Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(12): e0261660, 2021.
Article in English | MEDLINE | ID: mdl-34941939

ABSTRACT

Earlier, we proposed the "mechanosome" concept as a testable model for understanding how mechanical stimuli detected by cell surface adhesion molecules are transmitted to modulate gene expression inside cells. Here, for the first time we document a putative mechanosome involving Src, Pyk2 and MBD2 in MLO-Y4 osteocytes with high spatial resolution using FRET-FLIM. Src-Pyk2 complexes were concentrated at the periphery of focal adhesions and the peri-nuclear region. Pyk2-MBD2 complexes were located primarily in the nucleus and peri-nuclear region. Lifetime measurements indicated that Src and MBD2 did not interact directly. Finally, mechanical stimulation by fluid flow induced apparent accumulation of Src-Pyk2 protein complexes in the peri-nuclear/nuclear region, consistent with the proposed behavior of a mechanosome in response to a mechanical stimulus.


Subject(s)
DNA-Binding Proteins/metabolism , Focal Adhesion Kinase 2/metabolism , Osteocytes/metabolism , src-Family Kinases/metabolism , Animals , Cell Line , DNA-Binding Proteins/analysis , Fluorescence Resonance Energy Transfer , Focal Adhesion Kinase 2/analysis , Focal Adhesions/metabolism , Mechanotransduction, Cellular , Mice , Osteocytes/cytology , src-Family Kinases/analysis
2.
Biophys J ; 118(8): 1820-1829, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32191861

ABSTRACT

We report the use of pulsed interleaved excitation (PIE)-fluorescence lifetime imaging microscopy (FLIM) to measure the activities of two different biosensor probes simultaneously in single living cells. Many genetically encoded biosensors rely on the measurement of Förster resonance energy transfer (FRET) to detect changes in biosensor conformation that accompany the targeted cell signaling event. One of the most robust ways of quantifying FRET is to measure changes in the fluorescence lifetime of the donor fluorophore using FLIM. The study of complex signaling networks in living cells demands the ability to track more than one of these cellular events at the same time. Here, we demonstrate how PIE-FLIM can separate and quantify the signals from different FRET-based biosensors to simultaneously measure changes in the activity of two cell signaling pathways in the same living cells in tissues. The imaging system described here uses selectable laser wavelengths and synchronized detection gating that can be tailored and optimized for each FRET pair. Proof-of-principle studies showing simultaneous measurement of cytosolic calcium and protein kinase A activity are shown, but the PIE-FLIM approach is broadly applicable to other signaling pathways.


Subject(s)
Biosensing Techniques , Fluorescence Resonance Energy Transfer , Fluorescent Dyes , Light , Microscopy, Fluorescence
3.
iScience ; 20: 205-215, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31585407

ABSTRACT

Wnt signaling plays a key role in regulating bone remodeling. In vitro studies suggest that sclerostin's inhibitory action on Lrp5 is facilitated by the membrane-associated receptor Lrp4. We generated an Lrp4 R1170W knockin mouse model (Lrp4KI), based on a published mutation in patients with high bone mass (HBM). Lrp4KI mice have an HBM phenotype (assessed radiographically), including increased bone strength and formation. Overexpression of a Sost transgene had osteopenic effects in Lrp4-WT but not Lrp4KI mice. Conversely, sclerostin inhibition had blunted osteoanabolic effects in Lrp4KI mice. In a disuse-induced bone wasting model, Lrp4KI mice exhibit significantly less bone loss than wild-type (WT) mice. In summary, mice harboring the Lrp4-R1170W missense mutation recapitulate the human HBM phenotype, are less sensitive to altered sclerostin levels, and are protected from disuse-induced bone loss. Lrp4 is an attractive target for pharmacological targeting aimed at increasing bone mass and preventing bone loss due to disuse.

