Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Front Bioeng Biotechnol ; 12: 1308161, 2024.
Article in English | MEDLINE | ID: mdl-38433822

ABSTRACT

Background: Osteoadsorptive fluorogenic sentinel 3 (OFS-3) is a recently described compound that contains a bone-targeting bisphosphonate (BP) and cathepsin K (Ctsk)-triggered fluorescence signal. A prior study in a murine Achilles repair model demonstrated its effectiveness at targeting the site of tendon-to-bone repair, but the intrinsic effect of this novel bisphosphonate chaperone on tendon-to-bone healing has not been previously explored. We hypothesized that application of this bisphosphonate-fluorophore cargo conjugate would not affect the biomechanical properties or histologic appearance of tendon-bone repairs. Materials and Methods: Right hindlimb Achilles tendon-to-bone repair was performed on 12-week old male mice. Animals were divided into 2 groups of 18 each: 1) Achilles repair with OFS-3 applied directly to the repair site prior to closure, and 2) Achilles repair with saline applied prior to closure. Repaired hindlimbs from 12 animals per group were harvested at 6 weeks for biomechanical analysis with a custom 3D-printed jig. At 4 and 6 weeks, repaired hindlimbs from the remaining animals were assessed histologically using H&E, immunohistochemistry (IHC) staining for the presence of Ctsk, and second harmonic generation (SHG) imaging to evaluate collagen fibers. Results: At 6 weeks, there was no significant difference in failure load, stiffness, toughness, or displacement to failure between repaired hindlimbs that received OFS-3 versus saline. There was no difference in tissue healing on H&E or Ctsk staining on immunohistochemistry between animals that received OFS-3 versus saline. Finally, second harmonic generation imaging demonstrated no difference in collagen fiber parameters between the two groups. Conclusion: OFS-3 did not significantly affect the biomechanical properties or histologic appearance of murine Achilles tendon-to-bone repairs. This study demonstrates that OFS-3 can target the site of tendon-to-bone repair without causing intrinsic negative effects on healing. Further development of this drug delivery platform to target growth factors to the site of tendon-bone repair is warranted.

3.
J Orthop Res ; 41(10): 2250-2260, 2023 10.
Article in English | MEDLINE | ID: mdl-37087676

ABSTRACT

Tendon injuries are common and often treated surgically, however, current tendon repair healing results in poorly organized fibrotic tissue. While certain growth factors have been reported to improve both the strength and organization of the repaired enthesis, their clinical applicability is severely limited due to a lack of appropriate delivery strategies. In this study, we evaluated a recently developed fluorescent probe, Osteoadsorptive Fluorogenic Sentinel-3 that is composed of a bone-targeting bisphosphonate (BP) moiety linked to fluorochrome and quencher molecules joined via a cathepsin K-sensitive peptide sequence. Using a murine Achilles tendon-to-bone repair model, BP-based and/or Ctsk-coupled imaging probes were applied either locally or systemically. Fluorescence imaging was used to quantify the resultant signal in vivo. After tendon-bone repair, animals that received either local or systemic administration of imaging probes demonstrated significantly higher fluorescence signal at the repair site compared to the sham surgery group at all time points (p < 0.001), with signal peaking at 7-10 days after surgery. Our findings demonstrate the feasibility of using a novel BP-based targeting and Ctsk-activated delivery of molecules to the site of tendon-to-bone repair and creates a foundation for further development of this platform as an effective strategy to deliver bioactive molecules to sites of musculoskeletal injury.


