40 and BCP20/80 in patients undergoing MSFE. MATERIALS AND METHODS: Twenty patients undergoing MSFE were treated with BCP60/40 (n = 10) or BCP20/80 (n = 10). Bone and graft volumes were determined by micro-computed tomography and histomorphometrical analysis of biopsies of the augmented region. Osteoid volumes, number of osteoclasts, and blood vessels were determined by histomorphometrical analysis. The biopsies were taken 6.5 months (26 weeks) postoperatively prior to dental implant placement. RESULTS: Bone and osteoid volumes were 9.7% and 0.8% higher at the most cranial side of the BCP20/80 biopsies compared to the BCP60/40 biopsies. Graft volumes, number of osteoclasts, and blood vessels were similar in both groups. CONCLUSIONS: BCP20/80 showed enhanced osteogenic potential in patients undergoing MSFE compared to BCP60/40, due to either a faster bone remodeling rate or an earlier start of bone formation in BCP20/80-treated patients, suggesting that a higher TCP content positively contributes to the bone remodeling rate. Therefore, BCP20/80 might perform better, at least in the short term, as a scaffold for bone augmentation in the MSFE model than BCP60/40 as more bone is formed, and more osteoid is deposited at the cranial side in BCP20/80-treated patients compared to BCP60/40-treated patients. However, catch-up of BCP60/40 in the long term cannot be ruled out.


Subject(s)
Alveolar Process/diagnostic imaging , Bone Substitutes/therapeutic use , Hydroxyapatites/therapeutic use , Sinus Floor Augmentation/methods , Alveolar Process/pathology , Alveolar Process/surgery , Female , Humans , Male , Middle Aged , Radiography, Dental , Tissue Scaffolds , X-Ray Microtomography
14.
Curr Osteoporos Rep ; 15(5): 401-411, 2017 10.
Article in English | MEDLINE | ID: mdl-28891009

ABSTRACT

PURPOSE OF REVIEW: The bone is able to adapt its structure to mechanical signals via the bone remodeling process governed by mechanosensitive osteocytes. With aging, an imbalance in bone remodeling results in osteoporosis. In this review, we hypothesized that changes in lacunar morphology underlie the decreased bone mechanoresponsiveness to mechanical loading with aging. RECENT FINDINGS: Several studies have reported considerable variations in the shape of osteocytes and their lacunae with aging. Since osteocytes can sense matrix strain directly via their cell bodies, the variations in osteocyte morphology may cause changes in osteocyte mechanosensitivity. As a consequence, the load-adaptive response of osteocytes may change with aging, even when mechanical loading would remain unchanged. Though extensive quantitative data is lacking, evidence exists that the osteocyte lacunae are becoming smaller and more spherical with aging. Future dedicated studies might reveal whether these changes would affect osteocyte mechanosensation and the subsequent biological response, and whether this is (one of) the pathways involved in age-related bone loss.


Subject(s)
Aging/physiology , Bone Remodeling/physiology , Mechanotransduction, Cellular/physiology , Osteocytes/physiology , Osteoporosis/physiopathology , Stress, Mechanical , Humans
15.
Curr Osteoporos Rep ; 15(6): 555-563, 2017 12.
Article in English | MEDLINE | ID: mdl-29098573

ABSTRACT

PURPOSE OF REVIEW: Multiple dietary components have the potential to positively affect bone mineral density in early life and reduce loss of bone mass with aging. In addition, regular weight-bearing physical activity has a strong positive effect on bone through activation of osteocyte signaling. We will explore possible synergistic effects of dietary components and mechanical stimuli for bone health by identifying dietary components that have the potential to alter the response of osteocytes to mechanical loading. RECENT FINDINGS: Several (sub)cellular aspects of osteocytes determine their signaling towards osteoblasts and osteoclasts in response to mechanical stimuli, such as the osteocyte cytoskeleton, estrogen receptor α, the vitamin D receptor, and the architecture of the lacunocanalicular system. Potential modulators of these features include 1,25-dihydroxy vitamin D3, several forms of vitamin K, and the phytoestrogen genistein. Multiple dietary components potentially affect osteocyte function and therefore may have a synergistic effect on bone health when combined with a regime of physical activity.