4.
J Bone Miner Res ; 34(10): 1964-1975, 2019 10.
Article in English | MEDLINE | ID: mdl-31173667

ABSTRACT

Mechanical stimulation is a key regulator of bone mass, maintenance, and turnover. Wnt signaling is a key regulator of mechanotransduction in bone, but the role of ß-catenin-an intracellular signaling node in the canonical Wnt pathway-in disuse mechanotransduction is not defined. Using the ß-catenin exon 3 flox (constitutively active [CA]) mouse model, in conjunction with a tamoxifen-inducible, osteocyte-selective Cre driver, we evaluated the effects of degradation-resistant ß-catenin on bone properties during disuse. We hypothesized that if ß-catenin plays an important role in Wnt-mediated osteoprotection, then artificial stabilization of ß-catenin in osteocytes would protect the limbs from disuse-induced bone wasting. Two disuse models were tested: tail suspension, which models fluid shift, and botulinum-toxin (botox)-induced muscle paralysis, which models loss of muscle force. Tail suspension was associated with a significant loss of tibial bone mass and density, reduced architectural properties, and decreased bone formation indices in uninduced (control) mice, as assessed by dual-energy X-ray absorptiometry (DXA), micro-computed tomography (µCT), and histomorphometry. Activation of the ßcatCA allele in tail-suspended mice resulted in little to no change in those properties; ie, these mice were protected from bone loss. Similar protective effects were observed among botox-treated mice when the ßcatCA was activated. RNAseq analysis of altered gene regulation in tail-suspended mice yielded 35 genes, including Wnt11, Gli1, Nell1, Gdf5, and Pgf, which were significantly differentially regulated between tail-suspended ß-catenin stabilized mice and tail-suspended nonstabilized mice. Our findings indicate that selectively targeting/blocking of ß-catenin degradation in bone cells could have therapeutic implications in mechanically induced bone disease. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Mechanotransduction, Cellular , Osteocytes/metabolism , Osteogenesis , Tibia/metabolism , beta Catenin/metabolism , Animals , Bone Density , Mice , Mice, Transgenic , Osteocytes/pathology , Tibia/diagnostic imaging , Tibia/pathology , X-Ray Microtomography , beta Catenin/genetics
5.
Phytother Res ; 30(5): 790-6, 2016 May.
Article in English | MEDLINE | ID: mdl-26806653

ABSTRACT

Hydroxy-safflower yellow A (HSYA) is the major active component of safflower, a traditional Asia herbal medicine well known for its cardiovascular protective activities. The purpose of this study was to investigate the effect of HSYA on TNF-α-induced inflammatory responses in arterial endothelial cells (AECs) and to explore the mechanisms involved. The results showed that HSYA suppressed the up-regulation of ICAM-1 expression in TNF-α-stimulated AECs in a dose-dependent manner. High concentration (120 µM) HSYA significantly inhibited the TNF-α-induced adhesion of RAW264.7 cells to AECs. HSYA blocked the TNFR1-mediated phosphorylation and degradation of IκBα and also prevented the nuclear translocation of NF-κB p65. Moreover, HSYA reduced the cell surface level of TNFR1 and increased the content of sTNFR1 in the culture media. TNF-α processing inhibitor-0 (TAPI-0) prevented the HSYA inhibition of TNFR1-induced IκBα degradation, implying the occurrence of TNFR1 shedding. Furthermore, HSYA induced phosphorylation of TNF-α converting enzyme (TACE) at threonine 735, which is thought to be required for its activation. Conclusively, HSYA suppressed TNF-α-induced inflammatory responses in AECs, at least in part by inhibiting the TNFR1-mediated classical NF-κB pathway. TACE-mediated TNFR1 shedding can be involved in this effect. Our study provides new evidence for the antiinflammatory and anti-atherosclerotic effects of HSYA. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Chalcone/analogs & derivatives , Herbal Medicine/methods , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Chalcone/chemistry , Humans
6.
Mol Endocrinol ; 29(9): 1269-85, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26244796

ABSTRACT

PTH is an osteoanabolic for treating osteoporosis but its potency wanes. Disabling the transcription factor nuclear matrix protein 4 (Nmp4) in healthy, ovary-intact mice enhances bone response to PTH and bone morphogenetic protein 2 and protects from unloading-induced osteopenia. These Nmp4(-/-) mice exhibit expanded bone marrow populations of osteoprogenitors and supporting CD8(+) T cells. To determine whether the Nmp4(-/-) phenotype persists in an osteoporosis model we compared PTH response in ovariectomized (ovx) wild-type (WT) and Nmp4(-/-) mice. To identify potential Nmp4 target genes, we performed bioinformatic/pathway profiling on Nmp4 chromatin immunoprecipitation sequencing (ChIP-seq) data. Mice (12 w) were ovx or sham operated 4 weeks before the initiation of PTH therapy. Skeletal phenotype analysis included microcomputed tomography, histomorphometry, serum profiles, fluorescence-activated cell sorting and the growth/mineralization of cultured WT and Nmp4(-/-) bone marrow mesenchymal stem progenitor cells (MSPCs). ChIP-seq data were derived using MC3T3-E1 preosteoblasts, murine embryonic stem cells, and 2 blood cell lines. Ovx Nmp4(-/-) mice exhibited an improved response to PTH coupled with elevated numbers of osteoprogenitors and CD8(+) T cells, but were not protected from ovx-induced bone loss. Cultured Nmp4(-/-) MSPCs displayed enhanced proliferation and accelerated mineralization. ChIP-seq/gene ontology analyses identified target genes likely under Nmp4 control as enriched for negative regulators of biosynthetic processes. Interrogation of mRNA transcripts in nondifferentiating and osteogenic differentiating WT and Nmp4(-/-) MSPCs was performed on 90 Nmp4 target genes and differentiation markers. These data suggest that Nmp4 suppresses bone anabolism, in part, by regulating IGF-binding protein expression. Changes in Nmp4 status may lead to improvements in osteoprogenitor response to therapeutic cues.


