Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
1.
Periodontol 2000 ; 2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38073011

ABSTRACT

The oral squamous cell carcinoma (OSCC) 5 year survival rate of 41% has marginally improved in the last few years, with less than a 1% improvement per year from 2005 to 2017, with higher survival rates when detected at early stages. Based on histopathological grading of oral dysplasia, it is estimated that severe dysplasia has a malignant transformation rate of 7%-50%. Despite these numbers, oral dysplasia grading does not reliably predict its clinical behavior. Thus, more accurate markers predicting oral dysplasia progression to cancer would enable better targeting of these lesions for closer follow-up, especially in the early stages of the disease. In this context, molecular biomarkers derived from genetics, proteins, and metabolites play key roles in clinical oncology. These molecular signatures can help predict the likelihood of OSCC development and/or progression and have the potential to detect the disease at an early stage and, support treatment decision-making and predict treatment responsiveness. Also, identifying reliable biomarkers for OSCC detection that can be obtained non-invasively would enhance management of OSCC. This review will discuss biomarkers for OSCC that have emerged from different biological areas, including genomics, transcriptomics, proteomics, metabolomics, immunomics, and microbiomics.

3.
Stem Cells ; 40(9): 818-830, 2022 09 26.
Article in English | MEDLINE | ID: mdl-35728620

ABSTRACT

Nerve growth factor (NGF) is the best-characterized neurotrophin and is primarily recognized for its key role in the embryonic development of the nervous system and neuronal cell survival/differentiation. Recently, unexpected actions of NGF in bone regeneration have emerged as NGF is able to enhance the osteogenic differentiation of mesenchymal stem cells. However, little is known regarding how NGF signaling regulates osteogenic differentiation through epigenetic mechanisms. In this study, using human dental mesenchymal stem cells (DMSCs), we demonstrated that NGF mediates osteogenic differentiation through p75NTR, a low-affinity NGF receptor. P75NTR-mediated NGF signaling activates the JNK cascade and the expression of KDM4B, an activating histone demethylase, by removing repressive H3K9me3 epigenetic marks. Mechanistically, NGF-activated c-Jun binds to the KDM4B promoter region and directly upregulates KDM4B expression. Subsequently, KDM4B directly and epigenetically activates DLX5, a master osteogenic gene, by demethylating H3K9me3 marks. Furthermore, we revealed that KDM4B and c-Jun from the JNK signaling pathway work in concert to regulate NGF-mediated osteogenic differentiation through simultaneous recruitment to the promoter region of DLX5. We identified KDM4B as a key epigenetic regulator during the NGF-mediated osteogenesis both in vitro and in vivo using the calvarial defect regeneration mouse model. In conclusion, our study thoroughly elucidated the molecular and epigenetic mechanisms during NGF-mediated osteogenesis.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Animals , Cell Differentiation/genetics , Epigenesis, Genetic , Histone Demethylases/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Osteogenesis/genetics , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism
4.
Int J Oral Sci ; 14(1): 24, 2022 05 07.
Article in English | MEDLINE | ID: mdl-35525910

ABSTRACT

Aging of craniofacial skeleton significantly impairs the repair and regeneration of trauma-induced bony defects, and complicates dental treatment outcomes. Age-related alveolar bone loss could be attributed to decreased progenitor pool through senescence, imbalance in bone metabolism and bone-fat ratio. Mesenchymal stem cells isolated from oral bones (OMSCs) have distinct lineage propensities and characteristics compared to MSCs from long bones, and are more suited for craniofacial regeneration. However, the effect of epigenetic modifications regulating OMSC differentiation and senescence in aging has not yet been investigated. In this study, we found that the histone demethylase KDM4B plays an essential role in regulating the osteogenesis of OMSCs and oral bone aging. Loss of KDM4B in OMSCs leads to inhibition of osteogenesis. Moreover, KDM4B loss promoted adipogenesis and OMSC senescence which further impairs bone-fat balance in the mandible. Together, our data suggest that KDM4B may underpin the molecular mechanisms of OMSC fate determination and alveolar bone homeostasis in skeletal aging, and present as a promising therapeutic target for addressing craniofacial skeletal defects associated with age-related deteriorations.


Subject(s)
Aging , Facial Bones , Jumonji Domain-Containing Histone Demethylases , Mesenchymal Stem Cells , Osteogenesis , Osteoporosis , Cell Differentiation , Facial Bones/cytology , Facial Bones/physiology , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mesenchymal Stem Cells/cytology
5.
Periodontol 2000 ; 89(1): 99-113, 2022 06.
Article in English | MEDLINE | ID: mdl-35244945

ABSTRACT

Periodontitis and osteoporosis are prevalent inflammation-associated skeletal disorders that pose significant public health challenges to our aging population. Both periodontitis and osteoporosis are bone disorders closely associated with inflammation and aging. There has been consistent intrigue on whether a systemic skeletal disease such as osteoporosis will amplify the alveolar bone loss in periodontitis. A survey of the literature published in the past 25 years indicates that systemic low bone mineral density (BMD) is associated with alveolar bone loss, while recent evidence also suggests a correlation between clinical attachment loss and other parameters of periodontitis. Inflammation and its influence on bone remodeling play critical roles in the pathogenesis of both osteoporosis and periodontitis and could serve as the central mechanistic link between these disorders. Enhanced cytokine production and elevated inflammatory response exacerbate osteoclastic bone resorption while inhibiting osteoblastic bone formation, resulting in a net bone loss. With aging, accumulation of oxidative stress and cellular senescence drive the progression of osteoporosis and exacerbation of periodontitis. Vitamin D deficiency and smoking are shared risk factors and may mediate the connection between osteoporosis and periodontitis, through increasing oxidative stress and impairing host response to inflammation. With the connection between systemic and localized bone loss in mind, routine dental exams and intraoral radiographs may serve as a low-cost screening tool for low systemic BMD and increased fracture risk. Conversely, patients with fracture risk beyond the intervention threshold are at greater risk for developing severe periodontitis and undergo tooth loss. Various Food and Drug Administration-approved therapies for osteoporosis have shown promising results for treating periodontitis. Understanding the molecular mechanisms underlying their connection sheds light on potential therapeutic strategies that may facilitate co-management of systemic and localized bone loss.


Subject(s)
Alveolar Bone Loss , Osteoporosis, Postmenopausal , Osteoporosis , Periodontal Diseases , Periodontitis , Aged , Alveolar Bone Loss/complications , Bone Density/physiology , Female , Humans , Inflammation/complications , Osteoporosis/complications , Osteoporosis/drug therapy , Osteoporosis, Postmenopausal/complications , Osteoporosis, Postmenopausal/drug therapy , Periodontal Diseases/complications , Periodontal Diseases/therapy , Periodontitis/complications , Periodontitis/therapy
6.
Bone Res ; 10(1): 3, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34992221

ABSTRACT

Osteoporosis is a highly prevalent public health burden associated with an increased risk of bone fracture, particularly in aging women. Estrogen, an important medicinal component for the preventative and therapeutic treatment of postmenopausal osteoporosis, induces osteogenesis by activating the estrogen receptor signaling pathway and upregulating the expression of osteogenic genes, such as bone morphogenetic proteins (BMPs). The epigenetic regulation of estrogen-mediated osteogenesis, however, is still unclear. In this report, we found that estrogen significantly induced the expression of lysine-specific demethylase 6B (KDM6B) and that KDM6B depletion by shRNAs led to a significant reduction in the osteogenic potential of DMSCs. Mechanistically, upon estrogen stimulation, estrogen receptor-α (ERα) was recruited to the KDM6B promoter, directly enhancing KDM6B expression. Subsequently, KDM6B was recruited to the BMP2 and HOXC6 promoters, resulting in the removal of H3K27me3 marks and activating the transcription of BMP2 and HOXC6, the master genes of osteogenic differentiation. Furthermore, we found that estrogen enhanced DMSC osteogenesis during calvarial bone regeneration and that estrogen's pro-osteogenic effect was dependent on KDM6B in vivo. Taken together, our results demonstrate the vital role of the ERα/KDM6B regulatory axis in the epigenetic regulation of the estrogen-dependent osteogenic response.

8.
Int J Oral Sci ; 13(1): 36, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34782601

ABSTRACT

RNA sequencing (RNAseq) can reveal gene fusions, splicing variants, mutations/indels in addition to differential gene expression, thus providing a more complete genetic picture than DNA sequencing. This most widely used technology in genomics tool box has evolved from classic bulk RNA sequencing (RNAseq), popular single cell RNA sequencing (scRNAseq) to newly emerged spatial RNA sequencing (spRNAseq). Bulk RNAseq studies average global gene expression, scRNAseq investigates single cell RNA biology up to 20,000 individual cells simultaneously, while spRNAseq has ability to dissect RNA activities spatially, representing next generation of RNA sequencing. This article highlights these technologies, characteristic features and suitable applications in precision oncology.


Subject(s)
Neoplasms , Humans , Precision Medicine , Sequence Analysis, RNA , Exome Sequencing
9.
Int J Oral Sci ; 13(1): 24, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34341329

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC), an aggressive malignancy, is characterized by high morbidity and low survival rates with limited therapeutic options outside of regional surgery, conventional cytotoxic chemotherapy, and irradiation. Increasing studies have supported the synergistic role of the tumor microenvironment (TME) in cancer advancement. The immune system, in particular, plays a key role in surveillance against the initiation, development, and progression of HNSCC. The understanding of how neoplastic cells evolve and evade the immune system whether through self-immunogenicity manipulation, or expression of immunosuppressive mediators, provides the foundation for the development of advanced therapies. Furthermore, the crosstalk between cancer cells and the host immune system have a detrimental effect on the TME promoting angiogenesis, proliferation, and metastasis. This review provides a recent insight into the role of the key inflammatory cells infiltrating the TME, with a focus on reviewing immunological principles related to HNSCC, as cancer immunosurveillance and immune escape, including a brief overview of current immunotherapeutic strategies and ongoing clinical trials.


Subject(s)
Head and Neck Neoplasms , Tumor Microenvironment , Head and Neck Neoplasms/therapy , Humans , Immune Evasion , Squamous Cell Carcinoma of Head and Neck
10.
ACS Appl Mater Interfaces ; 13(30): 35342-35355, 2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34297530

ABSTRACT

Growth-factor-free bone regeneration remains a challenge in craniofacial engineering. Here, we engineered an osteogenic niche composed of a commercially modified alginate hydrogel and whitlockite microparticles (WHMPs), which impart tunable physicochemical properties that can direct osteogenesis of human gingival mesenchymal stem cells (GMSCs). Our in vitro studies demonstrate that WHMPs induce osteogenesis of GMSCs more effectively than previously demonstrated hydroxyapatite microparticles (HApMPs). Alginate-WHMP hydrogels showed higher elasticity without any adverse effects on the viability of the encapsulated GMSCs. Moreover, the alginate-WHMP hydrogels upregulate the mitogen-activated protein kinase (MAPK) pathway, which in turn orchestrates several osteogenic markers, such as RUNX2 and OCN, in the encapsulated GMSCs. Concurrent coculture studies with human osteoclasts demonstrate that GMSCs encapsulated in alginate-WHMP hydrogels downregulate osteoclastic activity, potentially due to release of Mg2+ ions from the WHMPs along with secretion of osteoprotegerin from the GMSCs. In vivo studies demonstrated that the GMSCs encapsulated in our osteogenic niche were able to promote bone repair in calvarial defects in murine models. Altogether, our results confirmed the development of a promising treatment modality for craniofacial bone regeneration based on an injectable growth-factor-free hydrogel delivery system.


Subject(s)
Bone Regeneration/drug effects , Calcium Phosphates/therapeutic use , Hydrogels/therapeutic use , Skull/drug effects , Alginates/therapeutic use , Animals , Cell Differentiation/drug effects , Cells, Immobilized , Gingiva/cytology , Humans , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteogenesis/drug effects , Rats, Sprague-Dawley , Tissue Engineering/methods
11.
Adv Sci (Weinh) ; 8(13): 2003376, 2021 07.
Article in English | MEDLINE | ID: mdl-34258151

ABSTRACT

Cancer stemness and immune evasion are closely associated, and play critical roles in tumor development and resistance to immunotherapy. However, little is known about the underlying molecular mechanisms that coordinate this association. Here, it is reported that elevated circular RNA FAT1 (circFAT1) in squamous cell carcinoma (SCC) unifies and regulates the positive association between cancer stemness and immune evasion by promoting STAT3 activation. circFAT1 knockdown (KD) reduces tumorsphere formation of SCC cells in vitro and tumor growth in vivo. Bioinformatic analysis reveals that circFAT1 KD impairs the cancer stemness signature and activates tumor cell-intrinsic immunity. Mechanistically, circFAT1 binding to STAT3 in the cytoplasm prevents STAT3 dephosphorylation by SHP1 and promotes STAT3 activation, resulting in inhibition of STAT1-mediated transcription. Moreover, circFAT1 KD significantly enhances PD1 blockade immunotherapy by promoting CD8+ cell infiltration into tumor microenvironment. Taken together, the results demonstrate that circFAT1 is an important regulator of cancer stemness and antitumor immunity.


Subject(s)
Cadherins/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , STAT3 Transcription Factor/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Animals , Cadherins/immunology , Cadherins/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Head and Neck Neoplasms/metabolism , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , RNA, Circular/genetics , RNA, Circular/immunology , RNA, Circular/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
12.
Nat Commun ; 12(1): 3974, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172737

ABSTRACT

Cancer stem cells (CSCs) play a critical role in invasive growth and metastasis of human head and neck squamous cell carcinoma (HNSCC). Although significant progress has been made in understanding the self-renewal and pro-tumorigenic potentials of CSCs, a key challenge remains on how to eliminate CSCs and halt metastasis effectively. Here we show that super-enhancers (SEs) play a critical role in the transcription of cancer stemness genes as well as pro-metastatic genes, thereby controlling their tumorigenic potential and metastasis. Mechanistically, we find that bromodomain-containing protein 4 (BRD4) recruits Mediators and NF-κB p65 to form SEs at cancer stemness genes such as TP63, MET and FOSL1, in addition to oncogenic transcripts. In vivo lineage tracing reveals that disrupting SEs by BET inhibitors potently inhibited CSC self-renewal and eliminated CSCs in addition to elimination of proliferating non-stem tumor cells in a mouse model of HNSCC. Moreover, disrupting SEs also inhibits the invasive growth and lymph node metastasis of human CSCs isolated from human HNSCC. Taken together, our results suggest that targeting SEs may serve as an effective therapy for HNSCC by eliminating CSCs.


Subject(s)
Enhancer Elements, Genetic , Head and Neck Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Lymphatic Metastasis/drug therapy , Lymphatic Metastasis/prevention & control , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Mice, Inbred C57BL , Mice, SCID , NF-kappa B/genetics , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Triazoles/pharmacology , Xenograft Model Antitumor Assays
13.
STAR Protoc ; 2(2): 100484, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33982017

ABSTRACT

BMI1-expressing cancer stem cells (CSCs) play a key role in the development, progression, therapy resistance, recurrence, and metastasis of head and neck squamous cell carcinoma (HNSCC). Here, we present a chemically-induced HNSCC mouse model, genetically and pathologically similar to human HNSCC. This protocol describes how to use genetic lineage tracing based on the Cre-loxP recombination strategy, which allows us to study the regulation and targeting of BMI1+ CSCs in primary tumors and lymph node metastases. For complete details on the use and execution of this protocol, please refer to Chen et al. (2017) and Jia et al. (2020).


Subject(s)
Head and Neck Neoplasms , Neoplasms, Experimental , Neoplastic Stem Cells , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins , Squamous Cell Carcinoma of Head and Neck , Animals , Cell Lineage/genetics , Cells, Cultured , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Immunohistochemistry , Male , Mice , Neoplastic Stem Cells/classification , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
14.
Cell Stem Cell ; 28(9): 1597-1613.e7, 2021 09 02.
Article in English | MEDLINE | ID: mdl-33945793

ABSTRACT

Immunosurveillance is a critical mechanism guarding against tumor development and progression. Checkpoint inhibitors have shown significant success in cancer treatment, but expression of key factors such as PD-L1 in putative cancer stem cell (CSC) populations in squamous cell carcinoma has been inconclusive, suggesting that CSCs may have developed other mechanisms to escape immune surveillance. Here we show that CSCs upregulate the immune checkpoint molecule CD276 (B7-H3) to evade host immune responses. CD276 is highly expressed by CSCs in mouse and human head and neck squamous cell carcinoma (HNSCC) and can be used to prospectively isolate tumorigenic CSCs. Anti-CD276 antibodies eliminate CSCs in a CD8+ T cell-dependent manner, inhibiting tumor growth and lymph node metastases in a mouse HNSCC model. Single-cell RNA sequencing (RNA-seq) showed that CD276 blockade remodels SCC heterogeneity and reduces epithelial-mesenchymal transition. These results show that CSCs utilize CD276 for immune escape and suggest that targeting CD276 may reduce CSCs in HNSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Animals , Cell Line, Tumor , Mice , Neoplastic Stem Cells , Squamous Cell Carcinoma of Head and Neck
16.
Mol Cell ; 81(10): 2148-2165.e9, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33743195

ABSTRACT

Developing strategies to activate tumor-cell-intrinsic immune response is critical for improving tumor immunotherapy by exploiting tumor vulnerability. KDM4A, as a histone H3 lysine 9 trimethylation (H3K9me3) demethylase, has been found to play a critical role in squamous cell carcinoma (SCC) growth and metastasis. Here we report that KDM4A inhibition promoted heterochromatin compaction and induced DNA replication stress, which elicited antitumor immunity in SCC. Mechanistically, KDM4A inhibition promoted the formation of liquid-like HP1γ puncta on heterochromatin and stall DNA replication, which activated tumor-cell-intrinsic cGAS-STING signaling through replication-stress-induced cytosolic DNA accumulation. Moreover, KDM4A inhibition collaborated with PD1 blockade to inhibit SCC growth and metastasis by recruiting and activating CD8+ T cells. In vivo lineage tracing demonstrated that KDM4A inhibition plus PD1 blockade efficiently eliminated cancer stem cells. Altogether, our results demonstrate that targeting KDM4A can activate anti-tumor immunity and enable PD1 blockade immunotherapy by aggravating replication stress in SCC cells.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , DNA Replication/genetics , Epigenesis, Genetic , Histone Demethylases/metabolism , Immunity/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Stress, Physiological/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Chemokines/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage/genetics , Epithelial Cells/metabolism , Gene Deletion , Humans , Lymphatic Metastasis , Mice, Transgenic , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Programmed Cell Death 1 Receptor/metabolism , Receptors, CXCR3/metabolism , Th1 Cells/immunology
18.
Nat Commun ; 12(1): 1293, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637721

ABSTRACT

Tendon injuries disrupt the balance between stability and mobility, causing compromised functions and disabilities. The regeneration of mature, functional tendons remains a clinical challenge. Here, we perform transcriptional profiling of tendon developmental processes to show that the extracellular matrix-associated protein periostin (Postn) contributes to the maintenance of tendon stem/progenitor cell (TSPC) functions and promotes tendon regeneration. We show that recombinant periostin (rPOSTN) promotes the proliferation and stemness of TSPCs, and maintains the tenogenic potentials of TSPCs in vitro. We also find that rPOSTN protects TSPCs against functional impairment during long-term passage in vitro. For in vivo tendon formation, we construct a biomimetic parallel-aligned collagen scaffold to facilitate TSPC tenogenesis. Using a rat full-cut Achilles tendon defect model, we demonstrate that scaffolds loaded with rPOSTN promote endogenous TSPC recruitment, tendon regeneration and repair with native-like hierarchically organized collagen fibers. Moreover, newly regenerated tendons show recovery of mechanical properties and locomotion functions.


Subject(s)
Biomimetics/methods , Cell Adhesion Molecules/genetics , Regeneration , Tissue Scaffolds , Achilles Tendon , Animals , Biomedical Engineering/methods , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Collagen , Gene Expression Profiling , Male , Rats , Rats, Sprague-Dawley , Regeneration/genetics , Stem Cells/metabolism , Tendon Injuries/therapy
19.
Cell Stem Cell ; 28(6): 1057-1073.e7, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33571444

ABSTRACT

Skeletal aging is a complex process, characterized by a decrease in bone formation, an increase in marrow fat, and stem cell exhaustion. Loss of H3K9me3, a heterochromatin mark, has been proposed to be associated with aging. Here, we report that loss of KDM4B in mesenchymal stromal cells (MSCs) exacerbated skeletal aging and osteoporosis by reducing bone formation and increasing marrow adiposity via increasing H3K9me3. KDM4B epigenetically coordinated ß-catenin/Smad1-mediated transcription by removing repressive H3K9me3. Importantly, KDM4B ablation impaired MSC self-renewal and promoted MSC exhaustion by inducing senescence-associated heterochromatin foci formation, providing a mechanistic explanation for stem cell exhaustion with aging. Moreover, while KDM4B was required for parathyroid hormone-mediated bone anabolism, KDM4B depletion accelerated bone loss and marrow adiposity induced by a high-fat diet. Our results suggest that the epigenetic rejuvenation and reversing bone-fat imbalance might be new strategies for preventing and treating skeletal aging and osteoporosis by activating KDM4B in MSCs.


Subject(s)
Mesenchymal Stem Cells , Bone Marrow , Bone Marrow Cells , Cell Differentiation , Osteogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...