Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell Mol Life Sci ; 80(6): 171, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37261512

RESUMEN

Imbalance of bone homeostasis induces bone degenerative diseases such as osteoporosis. Hedgehog (Hh) signaling plays critical roles in regulating the development of limb and joint. However, its unique role in bone homeostasis remained largely unknown. Here, we found that canonical Hh signaling pathway was gradually augmented during osteoclast differentiation. Genetic inactivation of Hh signaling in osteoclasts, using Ctsk-Cre;Smof/f conditional knockout mice, disrupted both osteoclast formation and subsequent osteoclast-osteoblast coupling. Concordantly, either Hh signaling inhibitors or Smo/Gli2 knockdown stunted in vitro osteoclast formation. Mechanistically, Hh signaling positively regulated osteoclast differentiation via transactivation of Traf6 and stabilization of TRAF6 protein. Then, we identified connective tissue growth factor (CTGF) as an Hh-regulatory bone formation-stimulating factor derived from osteoclasts, whose loss played a causative role in osteopenia seen in CKO mice. In line with this, recombinant CTGF exerted mitigating effects against ovariectomy induced bone loss, supporting a potential extension of local rCTGF treatment to osteoporotic diseases. Collectively, our findings firstly demonstrate that Hh signaling, which dictates osteoclast differentiation and osteoclast-osteoblast coupling by regulating TRAF6 and CTGF, is crucial for maintaining bone homeostasis, shedding mechanistic and therapeutic insights into the realm of osteoporosis.


Asunto(s)
Enfermedades Óseas Metabólicas , Resorción Ósea , Osteoporosis , Femenino , Ratones , Animales , Osteoclastos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Transducción de Señal , Osteoporosis/genética , Osteoporosis/metabolismo , Homeostasis , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Diferenciación Celular , Resorción Ósea/metabolismo
2.
Bioinformatics ; 37(17): 2699-2705, 2021 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-33705524

RESUMEN

MOTIVATION: Bio-entity Coreference Resolution focuses on identifying the coreferential links in biomedical texts, which is crucial to complete bio-events' attributes and interconnect events into bio-networks. Previously, as one of the most powerful tools, deep neural network-based general domain systems are applied to the biomedical domain with domain-specific information integration. However, such methods may raise much noise due to its insufficiency of combining context and complex domain-specific information. RESULTS: In this article, we explore how to leverage the external knowledge base in a fine-grained way to better resolve coreference by introducing a knowledge-enhanced Long Short Term Memory network (LSTM), which is more flexible to encode the knowledge information inside the LSTM. Moreover, we further propose a knowledge attention module to extract informative knowledge effectively based on contexts. The experimental results on the BioNLP and CRAFT datasets achieve state-of-the-art performance, with a gain of 7.5 F1 on BioNLP and 10.6 F1 on CRAFT. Additional experiments also demonstrate superior performance on the cross-sentence coreferences. AVAILABILITY AND IMPLEMENTATION: The source code will be made available at https://github.com/zxy951005/KB-CR upon publication. Data is avaliable at http://2011.bionlp-st.org/ and https://github.com/UCDenver-ccp/CRAFT/releases/tag/v3.1.3. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

3.
BMC Med Inform Decis Mak ; 22(1): 116, 2022 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-35501781

RESUMEN

BACKGROUND: Bio-entity Coreference Resolution (CR) is a vital task in biomedical text mining. An important issue in CR is the differential representation of identical mentions as their similar representations may make the coreference more puzzling. However, when extracting features, existing neural network-based models may bring additional noise to the distinction of identical mentions since they tend to get similar or even identical feature representations. METHODS: We propose a context-aware feature attention model to distinguish similar or identical text units effectively for better resolving coreference. The new model can represent the identical mentions based on different contexts by adaptively exploiting features, which enables the model reduce the text noise and capture the semantic information effectively. RESULTS: The experimental results show that the proposed model brings significant improvements on most of the baseline for coreference resolution and mention detection on the BioNLP dataset and CRAFT-CR dataset. The empirical studies further demonstrate its superior performance on the differential representation and coreferential link of identical mentions. CONCLUSIONS: Identical mentions impose difficulties on the current methods of Bio-entity coreference resolution. Thus, we propose the context-aware feature attention model to better distinguish identical mentions and achieve superior performance on both coreference resolution and mention detection, which will further improve the performance of the downstream tasks.