Subject(s)
Bone Resorption/drug therapy , CD8-Positive T-Lymphocytes/cytology , Nuclear Matrix-Associated Proteins/genetics , Osteoporosis/drug therapy , Parathyroid Hormone/therapeutic use , Transcription Factors/genetics , Animals , Bone Density/drug effects , Bone Diseases, Metabolic/prevention & control , Bone Morphogenetic Protein 2/metabolism , Bone Resorption/genetics , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Chromosome Mapping , Embryonic Stem Cells/cytology , Female , Genetic Therapy , Humans , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis/drug effects , Osteoporosis/genetics , Ovariectomy , Ovary/surgery
7.
J Cell Physiol ; 230(3): 578-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25160801

ABSTRACT

Recent studies suggest that megakaryocytes (MKs) may play a significant role in skeletal homeostasis, as evident by the occurrence of osteosclerosis in multiple MK related diseases (Lennert et al., 1975; Thiele et al., 1999; Chagraoui et al., 2006). We previously reported a novel interaction whereby MKs enhanced proliferation of osteoblast lineage/osteoprogenitor cells (OBs) by a mechanism requiring direct cell-cell contact. However, the signal transduction pathways and the downstream effector molecules involved in this process have not been characterized. Here we show that MKs contact with OBs, via beta1 integrin, activate the p38/MAPKAPK2/p90RSK kinase cascade in the bone cells, which causes Mdm2 to neutralizes p53/Rb-mediated check point and allows progression through the G1/S. Interestingly, activation of MAPK (ERK1/2) and AKT, collateral pathways that regulate the cell cycle, remained unchanged with MK stimulation of OBs. The MK-to-OB signaling ultimately results in significant increases in the expression of c-fos and cyclin A, necessary for sustaining the OB proliferation. Overall, our findings show that OBs respond to the presence of MKs, in part, via an integrin-mediated signaling mechanism, activating a novel response axis that de-represses cell cycle activity. Understanding the mechanisms by which MKs enhance OB proliferation will facilitate the development of novel anabolic therapies to treat bone loss associated with osteoporosis and other bone-related diseases.


Subject(s)
Cell Differentiation/genetics , Megakaryocytes/cytology , Osteoblasts/cytology , Signal Transduction/genetics , Cell Cycle/genetics , Cell Lineage , Cell Proliferation/genetics , Cells, Cultured , Humans , MAP Kinase Signaling System/genetics , Megakaryocytes/metabolism , Osteoblasts/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism
8.
PLoS One ; 9(5): e97942, 2014.
Article in English | MEDLINE | ID: mdl-24841674

ABSTRACT

Mechanical stimulation of the skeleton promotes bone gain and suppresses bone loss, ultimately resulting in improved bone strength and fracture resistance. The molecular mechanisms directing anabolic and/or anti-catabolic actions on the skeleton during loading are not fully understood. Identifying molecular mechanisms of mechanotransduction (MTD) signaling cascades could identify new therapeutic targets. Most research into MTD mechanisms is typically focused on understanding the signaling pathways that stimulate new bone formation in response to load. However, we investigated the structural, signaling and transcriptional molecules that suppress the stimulatory effects of loading. The high bone mass phenotype of mice with global deletion of either Pyk2 or Src suggests a role for these tyrosine kinases in repression of bone formation. We used fluid shear stress as a MTD stimulus to identify a novel Pyk2/Src-mediated MTD pathway that represses mechanically-induced bone formation. Our results suggest Pyk2 and Src function as molecular switches that inhibit MTD in our mechanically stimulated osteocyte culture experiments. Once activated by oscillatory fluid shear stress (OFSS), Pyk2 and Src translocate to and accumulate in the nucleus, where they associate with a protein involved in DNA methylation and the interpretation of DNA methylation patterns -methyl-CpG-binding domain protein 2 (MBD2). OFSS-induced Cox-2 and osteopontin expression was enhanced in Pyk2 KO osteoblasts, while inhibition of Src enhanced osteocalcin expression in response to OFSS. We found that Src kinase activity increased in the nucleus of osteocytes in response to OFSS and an interaction activated between Src (Y418) and Pyk2 (Y402) increased in response to OFSS. Thus, as a mechanism to prevent an over-reaction to physical stimulation, mechanical loading may induce the formation of a Src/Pyk2/MBD2 complex in the nucleus that functions to suppress anabolic gene expression.


