Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 282
Filter
1.
J Clin Invest ; 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39255038

ABSTRACT

Translation of mRNA to protein is tightly regulated by tRNAs, which are subject to various chemical modifications that maintain the structure, stability and function. Deficiency of tRNA N7-methylguanosine (m7G) modification in patients causes a type of primordial dwarfism, but the underlying mechanism remains unknown. Here we report the loss of m7G rewires cellular metabolism, leading to the pathogenesis of primordial dwarfism. Conditional deletion of the catalytic enzyme Mettl1 or missense mutation of the scaffold protein Wdr4 severely impaired endochondral bone formation and bone mass accrual. Mechanistically, Mettl1 knockout decreased abundance of m7G-modified tRNAs and inhibited translation of mRNAs relating to cytoskeleton and Rho GTPase signaling. Meanwhile, Mettl1 knockout enhanced cellular energy metabolism despite of incompetent proliferation and osteogenic commitment. Further exploration revealed that impaired Rho GTPase signaling upregulated branched-chain amino acid transaminase 1 (BCAT1) level that rewired cell metabolism and restricted intracellular α-ketoglutarate (αKG). Supplementation of αKG ameliorated the skeletal defect of Mettl1-deficient mice. In addition to the selective translation of metabolism-related mRNAs, we further revealed that Mettl1 knockout globally regulated translation via integrated stress response (ISR) and mammalian target of rapamycin complex 1 (mTORC1) signaling. Restoring translation either by targeting ISR or mTORC1 aggravated bone defects of Mettl1-deficient mice. Overall, our study unveils a critical role of m7G tRNA modification in bone development by regulating cellular metabolism, and indicates that suspension of translation initiation as quality control mechanism in response to tRNA dysregulation.

2.
J Clin Invest ; 134(17)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225088

ABSTRACT

The periosteum contains skeletal stem/progenitor cells that contribute to bone fracture healing. However, the in vivo identity of periosteal skeletal stem cells (P-SSCs) remains unclear, and membrane protein markers of P-SSCs that facilitate tissue engineering are needed. Here, we identified integral membrane protein 2A (Itm2a) enriched in SSCs using single-cell transcriptomics. Itm2a+ P-SSCs displayed clonal multipotency and self-renewal and sat at the apex of their differentiation hierarchy. Lineage-tracing experiments showed that Itm2a selectively labeled the periosteum and that Itm2a+ cells were preferentially located in the outer fibrous layer of the periosteum. The Itm2a+ cells rarely expressed CD34 or Osx, but expressed periosteal markers such as Ctsk, CD51, PDGFRA, Sca1, and Gli1. Itm2a+ P-SSCs contributed to osteoblasts, chondrocytes, and marrow stromal cells upon injury. Genetic lineage tracing using dual recombinases showed that Itm2a and Prrx1 lineage cells generated spatially separated subsets of chondrocytes and osteoblasts during fracture healing. Bone morphogenetic protein 2 (Bmp2) deficiency or ablation of Itm2a+ P-SSCs resulted in defects in fracture healing. ITM2A+ P-SSCs were also present in the human periosteum. Thus, our study identified a membrane protein marker that labels P-SSCs, providing an attractive target for drug and cellular therapy for skeletal disorders.


Subject(s)
Fracture Healing , Membrane Proteins , Periosteum , Animals , Periosteum/metabolism , Periosteum/cytology , Mice , Fracture Healing/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Stem Cells/metabolism , Stem Cells/cytology , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Fractures, Bone/pathology , Fractures, Bone/metabolism , Fractures, Bone/therapy , Fractures, Bone/genetics , Osteoblasts/metabolism , Osteoblasts/cytology , Cell Differentiation , Chondrocytes/metabolism , Chondrocytes/cytology , Male , Cell Lineage
3.
JCI Insight ; 9(16)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39171528

ABSTRACT

Obesity can increase the risk of bone fragility, even when bone mass is intact. This fragility stems from poor bone quality, potentially caused by deficiencies in bone matrix material properties. However, cellular and molecular mechanisms leading to obesity-related bone fragility are not fully understood. Using male mouse models of obesity, we discovered TGF-ß signaling plays a critical role in mediating the effects of obesity on bone. High-carbohydrate and high-fat diets increase TGF-ß signaling in osteocytes, which impairs their mitochondrial function, increases cellular senescence, and compromises perilacunar/canalicular remodeling and bone quality. By specifically inhibiting TGF-ß signaling in mouse osteocytes, some of the negative effects of high-fat and high-carbohydrate diets on bones, including the lacunocanalicular network, perilacunar/canalicular remodeling, senescence, and mechanical properties such as yield stress, were mitigated. DMP1-Cre-mediated deletion of TGF-ß receptor II also blunted adverse effects of high-fat and high-carbohydrate diets on energy balance and metabolism. These findings suggest osteocytes are key in controlling bone quality in response to high-fat and high-carbohydrate diets. Calibrating osteocyte function could mitigate bone fragility associated with metabolic diseases while reestablishing energy balance.


Subject(s)
Diet, High-Fat , Obesity , Osteocytes , Transforming Growth Factor beta , Animals , Osteocytes/metabolism , Diet, High-Fat/adverse effects , Mice , Transforming Growth Factor beta/metabolism , Male , Obesity/metabolism , Signal Transduction , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Bone Remodeling , Mice, Inbred C57BL , Disease Models, Animal , Bone and Bones/metabolism , Bone Density/drug effects , Dietary Carbohydrates/adverse effects , Dietary Carbohydrates/administration & dosage
4.
JCI Insight ; 9(16)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39171525

ABSTRACT

Type 2 diabetes (T2D) is on the rise worldwide and is associated with various complications in the oral cavity. Using an adult-onset diabetes preclinical model, we demonstrated profound periodontal alterations in T2D mice, including inflamed gingiva, disintegrated periodontal ligaments (PDLs), marked alveolar bone loss, and unbalanced bone remodeling due to decreased formation and increased resorption. Notably, we observed elevated levels of the Wnt signaling inhibitor sclerostin in the alveolar bone of T2D mice. Motivated by these findings, we investigated whether a sclerostin-neutralizing antibody (Scl-Ab) could rescue the compromised periodontium in T2D mice. Administering Scl-Ab subcutaneously once a week for 4 weeks, starting 4 weeks after T2D induction, led to substantial increases in bone mass. This effect was attributed to the inhibition of osteoclasts and promotion of osteoblasts in both control and T2D mice, effectively reversing the bone loss caused by T2D. Furthermore, Scl-Ab stimulated PDL cell proliferation, partially restored the PDL fibers, and mitigated inflammation in the periodontium. Our study thus established a T2D-induced periodontitis mouse model characterized by inflammation and tissue degeneration. Scl-Ab emerged as a promising intervention to counteract the detrimental effects of T2D on the periodontium, exhibiting limited side effects on other craniofacial hard tissues.


Subject(s)
Adaptor Proteins, Signal Transducing , Alveolar Bone Loss , Diabetes Mellitus, Type 2 , Animals , Mice , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Alveolar Bone Loss/prevention & control , Alveolar Bone Loss/etiology , Alveolar Bone Loss/pathology , Male , Periodontal Diseases/immunology , Antibodies, Neutralizing/pharmacology , Periodontal Ligament/pathology , Periodontal Ligament/drug effects , Disease Models, Animal , Diabetes Mellitus, Experimental/immunology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Mice, Inbred C57BL , Periodontitis/immunology , Periodontitis/pathology , Periodontitis/drug therapy , Bone Remodeling/drug effects
5.
JCI Insight ; 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39088272

ABSTRACT

Energy metabolism, through pathways such as oxidative phosphorylation (OxPhos) and glycolysis, plays a pivotal role in cellular differentiation and function. Our study investigates the impact of OxPhos disruption in cortical bone development by deleting Mitochondrial Transcription Factor A (TFAM). TFAM controls OxPhos by regulating the transcription of mitochondrial genes. The cortical bone, constituting the long bones' rigid shell, is sheathed by the periosteum, a connective tissue layer populated with skeletal progenitors that spawn osteoblasts, the bone-forming cells. TFAM-deficient mice presented with thinner cortical bone, spontaneous midshaft fractures, and compromised periosteal cell bioenergetics, characterized by reduced ATP levels. Additionally, they exhibited an enlarged periosteal progenitor cell pool with impaired osteoblast differentiation. Increasing Hypoxia-Inducible Factor 1a (HIF1) activity within periosteal cells significantly mitigated the detrimental effects induced by TFAM deletion. HIF1 is known to promote glycolysis in all cell types. Our findings underscore the indispensability of OxPhos for the proper accrual of cortical bone mass and indicate a compensatory mechanism between OxPhos and glycolysis in periosteal cells. The study opens new avenues for understanding the relationship between energy metabolism and skeletal health and suggests that modulating bioenergetic pathways may provide a therapeutic avenue for conditions characterized by bone fragility.

6.
J Clin Invest ; 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39196784

ABSTRACT

Chronic low back pain (LBP) can severely affect daily physical activity. Aberrant osteoclast-mediated resorption leads to porous endplates for the sensory innervation to cause LBP. Here, we report that the expression of proton-activated chloride (PAC) channel is induced during osteoclast differentiation in the porous endplates via a RANKL-NFATc1 signaling pathway. Extracellular acidosis evokes robust PAC currents in osteoclasts. An acidic environment of porous endplates and elevated PAC activation-enhanced osteoclast fusion provoke LBP. Further, we find that genetic knockout of PAC gene Pacc1 significantly reduces endplate porosity and spinal pain in a mouse LBP model, but it does not affect bone development or homeostasis of bone mass in adult mice. Moreover, both osteoclast bone resorptive compartment environment and PAC traffic from the plasma membrane to endosomes to form an intracellular organelle Cl channel have low pH around 5.0. The low pH environment activates PAC channel to increase sialyltransferase St3gal1 expression and sialylation of TLR2 in initiation of osteoclast fusion. Aberrant osteoclast-mediated resorption is also found in most skeletal disorders, including osteoarthritis, ankylosing spondylitis, rheumatoid arthritis, heterotopic ossification, enthesopathy. Thus, elevated Pacc1 expression and PAC activity could be a potential therapeutic target for LBP and osteoclast-associated pain.

7.
J Biomech ; 174: 112271, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39159585

ABSTRACT

Bone and cartilage tissues are physiologically dynamic organs that are systematically regulated by mechanical inputs. At cellular level, mechanical stimulation engages an intricate network where mechano-sensors and transmitters cooperate to manipulate downstream signaling. Despite accumulating evidence, there is a notable underutilization of available information, due to limited integration and analysis. In this context, we conceived an interactive web tool named MechanoBone to introduce a new avenue of literature-based discovery. Initially, we compiled a literature database by sourcing content from Pubmed and processing it through the Natural Language Toolkit project, Pubtator, and a custom library. We identified direct co-occurrence among entities based on existing evidence, archiving in a relational database via SQLite. Latent connections were then quantified by leveraging the Link Prediction algorithm. Secondly, mechanobiological pathway maps were generated, and an entity-pathway correlation scoring system was established through weighted algorithm based on our database, String, and KEGG, predicting potential functions of specific entities. Additionally, we established a mechanical circumstance-based exploration to sort genes by their relevance based on big data, revealing the potential mechanically sensitive factors in bone research and future clinical applications. In conclusion, MechanoBone enables: 1) interpreting mechanobiological processes; 2) identifying correlations and crosstalk among molecules and pathways under specific mechanical conditions; 3) connecting clinical applications with mechanobiological processes in bone research. It offers a literature mining tool with visualization and interactivity, facilitating targeted molecule navigation and prediction within the mechanobiological framework of bone-related cells, thereby enhancing knowledge sharing and big data analysis in the biomedical realm.


Subject(s)
Bone and Bones , Natural Language Processing , Humans , Bone and Bones/physiology , Algorithms , Tooth/physiology , Databases, Factual , Biomechanical Phenomena
8.
J Dent Res ; 103(9): 899-907, 2024 08.
Article in English | MEDLINE | ID: mdl-39104155

ABSTRACT

Macrophages are important regulators of bone remodeling, and M1 polarization is observed in the setting of medication-related osteonecrosis of the jaws (MRONJ). Here, we characterize the phenotype of macrophages during early stages of MRONJ development in zoledronate (ZA)-treated mice with periodontal disease and explore the role of rosiglitazone, a drug that has been reported to lower the M1/M2 macrophage ratio, in MRONJ burden. Mice received ZA, and experimental periodontal disease (EPD) was induced around their second left maxillary molar. The mice were euthanized 1, 2, or 4 wk later. Micro-computed tomography and histologic and immunohistochemical analyses were carried out. In a separate experiment, mice were treated with ZA in the absence or presence of rosiglitazone, EPD was induced for 5 wk, and the MRONJ burden was assessed. An M1 predilection was noted in ZA versus vehicle (Veh) mice at 1, 2, or 4 wk after ligature placement. M1 cells were found to be positive for MMP-13, and their presence coincided with disruption of the surrounding collagen network in ZA mice. Rosiglitazone caused a reversal in the M1/M2 polarization in Veh and ZA mice. Rosiglitazone did not cause significant radiographic changes 5 wk after EPD in Veh or ZA animals. Importantly, percentage osteonecrosis and bone exposure were decreased in the rosiglitazone-treated versus nontreated ZA sites 5 wk after EPD. Our data point to an important role of M1 macrophage polarization with an overexpression of MMP-13 in the early phases of MRONJ development and provide insight into the use of interventional approaches promoting an M2 phenotype as a preventative means to alleviate MRONJ burden.


Subject(s)
Bisphosphonate-Associated Osteonecrosis of the Jaw , Imidazoles , Macrophages , Rosiglitazone , Thiazolidinediones , X-Ray Microtomography , Zoledronic Acid , Animals , Mice , Rosiglitazone/pharmacology , Rosiglitazone/therapeutic use , Zoledronic Acid/pharmacology , Macrophages/drug effects , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Bisphosphonate-Associated Osteonecrosis of the Jaw/etiology , Imidazoles/pharmacology , Diphosphonates/pharmacology , Matrix Metalloproteinase 13/metabolism , Bone Density Conservation Agents/pharmacology , Disease Models, Animal , Phenotype , Male , Bone Remodeling/drug effects , Mice, Inbred C57BL , Periodontal Diseases , Collagen/metabolism
9.
JCI Insight ; 9(16)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990653

ABSTRACT

The neurofibromatosis type 1 (NF1) RASopathy is associated with persistent fibrotic nonunions (pseudarthrosis) in human and mouse skeletal tissue. Here, we performed spatial transcriptomics to define the molecular signatures occurring during normal endochondral healing following fracture in mice. Within the control fracture callus, we observed spatially restricted activation of morphogenetic pathways, such as TGF-ß, WNT, and BMP. To investigate the molecular mechanisms contributing to Nf1-deficient delayed fracture healing, we performed spatial transcriptomic analysis on a Postn-cre;Nf1fl/- (Nf1Postn) fracture callus. Transcriptional analyses, subsequently confirmed through phospho-SMAD1/5/8 immunohistochemistry, demonstrated a lack of BMP pathway induction in Nf1Postn mice. To gain further insight into the human condition, we performed spatial transcriptomic analysis of fracture pseudarthrosis tissue from a patient with NF1. Analyses detected increased MAPK signaling at the fibrocartilaginous-osseus junction. Similar to that in the Nf1Postn fracture, BMP pathway activation was absent within the pseudarthrosis tissue. Our results demonstrate the feasibility of delineating the molecular and tissue-specific heterogeneity inherent in complex regenerative processes, such as fracture healing, and reconstructing phase transitions representing endochondral bone formation in vivo. Furthermore, our results provide in situ molecular evidence of impaired BMP signaling underlying NF1 pseudarthrosis, potentially informing the clinical relevance of off-label BMP2 as a therapeutic intervention.


Subject(s)
Bone Morphogenetic Proteins , Fracture Healing , Neurofibromatosis 1 , Pseudarthrosis , Signal Transduction , Transcriptome , Animals , Pseudarthrosis/metabolism , Pseudarthrosis/genetics , Mice , Humans , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/complications , Neurofibromatosis 1/pathology , Fracture Healing/genetics , Fractures, Bone/metabolism , Fractures, Bone/genetics , Disease Models, Animal , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Gene Expression Profiling
10.
JCI Insight ; 9(16)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990647

ABSTRACT

Clarifying multifactorial musculoskeletal disorder etiologies supports risk analysis, development of targeted prevention, and treatment modalities. Deep learning enables comprehensive risk factor identification through systematic analyses of disease data sets but does not provide sufficient context for mechanistic understanding, limiting clinical applicability for etiological investigations. Conversely, multiscale biomechanical modeling can evaluate mechanistic etiology within the relevant biomechanical and physiological context. We propose a hybrid approach combining 3D explainable deep learning and multiscale biomechanical modeling; we applied this approach to investigate temporomandibular joint (TMJ) disorder etiology by systematically identifying risk factors and elucidating mechanistic relationships between risk factors and TMJ biomechanics and mechanobiology. Our 3D convolutional neural network recognized TMJ disorder patients through participant-specific morphological features in condylar, ramus, and chin. Driven by deep learning model outputs, biomechanical modeling revealed that small mandibular size and flat condylar shape were associated with increased TMJ disorder risk through increased joint force, decreased tissue nutrient availability and cell ATP production, and increased TMJ disc strain energy density. Combining explainable deep learning and multiscale biomechanical modeling addresses the "mechanism unknown" limitation undermining translational confidence in clinical applications of deep learning and increases methodological accessibility for smaller clinical data sets by providing the crucial biomechanical context.


Subject(s)
Deep Learning , Temporomandibular Joint Disorders , Humans , Risk Factors , Biomechanical Phenomena , Temporomandibular Joint Disorders/physiopathology , Temporomandibular Joint Disorders/pathology , Male , Female , Adult , Temporomandibular Joint/pathology , Temporomandibular Joint/physiopathology , Young Adult
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167333, 2024 10.
Article in English | MEDLINE | ID: mdl-38960054

ABSTRACT

Periodontitis, a common chronic inflammatory disease, epitomizes a significant impairment in the host immune system and an imbalance of bone metabolism. Macrophage polarization, a dynamic process dictated by the microenvironment, intricately contributes to the interplay between the immune system and bone remodeling, namely the osteoimmune system. Forkhead box protein O1 (FoxO1) has been shown to play a dramatic role in mediating oxidative stress, bone mass, as well as cellular metabolism. Nevertheless, the function and underlying mechanisms of FoxO1 in regulating macrophage polarization-mediated osteogenesis in periodontitis remain to be further elucidated. Here, we found that FoxO1 expression was closely linked to periodontitis, accompanied by aggravated inflammation. Notably, FoxO1 knockdown skewed macrophage polarization from M1 to the antiinflammatory M2 phenotype under inflammatory conditions, which rescued the impaired osteogenic potential. Mechanistically, we revealed that the enhancement of the transcription of peroxisome proliferator-activated receptor (PPAR) signaling in FoxO1-knockdown macrophages. In agreement with this contention, GW9662, a specific inhibitor of PPAR-γ signaling, greatly aggravated macrophage polarization from M2 to the M1 phenotype and attenuated osteogenic potential under inflammatory conditions. Additionally, PPAR-γ signaling agonist rosiglitazone (RSG) was applied to address ligature-induced periodontitis with attenuated inflammation. Our data lend conceptual credence to the function of FoxO1 in mediating macrophage polarization-regulated osteogenesis which serves as a novel therapeutic target for periodontitis.


Subject(s)
Forkhead Box Protein O1 , Macrophages , Osteogenesis , PPAR gamma , Periodontitis , Signal Transduction , PPAR gamma/metabolism , PPAR gamma/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Animals , Mice , Macrophages/metabolism , Periodontitis/metabolism , Periodontitis/pathology , Periodontitis/genetics , Male , Mice, Inbred C57BL , RAW 264.7 Cells , Rosiglitazone/pharmacology , Macrophage Activation
12.
J Clin Invest ; 134(15)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38885336

ABSTRACT

Osteogenesis imperfecta (OI) type V is the second most common form of OI, distinguished by hyperplastic callus formation and calcification of the interosseous membranes, in addition to the bone fragility. It is caused by a recurrent, dominant pathogenic variant (c.-14C>T) in interferon-induced transmembrane protein 5 (IFITM5). Here, we generated a conditional Rosa26-knockin mouse model to study the mechanistic consequences of the recurrent mutation. Expression of the mutant Ifitm5 in osteo-chondroprogenitor or chondrogenic cells resulted in low bone mass and growth retardation. Mutant limbs showed impaired endochondral ossification, cartilage overgrowth, and abnormal growth plate architecture. The cartilage phenotype correlates with the pathology reported in patients with OI type V. Surprisingly, expression of mutant Ifitm5 in mature osteoblasts caused no obvious skeletal abnormalities. In contrast, earlier expression in osteo-chondroprogenitors was associated with an increase in the skeletal progenitor cell population within the periosteum. Lineage tracing showed that chondrogenic cells expressing the mutant Ifitm5 had decreased differentiation into osteoblastic cells in diaphyseal bone. Moreover, mutant IFITM5 disrupted early skeletal homeostasis in part by activating ERK signaling and downstream SOX9 protein, and inhibition of these pathways partially rescued the phenotype in mutant animals. These data identify the contribution of a signaling defect altering osteo-chondroprogenitor differentiation as a driver in the pathogenesis of OI type V.


Subject(s)
Cell Differentiation , MAP Kinase Signaling System , Osteoblasts , Osteogenesis Imperfecta , SOX9 Transcription Factor , Animals , Female , Male , Mice , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Transgenic , Mutation , Osteoblasts/metabolism , Osteoblasts/pathology , Osteogenesis/genetics , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Osteogenesis Imperfecta/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Extracellular Signal-Regulated MAP Kinases
13.
J Dent Res ; 103(7): 723-733, 2024 07.
Article in English | MEDLINE | ID: mdl-38822570

ABSTRACT

A ligature-induced periodontitis model was established in wild-type and CD146CreERT2; RosatdTomato mice to explore the function of pericytes in alveolar bone formation. We found that during periodontitis progression and periodontal wound healing, CD146+/NG2+ pericytes were enriched in the periodontal tissue areas, which could migrate to the alveolar bone surface and colocalize with ALP+/OCN+ osteoblasts. Chemokine C-X-C motif receptor 4 (CXCR4) inhibition using AMD3100 blocked CD146-Cre+ pericyte migration and osteogenesis, as well as further exacerbated periodontitis-associated bone loss. Next, primary pericytes were sorted out by magnetic-activated cell sorting and demonstrated that C-X-C motif chemokine ligand 12 (CXCL12) promotes pericyte migration and osteogenesis via CXCL12-CXCR4-Rac1 signaling. Finally, the local administration of an adeno-associated virus for Rac1 overexpression in NG2+ pericytes promotes osteoblast differentiation of pericytes and increases alveolar bone volume in periodontitis. Thus, our results provided the evidence that pericytes may migrate and osteogenesis via the CXCL12-CXCR4-Rac1 axis during the pathological process of periodontitis.


Subject(s)
Cell Movement , Chemokine CXCL12 , Osteogenesis , Pericytes , Periodontitis , Receptors, CXCR4 , Animals , Osteogenesis/physiology , Cell Movement/physiology , Mice , Chemokine CXCL12/metabolism , Receptors, CXCR4/metabolism , Alveolar Bone Loss , Signal Transduction/physiology , rac1 GTP-Binding Protein/metabolism , Disease Models, Animal , CD146 Antigen , Osteoblasts , Cell Differentiation , Cyclams , Benzylamines
14.
JCI Insight ; 9(11)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38855864

ABSTRACT

The transcription factor SRY-related HMG box 9 (Sox9) is essential for chondrogenesis. Mutations in and around SOX9 cause campomelic dysplasia (CD) characterized by skeletal malformations. Although the function of Sox9 in this context is well studied, the mechanisms that regulate Sox9 expression in chondrocytes remain to be elucidated. Here, we have used genome-wide profiling to identify 2 Sox9 enhancers located in a proximal breakpoint cluster responsible for CD. Enhancer activity of E308 (located 308 kb 5' upstream) and E160 (located 160 kb 5' upstream) correlated with Sox9 expression levels, and both enhancers showed a synergistic effect in vitro. While single deletions in mice had no apparent effect, simultaneous deletion of both E308 and E160 caused a dwarf phenotype, concomitant with a reduction of Sox9 expression in chondrocytes. Moreover, bone morphogenetic protein 2-dependent chondrocyte differentiation of limb bud mesenchymal cells was severely attenuated in E308/E160 deletion mice. Finally, we found that an open chromatin region upstream of the Sox9 gene was reorganized in the E308/E160 deletion mice to partially compensate for the loss of E308 and E160. In conclusion, our findings reveal a mechanism of Sox9 gene regulation in chondrocytes that might aid in our understanding of the pathophysiology of skeletal disorders.


Subject(s)
Campomelic Dysplasia , Cell Differentiation , Chondrocytes , Chondrogenesis , SOX9 Transcription Factor , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Animals , Chondrocytes/metabolism , Mice , Campomelic Dysplasia/genetics , Campomelic Dysplasia/pathology , Campomelic Dysplasia/metabolism , Chondrogenesis/genetics , Cell Differentiation/genetics , Enhancer Elements, Genetic/genetics , Chromatin/metabolism , Chromatin/genetics , Gene Expression Regulation, Developmental , Mice, Knockout , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Humans , Bone Development/genetics
15.
Res Sq ; 2024 May 20.
Article in English | MEDLINE | ID: mdl-38826270

ABSTRACT

Background: Periodontal health in men with HIV remains understudied, despite suggestions of associations between HIV infection and gingival pocketing, periodontal attachment loss, and gingival inflammation. As antiretroviral therapy (ART) has improved the quality of life for people living with HIV (PLWH), aging-related risk factors and comorbidities, including periodontitis, have emerged. This study aims to assess alveolar bone height, gingival crevicular fluid (GCF) cytokines, and periodontal disease activity in men with and without HIV. Methods: Ninety-three men (50 HIV+, 43 HIV‒) aged 35-70 years were recruited from Columbia University Irving Medical Center clinics. Periodontal examination, GCF collection, and intraoral radiographs were conducted. Results: While no significant differences were observed in bleeding on probing, clinical attachment loss and pocket depths, men with HIV exhibited significantly greater alveolar crestal height on radiographs compared to men without HIV (HIV + 3.41+/-1.35 mm, HIV- 2.64+/-1.01 mm; p = 0.004), reflecting greater alveolar bone loss. GCF IL6 levels showed a trend towards elevation in men with HIV (HIV + 0.349+/-0.407 pg/ml, HIV- 0.220+/-0.228 pg/ml; p = 0.059). Conclusions: Men with HIV demonstrate increased alveolar bone loss compared to those without HIV, possibly mediated by elevated IL6 levels. These results underscore the importance of comprehensive oral health management in PLWH and highlight the need for further research understanding the mechanisms linking HIV infection, cytokine dysregulation, and periodontal health.

16.
JCI Insight ; 9(14)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38885330

ABSTRACT

Osteoporotic fractures are a major complication of long-term glucocorticoid therapy. Glucocorticoids transiently increase bone resorption, but they predominantly inhibit bone formation and induce osteocyte apoptosis, leading to bone loss. Current treatments of glucocorticoid-induced osteoporosis aim mainly at reducing bone resorption and are, therefore, inadequate. We previously showed that signaling via the NO/cGMP/protein kinase G pathway plays a key role in skeletal homeostasis. Here, we show that pharmacological PKG activation with the guanylyl cyclase-1 activator cinaciguat or expression of a constitutively active, mutant PKG2R242Q restored proliferation, differentiation, and survival of primary mouse osteoblasts exposed to dexamethasone. Cinaciguat treatment of WT mice or osteoblast-specific expression of PKG2R242Q in transgenic mice prevented dexamethasone-induced loss of cortical bone mass and strength. These effects of cinaciguat and PKG2R242Q expression were due to preserved bone formation parameters and osteocyte survival. The basis for PKG2's effects appeared to be through recovery of Wnt/ß-catenin signaling, which was suppressed by glucocorticoids but critical for proliferation, differentiation, and survival of osteoblast-lineage cells. Cinaciguat reduced dexamethasone activation of osteoclasts, but this did not occur in the PKG2R242Q transgenic mice, suggesting a minor role in osteoprotection. We propose that existing PKG-targeting drugs could represent a novel therapeutic approach to prevent glucocorticoid-induced osteoporosis.


Subject(s)
Cyclic GMP-Dependent Protein Kinases , Dexamethasone , Glucocorticoids , Mice, Transgenic , Osteoblasts , Osteoporosis , Wnt Signaling Pathway , Animals , Osteoporosis/chemically induced , Osteoporosis/metabolism , Osteoporosis/pathology , Mice , Glucocorticoids/adverse effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Dexamethasone/pharmacology , Dexamethasone/adverse effects , Wnt Signaling Pathway/drug effects , Cyclic GMP-Dependent Protein Kinases/metabolism , Cell Differentiation/drug effects , Osteocytes/metabolism , Osteocytes/drug effects , Osteogenesis/drug effects , Disease Models, Animal , Female , Cell Proliferation/drug effects , Bone Density/drug effects
17.
JCI Insight ; 9(12)2024 May 23.
Article in English | MEDLINE | ID: mdl-38781018

ABSTRACT

We present a transcriptomic analysis that provides a better understanding of regulatory mechanisms within the healthy and injured periosteum. The focus of this work is on characterizing early events controlling bone healing during formation of periosteal callus on day 3 after fracture. Building on our previous findings showing that induced Notch1 signaling in osteoprogenitors leads to better healing, we compared samples in which the Notch 1 intracellular domain is overexpressed by periosteal stem/progenitor cells, with control intact and fractured periosteum. Molecular mechanisms and changes in skeletal stem/progenitor cells (SSPCs) and other cell populations within the callus, including hematopoietic lineages, were determined. Notably, Notch ligands were differentially expressed in endothelial and mesenchymal populations, with Dll4 restricted to endothelial cells, whereas Jag1 was expressed by mesenchymal populations. Targeted deletion of Dll4 in endothelial cells using Cdh5CreER resulted in negative effects on early fracture healing, while deletion in SSPCs using α-smooth muscle actin-CreER did not impact bone healing. Translating these observations into a clinically relevant model of bone healing revealed the beneficial effects of delivering Notch ligands alongside the osteogenic inducer, BMP2. These findings provide insights into the regulatory mechanisms within the healthy and injured periosteum, paving the way for novel translational approaches to bone healing.


Subject(s)
Endothelial Cells , Fracture Healing , Jagged-1 Protein , Periosteum , Signal Transduction , Animals , Mice , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Endothelial Cells/metabolism , Periosteum/metabolism , Periosteum/cytology , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mesenchymal Stem Cells/metabolism , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Osteogenesis/genetics , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Male , Female , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics
18.
J Clin Invest ; 134(12)2024 May 16.
Article in English | MEDLINE | ID: mdl-38753433

ABSTRACT

Cells expressing features of senescence, including upregulation of p21 and p16, appear transiently following tissue injury, yet the properties of these cells or how they contrast with age-induced senescent cells remains unclear. Here, we used skeletal injury as a model and identified the rapid appearance following fracture of p21+ cells expressing senescence markers, mainly as osteochondroprogenitors (OCHs) and neutrophils. Targeted genetic clearance of p21+ cells suppressed senescence-associated signatures within the fracture callus and accelerated fracture healing. By contrast, p21+ cell clearance did not alter bone loss due to aging; conversely, p16+ cell clearance, known to alleviate skeletal aging, did not affect fracture healing. Following fracture, p21+ neutrophils were enriched in signaling pathways known to induce paracrine stromal senescence, while p21+ OCHs were highly enriched in senescence-associated secretory phenotype factors known to impair bone formation. Further analysis revealed an injury-specific stem cell-like OCH subset that was p21+ and highly inflammatory, with a similar inflammatory mesenchymal population (fibro-adipogenic progenitors) evident following muscle injury. Thus, intercommunicating senescent-like neutrophils and mesenchymal progenitor cells were key regulators of tissue repair in bone and potentially across tissues. Moreover, our findings established contextual roles of p21+ versus p16+ senescent/senescent-like cells that may be leveraged for therapeutic opportunities.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21 , Fracture Healing , Neutrophils , Animals , Male , Mice , Biomarkers/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Mesenchymal Stem Cells/metabolism , Neutrophils/metabolism , Neutrophils/pathology , Female
19.
JCI Insight ; 9(10)2024 May 22.
Article in English | MEDLINE | ID: mdl-38713511

ABSTRACT

While sclerostin-neutralizing antibodies (Scl-Abs) transiently stimulate bone formation by activating Wnt signaling in osteoblast lineage cells, they exert sustained inhibition of bone resorption, suggesting an alternate signaling pathway by which Scl-Abs control osteoclast activity. Since sclerostin can activate platelet-derived growth factor receptors (PDGFRs) in osteoblast lineage cells in vitro and PDGFR signaling in these cells induces bone resorption through M-CSF secretion, we hypothesized that the prolonged anticatabolic effect of Scl-Abs could result from PDGFR inhibition. We show here that inhibition of PDGFR signaling in osteoblast lineage cells is sufficient and necessary to mediate prolonged Scl-Ab effects on M-CSF secretion and osteoclast activity in mice. Indeed, sclerostin coactivates PDGFRs independently of Wnt/ß-catenin signaling inhibition, by forming a ternary complex with LRP6 and PDGFRs in preosteoblasts. In turn, Scl-Ab prevents sclerostin-mediated coactivation of PDGFR signaling and consequent M-CSF upregulation in preosteoblast cultures, thereby inhibiting osteoclast activity in preosteoblast/osteoclast coculture assays. These results provide a potential mechanism explaining the dissociation between anabolic and antiresorptive effects of long-term Scl-Ab.


Subject(s)
Adaptor Proteins, Signal Transducing , Bone Resorption , Osteoblasts , Osteoclasts , Receptors, Platelet-Derived Growth Factor , Signal Transduction , Animals , Osteoblasts/metabolism , Mice , Adaptor Proteins, Signal Transducing/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Antibodies, Neutralizing/pharmacology , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Cell Lineage , Osteogenesis/drug effects , Cell Differentiation
20.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 256-262, 2024 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-38645858

ABSTRACT

Runt-related transcription factor (RUNX1) is a transcription factor closely involved in hematopoiesis. RUNX1 gene mutation plays an essential pathogenic role in the initiation and development of hematological tumors, especially in acute myeloid leukemia. Recent studies have shown that RUNX1 is also involved in the regulation of bone development and the pathological progression of bone-related diseases. RUNX1 promotes the differentiation of mesenchymal stem cells into chondrocytes and osteoblasts and modulates the maturation and extracellular matrix formation of chondrocytes. The expression of RUNX1 in mesenchymal stem cells, chondrocytes, and osteoblasts is of great significance for maintaining normal bone development and the mass and quality of bones. RUNX1 also inhibits the differentiation and bone resorptive activities of osteoclasts, which may be influenced by sexual dimorphism. In addition, RUNX1 deficiency contributes to the pathogenesis of osteoarthritis, delayed fracture healing, and osteoporosis, which was revealed by the RUNX1 conditional knockout modeling in mice. However, the roles of RUNX1 in regulating the hypertrophic differentiation of chondrocytes, the sexual dimorphism of activities of osteoclasts, as well as bone loss in diabetes mellitus, senescence, infection, chronic inflammation, etc, are still not fully understood. This review provides a systematic summary of the research progress concerning RUNX1 in the field of bone biology, offering new ideas for using RUNX1 as a potential target for bone related diseases, especially osteoarthritis, delayed fracture healing, and osteoporosis.


Subject(s)
Bone Development , Cell Differentiation , Chondrocytes , Core Binding Factor Alpha 2 Subunit , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Humans , Animals , Bone Development/physiology , Bone Development/genetics , Chondrocytes/metabolism , Osteoblasts/metabolism , Osteoblasts/cytology , Osteoclasts/metabolism , Osteoclasts/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Bone Diseases/genetics , Bone Diseases/metabolism , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/etiology
SELECTION OF CITATIONS
SEARCH DETAIL