首页 > 最新文献

Bone Research最新文献

英文 中文
Aging impairs the osteocytic regulation of collagen integrity and bone quality 衰老会损害骨细胞对胶原蛋白完整性和骨骼质量的调节作用
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-26 DOI: 10.1038/s41413-023-00303-7
Charles A. Schurman, Serra Kaya, Neha Dole, Nadja M. Maldonado Luna, Natalia Castillo, Ryan Potter, Jacob P. Rose, Joanna Bons, Christina D. King, Jordan B. Burton, Birgit Schilling, Simon Melov, Simon Tang, Eric Schaible, Tamara Alliston

Poor bone quality is a major factor in skeletal fragility in elderly individuals. The molecular mechanisms that establish and maintain bone quality, independent of bone mass, are unknown but are thought to be primarily determined by osteocytes. We hypothesize that the age-related decline in bone quality results from the suppression of osteocyte perilacunar/canalicular remodeling (PLR), which maintains bone material properties. We examined bones from young and aged mice with osteocyte-intrinsic repression of TGFβ signaling (TβRIIocy−/−) that suppresses PLR. The control aged bone displayed decreased TGFβ signaling and PLR, but aging did not worsen the existing PLR suppression in male TβRIIocy−/− bone. This relationship impacted the behavior of collagen material at the nanoscale and tissue scale in macromechanical tests. The effects of age on bone mass, density, and mineral material behavior were independent of osteocytic TGFβ. We determined that the decline in bone quality with age arises from the loss of osteocyte function and the loss of TGFβ-dependent maintenance of collagen integrity.

骨质差是导致老年人骨骼脆弱的一个主要因素。建立和维持骨质量(与骨量无关)的分子机制尚不清楚,但认为主要由骨细胞决定。我们假设,与年龄相关的骨质下降是由于维持骨材料特性的骨细胞周围/关节重塑(PLR)受到抑制所致。我们对抑制 TGFβ 信号传导的骨细胞内在抑制(TβRIIocy-/-)的年轻小鼠和老年小鼠的骨骼进行了研究。对照组的老化骨骼显示出 TGFβ 信号传导和 PLR 的减少,但老化并没有加剧 TβRIIocy-/- 雄性骨骼中现有的 PLR 抑制。这种关系影响了胶原蛋白材料在宏观力学测试中的纳米级和组织级行为。年龄对骨质量、密度和矿物材料行为的影响与骨细胞 TGFβ 无关。我们确定,随着年龄的增长,骨质量的下降是由于成骨细胞功能的丧失和依赖于 TGFβ 的胶原蛋白完整性的维持的丧失造成的。
{"title":"Aging impairs the osteocytic regulation of collagen integrity and bone quality","authors":"Charles A. Schurman, Serra Kaya, Neha Dole, Nadja M. Maldonado Luna, Natalia Castillo, Ryan Potter, Jacob P. Rose, Joanna Bons, Christina D. King, Jordan B. Burton, Birgit Schilling, Simon Melov, Simon Tang, Eric Schaible, Tamara Alliston","doi":"10.1038/s41413-023-00303-7","DOIUrl":"https://doi.org/10.1038/s41413-023-00303-7","url":null,"abstract":"<p>Poor bone quality is a major factor in skeletal fragility in elderly individuals. The molecular mechanisms that establish and maintain bone quality, independent of bone mass, are unknown but are thought to be primarily determined by osteocytes. We hypothesize that the age-related decline in bone quality results from the suppression of osteocyte perilacunar/canalicular remodeling (PLR), which maintains bone material properties. We examined bones from young and aged mice with osteocyte-intrinsic repression of TGFβ signaling (<i>TβRII</i><sup><i>ocy−/−</i></sup>) that suppresses PLR. The control aged bone displayed decreased TGFβ signaling and PLR, but aging did not worsen the existing PLR suppression in male <i>TβRII</i><sup><i>ocy−/−</i></sup> bone. This relationship impacted the behavior of collagen material at the nanoscale and tissue scale in macromechanical tests. The effects of age on bone mass, density, and mineral material behavior were independent of osteocytic TGFβ. We determined that the decline in bone quality with age arises from the loss of osteocyte function and the loss of TGFβ-dependent maintenance of collagen integrity.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139967439","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}
引用次数: 0
Piezo1 expression in chondrocytes controls endochondral ossification and osteoarthritis development. 软骨细胞中 Piezo1 的表达控制着软骨内骨化和骨关节炎的发展。
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-23 DOI: 10.1038/s41413-024-00315-x
Laura J Brylka, Assil-Ramin Alimy, Miriam E A Tschaffon-Müller, Shan Jiang, Tobias Malte Ballhause, Anke Baranowsky, Simon von Kroge, Julian Delsmann, Eva Pawlus, Kian Eghbalian, Klaus Püschel, Astrid Schoppa, Melanie Haffner-Luntzer, David J Beech, Frank Timo Beil, Michael Amling, Johannes Keller, Anita Ignatius, Timur A Yorgan, Tim Rolvien, Thorsten Schinke

Piezo proteins are mechanically activated ion channels, which are required for mechanosensing functions in a variety of cell types. While we and others have previously demonstrated that the expression of Piezo1 in osteoblast lineage cells is essential for bone-anabolic processes, there was only suggestive evidence indicating a role of Piezo1 and/or Piezo2 in cartilage. Here we addressed the question if and how chondrocyte expression of the mechanosensitive proteins Piezo1 or Piezo2 controls physiological endochondral ossification and pathological osteoarthritis (OA) development. Mice with chondrocyte-specific inactivation of Piezo1 (Piezo1Col2a1Cre), but not of Piezo2, developed a near absence of trabecular bone below the chondrogenic growth plate postnatally. Moreover, all Piezo1Col2a1Cre animals displayed multiple fractures of rib bones at 7 days of age, which were located close to the growth plates. While skeletal growth was only mildly affected in these mice, OA pathologies were markedly less pronounced compared to littermate controls at 60 weeks of age. Likewise, when OA was induced by anterior cruciate ligament transection, only the chondrocyte inactivation of Piezo1, not of Piezo2, resulted in attenuated articular cartilage degeneration. Importantly, osteophyte formation and maturation were also reduced in Piezo1Col2a1Cre mice. We further observed increased Piezo1 protein abundance in cartilaginous zones of human osteophytes. Finally, we identified Ptgs2 and Ccn2 as potentially relevant Piezo1 downstream genes in chondrocytes. Collectively, our data do not only demonstrate that Piezo1 is a critical regulator of physiological and pathological endochondral ossification processes, but also suggest that Piezo1 antagonists may be established as a novel approach to limit osteophyte formation in OA.

压电蛋白是机械激活的离子通道,在多种细胞类型中需要机械感应功能。我们和其他研究人员之前已经证明,成骨细胞系细胞中 Piezo1 的表达对骨合成代谢过程至关重要,但只有提示性证据表明 Piezo1 和/或 Piezo2 在软骨中发挥作用。在此,我们探讨了软骨细胞表达机械敏感蛋白 Piezo1 或 Piezo2 是否以及如何控制生理性软骨内骨化和病理性骨关节炎(OA)的发生这一问题。软骨细胞特异性失活 Piezo1(Piezo1Col2a1Cre)而非 Piezo2 的小鼠在出生后软骨生长板下方几乎没有骨小梁。此外,所有 Piezo1Col2a1Cre 动物在 7 日龄时都会出现多发性肋骨骨折,骨折部位靠近生长板。虽然这些小鼠的骨骼生长只受到轻微影响,但与同窝对照组相比,这些小鼠在60周龄时的OA病变明显减轻。同样,当通过前十字韧带横断诱发 OA 时,只有 Piezo1 而不是 Piezo2 的软骨细胞失活才会导致关节软骨退化减轻。重要的是,Piezo1Col2a1Cre 小鼠的骨赘形成和成熟也有所减少。我们进一步观察到,人类骨质增生软骨区的 Piezo1 蛋白丰度增加。最后,我们发现 Ptgs2 和 Ccn2 可能是软骨细胞中与 Piezo1 相关的下游基因。总之,我们的数据不仅证明了 Piezo1 是生理和病理软骨内骨化过程的关键调控因子,还表明 Piezo1 拮抗剂可能成为限制 OA 骨质增生形成的一种新方法。
{"title":"Piezo1 expression in chondrocytes controls endochondral ossification and osteoarthritis development.","authors":"Laura J Brylka, Assil-Ramin Alimy, Miriam E A Tschaffon-Müller, Shan Jiang, Tobias Malte Ballhause, Anke Baranowsky, Simon von Kroge, Julian Delsmann, Eva Pawlus, Kian Eghbalian, Klaus Püschel, Astrid Schoppa, Melanie Haffner-Luntzer, David J Beech, Frank Timo Beil, Michael Amling, Johannes Keller, Anita Ignatius, Timur A Yorgan, Tim Rolvien, Thorsten Schinke","doi":"10.1038/s41413-024-00315-x","DOIUrl":"10.1038/s41413-024-00315-x","url":null,"abstract":"<p><p>Piezo proteins are mechanically activated ion channels, which are required for mechanosensing functions in a variety of cell types. While we and others have previously demonstrated that the expression of Piezo1 in osteoblast lineage cells is essential for bone-anabolic processes, there was only suggestive evidence indicating a role of Piezo1 and/or Piezo2 in cartilage. Here we addressed the question if and how chondrocyte expression of the mechanosensitive proteins Piezo1 or Piezo2 controls physiological endochondral ossification and pathological osteoarthritis (OA) development. Mice with chondrocyte-specific inactivation of Piezo1 (Piezo1<sup>Col2a1Cre</sup>), but not of Piezo2, developed a near absence of trabecular bone below the chondrogenic growth plate postnatally. Moreover, all Piezo1<sup>Col2a1Cre</sup> animals displayed multiple fractures of rib bones at 7 days of age, which were located close to the growth plates. While skeletal growth was only mildly affected in these mice, OA pathologies were markedly less pronounced compared to littermate controls at 60 weeks of age. Likewise, when OA was induced by anterior cruciate ligament transection, only the chondrocyte inactivation of Piezo1, not of Piezo2, resulted in attenuated articular cartilage degeneration. Importantly, osteophyte formation and maturation were also reduced in Piezo1<sup>Col2a1Cre</sup> mice. We further observed increased Piezo1 protein abundance in cartilaginous zones of human osteophytes. Finally, we identified Ptgs2 and Ccn2 as potentially relevant Piezo1 downstream genes in chondrocytes. Collectively, our data do not only demonstrate that Piezo1 is a critical regulator of physiological and pathological endochondral ossification processes, but also suggest that Piezo1 antagonists may be established as a novel approach to limit osteophyte formation in OA.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10891122/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139939631","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}
引用次数: 0
Interorgan communication in neurogenic heterotopic ossification: the role of brain-derived extracellular vesicles. 神经源性异位骨化中的器官间通信:脑源性细胞外囊泡的作用。
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-22 DOI: 10.1038/s41413-023-00310-8
Weicheng Lu, Jianfei Yan, Chenyu Wang, Wenpin Qin, Xiaoxiao Han, Zixuan Qin, Yu Wei, Haoqing Xu, Jialu Gao, Changhe Gao, Tao Ye, Franklin R Tay, Lina Niu, Kai Jiao

Brain-derived extracellular vesicles participate in interorgan communication after traumatic brain injury by transporting pathogens to initiate secondary injury. Inflammasome-related proteins encapsulated in brain-derived extracellular vesicles can cross the blood‒brain barrier to reach distal tissues. These proteins initiate inflammatory dysfunction, such as neurogenic heterotopic ossification. This recurrent condition is highly debilitating to patients because of its relatively unknown pathogenesis and the lack of effective prophylactic intervention strategies. Accordingly, a rat model of neurogenic heterotopic ossification induced by combined traumatic brain injury and achillotenotomy was developed to address these two issues. Histological examination of the injured tendon revealed the coexistence of ectopic calcification and fibroblast pyroptosis. The relationships among brain-derived extracellular vesicles, fibroblast pyroptosis and ectopic calcification were further investigated in vitro and in vivo. Intravenous injection of the pyroptosis inhibitor Ac-YVAD-cmk reversed the development of neurogenic heterotopic ossification in vivo. The present work highlights the role of brain-derived extracellular vesicles in the pathogenesis of neurogenic heterotopic ossification and offers a potential strategy for preventing neurogenic heterotopic ossification after traumatic brain injury. Brain-derived extracellular vesicles (BEVs) are released after traumatic brain injury. These BEVs contain pathogens and participate in interorgan communication to initiate secondary injury in distal tissues. After achillotenotomy, the phagocytosis of BEVs by fibroblasts induces pyroptosis, which is a highly inflammatory form of lytic programmed cell death, in the injured tendon. Fibroblast pyroptosis leads to an increase in calcium and phosphorus concentrations and creates a microenvironment that promotes osteogenesis. Intravenous injection of the pyroptosis inhibitor Ac-YVAD-cmk suppressed fibroblast pyroptosis and effectively prevented the onset of heterotopic ossification after neuronal injury. The use of a pyroptosis inhibitor represents a potential strategy for the treatment of neurogenic heterotopic ossification.

脑源性细胞外囊泡参与脑外伤后的器官间交流,运输病原体以引发二次损伤。包裹在脑源性细胞外囊泡中的炎症相关蛋白可穿过血脑屏障到达远端组织。这些蛋白质会引发炎症功能障碍,如神经源性异位骨化。由于发病机制相对不明,且缺乏有效的预防性干预策略,这种反复发作的病症对患者造成了极大的伤害。因此,为了解决这两个问题,我们建立了一个由脑外伤和腱鞘切除术联合诱发神经源性异位骨化的大鼠模型。损伤肌腱的组织学检查显示,异位钙化和成纤维细胞热解同时存在。研究人员在体外和体内进一步研究了脑源性细胞外囊泡、成纤维细胞热解和异位钙化之间的关系。静脉注射热凋亡抑制剂 Ac-YVAD-cmk 逆转了体内神经源性异位骨化的发展。本研究强调了脑源性细胞外囊泡在神经源性异位骨化发病机制中的作用,并为预防脑外伤后神经源性异位骨化提供了一种潜在的策略。脑外伤后会释放脑源性细胞外囊泡(BEVs)。这些脑源性细胞外囊泡含有病原体,并参与器官间的交流,从而引发远端组织的继发性损伤。肌腱腱鞘切除术后,成纤维细胞对 BEVs 的吞噬会诱发受伤肌腱的热凋亡,这是一种高度炎症性的溶解性程序性细胞死亡。成纤维细胞的热解导致钙和磷浓度增加,并创造了一种促进成骨的微环境。静脉注射成纤维细胞增殖抑制剂 Ac-YVAD-cmk 可抑制成纤维细胞增殖,有效防止神经元损伤后异位骨化的发生。使用热蛋白沉积抑制剂是治疗神经源性异位骨化的一种潜在策略。
{"title":"Interorgan communication in neurogenic heterotopic ossification: the role of brain-derived extracellular vesicles.","authors":"Weicheng Lu, Jianfei Yan, Chenyu Wang, Wenpin Qin, Xiaoxiao Han, Zixuan Qin, Yu Wei, Haoqing Xu, Jialu Gao, Changhe Gao, Tao Ye, Franklin R Tay, Lina Niu, Kai Jiao","doi":"10.1038/s41413-023-00310-8","DOIUrl":"10.1038/s41413-023-00310-8","url":null,"abstract":"<p><p>Brain-derived extracellular vesicles participate in interorgan communication after traumatic brain injury by transporting pathogens to initiate secondary injury. Inflammasome-related proteins encapsulated in brain-derived extracellular vesicles can cross the blood‒brain barrier to reach distal tissues. These proteins initiate inflammatory dysfunction, such as neurogenic heterotopic ossification. This recurrent condition is highly debilitating to patients because of its relatively unknown pathogenesis and the lack of effective prophylactic intervention strategies. Accordingly, a rat model of neurogenic heterotopic ossification induced by combined traumatic brain injury and achillotenotomy was developed to address these two issues. Histological examination of the injured tendon revealed the coexistence of ectopic calcification and fibroblast pyroptosis. The relationships among brain-derived extracellular vesicles, fibroblast pyroptosis and ectopic calcification were further investigated in vitro and in vivo. Intravenous injection of the pyroptosis inhibitor Ac-YVAD-cmk reversed the development of neurogenic heterotopic ossification in vivo. The present work highlights the role of brain-derived extracellular vesicles in the pathogenesis of neurogenic heterotopic ossification and offers a potential strategy for preventing neurogenic heterotopic ossification after traumatic brain injury. Brain-derived extracellular vesicles (BEVs) are released after traumatic brain injury. These BEVs contain pathogens and participate in interorgan communication to initiate secondary injury in distal tissues. After achillotenotomy, the phagocytosis of BEVs by fibroblasts induces pyroptosis, which is a highly inflammatory form of lytic programmed cell death, in the injured tendon. Fibroblast pyroptosis leads to an increase in calcium and phosphorus concentrations and creates a microenvironment that promotes osteogenesis. Intravenous injection of the pyroptosis inhibitor Ac-YVAD-cmk suppressed fibroblast pyroptosis and effectively prevented the onset of heterotopic ossification after neuronal injury. The use of a pyroptosis inhibitor represents a potential strategy for the treatment of neurogenic heterotopic ossification.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-02-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10881583/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139930013","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}
引用次数: 0
RANKL inhibition reduces lesional cellularity and Gαs variant expression and enables osteogenic maturation in fibrous dysplasia 抑制 RANKL 可降低纤维发育不良的病变细胞性和 Gαs 变异表达,并促进成骨成熟
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-20 DOI: 10.1038/s41413-023-00311-7
Luis F. de Castro, Jarred M. Whitlock, Zachary Michel, Kristen Pan, Jocelyn Taylor, Vivian Szymczuk, Brendan Boyce, Daniel Martin, Vardit Kram, Rebeca Galisteo, Kamran Melikov, Leonid V. Chernomordik, Michael T. Collins, Alison M. Boyce

Fibrous dysplasia (FD) is a rare, disabling skeletal disease for which there are no established treatments. Growing evidence supports inhibiting the osteoclastogenic factor receptor activator of nuclear kappa-B ligand (RANKL) as a potential treatment strategy. In this study, we investigated the mechanisms underlying RANKL inhibition in FD tissue and its likely indirect effects on osteoprogenitors by evaluating human FD tissue pre- and post-treatment in a phase 2 clinical trial of denosumab (NCT03571191) and in murine in vivo and ex vivo preclinical models. Histological analysis of human and mouse tissue demonstrated increased osteogenic maturation, reduced cellularity, and reduced expression of the pathogenic Gαs variant in FD lesions after RANKL inhibition. RNA sequencing of human and mouse tissue supported these findings. The interaction between osteoclasts and mutant osteoprogenitors was further assessed in an ex vivo lesion model, which indicated that the proliferation of abnormal FD osteoprogenitors was dependent on osteoclasts. The results from this study demonstrated that, in addition to its expected antiosteoclastic effect, denosumab reduces FD lesion activity by decreasing FD cell proliferation and increasing osteogenic maturation, leading to increased bone formation within lesions. These findings highlight the unappreciated role of cellular crosstalk between osteoclasts and preosteoblasts/osteoblasts as a driver of FD pathology and demonstrate a novel mechanism of action of denosumab in the treatment of bone disease.

TRIAL REGISTRATION: ClinicalTrials.gov NCT03571191

纤维发育不良(FD)是一种罕见的致残性骨骼疾病,目前尚无成熟的治疗方法。越来越多的证据表明,抑制破骨细胞生成因子核卡巴-B配体受体激活剂(RANKL)是一种潜在的治疗策略。在本研究中,我们通过评估在地诺单抗(NCT03571191)2 期临床试验中治疗前后的人类 FD 组织以及小鼠体内和体外临床前模型,研究了抑制 FD 组织中 RANKL 的机制及其对骨生成细胞可能产生的间接影响。对人类和小鼠组织的组织学分析表明,RANKL抑制后,FD病变中的成骨成熟度增加、细胞减少、致病性Gαs变体表达降低。人和小鼠组织的 RNA 测序也支持这些发现。在体内外病变模型中进一步评估了破骨细胞与突变型成骨细胞之间的相互作用,结果表明异常FD成骨细胞的增殖依赖于破骨细胞。这项研究的结果表明,除预期的抗破骨细胞作用外,地诺单抗还能通过减少 FD 细胞增殖和增加成骨细胞成熟来降低 FD 病变活性,从而增加病变内的骨形成。这些发现凸显了破骨细胞和前成骨细胞/成骨细胞之间的细胞串扰作为 FD 病理学驱动因素所起的未被重视的作用,并证明了地诺单抗治疗骨病的一种新的作用机制。
{"title":"RANKL inhibition reduces lesional cellularity and Gαs variant expression and enables osteogenic maturation in fibrous dysplasia","authors":"Luis F. de Castro, Jarred M. Whitlock, Zachary Michel, Kristen Pan, Jocelyn Taylor, Vivian Szymczuk, Brendan Boyce, Daniel Martin, Vardit Kram, Rebeca Galisteo, Kamran Melikov, Leonid V. Chernomordik, Michael T. Collins, Alison M. Boyce","doi":"10.1038/s41413-023-00311-7","DOIUrl":"https://doi.org/10.1038/s41413-023-00311-7","url":null,"abstract":"<p>Fibrous dysplasia (FD) is a rare, disabling skeletal disease for which there are no established treatments. Growing evidence supports inhibiting the osteoclastogenic factor receptor activator of nuclear kappa-B ligand (RANKL) as a potential treatment strategy. In this study, we investigated the mechanisms underlying RANKL inhibition in FD tissue and its likely indirect effects on osteoprogenitors by evaluating human FD tissue pre- and post-treatment in a phase 2 clinical trial of denosumab (NCT03571191) and in murine in vivo and ex vivo preclinical models. Histological analysis of human and mouse tissue demonstrated increased osteogenic maturation, reduced cellularity, and reduced expression of the pathogenic Gα<sub>s</sub> variant in FD lesions after RANKL inhibition. RNA sequencing of human and mouse tissue supported these findings. The interaction between osteoclasts and mutant osteoprogenitors was further assessed in an ex vivo lesion model, which indicated that the proliferation of abnormal FD osteoprogenitors was dependent on osteoclasts. The results from this study demonstrated that, in addition to its expected antiosteoclastic effect, denosumab reduces FD lesion activity by decreasing FD cell proliferation and increasing osteogenic maturation, leading to increased bone formation within lesions. These findings highlight the unappreciated role of cellular crosstalk between osteoclasts and preosteoblasts/osteoblasts as a driver of FD pathology and demonstrate a novel mechanism of action of denosumab in the treatment of bone disease.</p><p>TRIAL REGISTRATION: ClinicalTrials.gov NCT03571191</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139909288","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}
引用次数: 0
Author Correction: Ammonia promotes the proliferation of bone marrow-derived mesenchymal stem cells by regulating the Akt/mTOR/S6k pathway. 作者更正:氨通过调节 Akt/mTOR/S6k 通路促进骨髓间充质干细胞增殖
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-20 DOI: 10.1038/s41413-024-00314-y
Yu Liu, Xiangxian Zhang, Wei Wang, Ting Liu, Jun Ren, Siyuan Chen, Tianqi Lu, Yan Tie, Xia Yuan, Fei Mo, Jingyun Yang, Yuquan Wei, Xiawei Wei
{"title":"Author Correction: Ammonia promotes the proliferation of bone marrow-derived mesenchymal stem cells by regulating the Akt/mTOR/S6k pathway.","authors":"Yu Liu, Xiangxian Zhang, Wei Wang, Ting Liu, Jun Ren, Siyuan Chen, Tianqi Lu, Yan Tie, Xia Yuan, Fei Mo, Jingyun Yang, Yuquan Wei, Xiawei Wei","doi":"10.1038/s41413-024-00314-y","DOIUrl":"10.1038/s41413-024-00314-y","url":null,"abstract":"","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10879115/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139911969","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}
引用次数: 0
Insights and implications of sexual dimorphism in osteoporosis. 骨质疏松症中性畸形的见解和影响。
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-18 DOI: 10.1038/s41413-023-00306-4
Yuan-Yuan Zhang, Na Xie, Xiao-Dong Sun, Edouard C Nice, Yih-Cherng Liou, Canhua Huang, Huili Zhu, Zhisen Shen

Osteoporosis, a metabolic bone disease characterized by low bone mineral density and deterioration of bone microarchitecture, has led to a high risk of fatal osteoporotic fractures worldwide. Accumulating evidence has revealed that sexual dimorphism is a notable feature of osteoporosis, with sex-specific differences in epidemiology and pathogenesis. Specifically, females are more susceptible than males to osteoporosis, while males are more prone to disability or death from the disease. To date, sex chromosome abnormalities and steroid hormones have been proven to contribute greatly to sexual dimorphism in osteoporosis by regulating the functions of bone cells. Understanding the sex-specific differences in osteoporosis and its related complications is essential for improving treatment strategies tailored to women and men. This literature review focuses on the mechanisms underlying sexual dimorphism in osteoporosis, mainly in a population of aging patients, chronic glucocorticoid administration, and diabetes. Moreover, we highlight the implications of sexual dimorphism for developing therapeutics and preventive strategies and screening approaches tailored to women and men. Additionally, the challenges in translating bench research to bedside treatments and future directions to overcome these obstacles will be discussed.

骨质疏松症是一种以骨矿物质密度低和骨微结构退化为特征的代谢性骨病,在全球范围内导致致命性骨质疏松性骨折的风险很高。越来越多的证据表明,性别二形性是骨质疏松症的一个显著特征,在流行病学和发病机制方面存在性别差异。具体来说,女性比男性更容易患上骨质疏松症,而男性则更容易因该病致残或死亡。迄今为止,性染色体异常和类固醇激素通过调节骨细胞的功能,已被证实在很大程度上导致了骨质疏松症的性别双态性。了解骨质疏松症及其相关并发症的性别差异对于改进针对女性和男性的治疗策略至关重要。这篇文献综述主要探讨了骨质疏松症中性二态性的机制,主要是在老龄化患者、长期服用糖皮质激素和糖尿病人群中。此外,我们还强调了性双态性对开发适合女性和男性的治疗和预防策略及筛查方法的影响。此外,我们还将讨论将临床研究转化为床边治疗所面临的挑战,以及克服这些障碍的未来方向。
{"title":"Insights and implications of sexual dimorphism in osteoporosis.","authors":"Yuan-Yuan Zhang, Na Xie, Xiao-Dong Sun, Edouard C Nice, Yih-Cherng Liou, Canhua Huang, Huili Zhu, Zhisen Shen","doi":"10.1038/s41413-023-00306-4","DOIUrl":"10.1038/s41413-023-00306-4","url":null,"abstract":"<p><p>Osteoporosis, a metabolic bone disease characterized by low bone mineral density and deterioration of bone microarchitecture, has led to a high risk of fatal osteoporotic fractures worldwide. Accumulating evidence has revealed that sexual dimorphism is a notable feature of osteoporosis, with sex-specific differences in epidemiology and pathogenesis. Specifically, females are more susceptible than males to osteoporosis, while males are more prone to disability or death from the disease. To date, sex chromosome abnormalities and steroid hormones have been proven to contribute greatly to sexual dimorphism in osteoporosis by regulating the functions of bone cells. Understanding the sex-specific differences in osteoporosis and its related complications is essential for improving treatment strategies tailored to women and men. This literature review focuses on the mechanisms underlying sexual dimorphism in osteoporosis, mainly in a population of aging patients, chronic glucocorticoid administration, and diabetes. Moreover, we highlight the implications of sexual dimorphism for developing therapeutics and preventive strategies and screening approaches tailored to women and men. Additionally, the challenges in translating bench research to bedside treatments and future directions to overcome these obstacles will be discussed.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-02-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10874461/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139898428","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}
引用次数: 0
Spatial analysis of the osteoarthritis microenvironment: techniques, insights, and applications 骨关节炎微环境的空间分析:技术、见解和应用
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-04 DOI: 10.1038/s41413-023-00304-6
Xiwei Fan, Antonia Rujia Sun, Reuben S. E. Young, Isaac O. Afara, Brett R. Hamilton, Louis Jun Ye Ong, Ross Crawford, Indira Prasadam

Osteoarthritis (OA) is a debilitating degenerative disease affecting multiple joint tissues, including cartilage, bone, synovium, and adipose tissues. OA presents diverse clinical phenotypes and distinct molecular endotypes, including inflammatory, metabolic, mechanical, genetic, and synovial variants. Consequently, innovative technologies are needed to support the development of effective diagnostic and precision therapeutic approaches. Traditional analysis of bulk OA tissue extracts has limitations due to technical constraints, causing challenges in the differentiation between various physiological and pathological phenotypes in joint tissues. This issue has led to standardization difficulties and hindered the success of clinical trials. Gaining insights into the spatial variations of the cellular and molecular structures in OA tissues, encompassing DNA, RNA, metabolites, and proteins, as well as their chemical properties, elemental composition, and mechanical attributes, can contribute to a more comprehensive understanding of the disease subtypes. Spatially resolved biology enables biologists to investigate cells within the context of their tissue microenvironment, providing a more holistic view of cellular function. Recent advances in innovative spatial biology techniques now allow intact tissue sections to be examined using various -omics lenses, such as genomics, transcriptomics, proteomics, and metabolomics, with spatial data. This fusion of approaches provides researchers with critical insights into the molecular composition and functions of the cells and tissues at precise spatial coordinates. Furthermore, advanced imaging techniques, including high-resolution microscopy, hyperspectral imaging, and mass spectrometry imaging, enable the visualization and analysis of the spatial distribution of biomolecules, cells, and tissues. Linking these molecular imaging outputs to conventional tissue histology can facilitate a more comprehensive characterization of disease phenotypes. This review summarizes the recent advancements in the molecular imaging modalities and methodologies for in-depth spatial analysis. It explores their applications, challenges, and potential opportunities in the field of OA. Additionally, this review provides a perspective on the potential research directions for these contemporary approaches that can meet the requirements of clinical diagnoses and the establishment of therapeutic targets for OA.

骨关节炎(OA)是一种使人衰弱的退行性疾病,影响多个关节组织,包括软骨、骨、滑膜和脂肪组织。OA 表现出多种临床表型和不同的分子内型,包括炎症、代谢、机械、遗传和滑膜变异。因此,需要创新技术来支持有效诊断和精准治疗方法的开发。由于技术限制,对大块 OA 组织提取物的传统分析存在局限性,给区分关节组织中的各种生理和病理表型带来了挑战。这一问题导致标准化困难,阻碍了临床试验的成功。深入了解 OA 组织中细胞和分子结构的空间变化,包括 DNA、RNA、代谢物和蛋白质,以及它们的化学特性、元素组成和机械属性,有助于更全面地了解疾病亚型。空间解析生物学使生物学家能够在细胞组织微环境的背景下研究细胞,从而更全面地了解细胞功能。创新性空间生物学技术的最新进展使完整的组织切片现在可以利用基因组学、转录组学、蛋白质组学和代谢组学等各种组学透镜和空间数据进行研究。这种方法的融合为研究人员在精确的空间坐标上深入了解细胞和组织的分子组成和功能提供了重要依据。此外,先进的成像技术,包括高分辨率显微镜、高光谱成像和质谱成像,使生物分子、细胞和组织空间分布的可视化和分析成为可能。将这些分子成像结果与传统的组织组织学联系起来,有助于更全面地描述疾病表型。本综述总结了分子成像模式和深入空间分析方法的最新进展。它探讨了它们在 OA 领域的应用、挑战和潜在机遇。此外,本综述还对这些当代方法的潜在研究方向提供了一个视角,以满足临床诊断和确立 OA 治疗靶点的要求。
{"title":"Spatial analysis of the osteoarthritis microenvironment: techniques, insights, and applications","authors":"Xiwei Fan, Antonia Rujia Sun, Reuben S. E. Young, Isaac O. Afara, Brett R. Hamilton, Louis Jun Ye Ong, Ross Crawford, Indira Prasadam","doi":"10.1038/s41413-023-00304-6","DOIUrl":"https://doi.org/10.1038/s41413-023-00304-6","url":null,"abstract":"<p>Osteoarthritis (OA) is a debilitating degenerative disease affecting multiple joint tissues, including cartilage, bone, synovium, and adipose tissues. OA presents diverse clinical phenotypes and distinct molecular endotypes, including inflammatory, metabolic, mechanical, genetic, and synovial variants. Consequently, innovative technologies are needed to support the development of effective diagnostic and precision therapeutic approaches. Traditional analysis of bulk OA tissue extracts has limitations due to technical constraints, causing challenges in the differentiation between various physiological and pathological phenotypes in joint tissues. This issue has led to standardization difficulties and hindered the success of clinical trials. Gaining insights into the spatial variations of the cellular and molecular structures in OA tissues, encompassing DNA, RNA, metabolites, and proteins, as well as their chemical properties, elemental composition, and mechanical attributes, can contribute to a more comprehensive understanding of the disease subtypes. Spatially resolved biology enables biologists to investigate cells within the context of their tissue microenvironment, providing a more holistic view of cellular function. Recent advances in innovative spatial biology techniques now allow intact tissue sections to be examined using various -omics lenses, such as genomics, transcriptomics, proteomics, and metabolomics, with spatial data. This fusion of approaches provides researchers with critical insights into the molecular composition and functions of the cells and tissues at precise spatial coordinates. Furthermore, advanced imaging techniques, including high-resolution microscopy, hyperspectral imaging, and mass spectrometry imaging, enable the visualization and analysis of the spatial distribution of biomolecules, cells, and tissues. Linking these molecular imaging outputs to conventional tissue histology can facilitate a more comprehensive characterization of disease phenotypes. This review summarizes the recent advancements in the molecular imaging modalities and methodologies for in-depth spatial analysis. It explores their applications, challenges, and potential opportunities in the field of OA. Additionally, this review provides a perspective on the potential research directions for these contemporary approaches that can meet the requirements of clinical diagnoses and the establishment of therapeutic targets for OA.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139676905","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}
引用次数: 0
Age-related secretion of grancalcin by macrophages induces skeletal stem/progenitor cell senescence during fracture healing. 在骨折愈合过程中,巨噬细胞与年龄相关的颗粒钙素分泌会诱导骨骼干细胞/祖细胞衰老。
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-25 DOI: 10.1038/s41413-023-00309-1
Nan-Yu Zou, Ran Liu, Mei Huang, Yu-Rui Jiao, Jie Wei, Yangzi Jiang, Wen-Zhen He, Min Huang, Yi-Li Xu, Ling Liu, Yu-Chen Sun, Mi Yang, Qi Guo, Yan Huang, Tian Su, Ye Xiao, Wei-Shan Wang, Chao Zeng, Guang-Hua Lei, Xiang-Hang Luo, Chang-Jun Li

Skeletal stem/progenitor cell (SSPC) senescence is a major cause of decreased bone regenerative potential with aging, but the causes of SSPC senescence remain unclear. In this study, we revealed that macrophages in calluses secrete prosenescent factors, including grancalcin (GCA), during aging, which triggers SSPC senescence and impairs fracture healing. Local injection of human rGCA in young mice induced SSPC senescence and delayed fracture repair. Genetic deletion of Gca in monocytes/macrophages was sufficient to rejuvenate fracture repair in aged mice and alleviate SSPC senescence. Mechanistically, GCA binds to the plexin-B2 receptor and activates Arg2-mediated mitochondrial dysfunction, resulting in cellular senescence. Depletion of Plxnb2 in SSPCs impaired fracture healing. Administration of GCA-neutralizing antibody enhanced fracture healing in aged mice. Thus, our study revealed that senescent macrophages within calluses secrete GCA to trigger SSPC secondary senescence, and GCA neutralization represents a promising therapy for nonunion or delayed union in elderly individuals.