Subject(s)
Gene Expression Regulation/physiology , Mechanotransduction, Cellular/physiology , Multiprotein Complexes/metabolism , Osteocytes/physiology , Stress, Mechanical , Animals , Anthracenes , Blotting, Western , DNA Methylation/genetics , DNA Primers/genetics , DNA-Binding Proteins/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique , Focal Adhesion Kinase 2/metabolism , Mice , Multiprotein Complexes/biosynthesis , Osteocytes/metabolism , Real-Time Polymerase Chain Reaction , Shear Strength , src-Family Kinases/metabolism
9.
Am J Physiol Endocrinol Metab ; 306(8): E937-44, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24619882

ABSTRACT

Mechanical loading is an important regulator in skeletal growth, maintenance, and aging. Estrogen receptors have a regulatory role in mechanically induced bone adaptation. Estrogen receptor-α (ERα) is known to enhance load-induced bone formation, whereas ERß negatively regulates this process. We hypothesized that ERß regulates mechanical signaling in osteoblasts. We tested this hypothesis by subjecting primary calvarial cells isolated from wild-type and ERß-knockout mice (BERKO) to oscillatory fluid flow in the absence or presence of estradiol (E2). We found that the known responses to fluid shear stress, i.e., phosphorylation of the mitogen-activated protein kinase ERK and upregulation of COX-2 expression, were inhibited in BERKO cells in the absence of E2. Flow-induced increase in prostaglandin E2 (PGE2) release was not altered in BERKO cells in the absence of E2, but was increased when E2 was present. Additionally, immunofluorescence analysis and estrogen response element luciferase assays revealed increased ERα expression and flow- and ligand-induced nuclear translocation as well as transcriptional activity in BERKO cells in both the presence and absence of E2. Taken together, these data suggest that ERß plays both ligand-dependent and ligand-independent roles in mechanical signaling in osteoblasts. Furthermore, our data suggest that one mechanism by which ERß regulates mechanotransduction in osteoblasts may result from its inhibitory effect on ERα expression and function. Targeting estrogen receptors (e.g., inhibiting ERß) may represent an effective approach for prevention and treatment of age-related bone loss.


Subject(s)
Estrogen Receptor beta/physiology , Mechanotransduction, Cellular/genetics , Osteoblasts/metabolism , Animals , Cells, Cultured , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Estradiol/pharmacology , MAP Kinase Signaling System/drug effects , Mechanotransduction, Cellular/drug effects , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/drug effects , Phosphorylation/drug effects , Primary Cell Culture , Skull/cytology
10.
J Bone Miner Res ; 28(6): 1434-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23362087

ABSTRACT

Preclinical and clinical evidence from megakaryocyte (MK)-related diseases suggests that MKs play a significant role in maintaining bone homeostasis. Findings from our laboratories reveal that MKs significantly increase osteoblast (OB) number through direct MK-OB contact and the activation of integrins. We, therefore, examined the role of Pyk2, a tyrosine kinase known to be regulated downstream of integrins, in the MK-mediated enhancement of OBs. When OBs were co-cultured with MKs, total Pyk2 levels in OBs were significantly enhanced primarily because of increased Pyk2 gene transcription. Additionally, p53 and Mdm2 were both decreased in OBs upon MK stimulation, which would be permissive of cell cycle entry. We then demonstrated that OB number was markedly reduced when Pyk2-/- OBs, as opposed to wild-type (WT) OBs, were co-cultured with MKs. We also determined that MKs inhibit OB differentiation in the presence and absence of Pyk2 expression. Finally, given that MK-replete spleen cells from GATA-1-deficient mice can robustly stimulate OB proliferation and bone formation in WT mice, we adoptively transferred spleen cells from these mice into Pyk2-/- recipient mice. Importantly, GATA-1-deficient spleen cells failed to stimulate an increase in bone formation in Pyk2-/- mice, suggesting in vivo the important role of Pyk2 in the MK-induced increase in bone volume. Further understanding of the signaling pathways involved in the MK-mediated enhancement of OB number and bone formation will facilitate the development of novel anabolic therapies to treat bone loss diseases.


Subject(s)
Cell Differentiation/physiology , Focal Adhesion Kinase 2/metabolism , Megakaryocytes/enzymology , Osteoblasts/enzymology , Osteogenesis/physiology , Animals , Cells, Cultured , Coculture Techniques , Focal Adhesion Kinase 2/genetics , Megakaryocytes/cytology , Mice , Mice, Knockout , Osteoblasts/cytology , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Int J Mol Sci ; 13(11): 14385-400, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23203070

ABSTRACT

Live-cell microscopy is now routinely used to monitor the activities of the genetically encoded biosensor proteins that are designed to directly measure specific cell signaling events inside cells, tissues, or organisms. Most fluorescent biosensor proteins rely on Förster resonance energy transfer (FRET) to report conformational changes in the protein that occur in response to signaling events, and this is commonly measured with intensity-based ratiometric imaging methods. An alternative method for monitoring the activities of the FRET-based biosensor proteins is fluorescence lifetime imaging microscopy (FLIM). FLIM measurements are made in the time domain, and are not affected by factors that commonly limit intensity measurements. In this review, we describe the use of the digital frequency domain (FD) FLIM method for the analysis of FRET signals. We illustrate the methods necessary for the calibration of the FD FLIM system, and demonstrate the analysis of data obtained from cells expressing "FRET standard" fusion proteins. We then use the FLIM-FRET approach to monitor the changes in activities of two different biosensor proteins in specific regions of single living cells. Importantly, the factors required for the accurate determination and reproducibility of lifetime measurements are described in detail.