Subject(s)
Bone Density/physiology , Bone and Bones/physiology , Diet , Exercise/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Osteocytes/physiology , Resistance Training , Weight-Bearing/physiology , Bone and Bones/metabolism , Calcitriol , Estrogen Receptor alpha/metabolism , Genistein , Humans , Mechanotransduction, Cellular , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteocytes/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction , Vitamin K
16.
Artif Organs ; 41(6): 556-567, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27418522

ABSTRACT

Stability and antithrombotic functionality of endothelial cells on silicone hollow fibers (SiHFs) are critical in the development of biohybrid artificial lungs. Here we aimed to enhance endothelial cell retention and anti-thrombotic function by low (12 dyn/cm2 , 24 h) fluid shear stress ("flow") preconditioning of endothelial cells seeded on collagen-immobilized SiHFs. The response of endothelial cells without preconditioning (48 h static culture) and with preconditioning (24 h static culture followed by 24 h flow preconditioning) on hollow fibers to high fluid shear stress (30 dyn/cm2 , 1 h) was assessed in a parallel-plate flow chamber. Finite element (FE) modeling was used to simulate shear stress within the flow chamber. We found that collagen immobilization on hollow fibers using carbodiimide bonds provided sufficient stability to high shear stress. Flow preconditioning for 24 h before treatment with high shear stress for 1 h on collagen-immobilized hollow fibers increased cell retention (1.3-fold). The FE model showed that cell flattening due to flow preconditioning reduced maximum shear stress on cells by 32%. Flow preconditioning prior to exposure to high fluid shear stress enhanced the production of nitric oxide (1.3-fold) and prostaglandin I2 (1.2-fold). In conclusion, flow preconditioning of endothelial cells on collagen-immobilized SiHFs enhanced cell retention and antithrombotic function, which could significantly improve current biohybrid artificial lungs.


Subject(s)
Bioartificial Organs , Coated Materials, Biocompatible/chemistry , Collagen/chemistry , Endothelial Cells/cytology , Silicones/chemistry , Tissue Engineering/instrumentation , Cell Adhesion , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Equipment Design , Human Umbilical Vein Endothelial Cells , Humans , Hydrodynamics , Immobilized Proteins/chemistry , Lung/blood supply , Lung/cytology , Lung/physiology , Materials Testing , Nitric Oxide/metabolism , Prostaglandins/metabolism , Stress, Mechanical , Tissue Engineering/methods
17.
J Cell Physiol ; 231(6): 1283-90, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26505782

ABSTRACT

Insulin-like growth factor-1 (IGF-1) is anabolic for muscle by enhancing the rate of mRNA translation via activation of AKT and subsequent activation of the mammalian target of rapamycin complex 1 (mTOR), thereby increasing cellular protein production. IGF-1 is also anabolic for bone, but whether the mTOR pathway plays a role in the rate of bone matrix protein production by osteoblasts is unknown. We hypothesized that anabolic stimuli such as mechanical loading and IGF-1 stimulate protein synthesis in osteoblasts via activation of the AKT-mTOR pathway. MC3T3-E1 osteoblasts were either or not subjected for 1 h to mechanical loading by pulsating fluid flow (PFF) or treated with or without human recombinant IGF-1 (1-100 ng/ml) for 0.5-6 h, to determine phosphorylation of AKT and p70S6K (downstream of mTOR) by Western blot. After 4 days of culture with or without the mTOR inhibitor rapamycin, total protein, DNA, and gene expression were quantified. IGF-1 (100 ng/ml) reduced IGF-1 gene expression, although PFF enhanced IGF-1 expression. IGF-1 did not affect collagen-I gene expression. IGF-1 dose-dependently enhanced AKT and p70S6K phosphorylation at 2 and 6 h. PFF enhanced phosphorylation of AKT and p70S6K already within 1 h. Both IGF-1 and PFF enhanced total protein per cell by ∼30%, but not in the presence of rapamycin. Our results show that IGF-1 and PFF activate mTOR, thereby stimulating the rate of mRNA translation in osteoblasts. The known anabolic effect of mechanical loading and IGF-1 on bone may thus be partly explained by mTOR-mediated enhanced protein synthesis in osteoblasts.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Mechanotransduction, Cellular , Osteoblasts/drug effects , Protein Biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , 3T3 Cells , Animals , Cell Culture Techniques , Dose-Response Relationship, Drug , Enzyme Activation , Mice , Osteoblasts/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Physical Stimulation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pulsatile Flow , RNA, Messenger/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Time Factors
18.
N Engl J Med ; 369(16): 1529-36, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24088043

ABSTRACT

Plastin 3 (PLS3), a protein involved in the formation of filamentous actin (F-actin) bundles, appears to be important in human bone health, on the basis of pathogenic variants in PLS3 in five families with X-linked osteoporosis and osteoporotic fractures that we report here. The bone-regulatory properties of PLS3 were supported by in vivo analyses in zebrafish. Furthermore, in an additional five families (described in less detail) referred for diagnosis or ruling out of osteogenesis imperfecta type I, a rare variant (rs140121121) in PLS3 was found. This variant was also associated with a risk of fracture among elderly heterozygous women that was two times as high as that among noncarriers, which indicates that genetic variation in PLS3 is a novel etiologic factor involved in common, multi-factorial osteoporosis.


