Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.656
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Physiol Rev ; 102(1): 379-410, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34337974

RESUMO

Osteocytes, former osteoblasts encapsulated by mineralized bone matrix, are far from being passive and metabolically inactive bone cells. Instead, osteocytes are multifunctional and dynamic cells capable of integrating hormonal and mechanical signals and transmitting them to effector cells in bone and in distant tissues. Osteocytes are a major source of molecules that regulate bone homeostasis by integrating both mechanical cues and hormonal signals that coordinate the differentiation and function of osteoclasts and osteoblasts. Osteocyte function is altered in both rare and common bone diseases, suggesting that osteocyte dysfunction is directly involved in the pathophysiology of several disorders affecting the skeleton. Advances in osteocyte biology initiated the development of novel therapeutics interfering with osteocyte-secreted molecules. Moreover, osteocytes are targets and key distributors of biological signals mediating the beneficial effects of several bone therapeutics used in the clinic. Here we review the most recent discoveries in osteocyte biology demonstrating that osteocytes regulate bone homeostasis and bone marrow fat via paracrine signaling, influence body composition and energy metabolism via endocrine signaling, and contribute to the damaging effects of diabetes mellitus and hematologic and metastatic cancers in the skeleton.


Assuntos
Remodelação Óssea/fisiologia , Osteoclastos/citologia , Osteócitos/citologia , Osteogênese/fisiologia , Animais , Reabsorção Óssea/metabolismo , Diferenciação Celular/fisiologia , Humanos
2.
Development ; 151(17)2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39119717

RESUMO

Developing long bones alter their shape while maintaining uniform cortical thickness via coordinated activity of bone-forming osteoblasts and bone-resorbing osteoclasts at periosteal and endosteal surfaces, a process we designate trans-pairing. Two types of trans-pairing shift cortical bone in opposite orientations: peri-forming trans-pairing (peri-t-p) increases bone marrow space and endo-forming trans-pairing (endo-t-p) decreases it, via paired activity of bone resorption and formation across the cortex. Here, we focused on endo-t-p in growing bones. Analysis of endo-t-p activity in the cortex of mouse fibulae revealed osteoclasts under the periosteum compressed by muscles, and expression of RANKL in periosteal cells of the cambium layer. Furthermore, mature osteoblasts were localized on the endosteum, while preosteoblasts were at the periosteum and within cortical canals. X-ray tomographic microscopy revealed the presence of cortical canals more closely associated with endo- than with peri-t-p. Sciatic nerve transection followed by muscle atrophy and unloading induced circumferential endo-t-p with concomitant spread of cortical canals. Such canals likely supply the endosteum with preosteoblasts from the periosteum under endo-t-p, allowing bone shape to change in response to mechanical stress or nerve injury.


Assuntos
Osteoblastos , Osteoclastos , Periósteo , Animais , Osteoblastos/metabolismo , Osteoblastos/citologia , Periósteo/citologia , Periósteo/metabolismo , Osteoclastos/metabolismo , Osteoclastos/citologia , Camundongos , Desenvolvimento Ósseo , Osteogênese/fisiologia , Reabsorção Óssea/patologia , Osso Cortical , Ligante RANK/metabolismo , Camundongos Endogâmicos C57BL
3.
EMBO J ; 41(23): e111239, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36278281

RESUMO

Bone-derived mesenchymal stem cells (MSCs) reside in a hypoxic niche that maintains their differentiation potential. While hypoxia (low oxygen concentration) was reported to critically support stem cell function and osteogenesis, the molecular events triggering changes in stem cell fate decisions in response to normoxia (high oxygen concentration) remain elusive. Here, we study the impact of normoxia on mitochondrial-nuclear communication during stem cell differentiation. We show that normoxia-cultured murine MSCs undergo profound transcriptional alterations which cause irreversible osteogenesis defects. Mechanistically, high oxygen promotes chromatin compaction and histone hypo-acetylation, particularly on promoters and enhancers of osteogenic genes. Although normoxia induces metabolic rewiring resulting in elevated acetyl-CoA levels, histone hypo-acetylation occurs due to the trapping of acetyl-CoA inside mitochondria owing to decreased citrate carrier (CiC) activity. Restoring the cytosolic acetyl-CoA pool remodels the chromatin landscape and rescues the osteogenic defects. Collectively, our results demonstrate that the metabolism-chromatin-osteogenesis axis is perturbed upon exposure to high oxygen levels and identifies CiC as a novel, oxygen-sensitive regulator of the MSC function.


