Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Cell Biochem ; 117(4): 970-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26378628

ABSTRACT

CIZ/NMP4 (Cas interacting zinc finger protein, Nmp4, Zfp384) is a transcription factor that is known to regulate matrix related-proteins. To explore the possible pathophysiological role of CIZ/NMP4 in arthritis, we examined CIZ/NMP4 expression in articular cartilage in arthritis model. CIZ/NMP4 was expressed in the articular chondrocytes of mice at low levels while its expression was enhanced when arthritis was induced. Arthritis induction increased clinical score in wild type mice. In contrast, CIZ/NMP4 deficiency suppressed such rise in the levels of arthritis score and swelling of soft tissue. CIZ/NMP4 deficiency also reduced invasion of inflammatory cells in joint tissue. Quantitative PCR analyses of mRNA from joints revealed that arthritis-induced increase in expressions of IL-1ß was suppressed by CIZ/NMP4 deficiency. CIZ/NMP4 bound to IL-1ß promoter and activated its transcription. The increase in CIZ/NMP4 in arthritis was also associated with enhancement in bone resorption and cartilage matrix degradation. In fact, RANKL, a signaling molecule prerequisite for osteoclastogenesis and, MMP-3, a clinical marker for arthritis were increased in joints upon arthritis induction. In contrast, CIZ/NMP4 deficiency suppressed the arthritis-induced increase in bone resorption, expression of RANKL and MMP-3 mRNA. Thus, CIZ/NMP4 plays a role in the development of arthritis at least in part through regulation of key molecules related to the arthritis.


Subject(s)
Arthritis, Experimental/genetics , Cartilage, Articular/immunology , Matrix Metalloproteinase 3/immunology , Nuclear Matrix-Associated Proteins/immunology , RANK Ligand/immunology , Transcription Factors/immunology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoantibodies/biosynthesis , Bone Resorption , Cartilage, Articular/pathology , Chondrocytes/immunology , Chondrocytes/pathology , Female , Gene Expression Regulation , Glucose-6-Phosphate Isomerase/antagonists & inhibitors , Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/immunology , Immune Sera/administration & dosage , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Joints/immunology , Joints/pathology , Male , Matrix Metalloproteinase 3/genetics , Mice , Mice, Knockout , Nuclear Matrix-Associated Proteins/deficiency , Nuclear Matrix-Associated Proteins/genetics , Promoter Regions, Genetic , RANK Ligand/genetics , Severity of Illness Index , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic
2.
J Cell Physiol ; 229(10): 1353-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24604668

ABSTRACT

Bone metabolism is maintained via balanced repetition of bone resorption by osteoclasts and bone formation by osteoblasts. Osteoblastic cells are capable of conducting self-renewal and differentiation that are basically associated with cell-cycle transition to enable cell specification and bone formation. Osteoblasts are also migrating to fill the resorption cavity curved by osteoclasts during bone remodeling to maintain homeostasis of bone mass whose imbalance leads to osteoporosis. However, technical difficulties have hampered the research on the dynamic relationship between cell cycle and migration in osteoblasts. In this report, we overcome these problems by introducing fluorescent ubiquitination-based cell cycle indicator (FUCCI) reporter system in calvarial osteoblastic cells and reveal that the cells in G1 as well as S/G2 /M phase are migrating. Furthermore, the osteoblastic cells in S/G2 /M phase migrate faster than those in G1 phase. Interestingly, parathyroid hormone (PTH) as an anabolic agent enhances migration velocity of the cells. Mechanical stress, another anabolic signal, also enhances migration velocity. In contrast, in the presence of both PTH and mechanical stress, the migration velocity returns to the base line levels revealing the interaction between the two anabolic stimuli in the regulation of cell migration. Importantly, PTH and mechanical stress also interact when they regulate the transition of cell cycle. These data demonstrate that osteoblastic migration is linked to cell cycle and it is under the control of mechanical and chemical stimuli that coordinate to regulate bone mass.


Subject(s)
Biosensing Techniques , Bone Remodeling , Cell Cycle , Cell Movement , Cell Tracking/methods , Mechanotransduction, Cellular , Osteoblasts/metabolism , Parathyroid Hormone/metabolism , Animals , Cells, Cultured , Genes, Reporter , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Stress, Mechanical , Time Factors
3.
Proc Natl Acad Sci U S A ; 111(7): 2692-7, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550297

ABSTRACT

Osteoclastogenesis is under the control of posttranscriptional and transcriptional events. However, posttranscriptional regulation of osteoclastogenesis is incompletely understood. CNOT3 is a component of the CCR4 family that regulates mRNA stability, but its function in bone is not known. Here, we show that Cnot3 deficiency by deletion of a single allele induces osteoporosis. Cnot3 deficiency causes an enhancement in bone resorption in association with an elevation in bone formation, resulting in high-turnover type bone loss. At the cellular level, Cnot3 deficiency enhances receptor activator of NF-κB ligand (RANKL) effects on osteoclastogenesis in a cell-autonomous manner. Conversely, Cnot3 deficiency does not affect osteoblasts directly. Cnot3 deficiency does not alter RANKL expression but enhances receptor activator of NF-κB (RANK) mRNA expression in bone in vivo. Cnot3 deficiency promotes RANK mRNA stability about twofold in bone marrow cells of mice. Cnot3 knockdown also increases RANK mRNA expression in the precursor cell line for osteoclasts. Anti-CNOT3 antibody immunoprecipitates RANK mRNA. Cnot3 deficiency stabilizes luciferase reporter expression linked to the 3'-UTR fragment of RANK mRNA. In contrast, Cnot3 overexpression destabilizes the luciferase reporter linked to RANK 3'-UTR. In aged mice that exhibit severe osteoporosis, Cnot3 expression levels in bone are reduced about threefold in vivo. Surprisingly, Cnot3 deficiency in these aged mice further exacerbates osteoporosis, which also occurs via enhancement of osteoclastic activity. Our results reveal that CNOT3 is a critical regulator of bone mass acting on bone resorption through posttranscriptional down-regulation of RANK mRNA stability, at least in part, even in aging-induced osteoporosis.


Subject(s)
Bone Resorption/physiopathology , Gene Expression Regulation/physiology , Osteoporosis/physiopathology , RNA Stability/physiology , RNA, Messenger/metabolism , Transcription Factors/metabolism , Absorptiometry, Photon , Age Factors , Animals , Bone Density , DNA Primers/genetics , Imaging, Three-Dimensional , Luciferases , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Stability/genetics , RNA, Small Interfering/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
4.
Pathol Int ; 63(5): 272-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23714255

ABSTRACT

We report an atypical case of non-sebaceous lymphadenoma (NSL) of the parotid gland showing serous acinic cell differentiation. NSL is a rare benign salivary gland tumor with intermingled lymphoid and epithelial tissues without sebaceous differentiation. Since the first description of a case designated by Auclair et al. as 'non-sebaceous lymphadenoma' in 1991, to our best knowledge, only 37 cases have been reported, and no differentiation of tumor cells into serous acinic cell lineage has been described so far. In this paper, we present a case of NSL with serous acinic cell differentiation. The patient was a 78-year-old female with the complaint of a painless mass in the left parotid gland. The surgically resected tumor was encapsulated and measured 13 × 9 × 9 mm. Histologically, the tumor had the features of NSL, and an unusual finding of this case was the presence of epithelial cells with serous acinic cell differentiation. Dense cytoplasm packed with basophilic granules in those cells was positive in periodic acid Schiff reaction after diastase digestion (D-PAS), which was compatible with the feature of serous acinic cell differentiation. Possible differentiation of the epithelial component into serous acinic cell in this rare entity is warranted to avoid confusion in the diagnosis.


Subject(s)
Acinar Cells/pathology , Biomarkers, Tumor/metabolism , Lymphoma/pathology , Parotid Gland/pathology , Parotid Neoplasms/pathology , Acinar Cells/physiology , Aged , Female , Humans , Lymphoma/diagnostic imaging , Lymphoma/surgery , Parotid Gland/diagnostic imaging , Parotid Gland/surgery , Parotid Neoplasms/diagnostic imaging , Parotid Neoplasms/surgery , Radiography , Treatment Outcome
5.
Bone ; 54(1): 172-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23314072

ABSTRACT

Mechanical stress is known to alter bone mass and the loss of force stimuli leads to reduction of bone mass. However, molecules involved in this phenomenon are incompletely understood. As mechanical force would affect signaling events in cells, we focused on a calcium channel, TRPV4 regarding its role in the effects of force stimuli on calcium in osteoblasts. TRPV4 expression levels were enhanced upon differentiation of osteoblasts in culture. We found that BMP-2 treatment enhanced TRPV4 gene expression in a dose dependent manner. BMP-2 effects on TRPV4 expression were suppressed by inhibitors for transcription and new protein synthesis. In these osteoblasts, a TRPV4-selective agonist, 4α-PDD, enhanced calcium signaling and the effects of 4α-PDD were enhanced in differentiated osteoblasts compared to the control cells. Fluid flow, as a mechanical stimulation, induced intracellular calcium oscillation in wild type osteoblasts. In contrast, TRPV4 deficiency suppressed calcium oscillation significantly even when the cells were subjected to fluid flow. These data suggest that TRPV4 is involved in the flow-induced calcium signaling in osteoblasts.


Subject(s)
Calcium Signaling , Cell Differentiation , Osteoblasts/cytology , Osteoblasts/metabolism , Stress, Mechanical , TRPV Cation Channels/metabolism , Animals , Bone Morphogenetic Protein 2/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Culture Media/pharmacology , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rheology/drug effects , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
6.
J Cell Physiol ; 228(7): 1397-403, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23280595

ABSTRACT

Mechanical stress is an important signal to determine the levels of bone mass. Unloading-induced osteoporosis is a critical issue in bed-ridden patients and astronauts. Many molecules have been suggested to be involved in sensing mechanical stress in bone, though the mechanisms involved in this phenomenon are not fully understood. Nck1 is an adaptor protein known to mediate signaling from plasma membrane-activated receptors to cytosolic effectors regulating actin cytoskeleton remodeling. Nck1 has also been implicated in cellular responses to endoplasmic reticulum stress. In vitro, in case of cell stress the actin cytoskeleton is disrupted and in such cases Nck1 has been reported to enter the nucleus of the cells to mediate the nuclear actin polymerization. However, the role of Nck1 in vivo during the bone response to mechanical stimuli is unknown. The purpose of this study is to examine the role of Nck1 in unloading-induced bone loss in vivo. Sciatic and femoral nerve resection was conducted. Neurectomy-based unloading enhanced Nck1 gene expression in bone about twofold. Using the Nck1 deficient mice and control Nck1+/+, effects of neurectomy-based unloading on bone structure were examined. Unloading reduced bone volume in wild type mice by 30% whereas the levels in bone loss were exacerbated to 50% in Nck1 deficient mice due to neurectomy after 4 weeks. These data demonstrate that Nck1 gene deficiency accelerates the mechanical unloading-induced bone loss suggesting Nck1 to be a crucial molecule in mechanical stress mediated regulation in bone metabolism.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Bone Resorption/etiology , Oncogene Proteins/deficiency , 3T3 Cells , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Biomechanical Phenomena , Bone Resorption/pathology , Bone Resorption/physiopathology , Cell Nucleus/metabolism , Denervation , Femoral Nerve/surgery , Gene Expression , Hindlimb Suspension/adverse effects , Hindlimb Suspension/physiology , Humans , Locomotion , Male , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/etiology , Muscular Atrophy/physiopathology , Oncogene Proteins/genetics , Oncogene Proteins/physiology , Osteoblasts/metabolism , Osteoblasts/pathology , Sciatic Nerve/surgery
7.
J Biol Chem ; 287(40): 33545-53, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22773831

ABSTRACT

Bone development is a dynamic process that requires cell motility and morphological adaptation under the control of actin cytoskeleton. This actin cytoskeleton system is regulated by critical modulators including actin-binding proteins. Among them, profilin1 (Pfn1) is a key player to control actin fiber structure, and it is involved in a number of cellular activities such as migration. During the early phase of body development, skeletal stem cells and osteoblastic progenitor cells migrate to form initial rudiments for future skeletons. During this migration, these cells extend their process based on actin cytoskeletal rearrangement to locate themselves in an appropriate location within microenvironment. However, the role of Pfn1 in regulation of mesenchymal progenitor cells (MPCs) during skeletal development is incompletely understood. Here we examined the role of Pfn1 in skeletal development using a genetic ablation of Pfn1 in MPCs by using Prx1-Cre recombinase. We found that Pfn1 deficiency in MPCs caused complete cleft sternum. Notably, Pfn1-deficient mice exhibited an absence of trabecular bone in the marrow space of appendicular long bone. This phenotype is location-specific, as Pfn1 deficiency did not largely affect osteoblasts in cortical bone. Pfn1 deficiency also suppressed longitudinal growth of long bone. In vitro, Pfn1 deficiency induced retardation of osteoblastic cell migration. These observations revealed that Pfn1 is a critical molecule for the skeletal development, and this could be at least in part associated with the retardation of cell migration.