Asunto(s)
Minería de Datos , Semántica , Minería de Datos/métodos , Humanos , Redes Neurales de la Computación
4.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 33(6): 1133-8, 2016 Dec.
Artículo en Zh | MEDLINE | ID: mdl-29714978

RESUMEN

In order to establish the best procedure to store the femur samples from the biomechanical viewpoint,we compared the effects of different storage methods on the mechanical properties of mouse femurs.We obtained femurs surgically from twenty C57BL/6Jfemale mice,12 weeks old,and randomly divided them into 5groups,i.e.fresh control group,4% paraformaldehyde fixation group,4℃storage group,-20℃storage group and-80℃storage group,respectively,with five mice in each group.For the three low-temperature storage groups,each group was stored for 1week,2months,6months at their respective temperatures.After rewarming,three-point bending test was performed to test the load and deflection changes.The results showed that both the elastic modulus and deflection decreased significantly in the 4% paraformaldehyde group.The maximum load and elastic modulus of the samples in the 4 ℃ group after one week storage was significantly reduced;The mechanical properties were close to the fresh control group in the-20℃ group stored for 2months but the maximum load was also reduced after 6months.However,mechanical properties,such as elastic load,maximum load and elastic modulus,were not changed obviously in the-80 ℃ storage group.Accordingly,-80 ℃ cryopreservation had little influence on the mechanical properties of bone tissues,which proved that the temperature-80 ℃is a suitable one for long-term preservation.


Asunto(s)
Criopreservación , Módulo de Elasticidad , Fémur/fisiología , Estrés Mecánico , Animales , Fenómenos Biomecánicos , Densidad Ósea , Femenino , Ratones , Ratones Endogámicos C57BL
5.
Front Bioeng Biotechnol ; 12: 1356135, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38600948

RESUMEN

Introduction: Developmental engineering based on endochondral ossification has been proposed as a potential strategy for repairing of critical bone defects. Bone development is driven by growth plate-mediated endochondral ossification. Under physiological conditions, growth plate chondrocytes undergo compressive forces characterized by micro-mechanics, but the regulatory effect of micro-mechanical loading on endochondral bone formation has not been investigated. Methods: In this study, a periodic static compression (PSC) model characterized by micro-strain (with 0.5% strain) was designed to clarify the effects of biochemical/mechanical cues on endochondral bone formation. Hydrogel scaffolds loaded with bone marrow mesenchymal stem cells (BMSCs) were incubated in proliferation medium or chondrogenic medium, and PSC was performed continuously for 14 or 28 days. Subsequently, the scaffold pretreated for 28 days was implanted into rat femoral muscle pouches and femoral condylar defect sites. The chondrogenesis and bone defect repair were evaluated 4 or 10 weeks post-operation. Results: The results showed that PSC stimulation for 14 days significantly increased the number of COL II positive cells in proliferation medium. However, the chondrogenic efficiency of BMSCs was significantly improved in chondrogenic medium, with or without PSC application. The induced chondrocytes (ichondrocytes) spontaneously underwent hypertrophy and maturation, but long-term mechanical stimulation (loading for 28 days) significantly inhibited hypertrophy and mineralization in ichondrocytes. In the heterotopic ossification model, no chondrocytes were found and no significant difference in terms of mineral deposition in each group; However, 4 weeks after implantation into the femoral defect site, all scaffolds that were subjected to biochemical/mechanical cues, either solely or synergistically, showed typical chondrocytes and endochondral bone formation. In addition, simultaneous biochemical induction/mechanical loading significantly accelerated the bone regeneration. Discussion: Our findings suggest that microstrain mechanics, biochemical cues, and in vivo microenvironment synergistically regulate the differentiation fate of BMSCs. Meanwhile, this study shows the potential of micro-strain mechanics in the treatment of critical bone defects.