Subject(s)
Plastic Surgery Procedures , Tendon Injuries , Rats , Animals , Mice , Wound Healing , Rats, Sprague-Dawley , Tendon Injuries/diagnostic imaging , Tendon Injuries/drug therapy , Tendon Injuries/surgery , Tendons/surgery
4.
J Bone Miner Res ; 35(11): 2289-2300, 2020 11.
Article in English | MEDLINE | ID: mdl-32634285

ABSTRACT

CCN1/Cyr61 is a dynamically expressed matricellular protein that serves regulatory functions in multiple tissues. Previous studies from our laboratory demonstrated that CCN1 regulates bone maintenance. Using an osteoblast and osteocyte conditional knockout mouse model (Ccn1OCN ), we found a significant decrease in trabecular and cortical bone mass in vivo, in part through suppression of Wnt signaling since the expression of the Wnt antagonist sclerostin (SOST) is increased in osteoblasts lacking CCN1. It has been established that parathyroid hormone (PTH) signaling also suppresses SOST expression in bone. We therefore investigated the interaction between CCN1 and PTH-mediated responses in this study. We find that loss of Ccn1 in osteoblasts leads to impaired responsiveness to anabolic intermittent PTH treatment in Ccn1OCN mice in vivo and in osteoblasts from these mice in vitro. Analysis of Ccn1OCN mice demonstrated a significant decrease in parathyroid hormone receptor-1 (PTH1R) expression in osteoblasts in vivo and in vitro. We investigated the regulatory role of a non-canonical integrin-binding domain of CCN1 because several studies indicate that specific integrins are critical to mechanotransduction, a PTH-dependent response, in bone. These data suggest that CCN1 regulates the expression of PTH1R through interaction with the αvß3 and/or αvß5 integrin complexes. Osteoblasts that express a mutant form of CCN1 that cannot interact with αvß3/ß5 integrin demonstrate a significant decrease in mRNA and protein expression of both PTH1R and αv integrin. Overall, these data suggest that the αvß3/ß5-binding domain of CCN1 is required to endow PTH signaling with anabolic activity in bone cells. © 2020 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Cysteine-Rich Protein 61/physiology , Mechanotransduction, Cellular , Osteoblasts/cytology , Parathyroid Hormone , Animals , Mice , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1 , Wnt Signaling Pathway
5.
EMBO Rep ; 21(8): e48462, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32558157

ABSTRACT

At the neuromuscular junction (NMJ), lipoprotein-related receptor 4 (LRP4) mediates agrin-induced MuSK phosphorylation that leads to clustering of acetylcholine receptors (AChRs) in the postsynaptic region of the skeletal muscle. Additionally, the ectodomain of LRP4 is necessary for differentiation of the presynaptic nerve terminal. However, the molecules regulating LRP4 have not been fully elucidated yet. Here, we show that the CT domain of connective tissue growth factor (CTGF/CCN2) directly binds to the third beta-propeller domain of LRP4. CTGF/CCN2 enhances the binding of LRP4 to MuSK and facilitates the localization of LRP4 on the plasma membrane. CTGF/CCN2 enhances agrin-induced MuSK phosphorylation and AChR clustering in cultured myotubes. Ctgf-deficient mouse embryos (Ctgf-/- ) have small AChR clusters and abnormal dispersion of synaptic vesicles along the motor axon. Ultrastructurally, the presynaptic nerve terminals have reduced numbers of active zones and mitochondria. Functionally, Ctgf-/- embryos exhibit impaired NMJ signal transmission. These results indicate that CTGF/CCN2 interacts with LRP4 to facilitate clustering of AChRs at the motor endplate and the maturation of the nerve terminal.


Subject(s)
Connective Tissue Growth Factor , LDL-Receptor Related Proteins , Agrin/genetics , Agrin/metabolism , Animals , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Mice , Neuromuscular Junction/metabolism , Phosphorylation
6.
JCI Insight ; 5(13)2020 07 09.
Article in English | MEDLINE | ID: mdl-32484792

ABSTRACT

Heterotopic ossification (HO) is defined as abnormal differentiation of local stromal cells of mesenchymal origin, resulting in pathologic cartilage and bone matrix deposition. Cyr61, CTGF, Nov (CCN) family members are matricellular proteins that have diverse regulatory functions on cell proliferation and differentiation, including the regulation of chondrogenesis. However, little is known regarding CCN family member expression or function in HO. Here, a combination of bulk and single-cell RNA sequencing defined the dynamic temporospatial pattern of CCN family member induction within a mouse model of trauma-induced HO. Among CCN family proteins, Wisp1 (also known as Ccn4) was most upregulated during the evolution of HO, and Wisp1 expression corresponded with chondrogenic gene profile. Immunohistochemistry confirmed WISP1 expression across traumatic and genetic HO mouse models as well as in human HO samples. Transgenic Wisp1LacZ/LacZ knockin animals showed an increase in endochondral ossification in HO after trauma. Finally, the transcriptome of Wisp1-null tenocytes revealed enrichment in signaling pathways, such as the STAT3 and PCP signaling pathways, that may explain increased HO in the context of Wisp1 deficiency. In sum, CCN family members, and in particular Wisp1, are spatiotemporally associated with and negatively regulate trauma-induced HO formation.


Subject(s)
CCN Intercellular Signaling Proteins/metabolism , Ossification, Heterotopic/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cartilage/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Ossification, Heterotopic/pathology , RNA, Messenger/metabolism , Signal Transduction/physiology
7.
Bone ; 137: 115439, 2020 08.
Article in English | MEDLINE | ID: mdl-32442550

ABSTRACT

The ligands that comprise the Transforming Growth Factor ß superfamily highly govern the development of the embryonic growth plate. Members of this superfamily activate canonical TGFß and/or BMP (Bone Morphogenetic Protein) signaling pathways. How these pathways interact with one another is an area of active investigation. These two signaling pathways have been described to negatively regulate one another through crosstalk involving Smad proteins, the primary intracellular effectors of canonical signaling. More recently, a mechanism for regulation of the BMP pathway through TGFß and BMP receptor interactions has been described. Here in this review, we demonstrate examples of how TGFß is a gatekeeper of BMP action in the developing growth plate at both the receptor and transcriptional levels.


Subject(s)
Bone Morphogenetic Proteins , Growth Plate , Transforming Growth Factor beta , Animals , Bone Morphogenetic Proteins/metabolism , Growth Plate/metabolism , Humans , Signal Transduction , Smad Proteins , Transforming Growth Factor beta/metabolism
8.
Proc Natl Acad Sci U S A ; 116(31): 15570-15579, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31311865

ABSTRACT

The type I TGFß receptor TGFßRI (encoded by Tgfbr1) was ablated in cartilage. The resulting Tgfbr1Col2 mice exhibited lethal chondrodysplasia. Similar defects were not seen in mice lacking the type II TGFß receptor or SMADs 2 and 3, the intracellular mediators of canonical TGFß signaling. However, we detected elevated BMP activity in Tgfbr1Col2 mice. As previous studies showed that TGFßRI can physically interact with ACVRL1, a type I BMP receptor, we generated cartilage-specific Acvrl1 (Acvrl1Col2 ) and Acvrl1/Tgfbr1 (Acvrl1/Tgfbr1Col2) knockouts. Loss of ACVRL1 alone had no effect, but Acvrl1/Tgfbr1Col2 mice exhibited a striking reversal of the chondrodysplasia seen in Tgfbr1Col2 mice. Loss of TGFßRI led to a redistribution of the type II receptor ACTRIIB into ACVRL1/ACTRIIB complexes, which have high affinity for BMP9. Although BMP9 is not produced in cartilage, we detected BMP9 in the growth plate, most likely derived from the circulation. These findings demonstrate that the major function of TGFßRI in cartilage is not to transduce TGFß signaling, but rather to antagonize BMP signaling mediated by ACVRL1.