Subject(s)
Gene Expression Regulation, Developmental , Profilins/physiology , Alleles , Animals , Bone and Bones/metabolism , Cartilage/metabolism , Cell Movement , Cytoskeleton/metabolism , Genotype , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , Osteoblasts/cytology , Osteogenesis , Profilins/metabolism , RNA, Small Interfering/metabolism , Time Factors , Transfection , X-Ray Microtomography/methods
8.
Proc Natl Acad Sci U S A ; 109(19): 7433-8, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22538810

ABSTRACT

Parathyroid hormone (PTH), the major calcium-regulating hormone, and norepinephrine (NE), the principal neurotransmitter of sympathetic nerves, regulate bone remodeling by activating distinct cell-surface G protein-coupled receptors in osteoblasts: the parathyroid hormone type 1 receptor (PTHR) and the ß(2)-adrenergic receptor (ß(2)AR), respectively. These receptors activate a common cAMP/PKA signal transduction pathway mediated through the stimulatory heterotrimeric G protein. Activation of ß(2)AR via the sympathetic nervous system decreases bone formation and increases bone resorption. Conversely, daily injection of PTH (1-34), a regimen known as intermittent (i)PTH treatment, increases bone mass through the stimulation of trabecular and cortical bone formation and decreases fracture incidences in severe cases of osteoporosis. Here, we show that iPTH has no osteoanabolic activity in mice lacking the ß(2)AR. ß(2)AR deficiency suppressed both iPTH-induced increase in bone formation and resorption. We showed that the lack of ß(2)AR blocks expression of iPTH-target genes involved in bone formation and resorption that are regulated by the cAMP/PKA pathway. These data implicate an unexpected functional interaction between PTHR and ß(2)AR, two G protein-coupled receptors from distinct families, which control bone formation and PTH anabolism.


Subject(s)
Bone and Bones/drug effects , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Absorptiometry, Photon , Anabolic Agents/metabolism , Anabolic Agents/pharmacology , Animals , Bone Density/drug effects , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Female , Femur/drug effects , Femur/metabolism , Fluoresceins , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteogenesis/drug effects , Osteogenesis/genetics , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptors, Adrenergic, beta-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
9.
J Cell Physiol ; 227(7): 2807-12, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22307584

ABSTRACT

Tumor metastasis to bone is a serious pathological situation that causes severe pain, and deterioration in locomoter function. However, the mechanisms underlying tumor metastasis is still incompletely understood. CIZ/NMP4 is a nucleocytoplasmic shuttling protein and its roles in tumor cells have not been known. We, therefore, hypothesized the role of CIZ/NMP4 in B16 melanoma cells that metastasize to bone. CIZ/NMP4 is expressed in B16 cells. The CIZ/NMP4 expression levels are correlated to the metastatic activity in divergent types of melanoma cells. Overexpression of CIZ/NMP4 increased B16 cell migration in Trans-well assay. Conversely, siRNA-based knockdown of CIZ/NMP4 suppressed migratory activity of these cells. As RANKL promotes metastasis of tumor cells in bone, we tested its effect on CIZ in melanoma cells. RANKL treatment enhanced CIZ/NMP4 expression. This increase of CIZ by RANKL promoted migration. Conversely, we identified CIZ/NMP4 binding site in the promoter of RANKL. Furthermore, luciferase assay indicated that CIZ/NMP4 overexpression enhanced RANKL promoter activities, revealing a positive feedback loop of CIZ/NMP4 and RANKL in melanoma. These observations indicate that CIZ/NMP4 is critical regulator of metastasis of melanoma cells.