骨骼干/祖细胞(SSPC)衰老是随着年龄增长骨再生潜力下降的主要原因,但SSPC衰老的原因仍不清楚。在这项研究中,我们发现胼胝体中的巨噬细胞在衰老过程中会分泌包括粒钙蛋白(GCA)在内的前衰老因子,从而引发SSPC衰老并影响骨折愈合。向幼鼠局部注射人rGCA可诱导SSPC衰老并延迟骨折修复。遗传性删除单核细胞/巨噬细胞中的Gca足以使老龄小鼠的骨折修复恢复活力并缓解SSPC衰老。从机理上讲,GCA 与 plexin-B2 受体结合并激活 Arg2 介导的线粒体功能障碍,从而导致细胞衰老。SSPCs中Plxnb2的消耗会影响骨折愈合。给予 GCA 中和抗体可增强老年小鼠的骨折愈合。因此,我们的研究揭示了胼胝体中的衰老巨噬细胞会分泌 GCA 引发 SSPC 继发性衰老,而 GCA 中和是一种治疗老年人骨折不愈合或延迟愈合的有效方法。
{"title":"Age-related secretion of grancalcin by macrophages induces skeletal stem/progenitor cell senescence during fracture healing.","authors":"Nan-Yu Zou, Ran Liu, Mei Huang, Yu-Rui Jiao, Jie Wei, Yangzi Jiang, Wen-Zhen He, Min Huang, Yi-Li Xu, Ling Liu, Yu-Chen Sun, Mi Yang, Qi Guo, Yan Huang, Tian Su, Ye Xiao, Wei-Shan Wang, Chao Zeng, Guang-Hua Lei, Xiang-Hang Luo, Chang-Jun Li","doi":"10.1038/s41413-023-00309-1","DOIUrl":"10.1038/s41413-023-00309-1","url":null,"abstract":"<p><p>Skeletal stem/progenitor cell (SSPC) senescence is a major cause of decreased bone regenerative potential with aging, but the causes of SSPC senescence remain unclear. In this study, we revealed that macrophages in calluses secrete prosenescent factors, including grancalcin (GCA), during aging, which triggers SSPC senescence and impairs fracture healing. Local injection of human rGCA in young mice induced SSPC senescence and delayed fracture repair. Genetic deletion of Gca in monocytes/macrophages was sufficient to rejuvenate fracture repair in aged mice and alleviate SSPC senescence. Mechanistically, GCA binds to the plexin-B2 receptor and activates Arg2-mediated mitochondrial dysfunction, resulting in cellular senescence. Depletion of Plxnb2 in SSPCs impaired fracture healing. Administration of GCA-neutralizing antibody enhanced fracture healing in aged mice. Thus, our study revealed that senescent macrophages within calluses secrete GCA to trigger SSPC secondary senescence, and GCA neutralization represents a promising therapy for nonunion or delayed union in elderly individuals.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10808101/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139545566","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}
引用次数: 0
Transcriptional reprogramming during human osteoclast differentiation identifies regulators of osteoclast activity. 人类破骨细胞分化过程中的转录重编程确定了破骨细胞活性的调节因子。
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-24 DOI: 10.1038/s41413-023-00312-6
Morten S Hansen, Kaja Madsen, Maria Price, Kent Søe, Yasunori Omata, Mario M Zaiss, Caroline M Gorvin, Morten Frost, Alexander Rauch

Enhanced osteoclastogenesis and osteoclast activity contribute to the development of osteoporosis, which is characterized by increased bone resorption and inadequate bone formation. As novel antiosteoporotic therapeutics are needed, understanding the genetic regulation of human osteoclastogenesis could help identify potential treatment targets. This study aimed to provide an overview of transcriptional reprogramming during human osteoclast differentiation. Osteoclasts were differentiated from CD14+ monocytes from eight female donors. RNA sequencing during differentiation revealed 8 980 differentially expressed genes grouped into eight temporal patterns conserved across donors. These patterns revealed distinct molecular functions associated with postmenopausal osteoporosis susceptibility genes based on RNA from iliac crest biopsies and bone mineral density SNPs. Network analyses revealed mutual dependencies between temporal expression patterns and provided insight into subtype-specific transcriptional networks. The donor-specific expression patterns revealed genes at the monocyte stage, such as filamin B (FLNB) and oxidized low-density lipoprotein receptor 1 (OLR1, encoding LOX-1), that are predictive of the resorptive activity of mature osteoclasts. The expression of differentially expressed G-protein coupled receptors was strong during osteoclast differentiation, and these receptors are associated with bone mineral density SNPs, suggesting that they play a pivotal role in osteoclast differentiation and activity. The regulatory effects of three differentially expressed G-protein coupled receptors were exemplified by in vitro pharmacological modulation of complement 5 A receptor 1 (C5AR1), somatostatin receptor 2 (SSTR2), and free fatty acid receptor 4 (FFAR4/GPR120). Activating C5AR1 enhanced osteoclast formation, while activating SSTR2 decreased the resorptive activity of mature osteoclasts, and activating FFAR4 decreased both the number and resorptive activity of mature osteoclasts. In conclusion, we report the occurrence of transcriptional reprogramming during human osteoclast differentiation and identified SSTR2 and FFAR4 as antiresorptive G-protein coupled receptors and FLNB and LOX-1 as potential molecular markers of osteoclast activity. These data can help future investigations identify molecular regulators of osteoclast differentiation and activity and provide the basis for novel antiosteoporotic targets.