Subject(s)
Biosensing Techniques , Cell Tracking/methods , Microscopy, Fluorescence/methods , Animals , Humans
12.
Crit Rev Eukaryot Gene Expr ; 22(3): 205-18, 2012.
Article in English | MEDLINE | ID: mdl-23140162

ABSTRACT

Chronic degenerative diseases are increasing with the aging U.S. population. One consequence of this phenomenon is the need for long-term osteoporosis therapies. Parathyroid hormone (PTH), the only FDA-approved treatment that adds bone to the aged skeleton, loses its potency within two years of initial treatment but the mechanism regulating its limited "anabolic window" is unknown. We have discovered that disabling the nucleocytoplasmic shuttling transcription factor nuclear matrix protein 4/cas interacting zinc finger protein (Nmp4/CIZ) in mice extends the PTH bone-forming capacity. Nmp4 was discovered during our search for nuclear matrix transcription factors that couple this hormone's impact on osteoblast cytoskeletal and nuclear organization with its anabolic capacity. CIZ was independently discovered as a protein that associates with the focal adhesion-associated mechanosensor p130Cas. The Nmp4/CIZ-knockout (KO) skeletal phenotype exhibits a modestly enhanced bone mineral density but manifests an exaggerated response to both PTH and to BMP2 and is resistant to disuse-induced bone loss. The cellular basis of the global Nmp4/CIZ-KO skeletal phenotype remains to be elucidated but may involve an expansion of the bone marrow osteoprogenitor population along with modestly enhanced osteoblast and osteoclast activities supporting anabolic bone turnover. As a shuttling Cys(2)His(2) zinc finger protein, Nmp4/CIZ acts as a repressive transcription factor perhaps associated with epigenetic remodeling complexes, but the functional significance of its interaction with p130Cas is not known. Despite numerous remaining questions, Nmp4/CIZ provides insights into how the anabolic window is regulated, and itself may provide an adjuvant therapy target for the treatment of osteoporosis by extending PTH anabolic efficacy.


Subject(s)
Bone and Bones/physiology , Nuclear Matrix-Associated Proteins/metabolism , Parathyroid Hormone/physiology , Transcription Factors/metabolism , Animals , Bone Density , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Adhesion , Crk-Associated Substrate Protein/genetics , Crk-Associated Substrate Protein/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Humans , Mice , Nuclear Matrix-Associated Proteins/genetics , Osteoblasts/metabolism , Osteoclasts/metabolism , Parathyroid Hormone/pharmacology , Phenotype , Transcription Factors/genetics , Zinc Fingers/genetics
13.
J Cell Physiol ; 227(5): 1873-82, 2012 May.
Article in English | MEDLINE | ID: mdl-21732358

ABSTRACT

Intermittent parathyroid hormone (PTH) adds new bone to the osteoporotic skeleton; the transcription factor Nmp4/CIZ represses PTH-induced bone formation in mice and as a consequence is a potential drug target for improving hormone clinical efficacy. To explore the impact of Nmp4/CIZ on osteoblast phenotype, we immortalized bone marrow stromal cells from wildtype (WT) and Nmp4-knockout (KO) mice using murine telomerase reverse transcriptase. Clonal lines were initially chosen based on their positive staining for alkaline phosphatase and capacity for mineralization. Disabling Nmp4/CIZ had no gross impact on osteoblast phenotype development. WT and KO clones exhibited identical sustained growth, reduced population doubling times, extended maintenance of the mature osteoblast phenotype, and competency for differentiating toward the osteoblast and adipocyte lineages. Additional screening of the immortalized cells for PTH-responsiveness permitted further studies with single WT and KO clones. We recently demonstrated that PTH-induced c-fos femoral mRNA expression is enhanced in Nmp4-KO mice and in the present study we observed that hormone stimulated either an equivalent or modestly enhanced increase in c-fos mRNA expression in both primary null and KO clone cells depending on PTH concentration. The null primary osteoblasts and KO clone cells exhibited a transiently enhanced response to bone morphogenetic protein 2 (BMP2). The clones exhibited lower and higher expressions of the PTH receptor (Pthr1) and the BMP2 receptor (Bmpr1a, Alk3), respectively, as compared to primary cells. These immortalized cell lines will provide a valuable tool for disentangling the complex functional roles underlying Nmp4/CIZ regulation of bone anabolism.