Assuntos
Histonas , Osteogênese , Camundongos , Animais , Osteogênese/fisiologia , Acetilcoenzima A/metabolismo , Histonas/metabolismo , Diferenciação Celular/fisiologia , Mitocôndrias/metabolismo , Hipóxia/metabolismo , Oxigênio/metabolismo , Cromatina/metabolismo , Células Cultivadas
4.
Proc Natl Acad Sci U S A ; 120(22): e2220159120, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37216542

RESUMO

Osteolectin is a recently identified osteogenic growth factor that binds to Integrin α11 (encoded by Itga11), promoting Wnt pathway activation and osteogenic differentiation by bone marrow stromal cells. While Osteolectin and Itga11 are not required for the formation of the skeleton during fetal development, they are required for the maintenance of adult bone mass. Genome-wide association studies in humans reported a single-nucleotide variant (rs182722517) 16 kb downstream of Osteolectin associated with reduced height and plasma Osteolectin levels. In this study, we tested whether Osteolectin promotes bone elongation and found that Osteolectin-deficient mice have shorter bones than those of sex-matched littermate controls. Integrin α11 deficiency in limb mesenchymal progenitors or chondrocytes reduced growth plate chondrocyte proliferation and bone elongation. Recombinant Osteolectin injections increased femur length in juvenile mice. Human bone marrow stromal cells edited to contain the rs182722517 variant produced less Osteolectin and underwent less osteogenic differentiation than that of control cells. These studies identify Osteolectin/Integrin α11 as a regulator of bone elongation and body length in mice and humans.


Assuntos
Condrócitos , Osteogênese , Adulto , Camundongos , Animais , Humanos , Condrócitos/metabolismo , Osteogênese/fisiologia , Lâmina de Crescimento , Estudo de Associação Genômica Ampla , Osso e Ossos , Diferenciação Celular , Integrinas/metabolismo , Proliferação de Células
5.
Physiol Rev ; 98(4): 2431-2452, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30156494

RESUMO

Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the transforming growth factor-ß family of ligands. BMPs exhibit widespread utility and pleiotropic, context-dependent effects, and the strength and duration of BMP pathway signaling is tightly regulated at numerous levels via mechanisms operating both inside and outside the cell. Defects in the BMP pathway or its regulation underlie multiple human diseases of different organ systems. Yet much remains to be discovered about the BMP pathway in its original context, i.e., the skeleton. In this review, we provide a comprehensive overview of the intricacies of the BMP pathway and its inhibitors in bone development, homeostasis, and disease. We frame the content of the review around major unanswered questions for which incomplete evidence is available. First, we consider the gene regulatory network downstream of BMP signaling in osteoblastogenesis. Next, we examine why some BMP ligands are more osteogenic than others and what factors limit BMP signaling during osteoblastogenesis. Then we consider whether specific BMP pathway components are required for normal skeletal development, and if the pathway exerts endogenous effects in the aging skeleton. Finally, we propose two major areas of need of future study by the field: greater resolution of the gene regulatory network downstream of BMP signaling in the skeleton, and an expanded repertoire of reagents to reliably and specifically inhibit individual BMP pathway components.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Transdução de Sinais/fisiologia , Esqueleto/metabolismo , Esqueleto/fisiologia , Animais , Regulação da Expressão Gênica/fisiologia , Humanos , Osteogênese/fisiologia
6.
FASEB J ; 38(18): e70056, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39282872

RESUMO

Distraction osteogenesis is widely used for bone tissue engineering. Mechanical stimulation plays a central role in the massive tissue regeneration observed during distraction osteogenesis. Although distraction osteogenesis has been a boon for patients with bone defects, we still have limited knowledge about the intrinsic mechanotransduction that converts physical forces into biochemical signals capable of inducing cell behavior changes and new tissue formation. In this review, we summarize the findings for mechanoresponsive factors, including cells, genes, and signaling pathways, during the distraction osteogenesis different phases. These elements function for coupling of osteogenesis and angiogenesis via the Integrin-FAK, TGF-ß/BMP, Wnt/ß-catenin, Hippo, MAPK, PI3K/Akt, and HIF-1α signaling pathways in a mechanoresponsive niche. The available evidence further suggests the existence of a balance between the epithelial-mesenchymal transition and mesenchymal-epithelial transition under hypoxic stress. We also briefly summarize the current in silico simulation algorithms and propose several future research directions that may advance understanding of distraction osteogenesis in the era of bioinformation, particularly the integration of artificial intelligence models with reliable single-cell RNA sequencing datasets. The objective of this review is to utilize established knowledge to further optimize existing distraction protocols and to identify potential therapeutic targets.