6.
ACS Appl Mater Interfaces ; 15(13): 16380-16393, 2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-36961871

RESUMEN

Groove patterns are widely used in material surface modifications. However, the independent role of ditches/ridges in regulating fibrosis of soft tissues is not well-understood, especially the lack of linkage evidence in vitro and in vivo. Herein, two kinds of combinational microgroove chips with the gradient ditch/ridge width were fabricated by photolithography technology, termed R and G groups, respectively. In group R, the ridge width was 1, 5, 10, and 30 µm, with a ditch width of 30 µm; in group G, the groove width was 5, 10, 20, and 30 µm, and the ridge width was 5 µm. The effect of microgrooves on the morphology, proliferation, and expression of fibrous markers of stem cells was systematically investigated in vitro. Moreover, thicknesses of fibrous capsules were evaluated after chips were implanted into the muscular pouches of rats for 5 months. The results show that microgrooves have almost no effect on cell proliferation but significantly modulate the morphology of cells and focal adhesions (FAs) in vitro, as well as fibrosis differentiation. In particular, the differentiation of stem cells is attenuated after the intracellular force caused by stress fibers and FAs is interfered by drugs, such as rotenone and blebbistatin. Histological analysis shows that patterns of high intracellular force can apparently stimulate soft tissue fibrosis in vivo. This study not only reveals the specific rules and mechanisms of ditch/ridge regulating stem cell behaviors but also offers insight into tailoring implant surface patterns to induce controlled soft tissue fibrosis.


Asunto(s)
Señales (Psicología) , Adhesiones Focales , Ratas , Animales , Adhesiones Focales/fisiología , Células Madre , Propiedades de Superficie
7.
Bone Res ; 11(1): 64, 2023 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-38097598

RESUMEN

Given afferent functions, sensory nerves have recently been found to exert efferent effects and directly alter organ physiology. Additionally, several studies have highlighted the indirect but crucial role of sensory nerves in the regulation of the physiological function of osteoclasts. Nonetheless, evidence regarding the direct sensory nerve efferent influence on osteoclasts is lacking. In the current study, we found that high levels of efferent signals were transported directly from the sensory nerves into osteoclasts. Furthermore, sensory hypersensitivity significantly increased osteoclastic bone resorption, and sensory neurons (SNs) directly promoted osteoclastogenesis in an in vitro coculture system. Moreover, we screened a novel neuropeptide, Cyp40, using an isobaric tag for relative and absolute quantitation (iTRAQ). We observed that Cyp40 is the efferent signal from sensory nerves, and it plays a critical role in osteoclastogenesis via the aryl hydrocarbon receptor (AhR)-Ras/Raf-p-Erk-NFATc1 pathway. These findings revealed a novel mechanism regarding the influence of sensory nerves on bone regulation, i.e., a direct promoting effect on osteoclastogenesis by the secretion of Cyp40. Therefore, inhibiting Cyp40 could serve as a strategy to improve bone quality in osteoporosis and promote bone repair after bone injury.


Asunto(s)
Resorción Ósea , Osteogénesis , Humanos , Isomerasa de Peptidilprolil/metabolismo , Osteoclastos/metabolismo , Resorción Ósea/metabolismo
8.
Int J Surg Case Rep ; 99: 107569, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36108376

RESUMEN

INTRODUCTION AND IMPORTANCE: We used induced membrane combined with tissue-engineered bone (TEB) to repair the 14-cm juvenile ulnar defect formed after osteomyelitis debridement. The TEB was completely transformed into autologous bone after 4-year follow-up. CASE PRESENTATION: A 13-year-old male was hospitalized because of right ulna chronic osteomyelitis. After focal debridement, the total length of ular defect was 14 cm. Anti-infective bone cement was filled in the bone defect area. ß-Tricalcium phosphate (ß-TCP) was used as TEB scaffold. Autologous iliac bone marrow stromal cells (BMSCs) were cultured in vitro and were planted on ß-TCP scaffold to form TEB 3 weeks later. 47 months after implantation of TEB, the repaired ulna had continuous and smooth bone cortex, completely ossification of TEB, completely recanalization of medullary cavity. The upper limb function DASH score was 35. CLINICAL DISCUSSION: Masquelet put forward the concept of "induced membrane" and applied this technique on bone defects treatment formed after debridement of osteomyelitis. ß-Tricalcium phosphate (ß-TCP) is artificial bone materials commonly used in clinical. In this case, the seed cells used were autologous BMSCs and the culture medium was autologous serum. Cytokines promoting cell growth and differentiation were not used. CONCLUSION: The results of this case showed that TEB combined with induced membrane could repair ulna segmental bone defects as long as 14 cm in adolescents. This technique gives one alternative method to repair juvenile bone defects caused by osteomyelities of trauma. More clinical cases are needed to verify the effectiveness of this technique in the next.

9.
Mater Today Bio ; 16: 100342, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35847377

RESUMEN

Development of nano-laponite as bioinks based on cell-loaded hydrogels has recently attracted significant attention for promoting bone defect repairs and regeneration. However, the underlying mechanisms of the positive function of laponite in hydrogel was not fully explored. In this study, the effect of 3D bioprinted nano-laponite hydrogel construct on bone regeneration and the potential mechanism was explored in vitro and in vivo. In vitro analyses showed that the 3D construct protected encapsulated cells from shear stresses during bioprinting, promoted cell growth and cell spreading, and BMSCs at a density of 107/mL exhibited an optimal osteogenesis potential. Osteogenic differentiation and ectopic bone formation of BMSCs encapsulated inside the 3D construct were explored by determination of calcium deposition and x-ray, micro-CT analysis, respectively. RNA sequencing revealed that activation of PI3K/AKT signaling pathway of BMSCs inside the laponite hydrogel significantly upregulated expression of osteogenic related proteins. Expression of osteogenic proteins was significantly downregulated when the PI3K/AKT pathway was inhibited. The 3D bioprinted nano-laponite hydrogel construct exhibited a superior ability for bone regeneration in rat bones with defects compared with groups without laponite as shown by micro-CT and histological examination, while the osteogenesis activity was weakened by applications of a PI3K inhibitor. In summary, the 3D bioprinted nano-laponite hydrogel construct promoted bone osteogenesis by promoting cell proliferation, differentiation through activation of the PI3K/AKT signaling pathway.

10.
Bioact Mater ; 12: 185-197, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35310379

RESUMEN

The technique bottleneck of repairing large bone defects with tissue engineered bone is the vascularization of tissue engineered grafts. Although some studies have shown that extracellular vesicles (EVs) derived from bone marrow mesenchymal stem cells (BMSCs) promote bone healing and repair by accelerating angiogenesis, the effector molecules and the mechanism remain unclear, which fail to provide ideas for the future research and development of cell-free interventions. Here, we found that Nidogen1-enriched EV (EV-NID1) derived from BMSCs interferes with the formation and assembly of focal adhesions (FAs) by targeting myosin-10, thereby reducing the adhesion strength of rat arterial endothelial cells (RAECs) to the extracellular matrix (ECM), and enhancing the migration and angiogenesis potential of RAECs. Moreover, by delivery with composite hydrogel, EV-NID1 is demonstrated to promote angiogenesis and bone regeneration in rat femoral defects. This study identifies the intracellular binding target of EV-NID1 and further elucidates a novel approach and mechanism, thereby providing a cell-free construction strategy with precise targets for the development of vascularized tissue engineering products.

11.
Stem Cells Int ; 2022: 4363632, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35069747

RESUMEN

Schwann cells have been found to promote osteogenesis by an unclear molecular mechanism. To better understand how Schwann cells accelerate osteogenesis, RNA-Seq and LC-MS/MS were utilized to explore the transcriptomic and metabolic response of MC3T3-E1 to Schwann cells. Osteogenic differentiation was determined by ALP staining. Lentiviruses were constructed to alter the expression of Mif (macrophage migration inhibitory factor) in Schwann cells. Western blot (WB) analysis was employed to detect the protein expression. The results of this study show that Mif is essential for Schwann cells to promote osteogenesis, and its downstream CD74/FOXO1 is also involved in the promotion of Schwann cells on osteogenesis. Further, Schwann cells regulate amino acid metabolism and lipid metabolism in preosteoblasts. These findings unveil the mechanism for Schwann cells to promote osteogenesis where Mif is a key factor.

12.
Bone Res ; 10(1): 20, 2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35217644

RESUMEN

The circadian clock participates in maintaining homeostasis in peripheral tissues, including intervertebral discs (IVDs). Abnormal mechanical loading is a known risk factor for intervertebral disc degeneration (IDD). Based on the rhythmic daily loading pattern of rest and activity, we hypothesized that abnormal mechanical loading could dampen the IVD clock, contributing to IDD. Here, we investigated the effects of abnormal loading on the IVD clock and aimed to inhibit compression-induced IDD by targeting the core clock molecule brain and muscle Arnt-like protein-1 (BMAL1). In this study, we showed that BMAL1 KO mice exhibit radiographic features similar to those of human IDD and that BMAL1 expression was negatively correlated with IDD severity by systematic analysis based on 149 human IVD samples. The intrinsic circadian clock in the IVD was dampened by excessive loading, and BMAL1 overexpression by lentivirus attenuated compression-induced IDD. Inhibition of the RhoA/ROCK pathway by Y-27632 or melatonin attenuated the compression-induced decrease in BMAL1 expression. Finally, the two drugs partially restored BMAL1 expression and alleviated IDD in a diurnal compression model. Our results first show that excessive loading dampens the circadian clock of nucleus pulposus tissues via the RhoA/ROCK pathway, the inhibition of which potentially protects against compression-induced IDD by preserving BMAL1 expression. These findings underline the importance of the circadian clock for IVD homeostasis and provide a potentially effective therapeutic strategy for IDD.

13.
Stem Cell Res Ther ; 11(1): 77, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32085810

RESUMEN

BACKGROUND: Induced pluripotent stem cells (iPSCs) exhibit limitless pluripotent plasticity and proliferation capability to provide an abundant cell source for tissue regenerative medicine. Thus, inducing iPSCs toward a specific differentiation direction is an important scientific question. Traditionally, iPSCs have been induced to chondrocytes with the help of some small molecules within 21-36 days. To speed up the differentiation of iPSCs, we supposed to utilize bioactive ceramics to assist chondrogenic-induction process. METHODS: In this study, we applied ionic products (3.125~12.5 mg/mL) of the lithium-containing bioceramic (Li2Ca4Si4O13, L2C4S4) and individual Li+ (5.78~23.73 mg/L) in the direct chondrogenic differentiation of human iPSCs. RESULTS: Compared to pure chondrogenic medium and extracts of tricalcium phosphate (TCP), the extracts of L2C4S4 at a certain concentration range (3.125~12.5 mg/mL) significantly enhanced chondrogenic proteins Type II Collagen (COL II)/Aggrecan/ SRY-Box 9 (SOX9) synthesis and reduced hypertrophic protein type X collagen (COL X)/matrix metallopeptidase 13 (MMP13) production in iPSCs-derived chondrocytes within 14 days, suggesting that these newly generated chondrocytes exhibited favorable chondrocytes characteristics and maintained a low-hypertrophy state. Further studies demonstrated that the individual Li+ ions at the concentration range of 5.78~23.73 mg/L also accelerated the chondrogenic differentiation of iPSCs, indicating that Li+ ions played a pivotal role in chondrogenic differentiation process. CONCLUSIONS: These findings indicated that lithium-containing bioceramic with bioactive specific ionic components may be used for a promising platform for inducing iPSCs toward chondrogenic differentiation and cartilage regeneration.


Asunto(s)
Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Hipertrofia/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Litio/uso terapéutico , Medicina Regenerativa/métodos , Diferenciación Celular , Células Cultivadas , Humanos
14.
RSC Adv ; 10(43): 25652-25661, 2020 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-35518607

RESUMEN

Injectable hydrogels have long been gaining attention in the bone tissue engineering field owing to their ability to mix homogeneously with cells and therapeutic agents, minimally invasive administration, and seamless defect filling. Despite the advantages, the use of injectable hydrogels as cell delivery carriers is currently limited by the challenge of mimicking the natural microenvironment of the loaded cells, promoting cell proliferation, and enhancing bone regeneration. To overcome these problems, we aimed to develop an injectable and in situ-forming nanocomposite hydrogel composed of gelatin, alginate, and LAPONITE® to mimic the architecture and composition of the extracellular matrix. The encapsulated rat bone marrow mesenchymal stem cells (rBMSCs) survived in the nanocomposite hydrogel, and the gel promoted cell proliferation in vitro. Systematic in vivo research of the biomimetic hydrogel with or without cells was conducted in a critical-size (8 mm) rat bone defect model. The in vivo results proved that the hydrogel loaded with rBMSCs significantly promoted bone healing in rat calvarial defects, compared to the hydrogel without cells, and that the hydrogel did not provoked side effects on the recipients. Given these advantageous properties, the developed cell-loaded injectable nanocomposite hydrogel can greatly accelerate the bone healing in critical bone defects, thus providing a clinical potential candidate for orthopedic applications.

15.
Cell Rep ; 33(2): 108252, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33053358

RESUMEN

Osteogenic suppressors such as Sclerostin not only regulate skeletal development and regeneration but also serve as anti-osteoporosis drug targets. However, very few druggable suppressors have been identified due to limited understanding of the molecular mechanisms governing osteogenesis. Here, we show that fibroblast activation protein (Fap), a serine protease inhibited by the bone growth factor Osteolectin, is an osteogenic suppressor. Genetic deletion of Fap significantly ameliorates limb trabecular bone loss during aging. Pharmacological inhibition of Fap significantly promotes bone formation and inhibits bone resorption in wild-type mice by differentially regulating canonical Wnt and nuclear factor κB (NF-κB) pathways. Pharmacological inhibition of Fap promotes osteoblast differentiation, inhibits osteoclast differentiation, and significantly attenuates osteoporosis in ovariectomized mice. Epistasis analyses in zebrafish show that Osteolectin functions as an endogenous inhibitor of Fap to promote vertebrae mineralization. Taken together, we identify Fap as an important osteogenic suppressor and a potential drug target to treat osteoporosis.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Terapia Molecular Dirigida , Osteogénesis , Osteoporosis/tratamiento farmacológico , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Resorción Ósea/complicaciones , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/patología , Calcificación Fisiológica , Diferenciación Celular , Epistasis Genética , Eliminación de Gen , Células HEK293 , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/patología , Osteoporosis/complicaciones , Osteoporosis/diagnóstico por imagen , Osteoporosis/patología , Ovariectomía , Péptido Hidrolasas/metabolismo , Unión Proteica , Pez Cebra , Proteínas de Pez Cebra/metabolismo
16.
J Orthop Translat ; 21: 91-99, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32110508

RESUMEN

BACKGROUND/OBJECTIVE: Intervertebral disc degeneration (IDD) remains to be an intractable clinical challenge. Although IDD is characterised by loss of notochordal cells (NCs) and dysfunction of nucleus pulposus (NP) cells, little is known about the origin, heterogeneity, fate and maintenance of NCs and NP cells, which further stunts the therapeutic development. Thus, effective tools to spatially and temporally trace specific cell lineage and clarify cell functions in intervertebral disc (IVD) development and homoeostasis are urgently required. METHODS: In this study, NP specimens were obtained from 20 patients with degenerative disc disease or scoliosis. LepR-Cre mice was crossed with R26R-Tdtomato mice to generate LepR-Cre; R26R-Tdtomato mice, which enabled fate-mapping of NPs from embryo stage to late adult. LMNA G609G/G609G mice was used to determine the effect of premature-aging induced IDD on LepR NPs. X-ray imaging was used to measure lumber disc height of mice. RESULTS: Here, we provide the first evidence that the leptin receptor (LepR) is preferentially expressed in NCs at embryonic stages and notochord-derived cells in the postnatal IVD. By using R26R-Tdtomato fluorescent reporter mice, we systematically analysed the specificity of activity and targeting efficiency of leptin receptor-Cre (LepR-Cre) in IVD tissues from the embryonic stage E15.5 to 6-month-old LepR-Cre; Rosa26-Tdtomato (R26R-Tdtomato) mice. Specifically, LepR-Cre targets a distinct subpopulation of notochord-derived cells closely associated with disc homoeostasis. The percentage of LepR-expressing NP cells markedly decreases in the postnatal mouse IVD and, more importantly, in the human IVD with the progression of IDD. Moreover, both spine instability-induced and premature ageing-induced IDD mouse models display the phenotype of IDD with decreased percentage of LepR-expressing NP cells. These findings uncover a potential role of LepR-expressing notochord-derived cells in disc homoeostasis and open the gate for therapeutically targeting the NP cell subpopulation. CONCLUSION: In conclusion, our data prove LepR-Cre mice useful for mapping the fate of specific subpopulations of IVD cells and uncovering the underlying mechanisms of IDD. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: The translation potential of article is that we first identified LepR as a candidate marker of subpopulation of nucleus pulposus (NP) cells and provided LepR as a potential target for the treatment of intervertebral disc degeneration (IDD), which have certain profound significance.

17.
ACS Appl Mater Interfaces ; 12(14): 16058-16075, 2020 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-32182418

RESUMEN

Recent studies show that biomaterials are capable of regulating immune responses to induce a favorable osteogenic microenvironment and promote osteogenesis and angiogenesis. In this study, we investigated the effects of zinc silicate/nanohydroxyapatite/collagen (ZS/HA/Col) scaffolds on bone regeneration and angiogenesis and explored the related mechanism. We demonstrate that 10ZS/HA/Col scaffolds significantly enhanced bone regeneration and angiogenesis in vivo compared with HA/Col scaffolds. ZS/HA/Col scaffolds increased tartrate-resistant acid phosphatase (TRAP)-positive cells, nestin-positive bone marrow stromal cells (BMSCs) and CD31-positive neovessels, and expression of osteogenesis (Bmp-2 and Osterix) and angiogenesis-related (Vegf-α and Cd31) genes increased in nascent bone. ZS/HA/Col scaffolds with 10 wt % ZS activated the p38 signaling pathway in monocytes. The monocytes subsequently differentiated into TRAP+ cells and expressed higher levels of the cytokines SDF-1, TGF-ß1, VEGF-α, and PDGF-BB, which recruited BMSCs and endothelial cells (ECs) to the defect areas. Blocking the p38 pathway in monocytes reduced TRAP+ differentiation and cytokine secretion and resulted in a decrease in BMSC and EC homing and angiogenesis. Overall, these findings demonstrate that 10ZS/HA/Col scaffolds modulate monocytes and, thereby, create a favorable osteogenic microenvironment that promotes BMSC migration and differentiation and vessel formation by activating the p38 signaling pathway.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Colágeno/química , Durapatita/química , Nanopartículas/química , Silicatos/química , Compuestos de Zinc/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Diferenciación Celular/efectos de los fármacos , Quimiocina CXCL12/genética , Colágeno/síntesis química , Colágeno/farmacología , Durapatita/síntesis química , Durapatita/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/inmunología , Nestina/genética , Osteogénesis/efectos de los fármacos , Osteogénesis/inmunología , Impresión Tridimensional , Silicatos/síntesis química , Silicatos/farmacología , Fosfatasa Ácida Tartratorresistente/química , Andamios del Tejido/química , Compuestos de Zinc/síntesis química , Compuestos de Zinc/farmacología
18.
Mater Sci Eng C Mater Biol Appl ; 112: 110905, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32409059

RESUMEN

Three-dimensional (3D) bioprinting is an extremely convenient biofabrication technique for creating biomimetic tissue-engineered bone constructs and has promising applications in regenerative medicine. However, existing bioinks have shown low mechanical strength, poor osteoinductive ability, and lacking a suitable microenvironment for laden cells. Nanosilicate (nSi) has shown to be a promising biomaterial, due to its unique properties such as excellent biocompatibility, degrade into nontoxic products, and with osteoinductive properties, which has been used in bone bioprinting. However, the long term bone healing effects and associating risks, if any, of using nSi in tissue engineering bone scaffolds in vivo are unclear and require a more thorough assessment prior to practical use. Hence, a functional and biomimetic nanocomposite bioink composed of rat bone marrow mesenchymal stem cells (rBMSCs), nSi, gelatin and alginate for the 3D bioprinting of tissue-engineered bone constructs is firstly demonstrated, mimicking the structure of extracellular matrix, to create a conducive microenvironment for encapsulated cells. It is shown that the addition of nSi significantly increases the printability and mechanical strength of fabricated human-scale tissue or organ structures (up to 15 mm height) and induces osteogenic differentiation of the encapsulated rBMSCs in the absence of in vitro osteoinductive factors. A systematic in vivo research of the biomimetic nanocomposite bioink scaffolds is further demonstrated in a rat critical-size (8 mm) bone defect-repair model. The in vivo results demonstrate that the 3D bioprinted nanocomposite scaffolds can significantly promote the bone healing of the rat calvarial defects compared to other scaffolds without nSi or cells, and show rarely side effects on the recipients. Given the above advantageous properties, the 3D bioprinted nanocomposite scaffolds can greatly accelerate the bone healing in critical bone defects, thus providing a clinical potential candidate for orthopedic applications.


Asunto(s)
Materiales Biocompatibles/química , Bioimpresión/métodos , Hidrogeles/química , Nanocompuestos/química , Andamios del Tejido/química , Animales , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Enfermedades Óseas/patología , Enfermedades Óseas/terapia , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Impresión Tridimensional , Ratas , Ratas Sprague-Dawley , Medicina Regenerativa , Reología , Silicatos/química , Ingeniería de Tejidos
19.
Cell Prolif ; 53(1): e12725, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31746058

RESUMEN

OBJECTIVES: Activation of the sympathetic system and adrenergic ß-receptors following traumatic bone defects negatively impairs bone regeneration. Whether preventing ß-receptor activation could potentially improve bone defect repair is unknown. In this study, we investigated the effect of systematic administration and local delivery of propranolol through composite scaffolds on bone healing. MATERIALS AND METHODS: Collagen/PVA/propranolol/hydroxyapatite(CPPH)composite scaffolds were fabricated with 3D printing technique and characterized by scanning electron microscope (SEM). Micro-CT analysis and bone formation histology were performed to detect new bone formation. Osteogenic differentiation of bone marrow stromal cells (BMSCs) and osteoclastogenesis of bone marrow monocytes cultured with scaffolds extract were performed for further verification. RESULTS: Intraperitoneal injection of propranolol did not significantly improve bone repair, as indicated by micro-CT analysis and bone formation histology. However, CPPH scaffolds exhibited sustained release of propranolol in vitro and significantly enhanced bone regeneration compared with vehicle collagen/PVA/hydroxyapatite (CPH) scaffolds in vivo. Moreover, in vitro experiments indicated the scaffolds containing propranolol promoted the osteogenic differentiation and migration of rat BMSCs and inhibited osteoclastogenesis by preventing ß-receptor activation. CONCLUSIONS: This study demonstrates that local adrenergic ß-receptor blockade can effectively enhance the treatment of bone defects by stimulating osteogenic differentiation, inhibiting osteoclastogenesis and enhancing BMSCs migration.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Células de la Médula Ósea/metabolismo , Regeneración Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Propranolol/farmacología , Andamios del Tejido/química , Antagonistas Adrenérgicos beta/química , Animales , Células de la Médula Ósea/patología , Colágeno/química , Colágeno/farmacología , Implantes de Medicamentos/farmacología , Durapatita/química , Durapatita/farmacología , Masculino , Alcohol Polivinílico/química , Alcohol Polivinílico/farmacología , Propranolol/química , Ratas , Ratas Sprague-Dawley , Células del Estroma/metabolismo , Células del Estroma/patología
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 35(6): 505-511, 2019 Jun.
Artículo en Zh | MEDLINE | ID: mdl-31292054

RESUMEN

Objective To investigate the effects of fibroblast growth factor 2 (FGF-2) on the cytoskeleton and morphology of rat bone marrow mesenchymal stem cells (BMSCs). Methods Morphological and cytoskeleton changes of BMSCs were observed by scanning electron microscopy and rhodamine-phalloidin staining in TranswellTM co-culture system of rat vascular endothelial cells (RAECs) and BMSCs. The content of FGF-2 in cell supernatants were detected by ELISA, and the mRNA expression of FGF-2 in both conventional and co-cultured cells were evaluated by real-time quantitative PCR. NVP-BGJ398, an inhibitor of FGF-2 receptor was added into the co-culture system to block FGF-2 signal and its effect on BMSCs skeleton was observed. Recombinant FGF-2 was supplemented into the conventional medium of BMSCs to further verify the effect of exogenous FGF-2. Results After co-cultured with RAECs, BMSCs gradually stretched, contracted and formed a large number of filopodia. The content of FGF-2 increased in the co-culture system and was mainly secreted by RAECs. Cytoskeleton remodeling of BMSCs was significantly blocked by the inhibitor of FGF-2 receptor and the cells were mostly short spindle-shaped and arranged in a spiral pattern. Exogenous FGF-2 promoted the contraction and edge stretching of BMSCs, forming filopodia with staggered distribution. Conclusion FGF-2 secreted by RAECs induces cytoskeletal remodeling of BMSCs.


Asunto(s)
Citoesqueleto , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Madre Mesenquimatosas/citología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Células Madre Mesenquimatosas/efectos de los fármacos , Ratas , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA