Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 295(33): 11764-11775, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32587089

ABSTRACT

Macrophages play critical roles in homeostasis and inflammation. Macrophage polarization to either a pro-inflammatory or anti-inflammatory status is controlled by activating inflammatory signaling pathways. Ubiquitination is a posttranslational modification that regulates these inflammatory signaling pathways. However, the influence of protein ubiquitination on macrophage polarization has not been well studied. We hypothesized that the ubiquitination status of key proteins in inflammatory pathways contributes to macrophage polarization, which is regulated by itchy E3 ubiquitin ligase (ITCH), a negative regulator of inflammation. Using ubiquitin proteomics, we found that ubiquitination profiles are different among polarized murine macrophage subsets. Interestingly, interleukin-1α (IL-1α), an important pro-inflammatory mediator, was specifically ubiquitinated in lipopolysaccharide-induced pro-inflammatory macrophages, which was enhanced in ITCH-deficient macrophages. The ITCH-deficient macrophages had increased levels of the mature form of IL-1α and exhibited pro-inflammatory polarization, and reduced deubiquitination of IL-1α protein. Finally, IL-1α neutralization attenuated pro-inflammatory polarization of the ITCH-deficient macrophages. In conclusion, ubiquitination of IL-1α is associated with increased pro-inflammatory polarization of macrophages deficient in the E3 ligase ITCH.


Subject(s)
Interleukin-1alpha/metabolism , Macrophages/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Animals , Cells, Cultured , Female , Gene Deletion , Inflammation/genetics , Inflammation/metabolism , Macrophages/cytology , Male , Mice, Inbred C57BL , Ubiquitin/metabolism
2.
J Pathol ; 251(3): 323-335, 2020 07.
Article in English | MEDLINE | ID: mdl-32418202

ABSTRACT

The lymphatic system plays a crucial role in the maintenance of tissue fluid homeostasis and the immunological response to inflammation. The effects of lymphatic drainage dysfunction on periodontitis have not been well studied. Here we show that lymphatic vessel endothelial receptor 1 (LYVE1)+ /podoplanin (PDPN)+ lymphatic vessels (LVs) are increased in the periodontal tissues, with accumulation close to the alveolar bone surface, in two murine periodontitis models: rheumatoid arthritis (RA)-associated periodontitis and ligature-induced periodontitis. Further, PDPN+ /alpha-smooth muscle actin (αSMA)- lymphatic capillaries are increased, whereas PDPN+ /αSMA+ collecting LVs are decreased significantly in the inflamed periodontal tissues. Both mouse models of periodontitis have delayed lymph flow in periodontal tissues, increased TRAP-positive osteoclasts, and significant alveolar bone loss. Importantly, the local administration of adeno-associated virus for vascular endothelial growth factor C, the major growth factor that promotes lymphangiogenesis, increases the area and number of PDPN+ /αSMA+ collecting LVs, promotes local lymphatic drainage, and reduces alveolar bone loss in both models of periodontitis. Lastly, LYVE1+ /αSMA- lymphatic capillaries are increased, whereas LYVE1+ /αSMA+ collecting LVs are decreased significantly in gingival tissues of patients with chronic periodontitis compared with those of clinically healthy controls. Thus, our findings reveal an important role of local lymphatic drainage in periodontal inflammation-mediated alveolar bone loss. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Alveolar Bone Loss/prevention & control , Alveolar Process/metabolism , Chronic Periodontitis/therapy , Genetic Therapy , Lymph/metabolism , Lymphatic Vessels/metabolism , Maxilla/metabolism , Vascular Endothelial Growth Factor C/biosynthesis , Vascular Endothelial Growth Factor C/genetics , Alveolar Bone Loss/genetics , Alveolar Bone Loss/metabolism , Alveolar Bone Loss/pathology , Alveolar Process/pathology , Animals , Case-Control Studies , Chronic Periodontitis/genetics , Chronic Periodontitis/metabolism , Chronic Periodontitis/pathology , Disease Models, Animal , Humans , Lymphatic Vessels/pathology , Male , Maxilla/pathology , Mice, Inbred C57BL , Mice, Transgenic , Osteoclasts/metabolism , Osteoclasts/pathology , Tumor Necrosis Factor-alpha/genetics
3.
Am J Pathol ; 189(12): 2516-2530, 2019 12.
Article in English | MEDLINE | ID: mdl-31539516

ABSTRACT

NF-κB signals through canonical transcription factor p65 (RelA)/p50 and noncanonical avian reticuloendotheliosis viral oncogene related B (RelB)/p52 pathways. The RelA/p50 is involved in basal and inflammatory lymphangiogenesis. However, the role of RelB/p52 in lymphatic vessel biology is unknown. Herein, we investigated changes in lymphatic vessels (LVs) in mice deficient in noncanonical NF-κB signaling and the function of RelB in lymphatic endothelial cells (LECs). LVs were examined in Relb-/-, p52-/-, or control mice, and the gene expression profiles in LECs with RelB knockdown. Relb-/-, but not p52-/-, mice exhibited multiple LV abnormalities. They include the following: i) increased capillary vessel diameter, ii) reduced smooth muscle cell (SMC) coverage of mature vessels, iii) leakage, and iv) loss of active and passive lymphatic flow. Relb-/- mature LVs had thinner vessel walls, more apoptotic LECs and SMCs, and fewer LEC junctions. RelB knockdown LECs had decreased growth, survival, and adhesion, and dysregulated signaling pathways involving these cellular events. These results suggest that Relb-/- mice have abnormal LVs, mainly in mature vessels with reduced SMC coverage, leakage, and loss of contractions. RelB knockdown in LECs leads to reduced growth, survival, and adhesion. RelB plays a vital role in LEC-mediated LV maturation and function.


Subject(s)
Cell Proliferation , Endothelial Cells/pathology , Lymphatic Vessels/pathology , Transcription Factor RelB/physiology , Animals , Apoptosis , Cell Movement , Cells, Cultured , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B , Signal Transduction
4.
FASEB J ; 32(6): 3174-3183, 2018 06.
Article in English | MEDLINE | ID: mdl-29401595

ABSTRACT

Thy1 (CD90), a glycosylated, glycophosphatidylinositol-anchored membrane protein highly expressed by subsets of mesenchymal stem cells and fibroblasts, inhibits adipogenesis. The role of Thy1 on bone structure and function has been poorly studied and represents a major knowledge gap. Therefore, we analyzed the long bones of wild-type (WT) and Thy1 knockout (KO) mice with micro-computed tomography (micro-CT) and histomorphometry to compare changes in bone architecture and overall bone structure. micro-CT analysis of long bones revealed Thy1 KO and WT mice fed a high-fat diet demonstrated bone structural parameters at 4 mo that differed significantly between WT and KO mice. A significant reduction in trabecular bone volume was noted in Thy1 KO mice. The most prominent differences were observed in trabecular bone volume ratio and trabecular bone connectivity density. Consistent with micro-CT measurements, histomorphometric analysis also showed decreased bone volume in the obese Thy1 KO mice compared to obese WT mice. In vitro assays revealed that osteogenic conditions increased Thy1 expression during OB differentiation and absence of Thy1 attenuated osteoblastogenesis. Together, these findings support the concept that Thy1 serves as a major mechanistic link to regulate bone formation and negatively regulate adipogenesis.-Paine, A., Woeller, C. F., Zhang, H., Garcia-Hernandez, M. L., Huertas, N., Xing, L., Phipps, R. P., Ritchlin, C. T. Thy1 is a positive regulator of osteoblast differentiation and modulates bone homeostasis in obese mice.


Subject(s)
Cancellous Bone/metabolism , Cell Differentiation , Homeostasis , Obesity/metabolism , Osteoblasts/metabolism , Thy-1 Antigens/biosynthesis , Adipogenesis/genetics , Animals , Cancellous Bone/diagnostic imaging , Cancellous Bone/pathology , Mice , Mice, Knockout , Mice, Obese , Obesity/genetics , Obesity/pathology , Osteoblasts/pathology , Thy-1 Antigens/genetics , X-Ray Microtomography
5.
Semin Cell Dev Biol ; 38: 90-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25598390

ABSTRACT

Rheumatoid arthritis (RA) is a prevalent inflammatory joint disease with enigmatic flares, which causes swelling, pain, and irreversible connective tissue damage. Recently, it has been demonstrated in murine models of RA that the popliteal lymph node (PLN) is a biomarker of arthritic flare, as it "expands" in size and contrast enhancement during a prolonged asymptomatic phase, prior to when it "collapses" with accelerated synovitis and joint erosion. This PLN collapse is associated with adjacent knee flare, decreases in PLN volume and contrast enhancement, lymphatic pulse and pumping pressure, and an increase in PLN pressure. Currently, it is known that PLN collapse is accompanied by a translocation of B cells from the follicles to the sinuses, effectively clogging the lymphatic sinuses of the PLN, and that B cell depletion therapy ameliorates arthritic flare by eliminating these B cells and restoring passive lymphatic flow from inflamed joints. Here we review the technological advances that have launched this area of research, describe future directions to help elucidate the potential mechanism of PLN collapse, and speculate on clinical translation towards new diagnostics and therapies for RA.


Subject(s)
Arthritis, Rheumatoid/pathology , Lymph Nodes/pathology , Lymphatic System/physiology , Animals , Arthritis, Rheumatoid/immunology , B-Lymphocytes/immunology , Humans , Lymph Nodes/immunology , Lymphatic System/immunology , Symptom Flare Up
6.
Am J Physiol Cell Physiol ; 311(4): C673-C685, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27488671

ABSTRACT

Calponin is an actin cytoskeleton-associated protein that regulates motility-based cellular functions. Three isoforms of calponin are present in vertebrates, among which calponin 2 encoded by the Cnn2 gene is expressed in multiple types of cells, including blood cells from the myeloid lineage. Our previous studies demonstrated that macrophages from Cnn2 knockout (KO) mice exhibit increased migration and phagocytosis. Intrigued by an observation that monocytes and macrophages from patients with rheumatoid arthritis had increased calponin 2, we investigated anti-glucose-6-phosphate isomerase serum-induced arthritis in Cnn2-KO mice for the effect of calponin 2 deletion on the pathogenesis and pathology of inflammatory arthritis. The results showed that the development of arthritis was attenuated in systemic Cnn2-KO mice with significantly reduced inflammation and bone erosion than that in age- and stain background-matched C57BL/6 wild-type mice. In vitro differentiation of calponin 2-null mouse bone marrow cells produced fewer osteoclasts with decreased bone resorption. The attenuation of inflammatory arthritis was confirmed in conditional myeloid cell-specific Cnn2-KO mice. The increased phagocytotic activity of calponin 2-null macrophages may facilitate the clearance of autoimmune complexes and the resolution of inflammation, whereas the decreased substrate adhesion may reduce osteoclastogenesis and bone resorption. The data suggest that calponin 2 regulation of cytoskeleton function plays a novel role in the pathogenesis of inflammatory arthritis, implicating a potentially therapeutic target.


Subject(s)
Arthritis/genetics , Arthritis/pathology , Calcium-Binding Proteins/genetics , Inflammation/genetics , Inflammation/pathology , Macrophages/metabolism , Microfilament Proteins/genetics , Animals , Arthritis/metabolism , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Calcium-Binding Proteins/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Gene Deletion , Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/metabolism , Humans , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Monocytes/metabolism , Monocytes/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Phagocytosis/genetics , Phagocytosis/physiology , Calponins
7.
Calcif Tissue Int ; 96(4): 313-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25673503

ABSTRACT

Obesity is a severe health problem in children, afflicting several organ systems including bone. However, the role of obesity on bone homeostasis and bone cell function in children has not been studied in detail. Here we used young mice fed a high-fat diet (HFD) to model childhood obesity and investigate the effect of HFD on the phenotype of cells within the bone marrow environment. Five-week-old male mice were fed a HFD for 3, 6, and 12 weeks. Decreased bone volume was detected after 3 weeks of HFD treatment. After 6 and 12 weeks, HFD-exposed mice had less bone mass and increased osteoclast numbers. Bone marrow cells, but not spleen cells, from HFD-fed mice had increased osteoclast precursor frequency, elevated osteoclast formation, and bone resorption activity, as well as increased expression of osteoclastogenic regulators including RANKL, TNF, and PPAR-gamma. Bone formation rate and osteoblast and adipocyte numbers were also increased in HFD-fed mice. Isolated bone marrow cells also had a corresponding elevation in the expression of positive regulators of osteoblast and adipocyte differentiation. Our findings indicate that in juvenile mice, HFD-induced bone loss is mainly due to increased osteoclast bone resorption by affecting the bone marrow microenvironment. Thus, targeting osteoclast formation may present a new therapeutic approach for bone complications in obese children.


Subject(s)
Bone Marrow/pathology , Bone Resorption/physiopathology , Diet, High-Fat/adverse effects , Osteoclasts/cytology , Adipocytes/cytology , Animals , Biomarkers/blood , Blood Glucose/analysis , Bone Density , Bone Marrow/metabolism , Bone and Bones/pathology , Cell Differentiation , Cell Separation , Flow Cytometry , Leukocyte Common Antigens/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Osteoblasts/cytology , Osteoclasts/metabolism , PPAR gamma/metabolism , RANK Ligand/metabolism , Spleen/metabolism , Tumor Necrosis Factor-alpha/metabolism , X-Ray Microtomography
8.
J Biol Chem ; 288(31): 22359-68, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23782702

ABSTRACT

Itch is a ubiquitin E3 ligase that regulates protein stability. Itch(-/-) mice develop an autoimmune disease phenotype characterized by itchy skin and multiorgan inflammation. The role of Itch in the regulation of osteoclast function has not been examined. We report that Itch(-/-) bone marrow and spleen cells formed more osteoclasts than cells from WT littermates in response to receptor activator of NF-κB ligand (RANKL) and was associated with increased expression of the osteoclastogenic transcription factors c-fos and Nfatc1. Overexpression of Itch in Itch(-/-) cells rescued increased osteoclastogenesis. RANKL increased Itch expression, which can be blocked by a NF-κB inhibitor. The murine Itch promoter contains NF-κB binding sites. Overexpression of NF-κB p65 increased Itch expression, and RANKL promoted the binding of p65 onto the NF-κB binding sites in the Itch promoter. Itch(-/-) osteoclast precursors had prolonged RANKL-induced NF-κB activation and delayed TNF receptor-associated factor 6 (TRAF6) deubiquitination. In WT osteoclast precursors, Itch bound to TRAF6 and the deubiquitinating enzyme cylindromatosis. Adult Itch(-/-) mice had normal bone volume, but they had significantly increased LPS-induced osteoclastogenesis and bone resorption. Thus, Itch is a new RANKL target gene that is induced during osteoclastogenesis. Itch interacts with the deubiquitinating enzyme and is required for deubiquitination of TRAF6, thus limiting RANKL-induced osteoclast formation.


Subject(s)
Osteoclasts/cytology , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Base Sequence , DNA Primers , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Polymerase Chain Reaction , RANK Ligand/metabolism , Signal Transduction , Ubiquitination
9.
Blood ; 119(2): 540-50, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-21957195

ABSTRACT

Pancytopenia is a major cause of morbidity in acute myeloid leukemia (AML), yet its cause is unclear. Normal osteoblastic cells have been shown to support hematopoiesis. To define the effects of leukemia on osteoblastic cells, we used an immunocompetent murine model of AML. Leukemic mice had inhibition of osteoblastic cells, with decreased serum levels of the bone formation marker osteocalcin. Osteoprogenitor cells and endosteal-lining osteopontin(+) cells were reduced, and osteocalcin mRNA in CD45(-) marrow cells was diminished. This resulted in severe loss of mineralized bone. Osteoclasts were only transiently increased without significant increases in bone resorption, and their inhibition only partially rescued leukemia-induced bone loss. In vitro data suggested that a leukemia-derived secreted factor inhibited osteoblastic cells. Because the chemokine CCL-3 was recently reported to inhibit osteoblastic function in myeloma, we tested its expression in our model and in AML patients. Consistent with its potential novel role in leukemic-dependent bone loss, CCL-3 mRNA was significantly increased in malignant marrow cells from leukemic mice and from samples from AML patients. Based on these results, we propose that therapeutic mitigation of leukemia-induced uncoupling of osteoblastic and osteoclastic cells may represent a novel approach to promote normal hematopoiesis in patients with myeloid neoplasms.


Subject(s)
Chemokine CCL3/metabolism , Disease Models, Animal , Hematopoiesis , Leukemia, Myeloid/pathology , Osteoblasts/pathology , Osteoclasts/pathology , Animals , Blotting, Western , Bone Density Conservation Agents/pharmacology , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Resorption/drug therapy , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chemokine CCL3/genetics , Diphosphonates/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Imidazoles/pharmacology , Immunocompetence , Immunoenzyme Techniques , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Male , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism , Spleen/pathology , Zoledronic Acid
10.
Stem Cells ; 31(8): 1574-83, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23606569

ABSTRACT

Itch, a HECT family E3 ligase, affects numerous cell functions by regulating ubiquitination and proteasomal degradation of target proteins. However, the role of Itch in osteoblasts has not been investigated. We report that Itch(-/-) mice have significantly increased bone volume, osteoblast numbers, and bone formation rate. Using bone marrow stromal cells from Itch(-/-) mice and wild-type (WT) littermates as bone marrow mesenchymal precursor cells (BM-MPCs), we found that BM-MPCs from Itch(-/-) mice have compatible numbers of cells expressing mesenchymal stem cell markers. However, Itch(-/-) BM-MPCs grew faster in an in vitro culture, formed more CFU-F mesenchymal colonies, and exhibited increased osteoblast differentiation and decreased adipogenesis. Importantly, Itch(-/-) mesenchymal colony cells formed significantly more new bone in a tibial defect of recipient mice compared with WT cells. The expression levels of JunB, an AP-1 transcription factor that positively regulate osteoblast differentiation, were significantly increased in Itch(-/-) BM-MPCs when proteasome function is intact. In contrast, the amount of ubiquitinated JunB protein was markedly decreased in Itch(-/-) cells when proteasome function was blocked. Overexpression of WT Itch, but not an Itch ligase-inactive mutant, rescued differentiation defects of Itch(-/-) BM-MPCs. Itch(-/-) BM-MPCs had a similar role in immune modulation as WT cells. Thus, Itch negatively controls osteoblast differentiation from BM-MPCs through the regulation of proteasomal degradation of positive osteoblast regulator JunB protein. Itch is a potential new target for bone anabolic drug development to treat patients with bone loss.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Osteoblasts/cytology , Osteoblasts/enzymology , Stem Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Stem Cells/cytology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
11.
Arthritis Rheum ; 65(1): 130-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23002006

ABSTRACT

OBJECTIVE: B cell depletion therapy ameliorates rheumatoid arthritis by mechanisms that are incompletely understood. Arthritis flare in tumor necrosis factor (TNF)-transgenic mice is associated with efferent lymph node (LN) "collapse," triggered by B cell translocation into lymphatic spaces and decreased lymphatic drainage. The aim of this study was to examine whether the efficacy of B cell depletion therapy is associated with restoration of lymphatic drainage due to removal of obstructing nodal B cells. METHODS: We used contrast-enhanced magnetic resonance imaging, indocyanine green near-infrared imaging, and intravital immunofluorescence imaging to longitudinally assess synovitis, lymphatic flow, and cell migration in lymphatic vessels in TNF-transgenic mice. We conducted tests to determine whether the efficacy of B cell depletion therapy is associated with restoration of lymphatic draining and cell egress from arthritic joints. RESULTS: Unlike active lymphatics to normal and prearthritic knees, afferent lymphatic vessels to collapsed LNs in inflamed knees do not pulse. Intravital immunofluorescence imaging demonstrated that CD11b+ monocyte/macrophages in lymphatic vessels afferent to expanding LNs travel at high velocity (mean±SD 186±37 µm/second), while these cells are stationary in lymphatic vessels afferent to collapsed popliteal LNs. B cell depletion therapy for arthritis flares in TNF-transgenic mice significantly decreased knee synovium volume (by 50% from the baseline level) and significantly increased lymphatic clearance compared with placebo (P<0.05). This increased lymphatic drainage restored macrophage egress from inflamed joints without recovery of the lymphatic pulse. CONCLUSION: These results support a novel mechanism in which B cell depletion therapy for joint arthritis flares lessens inflammation by increasing lymphatic drainage and subsequent migration of cells and cytokines from the synovial space.


Subject(s)
Arthritis, Rheumatoid/therapy , Knee Joint/pathology , Lymphatic Vessels/pathology , Lymphocyte Depletion/methods , Synovitis/pathology , Animals , Arthritis, Rheumatoid/pathology , B-Lymphocytes , CD11b Antigen , Flow Cytometry , Immunohistochemistry , Lymph Nodes , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Microscopy, Fluorescence , Treatment Outcome
12.
J Zhejiang Univ Sci B ; 25(1): 38-50, 2024 Jan 15.
Article in English, Zh | MEDLINE | ID: mdl-38163665

ABSTRACT

Gorham-Stout disease (GSD) is a sporadic chronic disease characterized by progressive bone dissolution, absorption, and disappearance along with lymphatic vessel infiltration in bone-marrow cavities. Although the osteolytic mechanism of GSD has been widely studied, the cause of lymphatic hyperplasia in GSD is rarely investigated. In this study, by comparing the RNA expression profile of osteoclasts (OCs) with that of OC precursors (OCPs) by RNA sequencing, we identified a new factor, semaphorin 3A (Sema3A), which is an osteoprotective factor involved in the lymphatic expansion of GSD. Compared to OCPs, OCs enhanced the growth, migration, and tube formation of lymphatic endothelial cells (LECs), in which the expression of Sema3A is low compared to that in OCPs. In the presence of recombinant Sema3A, the growth, migration, and tube formation of LECs were inhibited, further confirming the inhibitory effect of Sema3A on LECs in vitro. Using an LEC-induced GSD mouse model, the effect of Sema3A was examined by injecting lentivirus-expressing Sema3A into the tibiae in vivo. We found that the overexpression of Sema3A in tibiae suppressed the expansion of LECs and alleviated bone loss, whereas the injection of lentivirus expressing Sema3A short hairpin RNA (shRNA) into the tibiae caused GSD-like phenotypes. Histological staining further demonstrated that OCs decreased and osteocalcin increased after Sema3A lentiviral treatment, compared with the control. Based on the above results, we propose that reduced Sema3A in OCs is one of the mechanisms contributing to the pathogeneses of GSD and that expressing Sema3A represents a new approach for the treatment of GSD.


Subject(s)
Lymphatic Vessels , Osteolysis, Essential , Semaphorin-3A , Animals , Mice , Endothelial Cells/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Osteolysis, Essential/metabolism , Osteolysis, Essential/pathology , Semaphorin-3A/metabolism
13.
J Orthop Translat ; 45: 66-74, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38511124

ABSTRACT

Background: The musculoskeletal system contains an extensive network of lymphatic vessels. Decreased lymph flow of the draining collecting lymphatics usually occurs in clinic after traumatic fractures. However, whether defects in lymphatic drainage can affect fracture healing is unclear. Methods: To investigate the effect of lymphatic dysfunction on fracture healing, we used a selective VEGFR3 tyrosine kinase inhibitor to treat tibial fractured mice for 5 weeks versus a vehicle-treated control. To ensure successfully establishing deceased lymphatic drainage model for fractured mice, we measured lymphatic clearance by near infrared indocyanine green lymphatic imaging (NIR-ICG) and the volume of the draining popliteal lymph nodes (PLNs) by ultrasound at the whole phases of fracture healing. In addition, hindlimb edema from day 0 to day 7 post-fracture, pain sensation by Hargreaves test at day 1 post-fracture, bone histomorphometry by micro-CT and callus composition by Alcian Blue-Hematoxylin/Orange G staining at day 14 post-fracture, and bone quality by biomechanical testing at day 35 post-fracture were applied to evaluate fracture healing. To promote fracture healing via increasing lymphatic drainage, we then treated fractured mice with anti-mouse podoplanin (PDPN) neutralizing antibody or isotype IgG antibody for 1 week to observe lymphatic drainage function and assess bone repair as methods described above. Results: Compared to vehicle-treated group, SAR-treatment group significantly decreased lymphatic clearance and the volume of draining PLNs. SAR-treatment group significantly increased soft tissue swelling, and reduced bone volume (BV)/tissue volume (TV), trabecular number (Tb.N), woven bone and biomechanical properties of fracture callus. In addition, anti-PDPN treated group significantly reduced the number of CD41+ platelets in PLNs and increased the number of pulsatile lymphatic vessels, lymphatic clearance and the volume of PLNs. Moreover, anti-PDPN treated group significantly reduced hindlimb edema and pain sensation and increased BV/TV, trabecular number (Tb.Th), woven bone and biomechanical properties of fracture callus. Conclusions: Inhibition of proper lymphatic drainage function delayed fracture healing. Use of a anti-PDPN neutralizing antibody reduced lymphatic platelet thrombosis (LPT), increased lymphatic drainage and improved fracture healing. The translational potential of this article: (1) We demonstrated lymphatic drainage function is crucial for fracture healing. (2) To unblock the lymphatic drainage and prevent the risk of bleeding and mortality by blood thinner, we demonstrated PDPN neutralizing antibody is a novel and safe way forward in the treatment of bone fracture healing by eliminating LPT and increasing lymphatic drainage.

14.
PLoS One ; 19(7): e0305623, 2024.
Article in English | MEDLINE | ID: mdl-38968295

ABSTRACT

BACKGROUND: Development of reliable disease activity biomarkers is critical for diagnostics, prognostics, and novel drug development. Although computed tomography (CT) is the gold-standard for quantification of bone erosions, there are no consensus approaches or rationales for utilization of specific outcome measures of erosive arthritis in complex joints. In the case of preclinical models, such as sexually dimorphic tumor necrosis factor transgenic (TNF-Tg) mice, disease severity is routinely quantified in the ankle through manual segmentation of the talus or small regions of adjacent bones primarily due to the ease in measurement. Herein, we sought to determine the particular hindpaw bones that represent reliable biomarkers of sex-dependent disease progression to guide future investigation and analysis. METHODS: Hindpaw micro-CT was performed on wild-type (n = 4 male, n = 4 female) and TNF-Tg (n = 4 male, n = 7 female) mice at monthly intervals from 2-5 (females) and 2-8-months (males) of age, since female TNF-Tg mice exhibit early mortality from cardiopulmonary disease at approximately 5-6-months. Further, 8-month-old WT (n = 4) and TNF-Tg males treated with anti-TNF monoclonal antibodies (n = 5) or IgG placebo isotype controls (n = 6) for 6-weeks were imaged with micro-CT every 3-weeks. For image analysis, we utilized our recently developed high-throughput and semi-automated segmentation strategy in Amira software. Synovial and osteoclast histology of ankle joints was quantified using Visiopharm. RESULTS: First, we demonstrated that the accuracy of automated segmentation, determined through analysis of ~9000 individual bones by a single user, was comparable in wild-type and TNF-Tg hindpaws before correction (79.2±8.9% vs 80.1±5.1%, p = 0.52). Compared to other bone compartments, the tarsal region demonstrated a sudden, specific, and significant bone volume reduction in female TNF-Tg mice, but not in males, by 5-months (4-months 4.3± 0.22 vs 5-months 3.4± 0.62 mm3, p<0.05). Specifically, the cuboid showed significantly reduced bone volumes at early timepoints compared to other tarsals (i.e., 4-months: Cuboid -24.1±7.2% vs Talus -9.0±5.9% of 2-month baseline). Additional bones localized to the anterolateral region of the ankle also exhibited dramatic erosions in the tarsal region of females, coinciding with increased synovitis and osteoclasts. In TNF-Tg male mice with severe arthritis, the talus and calcaneus exhibited the most sensitive response to anti-TNF therapy measured by effect size of bone volume change over treatment period. CONCLUSIONS: We demonstrated that sexually dimorphic changes in arthritic hindpaws of TNF-Tg mice are bone-specific, where the cuboid serves as a reliable early biomarker of erosive arthritis in female mice. Adoption of automated segmentation approaches in pre-clinical or clinical models has potential to translate quantitative biomarkers to monitor bone erosions in disease and evaluate therapeutic efficacy.


Subject(s)
Biomarkers , Mice, Transgenic , Tumor Necrosis Factor-alpha , X-Ray Microtomography , Animals , Female , Male , Mice , X-Ray Microtomography/methods , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Disease Models, Animal , Sex Factors , Mice, Inbred C57BL , Sex Characteristics
15.
Genesis ; 51(9): 667-75, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23775847

ABSTRACT

The troponin complex, which consists of three regulatory proteins (troponin C, troponin I, and troponin T), is known to regulate muscle contraction in skeletal and cardiac muscle, but its role in smooth muscle remains controversial. Troponin T3 (TnnT3) is a fast skeletal muscle troponin believed to be expressed only in skeletal muscle cells. To determine the in vivo function and tissue-specific expression of Tnnt3, we obtained the heterozygous Tnnt3+/flox/lacZ mice from Knockout Mouse Project (KOMP) Repository. Tnnt3(lacZ/+) mice are smaller than their WT littermates throughout development but do not display any gross phenotypes. Tnnt3(lacZ/lacZ) embryos are smaller than heterozygotes and die shortly after birth. Histology revealed hemorrhagic tissue in Tnnt3(lacZ/lacZ) liver and kidney, which was not present in Tnnt3(lacZ/+) or WT, but no other gross tissue abnormalities. X-gal staining for Tnnt3 promoter-driven lacZ transgene expression revealed positive staining in skeletal muscle and diaphragm and smooth muscle cells located in the aorta, bladder, and bronchus. Collectively, these findings suggest that troponins are expressed in smooth muscle and are required for normal growth and breathing for postnatal survival. Moreover, future studies with this mouse model can explore TnnT3 function in adult muscle function using the conditional-inducible gene deletion approach


Subject(s)
Muscle, Smooth/metabolism , Troponin T/metabolism , Animals , Kidney/abnormalities , Kidney/growth & development , Liver/abnormalities , Liver/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Muscle, Smooth/growth & development , Phenotype , Transgenes , Troponin , Troponin T/genetics
16.
Cell Metab ; 7(4): 283-5, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18396132

ABSTRACT

Enhanced and deficient immune responses are associated with abnormal bone homeostasis. A new study by Shinohara et al. (2008) shows that protein phosphorylation by the tyrosine kinases Bruton and Tec links immunity and bone as well as two signaling pathways in precursors of osteoclasts, the cells that degrade bone.


Subject(s)
Bone and Bones/cytology , Bone and Bones/immunology , Osteoblasts/metabolism , Osteoclasts/metabolism , Protein-Tyrosine Kinases/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Bone and Bones/enzymology , Cell Differentiation , Humans , Mice , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoclasts/cytology , Osteoclasts/enzymology
17.
Stem Cells ; 30(4): 709-18, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22290873

ABSTRACT

RelB and nuclear factor κB (NF-κB2) are the main effectors of NF-κB noncanonical signaling and play critical roles in many physiological processes. However, their role in hematopoietic stem/progenitor cell (HSPC) maintenance has not been characterized. To investigate this, we generated RelB/NF-κB2 double-knockout (dKO) mice and found that dKO HSPCs have profoundly impaired engraftment and self-renewal activity after transplantation into wild-type recipients. Transplantation of wild-type bone marrow cells into dKO mice to assess the role of the dKO microenvironment showed that wild-type HSPCs cycled more rapidly, were more abundant, and had developmental aberrancies: increased myeloid and decreased lymphoid lineages, similar to dKO HSPCs. Notably, when these wild-type cells were returned to normal hosts, these phenotypic changes were reversed, indicating a potent but transient phenotype conferred by the dKO microenvironment. However, dKO bone marrow stromal cell numbers were reduced, and bone-lining niche cells supported less HSPC expansion than controls. Furthermore, increased dKO HSPC proliferation was associated with impaired expression of niche adhesion molecules by bone-lining cells and increased inflammatory cytokine expression by bone marrow cells. Thus, RelB/NF-κB2 signaling positively and intrinsically regulates HSPC self-renewal and maintains stromal/osteoblastic niches and negatively and extrinsically regulates HSPC expansion and lineage commitment through the marrow microenvironment.


Subject(s)
Cellular Microenvironment , Hematopoietic Stem Cells/cytology , NF-kappa B/metabolism , Signal Transduction , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Size , Cellular Microenvironment/drug effects , Cytokines/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Signal Transduction/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Transcription Factor RelB/metabolism
18.
Arthritis Rheum ; 64(11): 3649-59, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22832945

ABSTRACT

OBJECTIVE: Glucocorticoid (GC) therapy is associated with increased risk of fracture in patients with rheumatoid arthritis (RA). To elucidate the cause of this increased risk, we examined the effects of chronic erosive inflammatory arthritis and GC treatment on bone quality, structure, and biomechanical properties in a murine model. METHODS: Mice with established arthritis and expressing human tumor necrosis factor α (TNFα) transgene (Tg) and their wild-type (WT) littermates were continually treated with GC (prednisolone 5 mg/kg/day via subcutaneous controlled-release pellet) or placebo for 14, 28, or 42 days. Microstructure, biomechanical properties, chemical composition, and morphology of the tibiae and lumbar vertebral bodies were assessed by micro-computed tomography, biomechanical testing, Raman spectroscopy, and histology, respectively. Serum markers of bone turnover were also determined. RESULTS: TNF-Tg and GC treatment additively decreased mechanical strength and stiffness in both the tibiae and the vertebral bodies. GC treatment in the TNF-Tg mice increased the ductility of tibiae under torsional loading. These changes were associated with significant alterations in the biochemical and structural composition of the mineral and organic components of the bone matrix, a decrease in osteoblast activity and bone formation, and an increase in osteoclast activity. CONCLUSION: Our findings indicate that the concomitant decrease in bone strength and increase in bone ductility associated with chronic inflammation and GC therapy, coupled with the significant changes in the bone quality and structure, may increase the susceptibility of the bone to failure under low-energy loading. This may explain the mechanism of symptomatic insufficiency fractures in patients with RA receiving GC therapy who do not have radiographic manifestations of fracture.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Fractures, Bone/pathology , Glucocorticoids/adverse effects , Prednisolone/adverse effects , Animals , Arthritis, Rheumatoid/epidemiology , Biomechanical Phenomena/drug effects , Bone Remodeling/drug effects , Disease Models, Animal , Fractures, Bone/epidemiology , Glucocorticoids/administration & dosage , Humans , Male , Mice , Mice, Transgenic , Prednisolone/administration & dosage , Risk Factors , Spectrum Analysis, Raman , Spine/diagnostic imaging , Spine/drug effects , Spine/pathology , Tibia/diagnostic imaging , Tibia/drug effects , Tibia/pathology , Transgenes/genetics , Tumor Necrosis Factor-alpha/genetics , X-Ray Microtomography
19.
Endocrinol Metab (Seoul) ; 38(5): 504-521, 2023 10.
Article in English | MEDLINE | ID: mdl-37749800

ABSTRACT

Maintenance of skeletal integrity requires the coordinated activity of multinucleated bone-resorbing osteoclasts and bone-forming osteoblasts. Osteoclasts form resorption lacunae on bone surfaces in response to cytokines by fusion of precursor cells. Osteoblasts are derived from mesenchymal precursors and lay down new bone in resorption lacunae during bone remodeling. Nuclear factorkappa B (NF-κB) signaling regulates osteoclast and osteoblast formation and is activated in osteoclast precursors in response to the essential osteoclastogenic cytokine, receptor activator of NF-κB ligand (RANKL), which can also control osteoblast formation through RANK-RANKL reverse signaling in osteoblast precursors. RANKL and some pro-inflammatory cytokines, including tumor necrosis factor (TNF), activate NF-κB signaling to positively regulate osteoclast formation and functions. However, these cytokines also limit osteoclast and osteoblast formation through NF-κB signaling molecules, including TNF receptor-associated factors (TRAFs). TRAF6 mediates RANKL-induced osteoclast formation through canonical NF-κB signaling. In contrast, TRAF3 limits RANKL- and TNF-induced osteoclast formation, and it restricts transforming growth factor ß (TGFß)-induced inhibition of osteoblast formation in young and adult mice. During aging, neutrophils expressing TGFß and C-C chemokine receptor type 5 (CCR5) increase in bone marrow of mice in response to increased NF-κB-induced CC motif chemokine ligand 5 (CCL5) expression by mesenchymal progenitor cells and injection of these neutrophils into young mice decreased bone mass. TGFß causes degradation of TRAF3, resulting in decreased glycogen synthase kinase-3ß/ß-catenin-mediated osteoblast formation and age-related osteoporosis in mice. The CCR5 inhibitor, maraviroc, prevented accumulation of TGFß+/CCR5+ neutrophils in bone marrow and increased bone mass by inhibiting bone resorption and increasing bone formation in aged mice. This paper updates current understanding of how NF-κB signaling is involved in the positive and negative regulation of cytokine-mediated osteoclast and osteoblast formation and activation with a focus on the role of TRAF3 signaling, which can be targeted therapeutically to enhance bone mass.


Subject(s)
NF-kappa B , Osteogenesis , Mice , Animals , NF-kappa B/metabolism , TNF Receptor-Associated Factor 3/metabolism , Ligands , Osteoclasts/metabolism , Osteoclasts/pathology , Transforming Growth Factor beta/metabolism
20.
Front Endocrinol (Lausanne) ; 14: 1090049, 2023.
Article in English | MEDLINE | ID: mdl-36875448

ABSTRACT

Cellular senescence plays important roles in age-related diseases, including musculoskeletal disorders. Senescent cells (SCs) exert a senescence-associated secretory phenotype (SASP) by producing SASP factors, some of which overlap with factors produced by inflammatory cells (Inf-Cs). However, the differences between SCs and Inf-Cs and how they interact with each other during fracture repair have not been well studied. Here, we analyzed single cell RNA sequencing data of aged mouse fracture callus stromal cells. We defined Inf-Cs as cells that express NF-κB Rela/Relb, SCs as cells that express the senescence genes, Cdkn1a, Cdkn2a or Cdkn2c, and inflammatory SCs (Inf-SCs) as cells that express both NF-κB and senescence genes. Differentially expressed genes and pathway analyses revealed that Inf-SCs and SCs had a similar gene expression profile and upregulated pathways that are related to DNA damage/oxidation-reduction and cellular senescence, while Inf-Cs expressed different gene signatures and pathways from SCs and Inf-SCs, mainly related to inflammation. Cellchat software analysis indicated that SCs and Inf-SCs are potential ligand-producing cells that affect Inf-Cs as target cells. Cell culture experiments demonstrated that SC conditioned medium promoted inflammatory gene expression by callus-derived mesenchymal progenitor cells, and Inf-Cs had reduced osteoblast differentiation capacity. In summary, we have identified three cell subclusters associated with inflammation and senescence in callus stromal cells, predicted potential effects of Inf-SCs and SCs on Inf-Cs by production of active ligands, and demonstrated that when mesenchymal progenitors acquire inflammatory phenotypes their osteogenic potential is reduced.


Subject(s)
Fractures, Bone , NF-kappa B , Animals , Mice , Stromal Cells , Osteogenesis , Cellular Senescence
SELECTION OF CITATIONS
SEARCH DETAIL