Assuntos
Mecanotransdução Celular , Osteogênese por Distração , Humanos , Osteogênese por Distração/métodos , Animais , Osteogênese/fisiologia , Regeneração Óssea/fisiologia , Transdução de Sinais , Engenharia Tecidual/métodos , Transição Epitelial-Mesenquimal/fisiologia
7.
FASEB J ; 38(19): e70079, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39340242

RESUMO

The jawbone periosteum, the easily accessible tissue responding to bone repair, has been overlooked in the recent development of cell therapy for jawbone defect reconstruction. Therefore, this study aimed to elucidate the in vitro and in vivo biological characteristics of jawbone periosteum-derived cells (jb-PDCs). For this purpose, we harvested the jb-PDCs from 8-week-old C57BL/6 mice. The in vitro cultured jb-PDCs (passages 1 and 3) contained skeletal stem/progenitor cells and exhibited clonogenicity and tri-lineage differentiation capacity. When implanted in vivo, the jb-PDCs (passage 3) showed evident ectopic bone formation after 4-week subcutaneous implantation, and active contribution to repair the critical-size jawbone defects in mice. Molecular profiling suggested that R-spondin 3 was strongly associated with the superior in vitro and in vivo osteogenic potentials of jb-PDCs. Overall, our study highlights the significance of comprehending the biological characteristics of the jawbone periosteum, which could pave the way for innovative cell-based therapies for the reconstruction of jawbone defects.


Assuntos
Diferenciação Celular , Arcada Osseodentária , Camundongos Endogâmicos C57BL , Osteogênese , Periósteo , Animais , Periósteo/citologia , Osteogênese/fisiologia , Camundongos , Arcada Osseodentária/citologia , Células Cultivadas , Masculino , Regeneração Óssea/fisiologia , Trombospondinas
8.
FASEB J ; 38(14): e23824, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39012304

RESUMO

The regenerative ability of limb bones after injury decreases during aging, but whether a similar phenomenon occurs in jawbones and whether autophagy plays a role in this process remain unclear. Through retrospective analysis of clinical data and studies on a mouse model of jawbone defects, we confirmed the presence of delayed or impaired bone regeneration in the jawbones of old individuals and mice. Subsequently, osteoblasts (OBs) derived from mouse jawbones were isolated, showing reduced osteogenesis in senescent osteoblasts (S-OBs). We observed a reduction in autophagy within both aged jawbones and S-OBs. Additionally, pharmacological inhibition of autophagy in normal OBs (N-OBs) led to cell aging and decreased osteogenesis, while autophagic activation reversed the aging phenotype of S-OBs. The activator rapamycin (RAPA) increased the autophagy level and bone regeneration in aged jawbones. Finally, we found that fatty acid-binding protein 3 (FABP3) was degraded by autolysosomes through its interaction with sequestosome 1 (P62/SQSTM1). Autophagy inhibition within senescent jawbones and S-OBs led to the excessive accumulation of FABP3, and FABP3 knockdown partially rescued the decreased osteogenesis in S-OBs and alleviated age-related compromised jawbone regeneration. In summary, we confirmed that autophagy inhibition plays an important role in delaying bone regeneration in aging jawbones. Autophagic activation or FABP3 knockdown can partially rescue the osteogenesis of S-OBs and the regeneration of aging jawbones, providing insight into jawbone aging.


Assuntos
Envelhecimento , Autofagia , Regeneração Óssea , Proteínas de Ligação a Ácido Graxo , Osteoblastos , Osteogênese , Animais , Feminino , Humanos , Masculino , Camundongos , Envelhecimento/fisiologia , Envelhecimento/metabolismo , Autofagia/fisiologia , Senescência Celular/fisiologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Arcada Osseodentária , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Osteogênese/fisiologia
9.
FASEB J ; 38(19): e70082, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39344592