Subject(s)
Cell Movement/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Nuclear Matrix-Associated Proteins/biosynthesis , RANK Ligand/metabolism , Transcription Factors/biosynthesis , Animals , Binding Sites , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Cell Adhesion/genetics , Feedback, Physiological , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Promoter Regions, Genetic , RANK Ligand/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Up-Regulation
10.
Auris Nasus Larynx ; 39(3): 270-4, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21794995

ABSTRACT

OBJECTIVE: Heat shock transcription factor 1 (HSF1) is a master regulator of heat shock response, and also inhibits expression of inflammatory cytokines directly or indirectly. Here, we examined effects of HSF1 activation on the expression of proinflammatory cytokines in mouse cochlea after exposure to noise. METHODS: Male CBA/N mice with normal Preyer's reflex were exposed to intense noise for 3h. Three hours after noise exposure, bilateral cochleae were removed and expression of major inflammatory cytokines was examined. RESULTS: We found that interleukin-6 (IL-6) and interleukin-1ß (IL-1ß) expression increased significantly after noise exposure, and the expression was suppressed significantly in mice administered with geranylgeranylacetone (GGA), which activates HSF1. Seven days after noise exposure, thresholds for auditory brainstem response were elevated, and GGA administration significantly suppressed this elevation. CONCLUSION: These results suggest that HSF1-mediated suppression of proinflammatory cytokines in the cochlea by GGA administration could be an important means of inner ear protection.


Subject(s)
Cochlea/drug effects , Cytokines/drug effects , DNA-Binding Proteins/metabolism , Diterpenes/pharmacology , Hearing Loss, Noise-Induced/metabolism , Inflammation/metabolism , Transcription Factors/metabolism , Animals , Auditory Threshold/drug effects , Cochlea/metabolism , Cytokines/metabolism , DNA-Binding Proteins/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Heat Shock Transcription Factors , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred CBA , Noise , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/drug effects
11.
Tissue Cell ; 44(1): 1-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22071216

ABSTRACT

Bone consists of type I collagen as a major protein with minor various matrix proteins. Type VI collagen is one of bone matrix proteins but its function is not known. We therefore examined the effects of type VI collagen deficiency on bone. 3D-µCT analysis revealed that type VI collagen deficiency reduced cancellous bone mass. Cortical bone mass was not affected. Type VI collagen deficiency distorted the shape of osteoblasts both in the cancellous bone and in the cambium layer of periosteal region. Furthermore, type VI collagen deficiency disorganized collagen arrangement. These data indicate that type VI collagen contributes to maintain bone mass.


Subject(s)
Bone Diseases, Metabolic/genetics , Collagen Type VI/genetics , Osteoblasts/pathology , Animals , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone Remodeling/genetics , Bone and Bones/metabolism , Bone and Bones/pathology , Collagen Type VI/deficiency , Collagen Type VI/physiology , Extracellular Matrix/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Polarization , Osteoclasts/pathology , X-Ray Microtomography
12.
Proc Natl Acad Sci U S A ; 108(43): 17767-72, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21990347

ABSTRACT

The sympathetic nervous system suppresses bone mass by mechanisms that remain incompletely elucidated. Using cell-based and murine genetics approaches, we show that this activity of the sympathetic nervous system requires osteopontin (OPN), a cytokine and one of the major members of the noncollagenous extracellular matrix proteins of bone. In this work, we found that the stimulation of the sympathetic tone by isoproterenol increased the level of OPN expression in the plasma and bone and that mice lacking OPN (OPN-KO) suppressed the isoproterenol-induced bone loss by preventing reduced osteoblastic and enhanced osteoclastic activities. In addition, we found that OPN is necessary for changes in the expression of genes related to bone resorption and bone formation that are induced by activation of the sympathetic tone. At the cellular level, we showed that intracellular OPN modulated the capacity of the ß2-adrenergic receptor to generate cAMP with a corresponding modulation of cAMP-response element binding (CREB) phosphorylation and associated transcriptional events inside the cell. Our results indicate that OPN plays a critical role in sympathetic tone regulation of bone mass and that this OPN regulation is taking place through modulation of the ß2-adrenergic receptor/cAMP signaling system.