Subject(s)
Fractures, Bone/genetics , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Osteoporosis/genetics , Adult , Animals , Bone Density/genetics , Bone Remodeling/genetics , Child , Child, Preschool , Female , Fractures, Bone/etiology , Genetic Diseases, X-Linked/genetics , Heterozygote , Humans , Male , Mutation , Osteoporosis/complications , Pedigree , Polymorphism, Single Nucleotide , Risk Factors , Young Adult , Zebrafish
19.
Calcif Tissue Int ; 98(6): 596-608, 2016 06.
Article in English | MEDLINE | ID: mdl-26887974

ABSTRACT

Bone remodeling can be disturbed in active rheumatoid arthritis (RA), possibly as a result of elevated levels of circulating inflammatory cytokines. Osteocyte-specific proteins and cytokines play a vital role in bone remodeling by orchestrating bone formation and/or bone resorption. Therefore, we aimed to investigate the effect of RA-serum or inflammatory cytokines on expression of human osteocyte-specific proteins and cytokines. Human trabecular bone chips were cultured with RA-serum or inflammatory cytokines for 7-days. Live-dead staining was performed to assess cell viability. Gene expression of osteocyte-specific proteins and cytokines was analyzed by qPCR. Immuno-staining was performed for osteocyte-specific markers. Approximately 60 % of the osteocytes on the bone chips were alive at day-7. Cells in or on the bone chips did express the gene for osteocyte markers SOST, FGF23, DMP1, and MEPE, and the cytokines IL-1ß, IL-6, and TNFα at day 0 and 7. Active RA-serum treatment enhanced IL-1ß, TNFα, SOST, and DKK1 gene expression. IL-1ß treatment enhanced IL-1ß, TNFα, IL-6, IL-8, FGF23, and SOST gene expression. TNFα treatment enhanced IL-1ß, TNFα, IL-6, IL-8, and FGF23 gene expression. IL-8 treatment enhanced TNFα, IL-8, and FGF23 gene expression. A combination of IL-1ß, IL-6, and TNFα treatment synergistically upregulated IL-1ß, IL-6, and IL-8 gene expression, as well as enhanced TNFα, OPG, SOST, and FGF23, and inhibited DKK1 gene expression. In conclusion, gene expression of human osteocyte-specific proteins and cytokines was affected by RA-serum, and exogenous recombinant cytokines treatment suggesting that osteocytes could provide a new target to prevent systemic inflammation-induced bone loss in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cytokines/metabolism , Inflammation/metabolism , Osteocytes/metabolism , Adult , Aged , Aged, 80 and over , Cells, Cultured , Female , Fibroblast Growth Factor-23 , Gene Expression , Humans , Immunohistochemistry , Male , Middle Aged , Real-Time Polymerase Chain Reaction
20.
Connect Tissue Res ; 57(3): 226-35, 2016 05.
Article in English | MEDLINE | ID: mdl-27050327

ABSTRACT

Delayed fracture healing is frequently experienced in patients with systemic inflammation such as during rheumatoid arthritis (RA). The reasons for this are diverse, but could also be caused by inflammatory cytokines and/or growth factors in serum from patients with active disease. We hypothesized that serum from patients with active RA contains circulating inflammatory factors that inhibit differentiation of osteochondrogenic precursors. Serum was obtained from 15 patients with active RA (active RA-sera) and from the same patients in clinical remission 1 year later (remission RA-sera; controls). The effect of active RA-sera on osteochondrogenic differentiation of chondrogenic ATDC5 cells and primary human periosteum-derived progenitor cells (HPDC) was determined in micromass culture. In ATDC5 cells, active RA-sera reduced Ki67 transcription levels by 40% and cartilage matrix accumulation by 14% at day 14, and Alp transcription levels by 16%, and matrix mineralization by 17% at day 21 compared with remission RA-sera. In HPDCs, active RA-sera inhibited metabolic activity by 8%, SOX9 transcription levels by 14%, and cartilage matrix accumulation by 7% at day 7 compared with remission RA-sera. In conclusion, sera from patients with active RA negatively affect differentiation of osteochondrogenic precursors, and as a consequence may contribute to delayed fracture healing in these patients.


Subject(s)
Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology , Cell Differentiation , Chondrogenesis , Osteogenesis , Serum/metabolism , Stem Cells/pathology , Biomarkers/metabolism , Calcification, Physiologic , Cartilage/metabolism , Cell Aggregation , Cell Line , Extracellular Matrix/metabolism , Female , Humans , Male , Middle Aged , Periosteum/pathology
SELECTION OF CITATIONS
SEARCH DETAIL