RESUMO

Nasal obstruction leads to a hypoxia condition throughout the entire body. In this study, the unilateral nasal obstruction (UNO) mouse model was established by blocking the left nostril of mice. The aim of this study was to investigate the effects of UNO-induced hypoxia on mandibular condyle in juvenile (3-week-old), adolescent (6-week-old) and adult (12-week-old) male C57BL/6J mice from the perspective of H-type angiogenesis coupling osteogenesis. Firstly, UNO exerted a significant inhibitory effect on weight gain in mice of all ages. However, only in adolescent mice did UNO have an obvious detrimental effect on femoral bone mass accrual. Subsequently, micro-computed tomography (CT) analysis of mandibular condylar bone mass revealed that UNO significantly retarded condylar head volume gain but increased condylar head trabecular number (Tb.N) in juvenile and adolescent mice. Furthermore, UNO promoted the ratio of proliferative layer to cartilage layer in condylar cartilage and facilitated the chondrocyte-to-osteoblast transformation in juvenile and adolescent mice. Moreover, although UNO enhanced the positive expression of hypoxia-inducible factor (HIF)-1α in the condylar subchondral bone of mice in all ages, an increase in H-type vessels and Osterix+ cells was only detected in juvenile and adolescent mice. In summary, on the one hand, in terms of condylar morphology, UNO has a negative effect on condylar growth, hindering the increase in condylar head volume in juvenile and adolescent mice. However, on the other hand, in terms of condylar microstructure, UNO has a positive effect on condylar osteogenesis, promoting the increase of condylar Tb.N, chondrocyte-to-osteoblast transformation, HIF-1α expression, H-type angiogenesis and Osterix+ cells in juvenile and adolescent mice. Although the changes in condylar morphology and microstructure caused by UNO have not yet been fully elucidated, these findings improve our current understanding of the effects of UNO on condylar bone homeostasis.


Assuntos
Côndilo Mandibular , Camundongos Endogâmicos C57BL , Obstrução Nasal , Osteogênese , Animais , Côndilo Mandibular/patologia , Côndilo Mandibular/metabolismo , Camundongos , Masculino , Osteogênese/fisiologia , Obstrução Nasal/fisiopatologia , Obstrução Nasal/patologia , Obstrução Nasal/metabolismo , Neovascularização Fisiológica/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Microtomografia por Raio-X , Condrócitos/metabolismo , Condrócitos/patologia , Osteoblastos/metabolismo , Angiogênese
10.
FASEB J ; 38(9): e23642, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38690719

RESUMO

Alterations to the human organism that are brought about by aging are comprehensive and detrimental. Of these, an imbalance in bone homeostasis is a major outward manifestation of aging. In older adults, the decreased osteogenic activity of bone marrow mesenchymal stem cells and the inhibition of bone marrow mesenchymal stem cell differentiation lead to decreased bone mass, increased risk of fracture, and impaired bone injury healing. In the past decades, numerous studies have reported the epigenetic alterations that occur during aging, such as decreased core histones, altered DNA methylation patterns, and abnormalities in noncoding RNAs, which ultimately lead to genomic abnormalities and affect the expression of downstream signaling osteoporosis treatment and promoter of fracture healing in older adults. The current review summarizes the impact of epigenetic regulation mechanisms on age-related bone homeostasis imbalance.


Assuntos
Envelhecimento , Osso e Ossos , Epigênese Genética , Homeostase , Humanos , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Osso e Ossos/metabolismo , Metilação de DNA , Osteoporose/genética , Osteoporose/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese/genética , Osteogênese/fisiologia , Histonas/metabolismo
11.
FASEB J ; 38(13): e23779, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38967255

RESUMO

Epigenetic modifications affect cell differentiation via transcriptional regulation. G9a/EHMT2 is an important epigenetic modifier that catalyzes the methylation of histone 3 lysine 9 (H3K9) and interacts with various nuclear proteins. In this study, we investigated the role of G9a in osteoclast differentiation. When we deleted G9a by infection of Cre-expressing adenovirus into bone marrow macrophages (BMMs) from G9afl/fl (Ehmt2fl/fl) and induced osteoclastic differentiation by the addition of macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL), the number of TRAP-positive multinucleated osteoclasts significantly increased compared with control. Furthermore, the mRNA expression of osteoclast markers, TRAP, and cathepsin K, and to a lesser extent, NFATc1, a critical transcription factor, increased in G9a KO cells. Infection of wild-type (WT) G9a-expressing adenovirus in G9a KO cells restored the number of TRAP-positive multinucleated cells. In G9a KO cells, increased nuclear accumulation of NFATc1 protein and decreased H3K9me2 accumulation were observed. Furthermore, ChIP experiments revealed that NFATc1 binding to its target, Ctsk promoter, was enhanced by G9a deletion. For in vivo experiments, we created G9a conditional knock-out (cKO) mice by crossing G9afl/fl mice with Rank Cre/+ (Tnfrsf11aCre/+) mice, in which G9a is deleted in osteoclast lineage cells. The trabecular bone volume was significantly reduced in female G9a cKO mice. The serum concentration of the C-terminal telopeptide of type I collagen (CTX), a bone-resorbing indicator, was higher in G9a cKO mice. In addition, osteoclasts differentiated from G9a cKO BMMs exhibited greater bone-resorbing activity. Our findings suggest that G9a plays a repressive role in osteoclastogenesis by modulating NFATc1 function.


