Bone ResearchPub Date : 2024-08-27DOI: 10.1038/s41413-024-00350-8
Kun Li, Bo Yang, Yingying Du, Yi Ding, Shihui Shen, Zhengwang Sun, Yun Liu, Yuhan Wang, Siyuan Cao, Wenjie Ren, Xiangyu Wang, Mengjuan Li, Yunpeng Zhang, Juan Wu, Wei Zheng, Wangjun Yan, Lei Li
{"title":"The HOXC10/NOD1/ERK axis drives osteolytic bone metastasis of pan-KRAS-mutant lung cancer.","authors":"Kun Li, Bo Yang, Yingying Du, Yi Ding, Shihui Shen, Zhengwang Sun, Yun Liu, Yuhan Wang, Siyuan Cao, Wenjie Ren, Xiangyu Wang, Mengjuan Li, Yunpeng Zhang, Juan Wu, Wei Zheng, Wangjun Yan, Lei Li","doi":"10.1038/s41413-024-00350-8","DOIUrl":"10.1038/s41413-024-00350-8","url":null,"abstract":"<p><p>While KRAS mutation is the leading cause of low survival rates in lung cancer bone metastasis patients, effective treatments are still lacking. Here, we identified homeobox C10 (HOXC10) as a lynchpin in pan-KRAS-mutant lung cancer bone metastasis. Through RNA-seq approach and patient tissue studies, we demonstrated that HOXC10 expression was dramatically increased. Genetic depletion of HOXC10 preferentially impeded cell proliferation and migration in vitro. The bioluminescence imaging and micro-CT results demonstrated that inhibition of HOXC10 significantly reduced bone metastasis of KRAS-mutant lung cancer in vivo. Mechanistically, the transcription factor HOXC10 activated NOD1/ERK signaling pathway to reprogram epithelial-mesenchymal transition (EMT) and bone microenvironment by activating the NOD1 promoter. Strikingly, inhibition of HOXC10 in combination with STAT3 inhibitor was effective against KRAS-mutant lung cancer bone metastasis by triggering ferroptosis. Taken together, these findings reveal that HOXC10 effectively alleviates pan-KRAS-mutant lung cancer with bone metastasis in the NOD1/ERK axis-dependent manner, and support further development of an effective combinatorial strategy for this kind of disease.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"12 1","pages":"47"},"PeriodicalIF":14.3,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11349752/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142079182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bone ResearchPub Date : 2024-08-26DOI: 10.1038/s41413-024-00349-1
Na Li, Baohong Shi, Zan Li, Jie Han, Jun Sun, Haitao Huang, Alisha R Yallowitz, Seoyeon Bok, Shuang Xiao, Zuoxing Wu, Yu Chen, Yan Xu, Tian Qin, Rui Huang, Haiping Zheng, Rong Shen, Lin Meng, Matthew B Greenblatt, Ren Xu
{"title":"Schnurri-3 inhibition rescues skeletal fragility and vascular skeletal stem cell niche pathology in the OIM model of osteogenesis imperfecta.","authors":"Na Li, Baohong Shi, Zan Li, Jie Han, Jun Sun, Haitao Huang, Alisha R Yallowitz, Seoyeon Bok, Shuang Xiao, Zuoxing Wu, Yu Chen, Yan Xu, Tian Qin, Rui Huang, Haiping Zheng, Rong Shen, Lin Meng, Matthew B Greenblatt, Ren Xu","doi":"10.1038/s41413-024-00349-1","DOIUrl":"10.1038/s41413-024-00349-1","url":null,"abstract":"<p><p>Osteogenesis imperfecta (OI) is a disorder of low bone mass and increased fracture risk due to a range of genetic variants that prominently include mutations in genes encoding type I collagen. While it is well known that OI reflects defects in the activity of bone-forming osteoblasts, it is currently unclear whether OI also reflects defects in the many other cell types comprising bone, including defects in skeletal vascular endothelium or the skeletal stem cell populations that give rise to osteoblasts and whether correcting these broader defects could have therapeutic utility. Here, we find that numbers of skeletal stem cells (SSCs) and skeletal arterial endothelial cells (AECs) are augmented in Col1a2<sup>oim/oim</sup> mice, a well-studied animal model of moderate to severe OI, suggesting that disruption of a vascular SSC niche is a feature of OI pathogenesis. Moreover, crossing Col1a2<sup>oim/oim</sup> mice to mice lacking a negative regulator of skeletal angiogenesis and bone formation, Schnurri 3 (SHN3), not only corrected the SSC and AEC phenotypes but moreover robustly corrected the bone mass and spontaneous fracture phenotypes. As this finding suggested a strong therapeutic utility of SHN3 inhibition for the treatment of OI, a bone-targeting AAV was used to mediate Shn3 knockdown, rescuing the Col1a2<sup>oim/oim</sup> phenotype and providing therapeutic proof-of-concept for targeting SHN3 for the treatment of OI. Overall, this work both provides proof-of-concept for inhibition of the SHN3 pathway and more broadly addressing defects in the stem/osteoprogenitor niche as is a strategy to treat OI.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"12 1","pages":"46"},"PeriodicalIF":14.3,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11345453/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142054853","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bone ResearchPub Date : 2024-08-21DOI: 10.1038/s41413-024-00351-7
Ziwei Luo, Wanyi Wei, Dawei Qiu, Zixia Su, Liangpu Liu, Honghai Zhou, Hao Cui, Li Yang
{"title":"Rejuvenation of BMSCs senescence by pharmacological enhancement of TFEB-mediated autophagy alleviates aged-related bone loss and extends lifespan in middle aged mice.","authors":"Ziwei Luo, Wanyi Wei, Dawei Qiu, Zixia Su, Liangpu Liu, Honghai Zhou, Hao Cui, Li Yang","doi":"10.1038/s41413-024-00351-7","DOIUrl":"10.1038/s41413-024-00351-7","url":null,"abstract":"<p><p>Bone marrow stromal/stem cells (BMSCs) are generally considered as common progenitors for both osteoblasts and adipocytes in the bone marrow, but show preferential differentiation into adipocytes rather than osteoblasts under aging, thus leading to senile osteoporosis. Accumulated evidences indicate that rejuvenation of BMSCs by autophagic enhancement delays bone aging. Here we synthetized and demonstrated a novel autophagy activator, CXM102 that could induce autophagy in aged BMSCs, resulting in rejuvenation and preferential differentiation into osteoblasts of BMSCs. Furthermore, CXM102 significantly stimulated bone anabolism, reduced marrow adipocytes, and delayed bone loss in middle-age male mice. Mechanistically, CXM102 promoted transcription factor EB (TFEB) nuclear translocation and favored osteoblasts formation both in vitro and in vivo. Moreover, CXM102 decreased serum levels of inflammation and reduced organ fibrosis, leading to a prolonger lifespan in male mice. Our results indicated that CXM102 could be used as an autophagy inducer to rejuvenate BMSCs and shed new lights on strategies for senile osteoporosis and healthyspan improvement.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"12 1","pages":"45"},"PeriodicalIF":14.3,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11336217/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142008252","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Coactivator-independent vitamin D receptor signaling causes severe rickets in mice, that is not prevented by a diet high in calcium, phosphate, and lactose","authors":"Stefanie Doms, Lieve Verlinden, Iris Janssens, Justine Vanhevel, Roy Eerlings, René Houtman, Shigeaki Kato, Chantal Mathieu, Brigitte Decallonne, Geert Carmeliet, Annemieke Verstuyf","doi":"10.1038/s41413-024-00343-7","DOIUrl":"https://doi.org/10.1038/s41413-024-00343-7","url":null,"abstract":"<p>The vitamin D receptor (VDR) plays a critical role in the regulation of mineral and bone homeostasis. Upon binding of 1α,25-dihydroxyvitamin D<sub>3</sub> to the VDR, the activation function 2 (AF2) domain repositions and recruits coactivators for the assembly of the transcriptional machinery required for gene transcription. In contrast to coactivator-induced transcriptional activation, the functional effects of coactivator-independent VDR signaling remain unclear. In humans, mutations in the AF2 domain are associated with hereditary vitamin D-resistant rickets, a genetic disorder characterized by impaired bone mineralization and growth. In the present study, we used mice with a systemic or conditional deletion of the VDR-AF2 domain (<i>Vdr</i><sup><i>ΔAF2</i></sup>) to study coactivator-independent VDR signaling. We confirm that ligand-induced transcriptional activation was disabled because the mutant VDR<sup>ΔAF2</sup> protein was unable to interact with coactivators. Systemic <i>Vdr</i><sup><i>ΔAF2</i></sup> mice developed short, undermineralized bones with dysmorphic growth plates, a bone phenotype that was more pronounced than that of systemic <i>Vdr</i> knockout (<i>Vdr</i><sup><i>−/−</i></sup>) mice. Interestingly, a rescue diet that is high in calcium, phosphate, and lactose, normalized this phenotype in <i>Vdr</i><sup><i>−/−</i></sup>, but not in <i>Vdr</i><sup><i>ΔAF2</i></sup> mice. However, osteoblast- and osteoclast-specific <i>Vdr</i><sup><i>ΔAF2</i></sup> mice did not recapitulate this bone phenotype indicating coactivator-independent VDR effects are more important in other organs. In addition, RNA-sequencing analysis of duodenum and kidney revealed a decreased expression of VDR target genes in systemic <i>Vdr</i><sup><i>ΔAF2</i></sup> mice, which was not observed in <i>Vdr</i><sup><i>−/−</i></sup> mice. These genes could provide new insights in the compensatory (re)absorption of minerals that are crucial for bone homeostasis. In summary, coactivator-independent VDR effects contribute to mineral and bone homeostasis.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"38 1","pages":""},"PeriodicalIF":12.7,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142007311","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bone ResearchPub Date : 2024-08-05DOI: 10.1038/s41413-024-00341-9
Lena Batoon, Amy Jean Koh, Susan Marie Millard, Jobanpreet Grewal, Fang Ming Choo, Rahasudha Kannan, Aysia Kinnaird, Megan Avey, Tatyana Teslya, Allison Robyn Pettit, Laurie K. McCauley, Hernan Roca
{"title":"Induction of osteoblast apoptosis stimulates macrophage efferocytosis and paradoxical bone formation","authors":"Lena Batoon, Amy Jean Koh, Susan Marie Millard, Jobanpreet Grewal, Fang Ming Choo, Rahasudha Kannan, Aysia Kinnaird, Megan Avey, Tatyana Teslya, Allison Robyn Pettit, Laurie K. McCauley, Hernan Roca","doi":"10.1038/s41413-024-00341-9","DOIUrl":"https://doi.org/10.1038/s41413-024-00341-9","url":null,"abstract":"<p>Apoptosis is crucial for tissue homeostasis and organ development. In bone, apoptosis is recognized to be a main fate of osteoblasts, yet the relevance of this process remains underexplored. Using our murine model with inducible Caspase 9, the enzyme that initiates intrinsic apoptosis, we triggered apoptosis in a proportion of mature osteocalcin (OCN<sup>+</sup>) osteoblasts and investigated the impact on postnatal bone development. Osteoblast apoptosis stimulated efferocytosis by osteal macrophages. A five-week stimulation of OCN<sup>+</sup> osteoblast apoptosis in 3-week-old male and female mice significantly enhanced vertebral bone formation while increasing osteoblast precursors. A similar treatment regimen to stimulate osterix<sup>+</sup> cell apoptosis had no impact on bone volume or density. The vertebral bone accrual following stimulation of OCN<sup>+</sup> osteoblast apoptosis did not translate in improved mechanical strength due to disruption of the lacunocanalicular network. The observed bone phenotype was not influenced by changes in osteoclasts but was associated with stimulation of macrophage efferocytosis and vasculature formation. Phenotyping of efferocytic macrophages revealed a unique transcriptomic signature and expression of factors including VEGFA. To examine whether macrophages participated in the osteoblast precursor increase following osteoblast apoptosis, macrophage depletion models were employed. Depletion of macrophages via clodronate-liposomes and the CD169-diphtheria toxin receptor mouse model resulted in marked reduction in leptin receptor<sup>+</sup> and osterix<sup>+</sup> osteoblast precursors. Collectively, this work demonstrates the significance of osteoblast turnover via apoptosis and efferocytosis in postnatal bone formation. Importantly, it exposes the potential of targeting this mechanism to promote bone anabolism in the clinical setting.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"1 1","pages":""},"PeriodicalIF":12.7,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892099","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bone ResearchPub Date : 2024-08-05DOI: 10.1038/s41413-024-00346-4
Bobin Mi, Yuan Xiong, Samuel Knoedler, Michael Alfertshofer, Adriana C. Panayi, Haixing Wang, Sien Lin, Gang Li, Guohui Liu
{"title":"Ageing-related bone and immunity changes: insights into the complex interplay between the skeleton and the immune system","authors":"Bobin Mi, Yuan Xiong, Samuel Knoedler, Michael Alfertshofer, Adriana C. Panayi, Haixing Wang, Sien Lin, Gang Li, Guohui Liu","doi":"10.1038/s41413-024-00346-4","DOIUrl":"https://doi.org/10.1038/s41413-024-00346-4","url":null,"abstract":"<p>Ageing as a natural irreversible process inherently results in the functional deterioration of numerous organ systems and tissues, including the skeletal and immune systems. Recent studies have elucidated the intricate bidirectional interactions between these two systems. In this review, we provide a comprehensive synthesis of molecular mechanisms of cell ageing. We further discuss how age-related skeletal changes influence the immune system and the consequent impact of immune system alterations on the skeletal system. Finally, we highlight the clinical implications of these findings and propose potential strategies to promote healthy ageing and reduce pathologic deterioration of both the skeletal and immune systems.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"82 1","pages":""},"PeriodicalIF":12.7,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892113","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bone ResearchPub Date : 2024-07-17DOI: 10.1038/s41413-024-00344-6
Yan Shao, Hongbo Zhang, Hong Guan, Chunyu Wu, Weizhong Qi, Lingfeng Yang, Jianbin Yin, Haiyan Zhang, Liangliang Liu, Yuheng Lu, Yitao Zhao, Sheng Zhang, Chun Zeng, Guiqing Wang, Xiaochun Bai, Daozhang Cai
{"title":"PDZK1 protects against mechanical overload-induced chondrocyte senescence and osteoarthritis by targeting mitochondrial function","authors":"Yan Shao, Hongbo Zhang, Hong Guan, Chunyu Wu, Weizhong Qi, Lingfeng Yang, Jianbin Yin, Haiyan Zhang, Liangliang Liu, Yuheng Lu, Yitao Zhao, Sheng Zhang, Chun Zeng, Guiqing Wang, Xiaochun Bai, Daozhang Cai","doi":"10.1038/s41413-024-00344-6","DOIUrl":"https://doi.org/10.1038/s41413-024-00344-6","url":null,"abstract":"<p>Mechanical overloading and aging are two essential factors for osteoarthritis (OA) development. Mitochondria have been identified as a mechano-transducer situated between extracellular mechanical signals and chondrocyte biology, but their roles and the associated mechanisms in mechanical stress-associated chondrocyte senescence and OA have not been elucidated. Herein, we found that PDZ domain containing 1 (PDZK1), one of the PDZ proteins, which belongs to the Na<sup>+</sup>/H<sup>+</sup> Exchanger (NHE) regulatory factor family, is a key factor in biomechanically induced mitochondrial dysfunction and chondrocyte senescence during OA progression. PDZK1 is reduced by mechanical overload, and is diminished in the articular cartilage of OA patients, aged mice and OA mice. <i>Pdzk1</i> knockout in chondrocytes exacerbates mechanical overload-induced cartilage degeneration, whereas intraarticular injection of adeno-associated virus-expressing PDZK1 had a therapeutic effect. Moreover, PDZK1 loss impaired chondrocyte mitochondrial function with accumulated damaged mitochondria, decreased mitochondrion DNA (mtDNA) content and increased reactive oxygen species (ROS) production. PDZK1 supplementation or mitoubiquinone (MitoQ) application alleviated chondrocyte senescence and cartilage degeneration and significantly protected chondrocyte mitochondrial functions. MRNA sequencing in articular cartilage from <i>Pdzk1</i> knockout mice and controls showed that PDZK1 deficiency in chondrocytes interfered with mitochondrial function through inhibiting Hmgcs2 by increasing its ubiquitination. Our results suggested that PDZK1 deficiency plays a crucial role in mediating excessive mechanical load-induced chondrocyte senescence and is associated with mitochondrial dysfunction. PDZK1 overexpression or preservation of mitochondrial functions by MitoQ might present a new therapeutic approach for mechanical overload-induced OA.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"23 1","pages":""},"PeriodicalIF":12.7,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141631314","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bone ResearchPub Date : 2024-07-11DOI: 10.1038/s41413-024-00338-4
Brenda Krishnacoumar, Martin Stenzel, Hilal Garibagaoglu, Yasunori Omata, Rachel L Sworn, Thea Hofmann, Natacha Ipseiz, Magdalena A Czubala, Ulrike Steffen, Antonio Maccataio, Cornelia Stoll, Christina Böhm, Martin Herrmann, Stefan Uderhardt, Robert H Jenkins, Philip R Taylor, Anika Grüneboom, Mario M Zaiss, Georg Schett, Gerhard Krönke, Carina Scholtysek
{"title":"Caspase-8 promotes scramblase-mediated phosphatidylserine exposure and fusion of osteoclast precursors.","authors":"Brenda Krishnacoumar, Martin Stenzel, Hilal Garibagaoglu, Yasunori Omata, Rachel L Sworn, Thea Hofmann, Natacha Ipseiz, Magdalena A Czubala, Ulrike Steffen, Antonio Maccataio, Cornelia Stoll, Christina Böhm, Martin Herrmann, Stefan Uderhardt, Robert H Jenkins, Philip R Taylor, Anika Grüneboom, Mario M Zaiss, Georg Schett, Gerhard Krönke, Carina Scholtysek","doi":"10.1038/s41413-024-00338-4","DOIUrl":"10.1038/s41413-024-00338-4","url":null,"abstract":"<p><p>Efficient cellular fusion of mononuclear precursors is the prerequisite for the generation of fully functional multinucleated bone-resorbing osteoclasts. However, the exact molecular factors and mechanisms controlling osteoclast fusion remain incompletely understood. Here we identify RANKL-mediated activation of caspase-8 as early key event during osteoclast fusion. Single cell RNA sequencing-based analyses suggested that activation of parts of the apoptotic machinery accompanied the differentiation of osteoclast precursors into mature multinucleated osteoclasts. A subsequent characterization of osteoclast precursors confirmed that RANKL-mediated activation of caspase-8 promoted the non-apoptotic cleavage and activation of downstream effector caspases that translocated to the plasma membrane where they triggered activation of the phospholipid scramblase Xkr8. Xkr8-mediated exposure of phosphatidylserine, in turn, aided cellular fusion of osteoclast precursors and thereby allowed generation of functional multinucleated osteoclast syncytia and initiation of bone resorption. Pharmacological blockage or genetic deletion of caspase-8 accordingly interfered with fusion of osteoclasts and bone resorption resulting in increased bone mass in mice carrying a conditional deletion of caspase-8 in mononuclear osteoclast precursors. These data identify a novel pathway controlling osteoclast biology and bone turnover with the potential to serve as target for therapeutic intervention during diseases characterized by pathologic osteoclast-mediated bone loss. Proposed model of osteoclast fusion regulated by caspase-8 activation and PS exposure. RANK/RANK-L interaction. Activation of procaspase-8 into caspase-8. Caspase-8 activates caspase-3. Active capase-3 cleaves Xkr8. Local PS exposure is induced. Exposed PS is recognized by the fusion partner. FUSION. PS is re-internalized.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"12 1","pages":"40"},"PeriodicalIF":3.784,"publicationDate":"2024-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11237014/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141578963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bone ResearchPub Date : 2024-07-10DOI: 10.1038/s41413-024-00342-8
Lifang Hu, Wei Chen, Airong Qian, Yi-Ping Li
{"title":"Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and disease","authors":"Lifang Hu, Wei Chen, Airong Qian, Yi-Ping Li","doi":"10.1038/s41413-024-00342-8","DOIUrl":"https://doi.org/10.1038/s41413-024-00342-8","url":null,"abstract":"<p>Wnts are secreted, lipid-modified proteins that bind to different receptors on the cell surface to activate canonical or non-canonical Wnt signaling pathways, which control various biological processes throughout embryonic development and adult life. Aberrant Wnt signaling pathway underlies a wide range of human disease pathogeneses. In this review, we provide an update of Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and diseases. The Wnt proteins, receptors, activators, inhibitors, and the crosstalk of Wnt signaling pathways with other signaling pathways are summarized and discussed. We mainly review Wnt signaling functions in bone formation, homeostasis, and related diseases, and summarize mouse models carrying genetic modifications of Wnt signaling components. Moreover, the therapeutic strategies for treating bone diseases by targeting Wnt signaling, including the extracellular molecules, cytosol components, and nuclear components of Wnt signaling are reviewed. In summary, this paper reviews our current understanding of the mechanisms by which Wnt signaling regulates bone formation, homeostasis, and the efforts targeting Wnt signaling for treating bone diseases. Finally, the paper evaluates the important questions in Wnt signaling to be further explored based on the progress of new biological analytical technologies.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"61 1","pages":""},"PeriodicalIF":12.7,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141566232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"PCLAF induces bone marrow adipocyte senescence and contributes to skeletal aging.","authors":"Lingqi Xie, Yalun Cheng, Biao Hu, Xin Chen, Yuze An, Zhuying Xia, Guangping Cai, Changjun Li, Hui Peng","doi":"10.1038/s41413-024-00337-5","DOIUrl":"10.1038/s41413-024-00337-5","url":null,"abstract":"<p><p>Bone marrow adipocytes (BMAds) affect bone homeostasis, but the mechanism remains unclear. Here, we showed that exercise inhibited PCNA clamp-associated factor (PCLAF) secretion from the bone marrow macrophages to inhibit BMAds senescence and thus alleviated skeletal aging. The genetic deletion of PCLAF in macrophages inhibited BMAds senescence and delayed skeletal aging. In contrast, the transplantation of PCLAF-mediated senescent BMAds into the bone marrow of healthy mice suppressed bone turnover. Mechanistically, PCLAF bound to the ADGRL2 receptor to inhibit AKT/mTOR signaling that triggered BMAds senescence and subsequently spread senescence among osteogenic and osteoclastic cells. Of note, we developed a PCLAF-neutralizing antibody and showed its therapeutic effects on skeletal health in old mice. Together, these findings identify PCLAF as an inducer of BMAds senescence and provide a promising way to treat age-related osteoporosis.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":"12 1","pages":"38"},"PeriodicalIF":14.3,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11222446/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141497131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}