破骨细胞生成和破骨细胞活性的增强导致了骨质疏松症的发生,骨质疏松症的特点是骨吸收增加和骨形成不足。由于需要新型抗骨质疏松症疗法,了解人类破骨细胞生成的基因调控有助于确定潜在的治疗靶点。本研究旨在概述人类破骨细胞分化过程中的转录重编程。破骨细胞由来自八名女性供体的 CD14+ 单核细胞分化而来。分化过程中的 RNA 测序发现了 8 980 个差异表达基因,这些基因分为 8 个时间模式,在不同供体之间保持一致。这些模式揭示了与绝经后骨质疏松症易感基因相关的不同分子功能,这些易感基因基于髂嵴活检组织的 RNA 和骨矿物质密度 SNPs。网络分析揭示了时间表达模式之间的相互依赖关系,并提供了对亚型特异性转录网络的深入了解。供体特异性表达模式揭示了单核细胞阶段的基因,如丝胶素B(FLNB)和氧化低密度脂蛋白受体1(OLR1,编码LOX-1),这些基因可预测成熟破骨细胞的吸收活性。在破骨细胞分化过程中,差异表达的G蛋白偶联受体的表达很强,而且这些受体与骨矿物质密度SNPs相关,这表明它们在破骨细胞分化和活性中起着关键作用。体外药理学调节补体5A受体1(C5AR1)、体生长激素受体2(SSTR2)和游离脂肪酸受体4(FFAR4/GPR120),体现了三种不同表达的G蛋白偶联受体的调控作用。激活 C5AR1 会促进破骨细胞的形成,而激活 SSTR2 则会降低成熟破骨细胞的吸收活性,激活 FFAR4 则会降低成熟破骨细胞的数量和吸收活性。总之,我们报告了人类破骨细胞分化过程中转录重编程的发生,并确定 SSTR2 和 FFAR4 为抗吸收性 G 蛋白偶联受体,FLNB 和 LOX-1 为破骨细胞活性的潜在分子标记。这些数据有助于未来的研究确定破骨细胞分化和活性的分子调控因子,并为新的抗骨质疏松靶标奠定基础。
{"title":"Transcriptional reprogramming during human osteoclast differentiation identifies regulators of osteoclast activity.","authors":"Morten S Hansen, Kaja Madsen, Maria Price, Kent Søe, Yasunori Omata, Mario M Zaiss, Caroline M Gorvin, Morten Frost, Alexander Rauch","doi":"10.1038/s41413-023-00312-6","DOIUrl":"10.1038/s41413-023-00312-6","url":null,"abstract":"<p><p>Enhanced osteoclastogenesis and osteoclast activity contribute to the development of osteoporosis, which is characterized by increased bone resorption and inadequate bone formation. As novel antiosteoporotic therapeutics are needed, understanding the genetic regulation of human osteoclastogenesis could help identify potential treatment targets. This study aimed to provide an overview of transcriptional reprogramming during human osteoclast differentiation. Osteoclasts were differentiated from CD14<sup>+</sup> monocytes from eight female donors. RNA sequencing during differentiation revealed 8 980 differentially expressed genes grouped into eight temporal patterns conserved across donors. These patterns revealed distinct molecular functions associated with postmenopausal osteoporosis susceptibility genes based on RNA from iliac crest biopsies and bone mineral density SNPs. Network analyses revealed mutual dependencies between temporal expression patterns and provided insight into subtype-specific transcriptional networks. The donor-specific expression patterns revealed genes at the monocyte stage, such as filamin B (FLNB) and oxidized low-density lipoprotein receptor 1 (OLR1, encoding LOX-1), that are predictive of the resorptive activity of mature osteoclasts. The expression of differentially expressed G-protein coupled receptors was strong during osteoclast differentiation, and these receptors are associated with bone mineral density SNPs, suggesting that they play a pivotal role in osteoclast differentiation and activity. The regulatory effects of three differentially expressed G-protein coupled receptors were exemplified by in vitro pharmacological modulation of complement 5 A receptor 1 (C5AR1), somatostatin receptor 2 (SSTR2), and free fatty acid receptor 4 (FFAR4/GPR120). Activating C5AR1 enhanced osteoclast formation, while activating SSTR2 decreased the resorptive activity of mature osteoclasts, and activating FFAR4 decreased both the number and resorptive activity of mature osteoclasts. In conclusion, we report the occurrence of transcriptional reprogramming during human osteoclast differentiation and identified SSTR2 and FFAR4 as antiresorptive G-protein coupled receptors and FLNB and LOX-1 as potential molecular markers of osteoclast activity. These data can help future investigations identify molecular regulators of osteoclast differentiation and activity and provide the basis for novel antiosteoporotic targets.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10806178/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139540980","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}
引用次数: 0
The role and applications of extracellular vesicles in osteoporosis 细胞外囊泡在骨质疏松症中的作用和应用
IF 12.7 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-23 DOI: 10.1038/s41413-023-00313-5
Fei Fang, Jie Yang, Jiahe Wang, Tiantian Li, Erxiang Wang, Demao Zhang, Xiaoheng Liu, Chenchen Zhou

Osteoporosis is a widely observed condition characterized by the systemic deterioration of bone mass and microarchitecture, which increases patient susceptibility to fragile fractures. The intricate mechanisms governing bone homeostasis are substantially impacted by extracellular vesicles (EVs), which play crucial roles in both pathological and physiological contexts. EVs derived from various sources exert distinct effects on osteoporosis. Specifically, EVs released by osteoblasts, endothelial cells, myocytes, and mesenchymal stem cells contribute to bone formation due to their unique cargo of proteins, miRNAs, and cytokines. Conversely, EVs secreted by osteoclasts and immune cells promote bone resorption and inhibit bone formation. Furthermore, the use of EVs as therapeutic modalities or biomaterials for diagnosing and managing osteoporosis is promising. Here, we review the current understanding of the impact of EVs on bone homeostasis, including the classification and biogenesis of EVs and the intricate regulatory mechanisms of EVs in osteoporosis. Furthermore, we present an overview of the latest research progress on diagnosing and treating osteoporosis by using EVs. Finally, we discuss the challenges and prospects of translational research on the use of EVs in osteoporosis.

骨质疏松症是一种被广泛观察到的疾病,其特点是骨量和微结构的系统性退化,这增加了患者对脆弱骨折的易感性。细胞外囊泡 (EVs) 对骨质平衡的复杂机制产生了重大影响,在病理和生理环境中都发挥着至关重要的作用。不同来源的细胞外囊泡对骨质疏松症有不同的影响。具体来说,成骨细胞、内皮细胞、肌细胞和间充质干细胞释放的EVs因其独特的蛋白质、miRNA和细胞因子载体而有助于骨形成。相反,破骨细胞和免疫细胞分泌的 EVs 会促进骨吸收,抑制骨形成。此外,将 EVs 用作诊断和管理骨质疏松症的治疗方法或生物材料也很有前景。在此,我们回顾了目前对 EVs 对骨稳态影响的理解,包括 EVs 的分类和生物生成以及 EVs 在骨质疏松症中错综复杂的调控机制。此外,我们还概述了利用 EVs 诊断和治疗骨质疏松症的最新研究进展。最后,我们讨论了利用 EVs 治疗骨质疏松症的转化研究面临的挑战和前景。
{"title":"The role and applications of extracellular vesicles in osteoporosis","authors":"Fei Fang, Jie Yang, Jiahe Wang, Tiantian Li, Erxiang Wang, Demao Zhang, Xiaoheng Liu, Chenchen Zhou","doi":"10.1038/s41413-023-00313-5","DOIUrl":"https://doi.org/10.1038/s41413-023-00313-5","url":null,"abstract":"<p>Osteoporosis is a widely observed condition characterized by the systemic deterioration of bone mass and microarchitecture, which increases patient susceptibility to fragile fractures. The intricate mechanisms governing bone homeostasis are substantially impacted by extracellular vesicles (EVs), which play crucial roles in both pathological and physiological contexts. EVs derived from various sources exert distinct effects on osteoporosis. Specifically, EVs released by osteoblasts, endothelial cells, myocytes, and mesenchymal stem cells contribute to bone formation due to their unique cargo of proteins, miRNAs, and cytokines. Conversely, EVs secreted by osteoclasts and immune cells promote bone resorption and inhibit bone formation. Furthermore, the use of EVs as therapeutic modalities or biomaterials for diagnosing and managing osteoporosis is promising. Here, we review the current understanding of the impact of EVs on bone homeostasis, including the classification and biogenesis of EVs and the intricate regulatory mechanisms of EVs in osteoporosis. Furthermore, we present an overview of the latest research progress on diagnosing and treating osteoporosis by using EVs. Finally, we discuss the challenges and prospects of translational research on the use of EVs in osteoporosis.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":12.7,"publicationDate":"2024-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139522581","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}
引用次数: 0
期刊
Bone Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1