Assuntos
Reabsorção Óssea , Diferenciação Celular , Histona-Lisina N-Metiltransferase , Fatores de Transcrição NFATC , Osteoclastos , Osteogênese , Animais , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição NFATC/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histona-Lisina N-Metiltransferase/genética , Camundongos , Osteoclastos/metabolismo , Reabsorção Óssea/metabolismo , Osteogênese/fisiologia , Camundongos Knockout , Ligante RANK/metabolismo , Camundongos Endogâmicos C57BL , Células Cultivadas
12.
FASEB J ; 38(17): e70011, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39250278

RESUMO

In clinical settings, addressing large bone defects remains a significant challenge for orthopedic surgeons. The use of genetically modified bone marrow mesenchymal stem cells (BMSCs) has emerged as a highly promising approach for these treatments. Signal peptide-CUB-EGF domain-containing protein 3 (SCUBE3) is a multifunctional secreted glycoprotein, the role of which remains unclear in human hBMSCs. This study used various experimental methods to elucidate the potential mechanism by which SCUBE3 influences osteogenic differentiation of hBMSCs in vitro. Additionally, the therapeutic efficacy of SCUBE3, in conjunction with porous GeLMA microspheres, was evaluated in vivo using a mouse bone defect model. Our findings indicate that SCUBE3 levels increase significantly during early osteogenic differentiation of hBMSCs, and that reducing SCUBE3 levels can hinder this differentiation. Overexpressing SCUBE3 elevated osteogenesis gene and protein levels and enhanced calcium deposition. Furthermore, treatment with recombinant human SCUBE3 (rhSCUBE3) protein boosted BMP2 and TGF-ß expression, activated mitophagy in hBMSCs, ameliorated oxidative stress, and restored osteogenic function through SMAD phosphorylation. In vivo, GELMA/OE treatment effectively accelerated bone healing in mice. In conclusion, SCUBE3 fosters osteogenic differentiation and mitophagy in hBMSCs by activating the BMP2/TGF-ß signaling pathway. When combined with engineered hydrogel cell therapy, it could offer valuable guidance for the clinical management of extensive bone defects.


Assuntos
Proteína Morfogenética Óssea 2 , Diferenciação Celular , Células-Tronco Mesenquimais , Mitofagia , Osteogênese , Transdução de Sinais , Fator de Crescimento Transformador beta , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Animais , Mitofagia/fisiologia , Camundongos , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Masculino
13.
FASEB J ; 38(17): e23892, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39230563

RESUMO

Mesenchymal stromal stem cells (MSCs) or skeletal stem cells (SSCs) play a major role in tissue repair due to their robust ability to differentiate into osteoblasts, chondrocytes, and adipocytes. Complex cell signaling cascades tightly regulate this differentiation. In osteogenic differentiation, Runt-related transcription factor 2 (RUNX2) and ALP activity are essential. Furthermore, during the latter stages of osteogenic differentiation, mineral formation mediated by the osteoblast occurs with the secretion of a collagenous extracellular matrix and calcium deposition. Activation of nuclear factor erythroid 2-related factor 2 (NRF2), an important transcription factor against oxidative stress, inhibits osteogenic differentiation and mineralization via modulation of RUNX2 function; however, the exact role of NRF2 in osteoblastogenesis remains unclear. Here, we demonstrate that NRF2 activation in human bone marrow-derived stromal cells (HBMSCs) suppressed osteogenic differentiation. NRF2 activation increased the expression of STRO-1 and KITLG (stem cell markers), indicating NRF2 protects HBMSCs stemness against osteogenic differentiation. In contrast, NRF2 activation enhanced mineralization, which is typically linked to osteogenic differentiation. We determined that these divergent results were due in part to the modulation of cellular calcium flux genes by NRF2 activation. The current findings demonstrate a dual role for NRF2 as a HBMSC maintenance factor as well as a central factor in mineralization, with implications therein for elucidation of bone formation and cellular Ca2+ kinetics, dystrophic calcification and, potentially, application in the modulation of bone formation.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais , Fator 2 Relacionado a NF-E2 , Osteoblastos , Osteogênese , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Diferenciação Celular/fisiologia , Osteoblastos/metabolismo , Osteoblastos/citologia , Calcificação Fisiológica/fisiologia , Células Cultivadas , Células da Medula Óssea/metabolismo , Células da Medula Óssea/citologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética
14.
FASEB J ; 38(20): e70126, 2024 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-39446097

RESUMO

Obesity, recognized as a risk factor for nonunion, detrimentally impacts bone health, with significant physical and economic repercussions for affected individuals. Nevertheless, the precise pathomechanisms by which obesity impairs fracture healing remain insufficiently understood. Multiple studies have identified neutrophil granulocytes as key players in the systemic immune response, being the predominant immune cells in early fracture hematomas. This study identified a previously unreported critical period for neutrophil infiltration into the callus. In vivo experiments demonstrated that diet-induced obesity (DIO) mice showed earlier neutrophil infiltration, along with increased formation of neutrophil extracellular traps (NETs), compared to control mice during the endochondral phase of fracture repair. Furthermore, Padi4 knockout was found to reduce NET formation and mitigate the fracture healing delays caused by high-fat diets. Mechanistically, in vitro analyses revealed that NETs, by activating NLRP3 inflammasomes, inhibited the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and concurrently promoted M1-like macrophage polarization. These findings establish a connection between NET formation during the endochondral phase and delayed fracture healing, suggesting that targeting NETs could serve as a promising therapeutic approach for addressing obesity-induced delays in fracture recovery.


Assuntos
Dieta Hiperlipídica , Armadilhas Extracelulares , Consolidação da Fratura , Camundongos Endogâmicos C57BL , Obesidade , Proteína-Arginina Desiminase do Tipo 4 , Animais , Armadilhas Extracelulares/metabolismo , Camundongos , Consolidação da Fratura/fisiologia , Obesidade/metabolismo , Proteína-Arginina Desiminase do Tipo 4/metabolismo , Dieta Hiperlipídica/efeitos adversos , Neutrófilos/metabolismo , Neutrófilos/imunologia , Camundongos Knockout , Masculino , Células-Tronco Mesenquimais/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Osteogênese/fisiologia , Macrófagos/metabolismo , Macrófagos/imunologia
15.
FASEB J ; 38(9): e23657, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38713087

RESUMO

The pathogenesis of osteoporosis (OP) is closely associated with the disrupted balance between osteogenesis and adipogenesis in bone marrow-derived mesenchymal stem cells (BMSCs). We analyzed published single-cell RNA sequencing (scRNA-seq) data to dissect the transcriptomic profiles of bone marrow-derived cells in OP, reviewing 56 377 cells across eight scRNA-seq datasets from femoral heads (osteoporosis or osteopenia n = 5, osteoarthritis n = 3). Seventeen genes, including carboxypeptidase M (CPM), were identified as key osteogenesis-adipogenesis regulators through comprehensive gene set enrichment, differential expression, regulon activity, and pseudotime analyses. In vitro, CPM knockdown reduced osteogenesis and promoted adipogenesis in BMSCs, while adenovirus-mediated CPM overexpression had the reverse effects. In vivo, intraosseous injection of CPM-overexpressing BMSCs mitigated bone loss in ovariectomized mice. Integrated scRNA-seq and bulk RNA sequencing analyses provided insight into the MAPK/ERK pathway's role in the CPM-mediated regulation of BMSC osteogenesis and adipogenesis; specifically, CPM overexpression enhanced MAPK/ERK signaling and osteogenesis. In contrast, the ERK1/2 inhibitor binimetinib negated the effects of CPM overexpression. Overall, our findings identify CPM as a pivotal regulator of BMSC differentiation, which provides new clues for the mechanistic study of OP.


Assuntos
Adipogenia , Carboxipeptidases , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais , Osteogênese , Análise de Célula Única , Animais , Feminino , Humanos , Camundongos , Carboxipeptidases/metabolismo , Carboxipeptidases/genética , Diferenciação Celular , Proteínas Ligadas por GPI , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Metaloendopeptidases , Camundongos Endogâmicos C57BL , Osteogênese/fisiologia , Osteogênese/genética , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Transcriptoma
16.
FASEB J ; 38(10): e23646, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38795328

RESUMO

Multiple regulatory mechanisms are in place to ensure the normal processes of bone metabolism, encompassing both bone formation and absorption. This study has identified chaperone-mediated autophagy (CMA) as a critical regulator that safeguards bone formation from the detrimental effects of excessive inflammation. By silencing LAMP2A or HSCA8, we observed a hindrance in the osteoblast differentiation of human bone marrow mesenchymal stem cells (hBMSCs) in vitro. To further elucidate the role of LAMP2A, we generated LAMP2A gene knockdown and overexpression of mouse BMSCs (mBMSCs) using adenovirus. Our results showed that LAMP2A knockdown led to a decrease in osteogenic-specific proteins, while LAMP2A overexpression favored the osteogenesis of mBMSCs. Notably, active-ß-catenin levels were upregulated by LAMP2A overexpression. Furthermore, we found that LAMP2A overexpression effectively protected the osteogenesis of mBMSCs from TNF-α, through the PI3K/AKT/GSK3ß/ß-catenin pathway. Additionally, LAMP2A overexpression significantly inhibited osteoclast hyperactivity induced by TNF-α. Finally, in a murine bone defect model, we demonstrated that controlled release of LAMP2A overexpression adenovirus by alginate sodium capsule efficiently protected bone healing from inflammation, as confirmed by imaging and histological analyses. Collectively, our findings suggest that enhancing CMA has the potential to safeguard bone formation while mitigating hyperactivity in bone absorption.


Assuntos
Autofagia Mediada por Chaperonas , Glicogênio Sintase Quinase 3 beta , Inflamação , Proteína 2 de Membrana Associada ao Lisossomo , Células-Tronco Mesenquimais , Osteogênese , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , beta Catenina , Animais , Osteogênese/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , beta Catenina/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Inflamação/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/genética , Transdução de Sinais , Masculino , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Diferenciação Celular , Osteoclastos/metabolismo
17.
Exp Cell Res ; 442(1): 114211, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39147261

RESUMO

Blood vessel growth and osteogenesis in the skeletal system are coupled; however, fundamental aspects of vascular function in osteoblast-to-osteocyte transition remain unclear. Our study demonstrates that vascular smooth muscle cells (VSMCs), but not endothelial cells, are sufficient to drive bone marrow mesenchymal stromal cell-derived osteoblast-to-osteocyte transition via ß-catenin signaling and exosome-mediated communication. We found that VSMC-derived exosomes are loaded with transcripts encoding proteins associated with the osteocyte phenotype and members of the WNT/ß-catenin signaling pathway. In contrast, endothelial cell-derived exosomes facilitated mature osteoblast differentiation by reprogramming the TGFB1 gene family and osteogenic transcription factors osterix (SP7) and RUNX2. Notably, VSMCs express significant levels of tetraspanins (CD9, CD63, and CD81) and drive the intracellular trafficking of exosomes with a lower membrane zeta potential than those from other cells. Additionally, the high ATP content within these exosomes supports mineralization mechanisms, as ATP is a substrate for alkaline phosphatase. Osteocyte function was further validated by RNA sequencing, revealing activity in genes related to intermittent mineralization and sonic hedgehog signaling, alongside a significant increase in TNFSF11 levels. Our findings unveil a novel role of VSMCs in promoting osteoblast-to-osteocyte transition, thus offering new insights into bone biology and homeostasis, as well as in bone-related diseases. Clinically, these insights could pave the way for innovative therapeutic strategies targeting VSMC-derived exosome pathways to treat bone-related disorders such as osteoporosis. By manipulating these signaling pathways, it may be possible to enhance bone regeneration and improve skeletal health in patients with compromised bone structure and function.


Assuntos
Exossomos , Músculo Liso Vascular , Osteoblastos , Osteócitos , Osteogênese , beta Catenina , Osteoblastos/metabolismo , Osteoblastos/citologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citologia , Exossomos/metabolismo , Animais , beta Catenina/metabolismo , beta Catenina/genética , Osteócitos/metabolismo , Osteócitos/citologia , Camundongos , Osteogênese/genética , Osteogênese/fisiologia , Miócitos de Músculo Liso/metabolismo , Diferenciação Celular , Humanos , Via de Sinalização Wnt , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Células Cultivadas , Transdução de Sinais , Camundongos Endogâmicos C57BL
18.
Cell Mol Life Sci ; 81(1): 310, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39066929

RESUMO

Anatomically connected bones and muscles determine movement of the body. Forces exerted on muscles are then turned to bones to promote osteogenesis. The crosstalk between muscle and bone has been identified as mechanotransduction previously. In addition to the mechanical features, bones and muscles are also secretory organs which interact closely with one another through producing myokines and osteokines. Moreover, besides the mechanical features, other factors, such as nutrition metabolism, physiological rhythm, age, etc., also affect bone-muscle crosstalk. What's more, osteogenesis and myogenesis within motor system occur almost in parallel. Pathologically, defective muscles are always detected in bone associated diseases and induce the osteopenia, inflammation and abnormal bone metabolism, etc., through biomechanical or biochemical coupling. Hence, we summarize the study findings of bone-muscle crosstalk and propose potential strategies to improve the skeletal or muscular symptoms of certain diseases. Altogether, functional improvement of bones or muscles is beneficial to each other within motor system.