Subject(s)
Bone Marrow Cells/physiology , Nuclear Matrix-Associated Proteins/genetics , Osteoblasts/physiology , Stromal Cells/physiology , Telomerase/metabolism , Transcription Factors/genetics , Adipocytes/cytology , Adipocytes/physiology , Animals , Bone Marrow Cells/cytology , Bone Morphogenetic Protein 2/pharmacology , Cell Line , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Matrix-Associated Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Phenotype , Stromal Cells/cytology , Telomerase/genetics , Transcription Factors/metabolism
14.
PLoS One ; 6(1): e16026, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21283581

ABSTRACT

Mechanotransduction, the process by which cells convert external mechanical stimuli such as fluid shear stress (FSS) into biochemical changes, plays a critical role in maintenance of the skeleton. We have proposed that mechanical stimulation by FSS across the surfaces of bone cells results in formation of unique signaling complexes called mechanosomes that are launched from sites of adhesion with the extracellular matrix and with other bone cells [1]. Deformation of adhesion complexes at the cell membrane ultimately results in alteration of target gene expression. Recently, we reported that focal adhesion kinase (FAK) functions as a part of a mechanosome complex that is required for FSS-induced mechanotransduction in bone cells. This study extends this work to examine the role of a second member of the FAK family of non-receptor protein tyrosine kinases, proline-rich tyrosine kinase 2 (Pyk2), and determine its role during osteoblast mechanotransduction. We use osteoblasts harvested from mice as our model system in this study and compared the contributions of Pyk2 and FAK during FSS induced mechanotransduction in osteoblasts. We exposed Pyk2(+/+) and Pyk2(-/-) primary calvarial osteoblasts to short period of oscillatory fluid flow and analyzed downstream activation of ERK1/2, and expression of c-fos, cyclooxygenase-2 and osteopontin. Unlike FAK, Pyk2 was not required for fluid flow-induced mechanotransduction as there was no significant difference in the response of Pyk2(+/+) and Pyk2(-/-) osteoblasts to short periods of fluid flow (FF). In contrast, and as predicted, FAK(-/-) osteoblasts were unable to respond to FF. These data indicate that FAK and Pyk2 have distinct, non-redundant functions in launching mechanical signals during osteoblast mechanotransduction. Additionally, we compared two methods of generating FF in both cell types, oscillatory pump method and another orbital platform method. We determined that both methods of generating FF induced similar responses in both primary calvarial osteoblasts and immortalized calvarial osteoblasts.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 2/metabolism , Mechanotransduction, Cellular , Osteoblasts/metabolism , Stress, Mechanical , Animals , Cells, Cultured , Mice , Rheology , Skull
15.
J Cell Physiol ; 226(4): 1044-51, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20857415

ABSTRACT

Fluid shear stress protects cells from TNF-α-induced apoptosis. Oscillatory fluid shear stress (OFSS) is generally perceived as physiologically relevant biophysical signal for bone cells. Here we identify several cellular mechanisms responsible for mediating the protective effects of OFSS against TNF-α-induced apoptosis in vitro. We found that exposure of MC3T3-E1 osteoblast-like cells to as little as 5 min of OFSS suppressed TNF-α-induced activation of caspase-3, cleavage of PARP and phosphorylation of histone. In contrast, H(2)O(2)-induced apoptosis was not inhibited by OFSS suggesting that OFSS might not be protecting cells from TNF-α-induced apoptosis via stimulation of global pro-survival signaling pathways. In support of this speculation, OFSS inhibition of TNF-α-induced apoptosis was unaffected by inhibitors of several pro-survival signaling pathways including pI3-kinase (LY294002), MAPK/ERK kinase (PD98059 or U0126), intracellular Ca2+ release (U73122), NO production (L-NAME), or protein synthesis (cycloheximide) that were applied to cells during exposure to OFSS and during TNF-α treatment. However, TNF-α-induced phosphorylation and degradation of IκBα was blocked by pre-exposure of cells to OFSS suggesting a more specific effect of OFSS on TNF-α signaling. We therefore focused on the mechanism of OFSS regulation of TNF-receptor 1 (TNFR1) signaling and found that OFSS (1) reduced the amount of receptor on the cell surface, (2) prevented the association of ubiquitinated RIP in TNFR1 complexes with TRADD and TRAF2, and (3) reduced TNF-α-induced IL-8 promoter activity in the nucleus. We conclude that the anti-apoptotic effect of OFSS is not mediated by activation of universal pro-survival signaling pathways. Rather, OFSS inhibits TNF-α-induced pro-apoptotic signaling which can be explained by the down-regulation of TNFR1 on the cell surface and blockade of TNFR1 downstream signaling by OFSS.