Subject(s)
Cartilage/metabolism , Growth Differentiation Factor 2/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Growth Differentiation Factor 2/genetics , Mice , Mice, Knockout , Receptor, Transforming Growth Factor-beta Type I/genetics
9.
Curr Top Dev Biol ; 133: 153-170, 2019.
Article in English | MEDLINE | ID: mdl-30902251

ABSTRACT

Synovial joints enable movement and protect the integrity of the articular cartilage. Joints form within skeletal condensations destined to undergo chondrogenesis. The suppression of this chondrogenic program in the interzone is the first morphological sign of joint formation. While we have a fairly good understanding of the essential roles of BMP and TGFß family members in promoting chondrogenic differentiation in developing skeletal elements, we know very little about how BMP activity is suppressed specifically within the interzone, a crucial step in joint development. The function of the BMP ligand Gdf5 has been especially difficult to decipher. On the one hand, Gdf5 is required to promote chondrogenesis of articular elements. On the other hand, Gdf5 is highly expressed in the joint interzone where chondrogenesis must be suppressed for the formation of many joints. Here we review the evidence that BMP signaling must be suppressed within the joint interzone for joint morphogenesis to progress, and consider how Gdf5 exerts its divergent effects on chondrogenesis and joint formation. We also consider how TGFß signaling impacts formation of the interzone. Finally, we propose a model whereby Gdf5 exerts distinct effects in the interzone vs. surrounding cartilage based on the repertoire of BMP receptors available in these tissues. Understanding how BMP antagonists and counteracting TGFß signals intersect with Gdf5 to sculpt the joint interzone is essential for understanding the origin of osteoarthritis and other diseases of joint tissues.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Joints/metabolism , Transforming Growth Factor beta/metabolism , Animals , Growth Differentiation Factor 5/genetics , Humans , Joints/embryology , Models, Biological , Signal Transduction
10.
Ear Nose Throat J ; 97(9): 306-313, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30273430

ABSTRACT

Although acute laryngitis is common, it is often managed by primary physicians. Therefore, video images documenting its signs are scarce. This series includes 7 professional voice users who previously had undergone baseline strobovideolaryngscopy (SVL) during routine examinations or during evaluations for other complaints and who returned with acute laryngitis. Sequential SVL showed not only the expected erythema, edema, cough, and dysphonia, but also new masses in 5 of the 7 subjects. All the signs returned to baseline. This series is reported to highlight the reversible structural changes that can be expected in patients with acute laryngitis and the value of conservative management.


Subject(s)
Laryngitis/diagnosis , Laryngoscopy/methods , Respiratory Tract Infections/diagnosis , Stroboscopy/methods , Acute Disease , Adolescent , Adult , Cough/diagnosis , Cough/etiology , Dysphonia/diagnosis , Dysphonia/etiology , Erythema/diagnosis , Erythema/etiology , Female , Humans , Laryngitis/complications , Laryngitis/therapy , Male , Respiratory Tract Infections/complications , Respiratory Tract Infections/therapy , Video Recording , Young Adult
11.
J Cell Commun Signal ; 12(1): 271, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29455304

ABSTRACT

In the original publication's title CCN5/WISP5 should have been CCN5/WISP2.

12.
J Cell Commun Signal ; 12(1): 265-270, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29396648

ABSTRACT

CCN5/WISP2 is part of the CCN family of matricellular proteins, but is distinct in that it lacks the C-terminal (CT) domain. Although CCN5 has been shown to impact cell proliferation and differentiation in vitro, its role in vivo is unclear. We therefore generated mice using ES cells developed by the Knockout Mouse Project (KOMP) in which exons 2-5, which encode the all of the conserved protein coding regions, are replaced by a lacZ cassette. Ccn5 LacZ/LacZ mice were viable and apparently normal. Based on previous studies showing that CCN5 impacts osteoblast proliferation and differentiation, we performed an analysis of adult bone phenotype. LacZ expression was examined in adult bone, and was found to be strong within the periosteum, but not in trabecular bone or bone marrow. Micro-CT analysis revealed no apparent changes in bone mineral density (BMD) or bone tissue volume (BV/TV) in Ccn5 LacZ/LacZ mice. These studies indicate that CCN5 is not required for normal bone formation, but they do not rule out a role in mechanotransduction or repair processes. The availability of Ccn5 LacZ mice enables studies of CCN5 expression and function in multiple tissues.

13.
J Bone Miner Res ; 33(6): 1076-1089, 2018 06.
Article in English | MEDLINE | ID: mdl-29351359

ABSTRACT

CYR61/CCN1 is a matricellular protein that resides in the extracellular matrix, but serves regulatory rather than structural roles. CYR61/CCN1 is found in mineralized tissues and has been shown to influence bone healing in vivo and osteogenic differentiation in vitro. In this study we generated Cyr61 bone-specific knockout mice to examine the physiological role of CYR61/CCN1 in bone development and maintenance in vivo. Extensive analysis of Cyr61 conditional knockout mice showed a significant decrease in both trabecular and cortical bone mass as compared to WT littermates. Our data suggest that CYR61/CCN1 exerts its effects on mature osteoblast/osteocyte function to modulate bone mass. Specifically, changes were observed in osteocyte/osteoblast expression of RankL, VegfA, and Sost. The increase in RankL expression was correlated with a significant increase in osteoclast number; decreased VegfA expression was correlated with a significant decrease in bone vasculature; increased Sost expression was associated with decreased Wnt signaling, as revealed by decreased Axin2 expression and increased adiposity in the bone marrow. Although the decreased number of vascular elements in bone likely contributes to the low bone mass phenotype in Cyr61 conditional knockout mice, this cannot explain the observed increase in osteoclasts and the decrease in Wnt signaling. We conducted in vitro assays using UMR-106 osteosarcoma cells to explore the role CYR61/CCN1 plays in modulating Sost mRNA and protein expression in osteocytes and osteoblasts. Overexpression of CYR61/CCN1 can suppress Sost expression in both control and Cyr61 knockout cells, and blocking Sost with siRNA can rescue Wnt responsiveness in Cyr61 knockout cells in vitro. Overall, our data suggest that CYR61/CCN1 modulates mature osteoblast and osteocyte function to regulate bone mass through angiogenic effects as well as by modulating Wnt signaling, at least in part through the Wnt antagonist Sost. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Bone and Bones/metabolism , Cysteine-Rich Protein 61/metabolism , Glycoproteins/metabolism , Adaptor Proteins, Signal Transducing , Adiposity , Animals , Bone Marrow/metabolism , Bone and Bones/blood supply , Cancellous Bone/metabolism , Cortical Bone/metabolism , Female , Gene Deletion , Intercellular Signaling Peptides and Proteins , Male , Mice , Models, Biological , Osteoblasts/metabolism , Osteocytes/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wnt Signaling Pathway
14.
Methods Mol Biol ; 1489: 325-345, 2017.
Article in English | MEDLINE | ID: mdl-27734387

ABSTRACT

Two developments have greatly facilitated the construction of CCN mutant mouse strains. The first is the availability of modified embryonic stem (ES) cells and mice developed through several large-scale government-sponsored research programs. The second is the advent of CRISPR/Cas9 technology. In this chapter, we describe the available mouse strains generated by gene targeting techniques and the CCN targeting vectors and genetically modified ES cells that are available for the generation of CCN mutant mice. Many of these mutant mouse lines and ES cells carry a ß-galactosidase reporter that can be used to track CCN expression, facilitating phenotypic analysis and revealing new sites of CCN action. Therefore, we also describe a method for ß-galactosidase staining.


Subject(s)
CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/metabolism , Gene Expression , beta-Galactosidase/genetics , Alleles , Animals , CRISPR-Cas Systems , Embryonic Stem Cells/metabolism , Gene Order , Gene Targeting , Genes, Reporter , Genetic Engineering , Genetic Loci , Genetic Vectors/genetics , Homologous Recombination , Male , Mice , Mice, Knockout , Mutagenesis
15.
J Cell Commun Signal ; 11(1): 39-48, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27766493

ABSTRACT

Treatment with rhBMP7 exerts profound protective effects in a wide variety of experimental models of renal disease. However, little is known about how these protective effects are mediated, and which cells in the kidney are targeted by exogenous rhBMP7 treatment. To determine if rhBMP7 increases glomerular and tubulointerstitial canonical BMP signaling, we performed Unilateral Ureteral Obstruction (UUO, a widely used obstructive nephropathy model) in mice reporting transcriptional activity downstream of canonical BMP signaling by the expression of GFP under the BMP Responsive Element of the Id1 promoter (BRE:gfp mice). We also analysed the impact of rhBMP7 treatment on severity of the UUO phenotype, on TGFß signaling, and on expression of CCN2 (CTGF). Despite profound protective effects with respect to morphological damage, macrophage infiltration, and fibrosis, no significant difference in GFP-expression was observed upon rhBMP7 administration. Also TGFß signalling was similar in rhBMP7 and vehicle treated mice, but CCN2 expression in obstructed kidneys was significantly reduced by rhBMP7 treatment. Of note, in heterozygous CCN2 mice (CCN2+/-) treatment with rhBMP7 did not (further) reduce the severity of kidney damage in the UUO-model. These data suggest that protection against obstructive nephropathy by exogenous rhBMP7 treatment relies primarily on non-canonical BMP signaling, and may be mediated in large part by downregulation of CCN2 expression.

16.
PLoS Genet ; 12(10): e1006352, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27741240

ABSTRACT

TGFßs act through canonical and non-canonical pathways, and canonical signals are transduced via Smad2 and Smad3. However, the contribution of canonical vs. non-canonical pathways in cartilage is unknown because the role of Smad2 in chondrogenesis has not been investigated in vivo. Therefore, we analyzed mice in which Smad2 is deleted in cartilage (Smad2CKO), global Smad3-/- mutants, and crosses of these strains. Growth plates at birth from all mutant strains exhibited expanded columnar and hypertrophic zones, linked to increased proliferation in resting chondrocytes. Defects were more severe in Smad2CKO and Smad2CKO;Smad3-/- (Smad2/3) mutant mice than in Smad3-/- mice, demonstrating that Smad2 plays a role in chondrogenesis. Increased levels of Ihh RNA, a key regulator of chondrocyte proliferation and differentiation, were seen in prehypertrophic chondrocytes in the three mutant strains at birth. In accordance, TGFß treatment decreased Ihh RNA levels in primary chondrocytes from control (Smad2fx/fx) mice, but inhibition was impaired in cells from mutants. Consistent with the skeletal phenotype, the impact on TGFß-mediated inhibition of Ihh RNA expression was more severe in Smad2CKO than in Smad3-/- cells. Putative Smad2/3 binding elements (SBEs) were identified in the proximal Ihh promoter. Mutagenesis demonstrated a role for three of them. ChIP analysis suggested that Smad2 and Smad3 have different affinities for these SBEs, and that the repressors SnoN and Ski were differentially recruited by Smad2 and Smad3, respectively. Furthermore, nuclear localization of the repressor Hdac4 was decreased in growth plates of Smad2CKO and double mutant mice. TGFß induced association of Hdac4 with Smad2, but not with Smad3, on the Ihh promoter. Overall, these studies revealed that Smad2 plays an essential role in the development of the growth plate, that both Smads 2 and 3 inhibit Ihh expression in the neonatal growth plate, and suggested they accomplish this by binding to distinct SBEs, mediating assembly of distinct repressive complexes.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , Chondrogenesis/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Animals , Cartilage/growth & development , Cartilage/metabolism , Chondrocytes/metabolism , Gene Expression Regulation, Developmental , Growth Plate/growth & development , Hedgehog Proteins/genetics , Histone Deacetylases/genetics , Mice , Promoter Regions, Genetic , Transforming Growth Factor beta/genetics
17.
PLoS Genet ; 12(3): e1005936, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27019229

ABSTRACT

Spondylocarpotarsal synostosis (SCT) is an autosomal recessive disorder characterized by progressive vertebral fusions and caused by loss of function mutations in Filamin B (FLNB). FLNB acts as a signaling scaffold by linking the actin cytoskleteon to signal transduction systems, yet the disease mechanisms for SCT remain unclear. Employing a Flnb knockout mouse, we found morphologic and molecular evidence that the intervertebral discs (IVDs) of Flnb-/-mice undergo rapid and progressive degeneration during postnatal development as a result of abnormal cell fate changes in the IVD, particularly the annulus fibrosus (AF). In Flnb-/-mice, the AF cells lose their typical fibroblast-like characteristics and acquire the molecular and phenotypic signature of hypertrophic chondrocytes. This change is characterized by hallmarks of endochondral-like ossification including alterations in collagen matrix, expression of Collagen X, increased apoptosis, and inappropriate ossification of the disc tissue. We show that conversion of the AF cells into chondrocytes is coincident with upregulated TGFß signaling via Smad2/3 and BMP induced p38 signaling as well as sustained activation of canonical and noncanonical target genes p21 and Ctgf. These findings indicate that FLNB is involved in attenuation of TGFß/BMP signaling and influences AF cell fate. Furthermore, we demonstrate that the IVD disruptions in Flnb-/-mice resemble aging degenerative discs and reveal new insights into the molecular causes of vertebral fusions and disc degeneration.


Subject(s)
Abnormalities, Multiple/genetics , Filamins/genetics , Intervertebral Disc Degeneration/genetics , Lumbar Vertebrae/abnormalities , Musculoskeletal Diseases/genetics , Scoliosis/congenital , Synostosis/genetics , Thoracic Vertebrae/abnormalities , Transforming Growth Factor beta/genetics , Abnormalities, Multiple/pathology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Chondrocytes/metabolism , Chondrocytes/pathology , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Disease Models, Animal , Growth Plate/growth & development , Growth Plate/pathology , Humans , Intervertebral Disc Degeneration/pathology , Lumbar Vertebrae/pathology , Mice , Mice, Knockout , Musculoskeletal Diseases/pathology , Scoliosis/genetics , Scoliosis/pathology , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Spine/growth & development , Spine/pathology , Synostosis/pathology , Thoracic Vertebrae/pathology
18.
J Cell Biochem ; 117(4): 927-37, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26364758

ABSTRACT

CCN2/connective tissue growth factor (CTGF) is a multi-functional molecule that promotes harmonized development and regeneration of cartilage through its matricellular interaction with a variety of extracellular biomolecules. Thus, deficiency in CCN2 supply profoundly affects a variety of cellular activities including basic metabolism. A previous study showed that the expression of a number of ribosomal protein genes was markedly enhanced in Ccn2-null chondrocytes. Therefore, in this study, we analyzed the impact of CCN2 on amino acid and protein metabolism in chondrocytes. Comparative metabolome analysis of the amino acids in Ccn2-null and wild-type mouse chondrocytes revealed stable decreases in the cellular levels of all of the essential amino acids. Unexpectedly, uptake of such amino acids was rather enhanced in Ccn2-null chondrocytes, and the addition of exogenous CCN2 to human chondrocytic cells resulted in decreased amino acid uptake. However, as expected, amino acid consumption by protein synthesis was also accelerated in Ccn2-null chondrocytes. Furthermore, we newly found that expression of two genes encoding two glycolytic enzymes, as well as the previously reported Eno1 gene, was repressed in those cells. Considering the impaired glycolysis and retained mitochondrial membrane potential in Ccn2-null chondrocytes, these findings suggest that Ccn2 deficiency induces amino acid shortage in chondrocytes by accelerated amino acid consumption through protein synthesis and acquisition of aerobic energy. Interestingly, CCN2 was found to capture such free amino acids in vitro. Under physiological conditions, CCN2 may be regulating the levels of free amino acids in the extracellular matrix of cartilage.


