Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
FASEB J ; 38(1): e23363, 2024 01.
Article in English | MEDLINE | ID: mdl-38085183

ABSTRACT

Intervertebral disc degeneration is a leading cause of chronic low back pain. Cell-based strategies that seek to treat disc degeneration by regenerating the central nucleus pulposus (NP) hold significant promise, but key challenges remain. One of these is the inability of therapeutic cells to effectively mimic the performance of native NP cells, which are unique amongst skeletal cell types in that they arise from the embryonic notochord. In this study, we use single cell RNA sequencing to demonstrate emergent heterogeneity amongst notochord-derived NP cells in the postnatal mouse disc. Specifically, we established the existence of progenitor and mature NP cells, corresponding to notochordal and chondrocyte-like cells, respectively. Mature NP cells exhibited significantly higher expression levels of extracellular matrix (ECM) genes including aggrecan, and collagens II and VI, along with elevated transforming growth factor-beta and phosphoinositide 3 kinase-protein kinase B signaling. Additionally, we identified Cd9 as a novel surface marker of mature NP cells, and demonstrated that these cells were localized to the NP periphery, increased in numbers with increasing postnatal age, and co-localized with emerging glycosaminoglycan-rich matrix. Finally, we used a goat model to show that Cd9+ NP cell numbers decrease with moderate severity disc degeneration, suggesting that these cells are associated with maintenance of the healthy NP ECM. Improved understanding of the developmental mechanisms underlying regulation of ECM deposition in the postnatal NP may inform improved regenerative strategies for disc degeneration and associated low back pain.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Low Back Pain , Nucleus Pulposus , Mice , Animals , Nucleus Pulposus/metabolism , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc/metabolism , Notochord/metabolism , Low Back Pain/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Sequence Analysis, RNA
2.
Stem Cells ; 37(5): 677-689, 2019 05.
Article in English | MEDLINE | ID: mdl-30681752

ABSTRACT

Long bone development involves the embryonic formation of a primary ossification center (POC) in the incipient diaphysis followed by postnatal development of a secondary ossification center (SOC) at each epiphysis. Studies have elucidated major basic mechanisms of POC development, but relatively little is known about SOC development. To gain insights into SOC formation, we used Col2-Cre Rosa-tdTomato (Col2/Tomato) reporter mice and found that their periarticular region contained numerous Tomato-positive lineage cells expressing much higher Tomato fluorescence (termed TomatoH ) than underlying epiphyseal chondrocytes (termed TomatoL ). With time, the TomatoH cells became evident at the SOC invagination site and cartilage canal, increased in number in the expanding SOC, and were present as mesenchymal lineage cells in the subchondral bone. These data were verified in two mouse lineage tracing models, Col2-CreER Rosa-tdTomato and Gli1-CreER Rosa-tdTomato. In vitro tests showed that the periarticular TomatoH cells from Col2/Tomato mice contained mesenchymal progenitors with multidifferentiation abilities. During canal initiation, the cells expressed vascular endothelial growth factor (VEGF) and migrated into epiphyseal cartilage ahead of individual or clusters of endothelial cells, suggesting a unique role in promoting vasculogenesis. Later during SOC expansion, chondrocytes in epiphyseal cartilage expressed VEGF, and angiogenic blood vessels preceded TomatoH cells. Gene expression analyses of microdissected samples revealed upregulation of MMPs in periarticular cells at the invagination site and suggested potential roles for novel kinase and growth factor signaling pathways in regulating SOC canal initiation. In summary, our data indicate that the periarticular region surrounding epiphyseal cartilage contains mesenchymal progenitors that initiate SOC development and form subchondral bone. Stem Cells 2019;37:677-689.


Subject(s)
Bone Development/genetics , Cell Differentiation/genetics , Mesenchymal Stem Cells , Osteogenesis/genetics , Animals , Cartilage/growth & development , Chondrocytes/cytology , Gene Expression Regulation, Developmental/genetics , Growth Plate/growth & development , Growth Plate/metabolism , Mice , Signal Transduction/genetics , Skull/growth & development , Skull/metabolism , Vascular Endothelial Growth Factor A/genetics , Zinc Finger Protein GLI1/genetics
3.
FASEB J ; 32(1): 52-62, 2018 01.
Article in English | MEDLINE | ID: mdl-28860152