Subject(s)
Bone and Bones/physiology , Osteopontin/metabolism , Sympathetic Nervous System/physiology , Analysis of Variance , Animals , Bone and Bones/metabolism , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer , Isoproterenol/pharmacology , Mice , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteopontin/deficiency , Receptors, Adrenergic, beta-2/metabolism , Sympathetic Nervous System/drug effects
13.
J Cell Biochem ; 112(12): 3525-30, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21866567

ABSTRACT

Loss of mechanical stress or unloading causes disuse osteoporosis that leads to fractures and deteriorates body function and affects mortality rate in aged population. This bone loss is due to reduction in osteoblastic bone formation and increase in osteoclastic bone resorption. MuRF1 is a muscle RING finger protein which is involved in muscle wasting and its expression is enhanced in the muscle of mice subjected to disuse condition such as hind limb unloading (HU). However, whether MuRF1 is involved in bone loss due to unloading is not known. We therefore examined the effects of MuRF1 deficiency on unloading-induced bone loss. We conducted hind limb unloading of MuRF1 KO mice and wild-type control mice. Unloading induced about 60% reduction in cancellous bone volume (BV/TV) in WT mice. In contrast, MuRF1 deficiency suppressed unloading-induced cancellous bone loss. The cortical bone mass was also reduced by unloading in WT mice. In contrast, MuRF1 deficiency suppressed this reduction in cortical bone mass. To understand whether the effects of MuRF1 deficiency suppress bone loss is on the side of bone formation or bone resorption, histomorphometry was conducted. Unloading reduced bone osteoblastic formation rate (BFR) in WT. In contrast, MuRF1 deficiency suppressed this reduction. Regarding bone resorption, unloading increased osteoclast number in WT. In contrast, MURF1 deficiency suppressed this osteoclast increase. These data indicated that the ring finger protein, MURF1 is involved in disuse-induced bone loss in both of the two major bone remodeling activities, osteoblastic bone formation and osteoclastic bone resorption.


Subject(s)
Muscle Proteins/physiology , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/genetics , Ubiquitin-Protein Ligases/physiology , Absorptiometry, Photon , Animals , Female , Hindlimb Suspension , Mice , Mice, Knockout , Muscle Proteins/genetics , Osteoblasts/pathology , Osteoclasts/pathology , Osteoporosis/pathology , Tomography, X-Ray Computed , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
14.
Cancer Sci ; 102(12): 2109-17, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21848808

ABSTRACT

The p210Bcr/Abl and p190Bcr/Abl fusion oncoproteins are known to cause chronic myelogenous leukemia (CML) and acute lymphoblastic leukemia (ALL). Bcr/Abl phosphorylates several proteins that can lead to leukemogenesis. Crk-associated substrate lymphocyte type (Cas-L)/human enhancer of filamentation-1 (HEF1)/neural precursor cell expressed, developmentally down-regulated 9 (NEDD9) is an adapter protein at focal adhesions known to be associated with solid tumor metastasis. Crk-associated substrate lymphocyte type has also been reported to be tyrosine phosphorylated by p190Bcr/Abl. We demonstrated that Cas-L was expressed in murine granulocytes, as well as in lymphocytes, and that Cas-L-deficient (Cas-L(-/-) ) granulocytes had increased migratory activity and decreased adhesiveness. To examine whether Cas-L was involved in leukemogenesis by p210Bcr/Abl, we generated Cas-L(-/-) p210Bcr/Abl transgenic mice. The mice displayed early development of myeloproliferative neoplasm seen in the chronic phase of CML, which resulted in the early death of the mice. Pathologically, increased infiltration of myeloid cells into several tissues was detected in the absence of Cas-L. In a hematopoietic reconstitution assay, Cas-L(-/-) p210Bcr/Abl transgenic cells showed a low population in the spleen, although only their myeloid cell population was normal. Thus, Cas-L seems to regulate the progression of CML in a negative way, presumably by attenuating extramedullary hyperplasia.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Myeloid Cells/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Cell Movement , Cell Proliferation , Crk-Associated Substrate Protein/metabolism , Disease Progression , Female , Fusion Proteins, bcr-abl/genetics , Genes, abl , Granulocytes/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemic Infiltration , Lymphocyte Count , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/pathology , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Neoplasm Metastasis , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
15.
Tissue Cell ; 43(3): 196-200, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21511320

ABSTRACT

Parathyroid hormone/parathyroid hormone-related protein receptor (PPR) signaling is known to be involved in tooth development. In bone, extracellular matrix protein osteopontin (OPN) is a negative regulator of PPR signaling in bone formation. However, the role of OPN in modulation of PPR action in tooth development is not understood. Therefore, we examined the tooth in double mutant mice. Constitutively active PPR was expressed specifically in the odontoblasts and osteoblasts (caPPR-tg) in the presence or absence of OPN. Radiographic analysis indicated that the length of the third molar (M3) and the incisor was decreased in the caPPR-tg mice compared to wild type, and such reduction in molar and incisor length was further enhanced in the absence of OPN (caPPR-tg OPN-KO). With respect to histology of incisors, caPPR-tg induced high cellularity and irregularity in odontoblastic shape and this was enhanced by the absence of OPN. These morphological observations suggest that OPN modulates PPR signaling that are involved in tooth formation.


Subject(s)
Odontoblasts/metabolism , Osteoblasts/metabolism , Osteopontin/deficiency , Receptor, Parathyroid Hormone, Type 1/physiology , Signal Transduction/physiology , Tooth/growth & development , Animals , Incisor/growth & development , Mice , Mice, Knockout , Parathyroid Hormone
16.
J Cell Biochem ; 112(2): 433-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268064

ABSTRACT

Per-1 is one of the clock genes and is known to regulate various biological events including bone mass determination. Parathyroid hormone is anabolic to bone while the mechanism of its action is not fully understood. Here, we examined the role of PTH on Per-1 gene expression under osteoblast specific PTH signaling. Constitutively active PTH receptor (caPPR) expressed specifically in osteoblasts in transgenic mice activates Per-1 gene expression in bone. This is specific as expression of other clock gene Bmal-1 is not affected by caPPR over-expression. Per-1 is also expressed in osteoblastic cell line. Interestingly, Per-1 expression is required for PTH signaling-induced CRE dependent transcription. This is forming a positive feed back loop in the anabolic action of PTH signaling and Per-1 in bone. These data indicate that PTH singling in osteoblasts activates Per-1 gene expression in vivo in association with its anabolic action in bone at least in part through the regulation of transcriptional events.


Subject(s)
Osteoblasts/metabolism , Parathyroid Hormone/metabolism , Period Circadian Proteins/genetics , ARNTL Transcription Factors/genetics , Absorptiometry, Photon , Animals , Bone Density/genetics , Cell Line , Chemokine CXCL12/genetics , Mice , Mice, Transgenic , Polymerase Chain Reaction , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , X-Ray Microtomography
17.
Arthritis Rheum ; 63(4): 1021-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21190246

ABSTRACT

OBJECTIVE: Regeneration of bone requires the combination of appropriate drugs and an appropriate delivery system to control cell behavior. However, the delivery of multiple drugs to heal bone is complicated by the availability of carriers. The aim of this study was to explore a new system for delivery of a selective EP4 receptor agonist (EP4A) in combination with low-dose bone morphogenetic protein 2 (BMP-2). METHODS: Combined delivery of EP4A and BMP-2 was carried out with a nanogel-based scaffold in the shape of a disc, to repair critical-size circle-shaped bone defects in calvariae that otherwise did not heal spontaneously. RESULTS: Combination treatment with EP4A and low-dose BMP-2 in nanogel efficiently activated bone cells to regenerate calvarial bone by forming both outer and inner cortical plates as well as bone marrow tissue to regenerate a structure similar to that of intact calvaria. EP4A enhanced low-dose BMP-2-induced cell differentiation and activation of transcription events in osteoblasts. CONCLUSION: These data indicate that combined delivery of EP4A and low-dose BMP-2 via nanogel-based hydrogel provides a new system for bone repair.


Subject(s)
Bone Diseases/drug therapy , Bone Morphogenetic Protein 2/therapeutic use , Intercellular Signaling Peptides and Proteins/therapeutic use , Polyethylene Glycols/therapeutic use , Polyethyleneimine/therapeutic use , Receptors, Prostaglandin E, EP4 Subtype/agonists , Alkaline Phosphatase/blood , Animals , Bone Diseases/physiopathology , Bone Morphogenetic Protein 2/pharmacology , Bone and Bones/drug effects , Bone and Bones/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Mice , Mice, Inbred ICR , Nanogels , Osteogenesis/drug effects , Osteogenesis/physiology , Phosphorylation , Polyethylene Glycols/pharmacology , Polyethyleneimine/pharmacology , Regeneration/drug effects , Regeneration/physiology , Smad4 Protein/metabolism , Tissue Scaffolds
18.
J Cell Physiol ; 226(3): 573-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21069746