Subject(s)
Amino Acids/metabolism , Cartilage/metabolism , Chondrocytes/metabolism , Connective Tissue Growth Factor/genetics , Metabolome , Animals , Biological Transport , Cartilage/cytology , Chondrocytes/cytology , Connective Tissue Growth Factor/metabolism , Embryo, Mammalian , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Microarray Analysis , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Primary Cell Culture , Protein Biosynthesis , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction
19.
Am J Pathol ; 185(11): 3090-101, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26348575

ABSTRACT

The hypoxia-inducible factor (Hif)-1α (Hif-1α) and Hif-2α (Epas1) have a critical role in both normal development and cancer. von Hippel Lindau (Vhl) protein, encoded by a tumor suppressor gene, is an E3 ubiquitin ligase that targets Hif-1α and Epas1 to the proteasome for degradation. To better understand the role of Vhl in the biology of mesenchymal cells, we analyzed mutant mice lacking Vhl in mesenchymal progenitors that give rise to the soft tissues that form and surround synovial joints. Loss of Vhl in mesenchymal progenitors of the limb bud caused severe fibrosis of the synovial joints and formation of aggressive masses with histologic features of mesenchymal tumors. Hif-1α and its downstream target connective tissue growth factor were necessary for the development of these tumors, which conversely still developed in the absence of Epas1, but at lower frequency. Human tumors of the soft tissue are a very complex and heterogeneous group of neoplasias. Our novel findings in genetically altered mice suggest that activation of the HIF signaling pathway could be an important pathogenetic event in the development and progression of at least a subset of these tumors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Fibrosis/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Signal Transduction , Soft Tissue Neoplasms/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Fibrosis/metabolism , Fibrosis/prevention & control , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/prevention & control , Synovial Membrane/metabolism , Synovial Membrane/pathology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
20.
J Biol Chem ; 290(22): 14004-18, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25864198

ABSTRACT

WISP1/CCN4 (hereafter referred to as WISP1), a member of the CCN family, is found in mineralized tissues and is produced by osteoblasts and their precursors. In this study, Wisp1-deficient (Wisp1(-/-)) mice were generated. Using dual-energy x-ray absorptiometry, we showed that by 3 months, the total bone mineral density of Wisp1(-/-) mice was significantly lower than that of WT mice. Further investigation by micro-computed tomography showed that female Wisp1(-/-) mice had decreased trabecular bone volume/total volume and that both male and female Wisp1(-/-) mice had decreased cortical bone thickness accompanied by diminished biomechanical strength. The molecular basis for decreased bone mass in Wisp1(-/-) mice arises from reduced bone formation likely caused by osteogenic progenitors that differentiate poorly compared with WT cells. Osteoclast precursors from Wisp1(-/-) mice developed more tartrate-resistant acid phosphatase-positive cells in vitro and in transplants, suggesting that WISP1 is also a negative regulator of osteoclast differentiation. When bone turnover (formation and resorption) was induced by ovariectomy, Wisp1(-/-) mice had lower bone mineral density compared WT mice, confirming the potential for multiple roles for WISP1 in controlling bone homeostasis. Wisp1(-/-) bone marrow stromal cells had reduced expression of ß-catenin and its target genes, potentially caused by WISP1 inhibition of SOST binding to LRP6. Taken together, our data suggest that the decreased bone mass found in Wisp1(-/-) mice could potentially be caused by an insufficiency in the osteodifferentiation capacity of bone marrow stromal cells arising from diminished Wnt signaling, ultimately leading to altered bone turnover and weaker biomechanically compromised bones.


Subject(s)
Bone Remodeling , Bone and Bones/metabolism , CCN Intercellular Signaling Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Signaling Pathway , Alleles , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Female , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , RNA, Messenger/metabolism , Receptors, LDL/metabolism , Recombination, Genetic , Stromal Cells/cytology , Tumor Suppressor Proteins/metabolism , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...