Assuntos
Osso e Ossos , Músculo Esquelético , Humanos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Músculo Esquelético/metabolismo , Animais , Osteogênese/fisiologia , Mecanotransdução Celular , Desenvolvimento Muscular
19.
Eur Heart J ; 45(37): 3871-3885, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-38976370

RESUMO

BACKGROUND AND AIMS: Valve interstitial cells (VICs) undergo a transition to intermediate state cells before ultimately transforming into the osteogenic cell population, which is a pivotal cellular process in calcific aortic valve disease (CAVD). Herein, this study successfully delineated the stages of VIC osteogenic transformation and elucidated a novel key regulatory role of lumican (LUM) in this process. METHODS: Single-cell RNA-sequencing (scRNA-seq) from nine human aortic valves was used to characterize the pathological switch process and identify key regulatory factors. The in vitro, ex vivo, in vivo, and double knockout mice were constructed to further unravel the calcification-promoting effect of LUM. Moreover, the multi-omic approaches were employed to analyse the molecular mechanism of LUM in CAVD. RESULTS: ScRNA-seq successfully delineated the process of VIC pathological transformation and highlighted the significance of LUM as a novel molecule in this process. The pro-calcification role of LUM is confirmed on the in vitro, ex vivo, in vivo level, and ApoE-/-//LUM-/- double knockout mice. The LUM induces osteogenesis in VICs via activation of inflammatory pathways and augmentation of cellular glycolysis, resulting in the accumulation of lactate. Subsequent investigation has unveiled a novel LUM driving histone modification, lactylation, which plays a role in facilitating valve calcification. More importantly, this study has identified two specific sites of histone lactylation, namely, H3K14la and H3K9la, which have been found to facilitate the process of calcification. The confirmation of these modification sites' association with the expression of calcific genes Runx2 and BMP2 has been achieved through ChIP-PCR analysis. CONCLUSIONS: The study presents novel findings, being the first to establish the involvement of lumican in mediating H3 histone lactylation, thus facilitating the development of aortic valve calcification. Consequently, lumican would be a promising therapeutic target for intervention in the treatment of CAVD.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica , Calcinose , Histonas , Lumicana , Osteogênese , Animais , Calcinose/genética , Calcinose/patologia , Calcinose/metabolismo , Valva Aórtica/patologia , Valva Aórtica/metabolismo , Lumicana/metabolismo , Lumicana/genética , Humanos , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/metabolismo , Estenose da Valva Aórtica/patologia , Camundongos , Osteogênese/genética , Osteogênese/fisiologia , Histonas/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Camundongos Knockout , Masculino , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/genética
20.
J Struct Biol ; 216(2): 108096, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38697586

RESUMO

The bone extracellular matrix consists of a highly organized collagen matrix that is mineralized with carbonated hydroxyapatite. Even though the structure and composition of bone have been studied extensively, the mechanisms underlying collagen matrix organization remain elusive. In this study, we used a 3D cell culture system in which osteogenic cells deposit and orient the collagen matrix that is subsequently mineralized. Using live fluorescence imaging combined with volume electron microscopy, we visualize the organization of the cells and collagen in the cell culture. We show that the osteogenically induced cells are organizing the collagen matrix during development. Based on the observation of tunnel-like structures surrounded by aligned collagen in the center of the culture, we propose that osteoblasts organize the deposited collagen during migration through the culture. Overall, we show that cell-matrix interactions are involved in collagen alignment during early-stage osteogenic differentiation and that the matrix is organized by the osteoblasts in the absence of osteoclast activity.


Assuntos
Diferenciação Celular , Colágeno , Matriz Extracelular , Osteoblastos , Osteogênese , Matriz Extracelular/metabolismo , Osteoblastos/metabolismo , Osteoblastos/citologia , Colágeno/metabolismo , Osteogênese/fisiologia , Animais , Técnicas de Cultura de Células em Três Dimensões/métodos , Camundongos , Osteoclastos/metabolismo , Osteoclastos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA