Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Nat Commun ; 15(1): 5094, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877020

ABSTRACT

Interactions between osteolineage cells and myeloid cells play important roles in maintaining skeletal homeostasis. Herein, we find that osteolineage cells transfer mitochondria to myeloid cells. Impairment of the transfer of mitochondria by deleting MIRO1 in osteolineage cells leads to increased myeloid cell commitment toward osteoclastic lineage cells and promotes bone resorption. In detail, impaired mitochondrial transfer from osteolineage cells alters glutathione metabolism and protects osteoclastic lineage cells from ferroptosis, thus promoting osteoclast activities. Furthermore, mitochondrial transfer from osteolineage cells to myeloid cells is involved in the regulation of glucocorticoid-induced osteoporosis, and glutathione depletion alleviates the progression of glucocorticoid-induced osteoporosis. These findings reveal an unappreciated mechanism underlying the interaction between osteolineage cells and myeloid cells to regulate skeletal metabolic homeostasis and provide insights into glucocorticoid-induced osteoporosis progression.


Subject(s)
Bone Resorption , Ferroptosis , Mitochondria , Myeloid Cells , Osteoclasts , Osteoporosis , Animals , Mitochondria/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Osteoclasts/metabolism , Myeloid Cells/metabolism , Osteoporosis/metabolism , Osteoporosis/pathology , Mice , Glucocorticoids/metabolism , Glutathione/metabolism , Mice, Inbred C57BL , Cell Differentiation , Mice, Knockout , Humans , Male
2.
Bioact Mater ; 39: 41-58, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38800718

ABSTRACT

Natural fracture healing is most efficient when the fine-tuned mechanical force and proper micromotion are applied. To mimick this micromotion at the fracture gap, a near-infrared-II (NIR-II)-activated hydrogel was fabricated by integrating two-dimensional (2D) monolayer Nb2C nanosheets into a thermally responsive poly(N-isopropylacrylamide) (NIPAM) hydrogel system. NIR-II-triggered deformation of the NIPAM/Nb2C hydrogel was designed to generate precise micromotion for co-culturing cells. It was validated that micromotion at 1/300 Hz, triggering a 2.37-fold change in the cell length/diameter ratio, is the most favorable condition for the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Moreover, mRNA sequencing and verification revealed that micromotion-induced augmentation was mediated by Piezo1 activation. Suppression of Piezo1 interrupts the mechano-sensitivity and abrogates osteogenic differentiation. Calvarial and femoral shaft defect models were established to explore the biocompatibility and osteoinductivity of the Micromotion Biomaterial. A series of research methods, including radiography, micro-CT scanning, and immunohistochemical staining have been performed to evaluate biosafety and osteogenic efficacy. The in vivo results revealed that tunable micromotion strengthens the natural fracture healing process through the sequential activation of endochondral ossification, promotion of neovascularization, initiation of mineral deposition, and combinatory acceleration of full-thickness osseous regeneration. This study demonstrated that Micromotion Biomaterials with controllable mechanophysical characteristics could promote the osteogenic differentiation of BMSCs and facilitate full osseous regeneration. The design of NIPAM/Nb2C hydrogel with highly efficient photothermal conversion, specific features of precisely controlled micromotion, and bionic-mimicking bone-repair capabilities could spark a new era in the field of regenerative medicine.

3.
J Adv Res ; 2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38246244

ABSTRACT

BACKGROUND: Endothelial cells (ECs) and pericytes (PCs) are crucial components of the vascular system, with ECs lining the inner layer of blood vessels and PCs surrounding capillaries to regulate blood flow and angiogenesis. Intercellular communication between ECs and PCs is vital for the formation, stability, and function of blood vessels. Various signaling pathways, such as the vascular endothelial growth factor/vascular endothelial growth factor receptor pathway and the platelet-derived growth factor-B/platelet-derived growth factor receptor-ß pathway, play roles in communication between ECs and PCs. Dysfunctional communication between these cells is associated with various diseases, including vascular diseases, central nervous system disorders, and certain types of cancers. AIM OF REVIEW: This review aimed to explore the diverse roles of ECs and PCs in the formation and reshaping of blood vessels. This review focused on the essential signaling pathways that facilitate communication between these cells and investigated how disruptions in these pathways may contribute to disease. Additionally, the review explored potential therapeutic targets, future research directions, and innovative approaches, such as investigating the impact of EC-PCs in novel systemic diseases, addressing resistance to antiangiogenic drugs, and developing novel antiangiogenic medications to enhance therapeutic efficacy. KEY SCIENTIFIC CONCEPTS OF REVIEW: Disordered EC-PC intercellular signaling plays a role in abnormal blood vessel formation, thus contributing to the progression of various diseases and the development of resistance to antiangiogenic drugs. Therefore, studies on EC-PC intercellular interactions have high clinical relevance.

4.
FEBS J ; 290(16): 4074-4091, 2023 08.
Article in English | MEDLINE | ID: mdl-37042280

ABSTRACT

Osteocytes are the terminally differentiated bone cells resulted from bone formation. Although there are two distinct processes of bone formation, intramembranous and endochondral ossifications contributing to the formation of calvarial and long bones, it is not clear whether the distinct pathways determine the differences between calvaria and femoral cortical bone derived osteocytes. In the present study, we employed confocal structured illumination microscopy and mRNA-sequencing analysis to characterize the morphologic and transcriptomic expression of osteocytes from murine calvaria and mid-shaft femoral cortical bone. Structured illumination microscopy and geometric modelling showed round shaped and irregularly scattered calvarial osteocytes compared to spindle shaped and orderly arrayed cortical osteocytes. mRNA-sequencing analysis indicated different transcriptomic profiles between calvarial and cortical osteocytes and provided evidence that mechanical response of osteocytes may contribute to geometrical differences. Furthermore, transcriptomic analysis showed that these two groups of osteocytes come from distinct pathways with 121 ossification-related genes differentially expressed. Analysis of correlation between ossification and osteocyte geometries via a Venn diagram showed that several genes related to ossification, cytoskeleton organization and dendrite development were differentially expressed between calvarial and cortical osteocytes. Finally, we demonstrated that aging disrupted the organization of dendrites and cortical osteocytes but had no significant effects on calvarial osteocytes. Together, we conclude that calvarial and cortical osteocytes are different in various aspects, which is probably the consequence of their distinct pathways of ossification.


Subject(s)
Osteocytes , Skull , Animals , Mice , Osteocytes/metabolism , Gene Expression , RNA, Messenger/metabolism , Aging/genetics
5.
Zhongguo Gu Shang ; 36(3): 279-83, 2023 Mar 25.
Article in Chinese | MEDLINE | ID: mdl-36946024

ABSTRACT

American Academy of Orthopaedic Surgeons (AAOS) just released the up-to-date , which has become the principles to care hip fractures in the elderly. In comparison to the Guideline 2014, considerable changes are made in terms of guideline composition and focused items. The interval of 7 years yielded dramatic progress in the care of geriatric hip fractures, including the recommendation of cemented femoral stems in hip arthroplasty due to displaced femoral neck fractures, cephalomedullary device for unstable intertrochanteric fractures and tranexamic acid to reduce blood loss and blood transfusion. Additionally, the individualized properties of the elderly with hip fractures should be noted to balance an early operation within 24 and 48 hours and patient safety. The interpretation of is helpful to comprehensively understand the progress of the care of geriatric hip fractures, thus to make orthopaedic surgeons master the key points of clinical practice, and to improve the quality of operations and decrease perioperative complications.


Subject(s)
Arthroplasty, Replacement, Hip , Femoral Neck Fractures , Hip Fractures , Orthopedic Surgeons , Aged , Humans , Femoral Neck Fractures/surgery , Fracture Fixation, Internal , Hip Fractures/surgery , Treatment Outcome , United States , Practice Guidelines as Topic
6.
Commun Biol ; 6(1): 197, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36804475

ABSTRACT

The brain-bone regulatory system regulates skeletal homeostasis via bioactive neuropeptides, yet the underlying mechanism remains elusive. Here, we report the role of the neuropeptide VF (NPVF, VPNLPQRF-NH2) in enhancing both angiogenesis and osteogenesis in a rat skeletal system and the potential pathways involved. An in vitro study revealed that NPVF not only promotes migration and angiogenesis of human umbilical vein endothelial cells (HUVECs) by activating NPFFR1, which leads to upregulation of miR-181c-3p and downregulation of Argonaute1 (AGO1), but also mediates osteogenic differentiation of bone mesenchymal stem cells (BMSCs) via the Wnt/ß-catenin signaling pathway. To improve the stability and bioavailability and thus efficacy of NPVF as a promoter of in vivo bone regeneration, we genetically engineered amyloid-NPVF-fusion proteins and utilized them as self-assembling nanofiber coatings to treat bone defects in a rat calvarial defect model. We found that a porous hydroxyapatite scaffold loaded with the NPVF peptide-fused amyloid coating substantially enhanced angiogenesis and site-specific fresh bone in-growth when implanted in calvarial defects. Taken together, our work uncovered a previously undefined crosstalk between the brain and bone by unveiling the role of NPVF in bone tissue and demonstrated a viable method for promoting bone tissue repairs based upon self-assembling NPVF-containing protein coatings.


Subject(s)
Neuropeptides , Osteogenesis , Rats , Humans , Animals , Bone Regeneration , Neuropeptides/genetics , Neuropeptides/metabolism , Bone and Bones/metabolism , Human Umbilical Vein Endothelial Cells/metabolism
7.
J Orthop Translat ; 38: 229-240, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36474855

ABSTRACT

Background: Glucocorticoid (GC) is one of frequently used anti-inflammatory agents, but its administration is unfortunately accompanied with bone loss. Although sporadic studies indicated that osteocytes are subject to a series of pathological changes under GC stress, including overexpression of cathepsin K, the definite role of osteocytes in GC-induced bone loss remains largely unclear. Methods: Gene expression of Ctsk and protein levels of cathepsin K were assessed in MLO-Y4 cell lines exposed to dexamethasone (Dex) of different time (0, 12, 24 hours) and dose (0, 10-8 and 10-6 M) courses by RT-qPCR and western blotting, respectively. Confocal imaging and immunostaining were then performed to evaluate the effects of osteocyte-derived cathepsin K on type I collagen in a primary osteocyte ex vivo culture system. MitoTracker Red was used to stain mitochondria for mitochondria morphology assessment and JC-1 assay was employed to evaluate the mitochondria membrane potential in MLO-Y4 cells following Dex treatment. Activation of PINK1-mediated mitophagy was evaluated by immunostaining of the PINK1 protein and CytoID assay. Mdivi-1 was used to inhibit mitophagy and siRNAs were used for the inhibition of Pink1 and Atg5. Results: GC triggered osteocytes to produce excessive cathepsin K which in turn led to the degradation of type I collagen in the extracellular matrix in a primary osteocyte ex vivo culture system. Meanwhile, GC administration increased mitochondrial fission and membrane depolarization in osteocytes. Further, the activation of PINK1-mediated mitophagy was demonstrated to be responsible for the diminishment of dysfunctional mitochondria in osteocytes. Examination of relationship between mitophagy and cathepsin K production revealed that inhibition of mitophagy via knocking down Pink1 gene abolished the GC-triggered cathepsin K production. Interestingly, GC's activation effect towards cathepsin K via mitophagy was found to be independent on the canonical autophagy as this effect was not impeded when inhibiting the canonical autophagy via Atg5 suppression. Conclusion: GC-induced PINK1-mediated mitophagy substantially modulates the production of cathepsin K in osteocytes, which could be an underlying mechanism by which osteocytes contribute to the extracellular matrix degradation during bone loss. The Translational potential of this article: Findings of the current study indicate a possible role of osteocyte mitophagy in GC-induced bone loss, which provides a potential therapeutic approach to alleviate GC-induced osteoporosis by targeting PINK1-mediated osteocytic mitophagy.

8.
J Bone Joint Surg Am ; 104(23): 2108-2116, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36325763

ABSTRACT

BACKGROUND: There is currently no ideal treatment for osteochondral lesions of the femoral head (OLFH) in young patients. METHODS: We performed a 1-year single-arm study and 2 additional years of follow-up of patients with a large (defined as >3 cm 2 ) OLFH treated with insertion of autologous costal cartilage graft (ACCG) to restore femoral head congruity after lesion debridement. Twenty patients ≤40 years old who had substantial hip pain and/or dysfunction after nonoperative treatment were enrolled at a single center. The primary outcome was the change in Harris hip score (HHS) from baseline to 12 months postoperatively. Secondary outcomes included the EuroQol visual analogue scale (EQ VAS), hip joint space width, subchondral integrity on computed tomography scanning, repair tissue status evaluated with the Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) score, and evaluation of cartilage biochemistry by delayed gadolinium-enhanced magnetic resonance imaging of cartilage (dGEMRIC) and T2 mapping. RESULTS: All 20 enrolled patients (31.02 ± 7.19 years old, 8 female and 12 male) completed the initial study and the 2 years of additional follow-up. The HHS improved from 61.89 ± 6.47 at baseline to 89.23 ± 2.62 at 12 months and 94.79 ± 2.72 at 36 months. The EQ VAS increased by 17.00 ± 8.77 at 12 months and by 21.70 ± 7.99 at 36 months (p < 0.001 for both). Complete integration of the ACCG with the bone was observed by 12 months in all 20 patients. The median MOCART score was 85 (interquartile range [IQR], 75 to 95) at 12 months and 75 (IQR, 65 to 85) at the last follow-up (range, 24 to 38 months). The ACCG demonstrated magnetic resonance properties very similar to hyaline cartilage; the median ratio between the relaxation times of the ACCG and recipient cartilage was 0.95 (IQR, 0.90 to 0.99) at 12 months and 0.97 (IQR, 0.92 to 1.00) at the last follow-up. CONCLUSIONS: ACCG is a feasible method for improving hip function and quality of life for at least 3 years in young patients who were unsatisfied with nonoperative treatment of an OLFH. Promising long-term outcomes may be possible because of the good integration between the recipient femoral head and the implanted ACCG. LEVEL OF EVIDENCE: Therapeutic Level IV . See Instructions for Authors for a complete description of levels of evidence.


Subject(s)
Costal Cartilage , Humans , Female , Male , Adult , Young Adult , Femur Head/diagnostic imaging , Femur Head/surgery , Quality of Life
9.
Elife ; 112022 10 28.
Article in English | MEDLINE | ID: mdl-36305580

ABSTRACT

The skeletal system contains a series of sophisticated cellular lineages arising from the mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) that determine the homeostasis of bone and bone marrow. Here, we reasoned that osteocyte may exert a function in regulation of these lineage cell specifications and tissue homeostasis. Using a mouse model of conditional deletion of osteocytes by the expression of diphtheria toxin subunit α in dentin matrix protein 1 (DMP1)-positive osteocytes, we demonstrated that partial ablation of DMP1-positive osteocytes caused severe sarcopenia, osteoporosis, and degenerative kyphosis, leading to shorter lifespan in these animals. Osteocytes reduction altered mesenchymal lineage commitment, resulting in impairment of osteogenesis and induction of osteoclastogensis. Single-cell RNA sequencing further revealed that hematopoietic lineage was mobilized toward myeloid lineage differentiation with expanded myeloid progenitors, neutrophils, and monocytes, while the lymphopoiesis was impaired with reduced B cells in the osteocyte ablation mice. The acquisition of a senescence-associated secretory phenotype (SASP) in both osteogenic and myeloid lineage cells was the underlying cause. Together, we showed that osteocytes play critical roles in regulation of lineage cell specifications in bone and bone marrow through mediation of senescence.


A hallmark of aging is the weakening of our muscles and bones, which become more fragile as we get older. These gradual changes can result in a humpback and muscle shrinking among other conditions. At the same time little is known about what role osteocytes ­ the most abundant type of bone cell ­ play in the process of bone and muscle aging. One way to investigate the role of osteocytes in aging is to remove them and observe what happens to nearby cells as they age. To achieve this Ding, Gao, Gao et al. genetically altered mice so that they would carry and activate a gene called DTA in their osteocytes. DTA is a gene derived from the bacterium that causes diphtheria, and when it is activated, it produces a toxin that accumulates in cells, eventually killing them. In the mice line developed by Ding, Gao, Gao et al. DTA slowly killed osteocytes, leading to adult mice lacking most of their osteocyte population that have a normal embryonic development. This is important because the fact that the mice develop normally before birth allowed the team to rule out embryonic defects when looking at their results. Ding, Gao, Gao et al. found that, without enough osteocytes, the nearby bone and bone marrow cells aged faster than expected. Indeed, the skeleton and muscles of adult mice was severely affected by the loss of osteocytes, leading to fragile bones with lower mass and muscle shrinking. These mice looked old in their young age and died earlier. At the cellular level, the removal of osteocytes impaired the formation of osteoblasts, the cells that are responsible for making bones. It also led to an increase in the numbers of osteoclasts ­ the cells that destroy bone tissue to repair it and maintain it ­ and fat tissue cells. Furthermore, cells in the bone marrow, which go on to make white blood cells, were also affected. The mechanisms through which osteocytes affect the growth of these other cells is yet to be fully understood. However, Ding, Gao, Gao et al. did observe that these cells acquired traits characteristic of aging cells, implying that osteocytes have a role in regulating cellular aging or senescence. Among these senescence traits is the increased production and secretion of molecules that interact with the immune system, a feature known as the 'senescence-associated secretory phenotype'. Overall, the results of Ding, Gao, Gao et al. suggest that reducing the number of osteocytes in mice leads to faster bone aging and affects the balance of the different cell types required for healthy bone and bone marrow growth. Future research could focus on finding drugs that allow osteocytes to keep performing their role during aging, and thus help maintain bone health. The findings of Ding, Gao, Gao et al. also suggest that osteocytes may be playing a previously underappreciated role in age-related diseases, which warrants further investigation.


Subject(s)
Osteoblasts , Osteocytes , Animals , Osteocytes/metabolism , Osteoblasts/metabolism , Bone Marrow , Bone and Bones , Osteogenesis/physiology
10.
Exp Mol Med ; 54(6): 765-776, 2022 06.
Article in English | MEDLINE | ID: mdl-35672449

ABSTRACT

Classical monocytes are commonly involved in the innate inflammatory response and are the progenitors of osteoclasts. Excess endogenous glucocorticoids (GCs) can increase the levels of classical monocytes in blood and bone marrow. The role of this cell population in high-dose exogenous GC-induced osteoporosis (GIOP) remains to be elucidated. In this study, GIOP was established in rats and mice by daily methylprednisolone injection, and monocyte subsets were analyzed by flow cytometry. We demonstrated that classical monocytes accumulate in bone marrow during GIOP. Similarly, the monocyte proportion among bone marrow nucleated cells was also increased in patients with steroid treatment history. We sorted classical monocytes and analyzed their transcriptional profile in response to GCs by RNA sequencing. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that classical monocytes isolated from GC-treated rats exhibited osteoclast differentiation potential. Deletion of classical monocytes by clodronate liposome treatment prevented GIOP via inhibition of osteoclastogenesis and restoration of CD31HiendomucinHi vessels. Regarding the molecular mechanism, classical monocytes express high levels of glucocorticoid receptors. In vitro treatment with GCs increased both the percentage and absolute number of monocytes and promoted their proliferation. In summary, classical monocytes mediated GC-induced bone loss and are a potential target for therapeutic intervention in GIOP treatment.


Subject(s)
Glucocorticoids , Osteoporosis , Animals , Glucocorticoids/adverse effects , Mice , Monocytes/metabolism , Osteoclasts/metabolism , Osteogenesis , Osteoporosis/chemically induced , Rats
11.
Carbohydr Polym ; 291: 119575, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35698339

ABSTRACT

Nacre, exhibiting excellent mechanical strengths due to its hierarchical structures, becomes a potential model to design bone implants. Herein, we firstly fabricated nacre-mimetic hydroxyapatite/chitosan/gelatin (HA/CS/Gel) layered scaffolds and incorporated substance P (SP) peptides. The CS scaffolds with a layered architecture could regulate the deposition of HA nanoplates on the interlamellar CS sheets by using CaCO3 as precursors. The HA/CS/Gel layered scaffolds exhibited a great flexure strength of 11.42 MPa due to a brick-and-mortar structure. The biocompatible components, layered macropores and SP peptides in the HA/CS/Gel therapeutic scaffolds facilitated the spreading and proliferation mesenchymal stem cells (MSCs). Notably, the incorporation of SP peptides induced MSC osteogenic differentiation and extracellular matrix mineralization. Rabbit knee subchondral defect models further proved that the HA/CS/Gel-SP layered scaffolds promoted in vivo subchondral bone regeneration. Hence, the combination of nacre-mimetic bone implants and therapeutic drugs may become an attractive strategy for subchondral bone regeneration.


Subject(s)
Chitosan , Nacre , Animals , Biocompatible Materials/chemistry , Bone Regeneration , Cells, Cultured , Chitosan/chemistry , Durapatite/chemistry , Gelatin/chemistry , Osteogenesis , Rabbits , Substance P/pharmacology , Tissue Engineering , Tissue Scaffolds/chemistry
12.
Bone Joint Res ; 11(6): 386-397, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35730670

ABSTRACT

AIMS: Alcoholism is a well-known detrimental factor in fracture healing. However, the underlying mechanism of alcohol-inhibited fracture healing remains poorly understood. METHODS: MicroRNA (miR) sequencing was performed on bone mesenchymal stem cells (BMSCs). The effects of alcohol and miR-19a-3p on vascularization and osteogenic differentiation were analyzed in vitro using BMSCs and human umbilical vein endothelial cells (HUVECs). An in vivo alcohol-fed mouse model of femur fracture healing was also established, and radiological and histomorphometric analyses were used to evaluate the role of miR-19a-3p. The binding of miR-19a-3p to forkhead box F2 (FOXF2) was analyzed using a luciferase reporter assay. RESULTS: miR-19a-3p was identified as one of the key regulators in the osteogenic differentiation of BMSCs, and was found to be downregulated in the alcohol-fed mouse model of fracture healing. In vitro, miR-19a-3p expression was downregulated after ethanol administration in both BMSCs and HUVECs. Vascularization and osteogenic differentiation were independently suppressed by ethanol and reversed by miR-19a-3p. In addition, the luciferase reporter assay showed that FOXF2 is the direct binding target of miR-19a-3p. In vivo, miR-19a-3p agomir stimulated callus transformation and improved the alcohol-impaired fracture healing. CONCLUSION: This study is the first to demonstrate that the miR-19a-3p/FOXF2 axis has a pivotal role in alcohol-impaired fracture healing, and may be a potential therapeutic target. Cite this article: Bone Joint Res 2022;11(6):386-397.

13.
Cell Prolif ; 55(6): e13252, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35567426

ABSTRACT

OBJECTIVES: Osteonecrosis of the femoral head (ONFH) is a devastating disease characterized by destructive bone structures, enlarged adipocyte accumulation and impaired vascularization. The aldehyde dehydrogenase 2 (ALDH 2) is the limiting enzyme for ethanol metabolism with many physiological functions. The aim was investigated the potential protective role of activated ALDH 2 by Alda-1 for ethanol-induced ONFH. MATERIALS AND METHODS: The ethanol-induced ONFH in rat was performed to explore the protective of Alda-1 by various experimental methods. Subsequently, the effect of Alda-1 and ethanol on the osteogenic and adipogenic differentiation was investigated via multiple cellular and molecular methods. Finally, the effect of Alda-1 and ethanol on the neo-vascularization was detected in Human umbilical vein endothelial cells (HUVECs) and ONFH model. RESULTS: Firstly, radiographical and pathological measurements indicated that alda-1 protected ethanol-induced ONFH. Moreover, ethanol significantly inhibited the proliferation and osteogenic differentiation of BMSCs, whereas Alda-1 could distinctly rescue it by PI3K/AKT signalling. Secondly, ethanol remarkably promoted the lipid vacuoles formation of BMSCs, while Alda-1 significantly retarded it on BMSCs by AMPK signalling pathway. Finally, ethanol significantly inhibited proliferation and growth factor level resulting in reduced angiogenesis, whereas Alda-1 could rescue the effect of ethanol. Additionally, Alda-1 significantly reduced the occurrence of ONFH and promoted vessel number and distribution in alcoholic ONFH. CONCLUSIONS: Alda-1 activation of ALDH 2 was highly demonstrated to protect ethanol-induced ONFH by triggering new bone formation, reducing adipogenesis and stimulating vascularization.


Subject(s)
Femur Head Necrosis , Femur Head , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase/pharmacology , Animals , Ethanol/toxicity , Femur Head/metabolism , Femur Head Necrosis/chemically induced , Femur Head Necrosis/prevention & control , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Osteogenesis , Phosphatidylinositol 3-Kinases/metabolism , Rats
14.
Geroscience ; 44(4): 2139-2155, 2022 08.
Article in English | MEDLINE | ID: mdl-35545739

ABSTRACT

Aging is a progressive loss of physiological function and increased susceptibility to major pathologies. Degenerative diseases in both brain and bone including Alzheimer disease (AD) and osteoporosis are common in aging groups. TERC is RNA component of telomerase, and its deficiency accelerates aging-related phenotypes including impaired life span, organ failure, bone loss, and brain dysfunction. In this study, we investigated the traits of bone marrow-brain cross-tissue communications in young mice, natural aging mice, and premature aging (TERC deficient, TERC-KO) mice by single-cell transcriptome sequencing. Differentially expressed gene analysis of brain as well as bone marrow between premature aging mouse and young mouse demonstrated aging-related inflammatory response and suppression of neuron development. Further analysis of senescence-associated secretory phenotype (SASP) landscape indicated that TERC-KO perturbation was enriched in oligodendrocyte progenitor cells (OPCs) and hematopoietic stem and progenitor cells (HSPC). Series of inflammatory associated myeloid cells was activated in premature aging mice brain and bone marrow. Cross-tissue comparison of TERC-KO mice brain and bone marrow illustrated obvious ligand-receptor communications between brain glia cells, macrophages, and bone marrow myeloid cells in premature aging-induced inflammation. Enrichment of co-regulation modules between brain and bone marrow identified premature aging response genes such as Dusp1 and Ifitm3. Our study provides a rich resource for understanding premature aging-associated perturbation in brain and bone marrow and supporting myeloid cells and endothelial cells as promising therapy targeting for age-related brain-bone diseases.