Subject(s)
Osteoblasts/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Rheology , Signal Transduction , Stress, Mechanical , Animals , Apoptosis/drug effects , Calcium Signaling/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Endocytosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Peroxide/pharmacology , I-kappa B Proteins/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-KappaB Inhibitor alpha , Nitric Oxide/biosynthesis , Osteoblasts/drug effects , Osteoblasts/enzymology , Promoter Regions, Genetic/genetics , Protein Biosynthesis/drug effects , Rheology/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination/drug effects
16.
Sci Signal ; 3(153): pe51, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21177492

ABSTRACT

Understanding the molecular mechanisms that mediate the response of cells to mechanical stimuli, the process known as mechanotransduction, has emerged as a research topic with relevance to human health and disease. Mechanotransduction in bone is particularly relevant because the mammalian skeleton remodels to adapt to its loading environment The mechanosome hypothesis has been proposed to explain how mechanical signals detected at the bone cell membrane are converted into changes in transcription of target genes. In one model, adhesion complexes at the surface of the sensor cell activate multiprotein complexes (mechanosomes) that include both proteins involved in adhesion and transcription factors that move to the nucleus and regulate transcriptional activity of target genes. New work has identified a previously unknown mechanotransduction complex-consisting of nitric oxide (NO), cyclic guanosine monophosphate (cGMP), protein kinase G II, SHP-1, and SHP-2-that associates with ß3 integrins through Src. This complex regulates gene expression in response to fluid flow and has several of the necessary elements of a mechanosome complex. These findings beg the question of just how extensive the mechanosome network is and how mechanosomes interact with other signal transduction pathways that also respond to mechanical load.


Subject(s)
Bone and Bones/physiology , Gene Expression Regulation/physiology , Mechanotransduction, Cellular/physiology , Multiprotein Complexes/metabolism , Biomechanical Phenomena , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Humans , Models, Biological , Transcription Factors/metabolism
17.
Bone ; 47(1): 74-82, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20353839

ABSTRACT

When bone is mechanically loaded fluid shear stress (FSS) is generated as a result of the movement of interstitial fluid across the membranes of osteoblasts and osteocytes. This external mechanical loading stimulates changes in the activity of cytoplasmic signaling molecules and alters gene expression in bone cells. This process, referred to as mechanotransduction, is vital for maintaining bone health in vivo by regulating the balance between bone formation and bone resorption. This current study focuses on the role of focal adhesions, sites of integrin-mediated cellular attachment to the extracellular matrix, and their proposed function as mechanosensors in bone cells. We examined the role of a key component of focal adhesions and of mechanotransduction, focal adhesion kinase (FAK) in regulation of FSS- and tumor necrosis factor-alpha (TNF-alpha)-induced activation of nuclear factor-kappa B (NF-kappaB) signaling in osteoblasts. Immortalized FAK(+/+) and FAK(-)(/)(-) osteoblasts were exposed to periods of oscillatory fluid shear stress (OFF) and NF-kappaB activation was analyzed. We determined that FAK is required for OFF-induced nuclear translocation and activation of NF-kappaB in osteoblasts. In addition we found that OFF-induced phosphorylation of the IkappaB kinases (IKKalpha/beta) in both FAK(+/+) and FAK(-/-) osteoblasts, but only FAK(+/+) osteoblasts demonstrated the resulting degradation of NF-kappaB inhibitors IkappaBalpha and IkappaBbeta. OFF did not induce the degradation of IkappaBepsilon or the processing of p105 in either FAK(+/+) and FAK(-/-) osteoblasts. To compare the role of FAK in mediating OFF-induced mechanotransduction to the well characterized activation of NF-kappaB by inflammatory cytokines, we exposed FAK(+/+) and FAK(-/-) osteoblasts to TNF-alpha. Interestingly, FAK was not required for TNF-alpha induced NF-kappaB activation in osteoblasts. In addition we determined that TNF-alpha treatment did not induce the degradation of IkappaBbeta as did OFF. These data indicate a novel relationship between FAK and NF-kappaB activation in osteoblast mechanotransduction and demonstrates that the mechanism of FSS-induced NF-kappaB activation in osteoblasts differs from the well characterized TNF-alpha-induced activation.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Osteoblasts/drug effects , Osteoblasts/enzymology , Rheology , Stress, Mechanical , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Focal Adhesion Protein-Tyrosine Kinases/deficiency , I-kappa B Proteins/metabolism , Mice , NF-KappaB Inhibitor alpha , NF-kappa B p50 Subunit/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Rheology/drug effects , Transcription Factor RelA/genetics
18.
J Cell Physiol ; 223(2): 435-41, 2010 May.
Article in English | MEDLINE | ID: mdl-20112285