ABSTRACT

Schnurri (Shn)-2 is a transcriptional modulator of bone formation and bone resorption and its deficiency causes low turnover state with higher cancellous bone mass due to the defects in osteoclasts that exceeds the defects in osteoblasts in mice. We addressed whether such low turnover of bone remodeling in Shn2 deficiency may be modulated in the absence of estrogen that induces high turnover state in vivo. Ovariectomy reduced bone mass in wild type compared to sham operated control mice and such reduction in bone mass was also observed in Shn2 deficient mice. However, due to the high levels of basal bone mass in Shn2 deficient mice, the bone mass levels after ovariectomy were still comparable to sham operated wild-type mice. Analysis indicated that estrogen depletion increased bone resorption at similar levels in wild type and Shn2 deficient mice though the basal levels of osteoclast number was slightly lower in Shn2-deficient mice. In contrast, basal levels of bone marrow cell mineralization in cultures were low in Shn2-deficeint mice while estrogen depletion increased the mineralization levels to those that were comparable to sham wild type. This indicates that Shn2-deficient mice maintain bone mass at the levels comparable to wild-type sham mice even after ovariectomy-induced bone loss and this correlates with the high levels of mineralization activity in bone marrow cells after ovariectomy.


Subject(s)
Bone Resorption/metabolism , DNA-Binding Proteins/deficiency , Ovariectomy , Animals , Bone Resorption/pathology , Bone Resorption/physiopathology , Calcification, Physiologic , Cell Count , DNA-Binding Proteins/metabolism , Enzyme Assays , Female , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Organ Size , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis
19.
Clin Calcium ; 20(12): 1801-5, 2010 Dec.
Article in Japanese | MEDLINE | ID: mdl-21123931

ABSTRACT

The relationship between bone and nervous system has been considered based on clinical observations such as Reflex Sympathetic Dystrophy (RSD) or ectopic bone formation associated with spinal cord injury. Sympathtic nervous tone has been reported to control both bone formation and bone resorption. Unloading induces bone loss due to an increase in bone resorption and decrease in bone formation. Both of these two arms are shown to be influenced by sympathetic tone. In addition, cannabinoid receptor has been observed to be involved in regulation of bone mass. Psychiatric diseases such as depression has also been suggested to linked to the alteration in the levels of bone mass. These observations together point to importance of the relationship between bone mass and nervous system.


Subject(s)
Bone Density , Bone and Bones/metabolism , Sympathetic Nervous System/physiology , Animals , Bone Resorption/etiology , Depression/metabolism , Humans , Mice , Nerve Endings/physiology , Osteogenesis , Receptors, Cannabinoid/physiology , Reflex Sympathetic Dystrophy/etiology , Signal Transduction/physiology
20.
J Cell Biochem ; 105(1): 121-8, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18465784

ABSTRACT

Crk-associated substrate lymphocyte type (Cas-L) protein, also known as human enhancer of filamentation 1 (Hef1) or neural precursor cell-expressed, developmentally down-regulated gene 9 (Nedd9), belongs to the Cas family of adapter proteins, which are involved in integrin signaling. Previous reports showed that Cas-L is expressed preferentially in lymphocytes and epithelial cells. Cas-L mediates signals from integrins, T-cell receptors, B-cells receptors, and transforming growth factor beta, leading to cell movement and cell division. Here, we report the expression of Cas-L in neutrophils. Cas-L was tyrosine-phosphorylated when human neutrophils were stimulated by fMLP, tumor necrosis factor-alpha (TNF), or lipopolysaccharide. The tyrosine phosphorylation of Cas-L in fMLP- or TNF- stimulated neutrophils was further enhanced by adhesion of the cells to their substrates. Cas-L was found to be localized at focal adhesions in stimulated neutrophils based on immunofluorescence microscopy. These findings suggest that Cas-L is one of the targets of inflammatory cytokines and is also modulated by cell adhesion process in neutrophils.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neutrophils/metabolism , Phosphoproteins/metabolism , Phosphotyrosine/metabolism , Cell Adhesion , Cell Line , Humans , Lipopolysaccharides/pharmacology , Neutrophils/cytology , Neutrophils/drug effects , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...