ABSTRACT

Bone atrophy and its related fragility fractures are frequent, late side effects of radiotherapy in cancer survivors and have a detrimental impact on their quality of life. In another study, we showed that parathyroid hormone 1-34 and anti-sclerostin antibody attenuates radiation-induced bone damage by accelerating DNA repair in osteoblasts. DNA damage responses are partially regulated by the ubiquitin proteasome pathway. In the current study, we examined whether proteasome inhibitors have similar bone-protective effects against radiation damage. MG132 treatment greatly reduced radiation-induced apoptosis in cultured osteoblastic cells. This survival effect was owing to accelerated DNA repair as revealed by γH2AX foci and comet assays and to the up-regulation of Ku70 and DNA-dependent protein kinase, catalytic subunit, essential DNA repair proteins in the nonhomologous end-joining pathway. Administration of bortezomib (Bzb) reversed the loss of trabecular bone structure and strength in mice at 4 wk after focal radiation. Histomorphometry revealed that Bzb significantly increased the number of osteoblasts and activity in the irradiated area and suppressed the number and activity of osteoclasts, regardless of irradiation. Two weeks of Bzb treatment accelerated DNA repair in bone-lining osteoblasts and thus promoted their survival. Meanwhile, it also inhibited bone marrow adiposity. Taken together, we demonstrate a novel role of proteasome inhibitors in treating radiation-induced osteoporosis.-Chandra, A., Wang, L., Young, T., Zhong, L., Tseng, W.-J., Levine, M. A., Cengel, K., Liu, X. S., Zhang, Y., Pignolo, R. J., Qin, L. Proteasome inhibitor bortezomib is a novel therapeutic agent for focal radiation-induced osteoporosis.


Subject(s)
Bortezomib/pharmacology , Osteoporosis/drug therapy , Proteasome Inhibitors/pharmacology , Radiation Injuries/drug therapy , Radiation-Protective Agents/pharmacology , 3T3 Cells , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bone Remodeling/drug effects , Bone Remodeling/radiation effects , Cell Line , Cell Survival/drug effects , DNA Repair/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/drug effects , Osteoblasts/pathology , Osteoblasts/radiation effects , Osteoporosis/metabolism , Osteoporosis/pathology , Radiation Injuries/metabolism , Radiation Injuries/pathology , X-Ray Microtomography
4.
Biomacromolecules ; 19(1): 94-102, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29211452

ABSTRACT

We investigated the effects of different oxygen tension (21% and 2.5% O2) on the chondrogenesis of different cell systems cultured in pH-degradable PVA hydrogels, including human articular chondrocytes (hACs), human mesenchymal stem cells (hMSCs), and their cocultures with a hAC/hMSC ratio of 20/80. These hydrogels were prepared with vinyl ether acrylate-functionalized PVA (PVA-VEA) and thiolated PVA-VEA (PVA-VEA-SH) via Michael-type addition reaction. The rheology tests determined the gelation of the hydrogels was controlled within 2-7 min, dependent on the polymer concentrations. The different cell systems were cultured in the hydrogel scaffolds for 5 weeks, and the safranin O and GAG assay showed that hypoxia (2.5% O2) greatly promoted the cartilage matrix production with an order of hAC > hAC/hMSC > hMSC. The real time quantitative PCR (RT-PCR) revealed that the hMSC group exhibited the highest hypertrophic marker gene expression (COL10A1, ALPL, MMP13) as well as the dedifferentiated marker gene expression (COL1A1) under normoxia conditions (21% O2), while these expressions were greatly inhibited by coculturing with a 20% amount of hACs and significantly further repressed under hypoxia conditions, which was comparative to the sole hAC group. The enzyme-linked immunosorbent assay (ELISA) also showed that coculture of hMSC/hAC greatly reduced the catabolic gene expression of MMP1 and MMP3 compared with the hMSC group. It is obvious that the hypoxia conditions promoted the chondrogenesis of hMSC by adding a small amount of hACs, and also effectively inhibited their hypotrophy. We are convinced that coculture of hAC/hMSC using in situ forming hydrogel scaffolds is a promising approach to producing cell source for cartilage engineering without the huge needs of primary chondrocyte harvest and expansion.


Subject(s)
Cell Hypoxia , Chondrocytes/cytology , Chondrogenesis , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Tissue Scaffolds , Alkaline Phosphatase/metabolism , Biocompatible Materials/chemistry , Cartilage, Articular/cytology , Cartilage, Articular/enzymology , Cartilage, Articular/metabolism , Chondrocytes/enzymology , Chondrocytes/metabolism , Coculture Techniques , Collagen/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression , Glycosaminoglycans/metabolism , Humans , Matrix Metalloproteinase 13/genetics , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/metabolism , Middle Aged , Phenazines/metabolism , Polyvinyl Alcohol/chemistry
5.
Int J Mol Sci ; 19(2)2018 Feb 13.
Article in English | MEDLINE | ID: mdl-29438298

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent cells, mainly from bone marrow, and an ideal source of cells in bone and cartilage tissue engineering. A study of the chondrogenic differentiation of MSCs is of particular interest for MSCs-based cartilage regeneration. In this study, we aimed to optimize the conditions for the chrondogenic differentiation of MSCs by regulating WNT signaling using the small molecule WNT inhibitor PKF118-310 and activator BIO. Human mesenchymal stem cells (hMSCs) were isolated from bone marrow aspirates and cultured in hMSCs proliferation medium. Pellet culture was subsequently established for three-dimensional chondrogenic differentiation of 5 weeks. WNT signaling was increased by the small molecule glycogen synthase kinase-3 inhibitor 6-bromoindirubin-3-oxim (BIO) and decreased by the WNT inhibitor PKF118-310 (PKF). The effects of BIO and PKF on the chondrogenesis of hMSCs was examined by real-time PCR, histological methods, and ELISA. We found that activation of canonical WNT-signaling by BIO significantly downregulated the expression of cartilage-specific genes SOX9, COL2A1, and ACAN, and matrix metalloproteinase genes MMP1/3/9/13, but increased ADAMTS 4/5. Inhibition of WNT signaling by PKF increased the expression of SOX9, COL2A1, ACAN, and MMP9, but decreased MMP13 and ADAMTS4/5. In addition, a high level of WNT signaling induced the expression of hypertrophic markers COL10A1, ALPL, and RUNX2, the dedifferentiation marker COL1A1, and glycolysis genes GULT1 and PGK1. Deposition of glycosaminoglycan (GAG) and collagen type II in the pellet matrix was significantly lost in the BIO-treated group and increased in the PKF-treated group. The protein level of COL10A1 was also highly induced in the BIO group. Interestingly, BIO decreased the number of apoptotic cells while PKF significantly induced apoptosis during chondrogenesis. The natural WNT antagonist DKK1 and the protein level of MMP1 in the pellet culture medium were decreased after PKF treatment. All of these chondrogenic effects appeared to be mediated through the canonical WNT signaling pathway, since the target gene Axin2 and other WNT members, such as TCF4 and ß-catenin, were upregulated by BIO and downregulated by PKF, respectively, and BIO induced nuclear translocation of ß-catenin while PKF inhibited ß-catenin translocation into the nucleus. We concluded that addition of BIO to a chondrogenic medium of hMSCs resulted in a loss of cartilage formation, while PKF induced chondrogenic differentiation and cartilage matrix deposition and inhibited hypertrophic differentiation. However, BIO promoted cell survival by inhibiting apoptosis while PKF induced cell apoptosis. This result indicates that either an overexpression or overinhibition of WNT signaling to some extent causes harmful effects on chondrogenic differentiation. Cartilage tissue engineering could benefit from the adjustment of the critical level of WNT signaling during chondrogenesis of hMSC.


Subject(s)
Cell Differentiation , Indoles/pharmacology , Mesenchymal Stem Cells/cytology , Oximes/pharmacology , Pyrimidinones/pharmacology , Triazines/pharmacology , Wnt Signaling Pathway , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen/genetics , Collagen/metabolism , HEK293 Cells , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Int J Mol Sci ; 18(11)2017 Nov 22.
Article in English | MEDLINE | ID: mdl-29165387

ABSTRACT

Interleukin 1 beta (IL1ß) and Wingless-Type MMTV Integration Site Family (WNT) signaling are major players in Osteoarthritis (OA) pathogenesis. Despite having a large functional overlap in OA onset and development, the mechanism of IL1ß and WNT crosstalk has remained largely unknown. In this study, we have used a combination of computational modeling and molecular biology to reveal direct or indirect crosstalk between these pathways. Specifically, we revealed a mechanism by which IL1ß upregulates WNT signaling via downregulating WNT antagonists, DKK1 and FRZB. In human chondrocytes, IL1ß decreased the expression of Dickkopf-1 (DKK1) and Frizzled related protein (FRZB) through upregulation of nitric oxide synthase (iNOS), thereby activating the transcription of WNT target genes. This effect could be reversed by iNOS inhibitor 1400W, which restored DKK1 and FRZB expression and their inhibitory effect on WNT signaling. In addition, 1400W also inhibited both the matrix metalloproteinase (MMP) expression and cytokine-induced apoptosis. We concluded that iNOS/NO play a pivotal role in the inflammatory response of human OA through indirect upregulation of WNT signaling. Blocking NO production may inhibit the loss of the articular phenotype in OA by preventing downregulation of the expression of DKK1 and FRZB.


Subject(s)
Chondrocytes/metabolism , Gene Expression Regulation , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-1beta/metabolism , Nitric Oxide/metabolism , Wnt Signaling Pathway , Cartilage , Humans , Interleukin-1beta/pharmacology , Intracellular Signaling Peptides and Proteins , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , beta Catenin/metabolism
7.
Int J Mol Sci ; 17(7)2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27428952

ABSTRACT

Osteoarthritis (OA) is a multifactorial disease characterized by gradual degradation of joint cartilage. This study aimed to quantify major pathogenetic factors during OA progression in human cartilage. Cartilage specimens were isolated from OA patients and scored 0-5 according to the Osteoarthritis Research Society International (OARSI) guidelines. Protein and gene expressions were measured by immunohistochemistry and qPCR, respectively. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays were used to detect apoptotic cells. Cartilage degeneration in OA is a gradual progress accompanied with gradual loss of collagen type II and a gradual decrease in mRNA expression of SOX9, ACAN and COL2A1. Expression of WNT antagonists DKK1 and FRZB was lost, while hypertrophic markers (RUNX2, COL10A1 and IHH) increased during OA progression. Moreover, DKK1 and FRZB negatively correlated with OA grading, while RUNX2 and IHH showed a significantly positive correlation with OA grading. The number of apoptotic cells was increased with the severity of OA. Taken together, our results suggested that genetic profiling of the gene expression could be used as markers for staging OA at the molecular level. This helps to understand the molecular pathology of OA and may lead to the development of therapies based on OA stage.


Subject(s)
Apoptosis , Biomarkers/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Aged , Blotting, Western , Cartilage, Articular/cytology , Cell Proliferation , Cells, Cultured , Female , Gene Expression Regulation , Humans , Immunoenzyme Techniques , Male , Osteoarthritis/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
Int J Mol Sci ; 16(8): 19225-47, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26287176

ABSTRACT

Hypertrophic differentiation of chondrocytes is a main barrier in application of mesenchymal stem cells (MSCs) for cartilage repair. In addition, hypertrophy occurs occasionally in osteoarthritis (OA). Here we provide a comprehensive review on recent literature describing signal pathways in the hypertrophy of MSCs-derived in vitro differentiated chondrocytes and chondrocytes, with an emphasis on the crosstalk between these pathways. Insight into the exact regulation of hypertrophy by the signaling network is necessary for the efficient application of MSCs for articular cartilage repair and for developing novel strategies for curing OA. We focus on articles describing the role of the main signaling pathways in regulating chondrocyte hypertrophy-like changes. Most studies report hypertrophic differentiation in chondrogenesis of MSCs, in both human OA and experimental OA. Chondrocyte hypertrophy is not under the strict control of a single pathway but appears to be regulated by an intricately regulated network of multiple signaling pathways, such as WNT, Bone morphogenetic protein (BMP)/Transforming growth factor-ß (TGFß), Parathyroid hormone-related peptide (PTHrP), Indian hedgehog (IHH), Fibroblast growth factor (FGF), Insulin like growth factor (IGF) and Hypoxia-inducible factor (HIF). This comprehensive review describes how this intricate signaling network influences tissue-engineering applications of MSCs in articular cartilage (AC) repair, and improves understanding of the disease stages and cellular responses within an OA articular joint.


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/pathology , Mesenchymal Stem Cells/pathology , Osteoarthritis/pathology , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Chondrogenesis , Humans , Hypertrophy/metabolism , Hypertrophy/pathology , Mesenchymal Stem Cells/metabolism , Osteoarthritis/metabolism , Signal Transduction
9.
J Biol Chem ; 288(24): 17552-8, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23603903

ABSTRACT

T cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors are downstream effectors of Wnt/ß-catenin signaling, which has been implicated in the development and progression of osteoarthritis (OA). This study aimed to investigate the role of TCF/LEF transcription factors in human articular chondrocytes. Primary human osteoarthritic cartilage predominantly expressed TCF4 and to a lesser extent, LEF1 and TCF3 mRNA. Overexpression of TCF4, but not of TCF3 or LEF1, induced MMP-1, -3, and -13 expression and generic MMP activity in human chondrocytes. This was due to potentiating NF-κB signaling by a protein-protein interaction between TCF4 and NF-κB p65 activating established NF-κB target genes such as MMPs and IL-6. LEF1 competed with TCF4 for binding to NF-κB p65. IκB-α was able to counteract the effect of TCF4 on NF-κB target gene expression. Finally, we showed that TCF4 mRNA expression was elevated in OA cartilage compared with healthy cartilage and induced chondrocyte apoptosis at least partly through activating caspase 3/7. Our findings suggest that increased TCF4 expression may contribute to cartilage degeneration in OA by augmenting NF-κB signaling.


Subject(s)
Apoptosis , Chondrocytes/metabolism , Transcription Factor 7-Like 2 Protein/physiology , Transcription Factor RelA/metabolism , Aged , Aged, 80 and over , Cartilage, Articular/pathology , Cells, Cultured , Gene Expression , HEK293 Cells , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/metabolism , Middle Aged , NF-kappa B/metabolism , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation
10.
Elife ; 122023 Dec 11.
Article in English | MEDLINE | ID: mdl-38079220

ABSTRACT

Insufficient bone fracture repair represents a major clinical and societal burden and novel strategies are needed to address it. Our data reveal that the transforming growth factor-ß superfamily member Activin A became very abundant during mouse and human bone fracture healing but was minimally detectable in intact bones. Single-cell RNA-sequencing revealed that the Activin A-encoding gene Inhba was highly expressed in a unique, highly proliferative progenitor cell (PPC) population with a myofibroblast character that quickly emerged after fracture and represented the center of a developmental trajectory bifurcation producing cartilage and bone cells within callus. Systemic administration of neutralizing Activin A antibody inhibited bone healing. In contrast, a single recombinant Activin A implantation at fracture site in young and aged mice boosted: PPC numbers; phosphorylated SMAD2 signaling levels; and bone repair and mechanical properties in endochondral and intramembranous healing models. Activin A directly stimulated myofibroblastic differentiation, chondrogenesis and osteogenesis in periosteal mesenchymal progenitor culture. Our data identify a distinct population of Activin A-expressing PPCs central to fracture healing and establish Activin A as a potential new therapeutic tool.


Subject(s)
Activins , Bony Callus , Fracture Healing , Mice , Humans , Animals , Fracture Healing/genetics , Osteogenesis , Stem Cells , Cell Differentiation
11.
bioRxiv ; 2023 May 22.
Article in English | MEDLINE | ID: mdl-37292597

ABSTRACT

Intervertebral disc degeneration is a leading cause of chronic low back pain. Cell-based strategies that seek to treat disc degeneration by regenerating the central nucleus pulposus hold significant promise, but key challenges remain. One of these is the inability of therapeutic cells to effectively mimic the performance of native nucleus pulposus cells, which are unique amongst skeletal cell types in that they arise from the embryonic notochord. In this study we use single cell RNA sequencing to demonstrate emergent heterogeneity amongst notochord-derived nucleus pulposus cells in the postnatal mouse disc. Specifically, we established the existence of early and late stage nucleus pulposus cells, corresponding to notochordal progenitor and mature cells, respectively. Late stage cells exhibited significantly higher expression levels of extracellular matrix genes including aggrecan, and collagens II and VI, along with elevated TGF-ß and PI3K-Akt signaling. Additionally, we identified Cd9 as a novel surface marker of late stage nucleus pulposus cells, and demonstrated that these cells were localized to the nucleus pulposus periphery, increased in numbers with increasing postnatal age, and co-localized with emerging glycosaminoglycan-rich matrix. Finally, we used a goat model to show the Cd9+ nucleus pulposus cell numbers decrease with moderate severity disc degeneration, suggesting that these cells are associated with maintenance of the healthy nucleus pulposus extracellular matrix. Improved understanding of the developmental mechanisms underlying regulation of ECM deposition in the postnatal NP may inform improved regenerative strategies for disc degeneration and associated low back pain.

12.
Elife ; 122023 02 13.
Article in English | MEDLINE | ID: mdl-36779854

ABSTRACT

Colony-stimulating factor 1 (Csf1) is an essential growth factor for osteoclast progenitors and an important regulator for bone resorption. It remains elusive which mesenchymal cells synthesize Csf1 to stimulate osteoclastogenesis. We recently identified a novel mesenchymal cell population, marrow adipogenic lineage precursors (MALPs), in bone. Compared to other mesenchymal subpopulations, MALPs expressed Csf1 at a much higher level and this expression was further increased during aging. To investigate its role, we constructed MALP-deficient Csf1 CKO mice using AdipoqCre. These mice had increased femoral trabecular bone mass, but their cortical bone appeared normal. In comparison, depletion of Csf1 in the entire mesenchymal lineage using Prrx1Cre led to a more striking high bone mass phenotype, suggesting that additional mesenchymal subpopulations secrete Csf1. TRAP staining revealed diminished osteoclasts in the femoral secondary spongiosa region of Csf1 CKOAdipoq mice, but not at the chondral-osseous junction nor at the endosteal surface of cortical bone. Moreover, Csf1 CKOAdipoq mice were resistant to LPS-induced calvarial osteolysis. Bone marrow cellularity, hematopoietic progenitors, and macrophages were also reduced in these mice. Taken together, our studies demonstrate that MALPs synthesize Csf1 to control bone remodeling and hematopoiesis.


Subject(s)
Bone Marrow , Osteoclasts , Mice , Animals , Osteoclasts/metabolism , Bone Marrow/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Bone and Bones/metabolism , Hematopoiesis
13.
Article in English | MEDLINE | ID: mdl-36429619

ABSTRACT

Disasters such as rock bursts and mine earthquakes became increasingly serious with the increase in mining depth in Erdos Coal Field and became serious problems that restrict high-strength continuous mining of coal mines. In this study, strata movement and energy polling distribution of ultrathick weak-bonding sandstone layers were controlled by the local filling−caving multi-faces coordinated mining technique, which was based on the analysis of subsidence and overlying structural characteristics in the Yingpanhao mining area. Moreover, the influencing factors and the control effect laws were investigated. Surface subsidence and energy polling distribution control effects of different mining modes were compared, which confirmed the superiority of local filling based on the main key stratum. According to the results, the maximum surface subsidence velocity of the first mining face was 1.24 mm/d, which indicates the presence of a logistic functional relationship between the mining degree and subsidence factors. When the mining degree was close to full mining, the practical surface subsidence was smaller than the corresponding logistic functional value. The largest influencing factor for the strata movement control effect of partial filling mining based on the main key stratum was the width of the caving face, followed by the filling ratio, section pillar width, and width of the filling face, successively. With respect to the influencing degree on the energy polling distribution of partial filling mining based on the main key stratum, the order followed as section pillar width > filling ratio > caving working face > width of backfilling working face. Additionally, the comparative analysis from the perspectives of control effect, resource utilization, and cost-effectiveness demonstrated that partial filling mining based on the main key stratum was one of the techniques with high cost-effectiveness in controlling strata movement and relieving rock bursts, mining earthquakes, and subsidence disasters.

14.
Biomaterials ; 283: 121437, 2022 04.
Article in English | MEDLINE | ID: mdl-35247635

ABSTRACT

Oxidative stress and the reactive oxygen species (ROS) have important roles in osteoarthritis (OA) development and progression. Scavenging ROS by exogenous antioxidant enzymes could be a promising approach for OA treatment. However, the direct use of antioxidant enzymes, such as superoxide dismutase (SOD), is challenging due to a lack of effective drug delivery system to knee joints. This study utilized a highly efficient antioxidative nanoparticle based on SOD-loaded porous polymersome nanoparticles (SOD-NPs) for delivery of SOD to mouse knee joints. The resultant SOD-NPs had prolonged mouse joint retention time with predominant accumulation in synovium but not in articular cartilage. Examining human synovial explants revealed that SOD-NPs minimize oxidative damages induced by OA-like insults. Intra-articular injections of SOD-NPs in mice receiving OA surgery were effective in attenuating OA initiation and preventing its further progression. Mechanistically, SOD-NPs reduced ROS production and the synthesis of catabolic proteases in both articular cartilage and synovium. Hence, our work demonstrates the therapeutic potential of SOD-NPs and indicate that targeting synovium holds a great promise for OA therapy.


Subject(s)
Cartilage, Articular , Nanoparticles , Osteoarthritis , Animals , Antioxidants/metabolism , Cartilage, Articular/metabolism , Mice , Nanoparticles/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Porosity , Superoxide Dismutase/metabolism , Synovial Membrane/metabolism
15.
JCI Insight ; 7(7)2022 04 08.
Article in English | MEDLINE | ID: mdl-35393948

ABSTRACT

Radiation causes a collapse of bone marrow cells and elimination of microvasculature. To understand how bone marrow recovers after radiation, we focused on mesenchymal lineage cells that provide a supportive microenvironment for hematopoiesis and angiogenesis in bone. We recently discovered a nonproliferative subpopulation of marrow adipogenic lineage precursors (MALPs) that express adipogenic markers with no lipid accumulation. Single-cell transcriptomic analysis revealed that MALPs acquire proliferation and myofibroblast features shortly after radiation. Using an adipocyte-specific Adipoq-Cre, we validated that MALPs rapidly and transiently expanded at day 3 after radiation, coinciding with marrow vessel dilation and diminished marrow cellularity. Concurrently, MALPs lost most of their cell processes, became more elongated, and highly expressed myofibroblast-related genes. Radiation activated mTOR signaling in MALPs that is essential for their myofibroblast conversion and subsequent bone marrow recovery at day 14. Ablation of MALPs blocked the recovery of bone marrow vasculature and cellularity, including hematopoietic stem and progenitors. Moreover, VEGFa deficiency in MALPs delayed bone marrow recovery after radiation. Taken together, our research demonstrates a critical role of MALPs in mediating bone marrow repair after radiation injury and sheds light on a cellular target for treating marrow suppression after radiotherapy.


Subject(s)
Bone Marrow , Myofibroblasts , Adipogenesis , Bone Marrow Cells , Cell Differentiation
16.
J Bone Miner Res ; 37(12): 2498-2511, 2022 12.
Article in English | MEDLINE | ID: mdl-36178273

ABSTRACT

Posttraumatic osteoarthritis (PTOA) results in joint pain, loss of joint function, and impaired quality of daily life in patients with limited treatment options. We previously demonstrated that epidermal growth factor receptor (EGFR) signaling is essential for maintaining chondroprogenitors during articular cartilage development and homeostasis. Here, we used a nonsurgical, loading-induced PTOA mouse model to investigate the protective action of EGFR signaling. A single bout of cyclic tibial loading at a peak force of 6 N injured cartilage at the posterior aspect of lateral femoral condyle. Similar loading at a peak force of 9 N ruptured the anterior cruciate ligament, causing additional cartilage damage at the medial compartment and ectopic cartilage formation in meniscus and synovium. Constitutively overexpression of an EGFR ligand, heparin binding EGF-like growth factor (HBEGF), in chondrocytes significantly reduced cartilage injury length, synovitis, and pain after 6 N loading and mitigated medial side cartilage damage and ectopic cartilage formation after 9 N loading. Mechanistically, overactivation of EGFR signaling protected chondrocytes from loading-induced apoptosis and loss of proliferative ability and lubricant synthesis. Overexpressing HBEGF in adult cartilage starting right before 6 N loading had similar beneficial effects. In contrast, inactivating EGFR in adult cartilage led to accelerated PTOA progression with elevated cartilage Mankin score and synovitis score and increased ectopic cartilage formation. As a therapeutic approach, we constructed a nanoparticle conjugated with the EGFR ligand TGFα. Intra-articular injections of this nanoconstruct once every 3 weeks for 12 weeks partially mitigated PTOA symptoms in cartilage and synovium after 6 N loading. Our findings demonstrate the anabolic actions of EGFR signaling in maintaining articular cartilage during PTOA development and shed light on developing a novel nanomedicine for PTOA. © 2022 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
ErbB Receptors , Osteoarthritis , Animals , Mice , Cartilage, Articular/metabolism , ErbB Receptors/metabolism , Ligands , Osteoarthritis/metabolism , Synovitis/metabolism
17.
J Bone Miner Res ; 37(5): 1012-1023, 2022 05.
Article in English | MEDLINE | ID: mdl-35191092

ABSTRACT

The uppermost superficial zone of articular cartilage is the first line of defense against the initiation of osteoarthritis (OA). We previously used Col2-Cre to demonstrate that epidermal growth factor receptor (EGFR), a tyrosine kinase receptor, plays an essential role in maintaining superficial chondrocytes during articular cartilage development. Here, we showed that EGFR activity in the articular cartilage decreased as mice age. In mouse and human OA samples, EGFR activity was initially reduced at the superficial layer and then resurged in cell clusters within the middle and deep zone in late OA. To investigate the role of EGFR signaling in postnatal and adult cartilage, we constructed an inducible mouse model with cartilage-specific EGFR inactivation (Aggrecan-CreER EgfrWa5/flox , Egfr iCKO). EdU incorporation revealed that postnatal Egfr iCKO mice contained fewer slow-cycling cells than controls. EGFR deficiency induced at 3 months of age reduced cartilage thickness and diminished superficial chondrocytes, in parallel to alterations in lubricin production, cell proliferation, and survival. Furthermore, male Egfr iCKO mice developed much more severe OA phenotypes, including cartilage erosion, subchondral bone plate thickening, cartilage degeneration at the lateral site, and mechanical allodynia, after receiving destabilization of the medial meniscus (DMM) surgery. Similar OA phenotypes were also observed in female iCKO mice. Moreover, tamoxifen injections of iCKO mice at 1 month post-surgery accelerated OA development 2 months later. In summary, our data demonstrated that chondrogenic EGFR signaling maintains postnatal slow-cycling cells and plays a critical role in adult cartilage homeostasis and OA progression. © 2022 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Cartilage, Articular , ErbB Receptors , Osteoarthritis , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , ErbB Receptors/metabolism , Female , Homeostasis , Male , Mice , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology
18.
Methods Mol Biol ; 2221: 29-39, 2021.
Article in English | MEDLINE | ID: mdl-32979196

ABSTRACT

Bone marrow mesenchymal stem cells (MSCs) are promising therapeutic tools for tissue repair and treatment of a number of human diseases. As a result, there is substantial interest in characterizing and expanding these cells to uncover their therapeutic potential. Bone marrow mesenchymal progenitors, containing both MSCs and their proliferative progeny, are commonly isolated from the central region of rodent long bones. However, challenges exist in expanding these central mesenchymal progenitors in culture. We have designed an enzymatic digestion protocol to isolate mesenchymal progenitors within rodent long bones that resides close to the bone surface, which we termed endosteal mesenchymal progenitors. These cells are more metabolically active and more responsive to external stimuli compared to central mesenchymal progenitors. Therefore, they represent a biologically important target for MSC research. This chapter describes the approach in detail how to isolate and culture endosteal mesenchymal progenitors as well as their central counterparts from rodent long bones.


Subject(s)
Bone Marrow Cells/cytology , Bone and Bones/cytology , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Animals , Cell Proliferation , Cell Separation , Cells, Cultured , Mice, Inbred C57BL , Rats, Sprague-Dawley
19.
Best Pract Res Clin Endocrinol Metab ; 35(4): 101518, 2021 07.
Article in English | MEDLINE | ID: mdl-33812853

ABSTRACT

Bone marrow mesenchymal stromal cells are a highly heterogenic cell population containing mesenchymal stem cells as well as other cell types. With the advance of single cell transcriptome analysis, several recent reports identified a prominent subpopulation of mesenchymal stromal cells that specifically express adipocyte markers but do not contain lipid droplets. We name this cell type marrow adipogenic lineage precursor, MALP, and consider it as a major cellular component of marrow adipose tissue. Here, we review the discovery of MALPs and summarize their unique features and regulatory roles in bone. We further discuss how these findings advance our understanding of bone remodeling, mesenchymal niche regulation of hematopoiesis, and marrow vasculature maintenance.


Subject(s)
Bone Marrow , Mesenchymal Stem Cells , Adipocytes , Adipose Tissue , Bone and Bones , Cell Differentiation , Cell Lineage , Humans
20.
Cartilage ; 12(4): 496-504, 2021 10.
Article in English | MEDLINE | ID: mdl-30947534

ABSTRACT

OBJECTIVE: To investigate the presence of WNT antagonists Dickkopf-related protein 1 (DKK1), Frizzled-related protein (FRZB) and BMP antagonist Gremlin 1 (GREM1) in synovial fluid (SF) and serum, respectively, from end-stage knee osteoarthritis (OA) patients, and correlate their expression with other markers of OA. DESIGN: In a cross-sectional study, SF and serum were collected from OA patients (n = 132). The concentrations of DKK1, FRZB and GREM1 in SF and serum were determined using immunoassays. Correlation measurements were performed between groups and previously assessed disease markers, such as synovium nitric oxide (NO), inerleukin-1ß (IL1ß), tumor necrosis factor-α (TNFα), and prostaglandin E2 (PGE2). RESULTS: The OA patients with the celecoxib treatment till surgery have higher median SF FRZB values compared with the control (no treatment); the celecoxib 3-days before surgery stopped treatment group has higher median serum FRZB values than the control and the naproxen treatment group. The combinational analysis of SF DKK1 and SF FRZB negatively correlated with macroscopic cartilage scores and histological synovium scores in OA patients. The expression of DKK1 and FRZB in SF showed the same expression trend as their expression in serum. Furthermore, the SF concentration of DKK1 was positively correlated with FRZB in both SF and serum. In contrast, it was negatively correlated with synovium NO and IL1ß. SF FRZB was negatively correlated with synovium NO, IL1ß, cartilage PGE2, and age. CONCLUSIONS: Our findings suggest DKK1 and FRZB were negatively correlated with OA severity and multiple pro-inflammatory cytokines. Our data indicate that DKK1 and FRZB can be joint disease-specific biomarkers.


Subject(s)
Dinoprostone , Osteoarthritis, Knee , Celecoxib , Cross-Sectional Studies , Humans , Inflammation , Intracellular Signaling Peptides and Proteins
SELECTION OF CITATIONS
SEARCH DETAIL