ABSTRACT

Cellular mechanotransduction, the process of converting mechanical signals into biochemical responses within cells, is a critical aspect of bone health. While the effects of mechanical loading on bone are well recognized, elucidating the specific molecular pathways involved in the processing of mechanical signals by bone cells represents a challenge and an opportunity to identify therapeutic strategies to combat bone loss. In this study we have for the first time examined the relationship between the nucleocytoplasmic shuttling transcription factor nuclear matrix protein-4/cas interacting zinc finger protein (Nmp4/CIZ) and beta-catenin signaling in response to a physiologic mechanical stimulation (oscillatory fluid shear stress, OFSS) in osteoblasts. Using calvaria-derived osteoblasts from Nmp4-deficient and wild-type mice, we found that the normal translocation of beta-catenin to the nucleus in osteoblasts that is induced by OFSS is enhanced when Nmp4/CIZ is absent. Furthermore, we found that other aspects of OFSS-induced mechanotransduction generally associated with the beta-catenin signaling pathway, including ERK, Akt, and GSK3beta activity, as well as expression of the beta-catenin-responsive protein cyclin D1 are also enhanced in cells lacking Nmp4/CIZ. Finally, we found that in the absence of Nmp4/CIZ, OFSS-induced cytoskeletal reorganization and the formation of focal adhesions between osteoblasts and the extracellular substrate is qualitatively enhanced, suggesting that Nmp4/CIZ may reduce the sensitivity of bone cells to mechanical stimuli. Together these results provide experimental support for the concept that Nmp4/CIZ plays an inhibitory role in the response of bone cells to mechanical stimulation induced by OFSS.


Subject(s)
Mechanotransduction, Cellular/physiology , Nuclear Matrix-Associated Proteins/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Bone Resorption/metabolism , Bone Resorption/physiopathology , Bone Resorption/prevention & control , Cell Adhesion/physiology , Cells, Cultured , Cyclin D1/metabolism , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesions/physiology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Mice , Mice, Knockout , Nuclear Matrix-Associated Proteins/genetics , Osteoblasts/cytology , Physical Stimulation , Protein Transport/physiology , Proto-Oncogene Proteins c-akt/metabolism , Stress, Mechanical , Transcription Factors/genetics , beta Catenin/genetics
19.
Clin Rev Bone Miner Metab ; 8(4): 213-223, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21479153

ABSTRACT

We introduced the mechanosome hypothesis in 2003 as a heuristic model for investigating mechanotransduction in bone (Pavalko et al., J Cell Biochem, 2003, 88(1):104-112). This model suggested specific approaches for investigating how mechanical information is conveyed from the membrane of the sensor bone cell to the target genes and how this transmitted information from the membrane is converted into changes in transcription. The key concepts underlying the mechanosome hypothesis are that load-induced deformation of bone deforms the sensor cell membrane; embedded in the membrane are the focal adhesion and cadherin-catenin complexes, which in turn are physically connected to the chromatin via a solid-state scaffold. The physical stimulation of the membrane launches multiprotein complexes (mechanosomes) from the adhesion platforms while concomitantly tugging target genes into position for contact with the incoming mechanosomes, the carriers of the mechanical information to the nucleus. The mechanosome is comprised of an adhesion-associated protein and a nucleocytoplasmic shuttling transcription factor. Upon arrival at the target gene, mechanosomes alter DNA conformation and thus influence the interactions between trans-acting proteins along the gene, changing gene activity. Here, we update significant progress related to the mechanosome concept since publication of our original hypothesis. The launching of adhesion- and cytoskeletal-associated proteins into the nucleus toward target genes appears to be a common mechanism for regulating cell response to changes in its mechanical microenvironment.

20.
J Bone Miner Res ; 24(3): 411-24, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19016591

ABSTRACT

Mechanical loading of bone is important for maintenance of bone mass and structural stability of the skeleton. When bone is mechanically loaded, movement of fluid within the spaces surrounding bone cells generates fluid shear stress (FSS) that stimulates osteoblasts, resulting in enhanced anabolic activity. The mechanisms by which osteoblasts convert the external stimulation of FSS into biochemical changes, a process known as mechanotransduction, remain poorly understood. Focal adhesions are prime candidates for transducing external stimuli. Focal adhesion kinase (FAK), a nonreceptor tyrosine kinase found in focal adhesions, may play a key role in mechanotransduction, although its function has not been directly examined in osteoblasts. We examined the role of FAK in osteoblast mechanotransduction using short interfering RNA (siRNA), overexpression of a dominant negative FAK, and FAK(-/-) osteoblasts to disrupt FAK function in calvarial osteoblasts. Osteoblasts were subjected to varying periods oscillatory fluid flow (OFF) from 5 min to 4 h, and several physiologically important readouts of mechanotransduction were analyzed including: extracellular signal-related kinase 1/2 phosphorylation, upregulation of c-fos, cyclooxygenase-2, and osteopontin, and release of prostaglandin E(2). Osteoblasts with disrupted FAK signaling exhibited severely impaired mechanical responses in all endpoints examined. These data indicate the importance of FAK for both short and long periods of FSS-induced mechanotransduction in osteoblasts.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mechanotransduction, Cellular , Osteoblasts/cytology , Osteoblasts/enzymology , Stress, Mechanical , Animals , Cyclooxygenase 2/biosynthesis , Dinoprostone/metabolism , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/deficiency , Mechanotransduction, Cellular/drug effects , Mice , Osteoblasts/drug effects , Osteopontin/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , RNA, Small Interfering/metabolism , Rats , Rheology , Time Factors , Transfection , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...