Subject(s)
Aging, Premature , Bone Marrow , Mice , Animals , Bone Marrow/pathology , Transcriptome , Aging, Premature/genetics , Endothelial Cells , Brain
15.
ACS Infect Dis ; 8(5): 918-927, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35410468

ABSTRACT

Osteomyelitis is a Staphylococcus aureus-caused bone infection. In this study, the effects of miR-146a on osteomyelitis were evaluated. Using the osteoblast cell model and S. aureus-induced osteomyelitis mice model, we monitored the miR-146 expression and explored the effects of miR-146a on cell proliferation of osteoblasts, bone remodeling, osteoclastogenesis, inflammatory cytokine production, and bacterial burden. Upregulated miR-146a was found in mice with S. aureus-induced osteomyelitis. miR-146a attenuated S. aureus-induced cell loss of osteoblasts, rescued the expression of osteogenic markers, altered the bone remodeling, and inhibited inflammatory cytokine production and osteoclastogenesis. miR-146a knockout mice had higher S. aureus burden. In conclusion, miR-146a protects against S. aureus-induced osteomyelitis by regulating inflammation and osteogenesis.


Subject(s)
MicroRNAs , Osteomyelitis , Staphylococcal Infections , Animals , Cytokines , Inflammation , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Osteogenesis , Osteomyelitis/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus
17.
J Orthop Surg Res ; 17(1): 108, 2022 Feb 20.
Article in English | MEDLINE | ID: mdl-35184732

ABSTRACT

OBJECTIVES: Internal fixation with multiple cannulated compression screws is an optional treatment for femoral neck fracture. Recently, fully threaded cannulated compression screws (FTCCS) have been introduced to fix fresh femoral neck fractures (FNF). The purpose of this study was to investigate the effectiveness of FTCCS. PATIENTS AND METHODS: Patients with FNF fixed by multiple FTCCS from February 1st, 2014 to August 31st, 2017 were included in this study. They were followed for at least 12 months postoperatively. Nonunion, osteonecrosis of the femoral head (ONFH), fixation failure, reoperation, and femoral neck shortening (FNS) were used to evaluate the outcomes. Risk factors including age, sex, fracture side, fracture displacement, fracture stability, fixation configuration, and screw numbers were analyzed. RESULTS: A total of 113 patients including 67 males and 46 females with an average age of 48.4 ± 13.4 years were included. The mean duration of follow-up was 27.1 months (range: 12-51 months). The incidence of nonunion, ONFH, fixation failure, and reoperation was 15.9%, 22.1%, 8.8%, and 24.8%, respectively. The rates of nonunion and reoperation were significantly higher in displaced fractures and unstable fractures. And patients with an unstable fracture had a higher risk of internal fixation failure. The median length of FNS was 2.9 mm (interquartile range: 0.9-6.5 mm, range: 0-17.5 mm). Age was a significant risk factor for FNS. CONCLUSIONS: The screw fixation method with FTCCS provided encouraging clinical results which may be a rational choice for the treatment of fresh FNF. Displaced fractures and unstable fractures were attributed to the higher incidence of complications. TRIAL REGISTRATION: ChiCTR, ChiCTR1800017200. Registered 17 July 2018-Retrospectively registered, http: www.chictr.org.cn/showprojen.aspx?proj=29182 .


Subject(s)
Bone Screws , Femoral Neck Fractures/surgery , Femur Neck/surgery , Fracture Fixation, Internal/methods , Adult , Aged , Female , Femoral Neck Fractures/diagnostic imaging , Femur Neck/diagnostic imaging , Fracture Fixation, Internal/adverse effects , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors , Treatment Outcome
18.
Adv Biol (Weinh) ; 6(5): e2200007, 2022 05.
Article in English | MEDLINE | ID: mdl-35195371

ABSTRACT

In humans, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can cause medical complications across various tissues and organs. Despite the advances to understanding the pathogenesis of SARS-CoV-2, its tissue tropism and interactions with host cells have not been fully understood. Existing clinical data have revealed disordered calcium and phosphorus metabolism in Coronavirus Disease 2019 (COVID-19) patients, suggesting possible infection or damage in the human skeleton system by SARS-CoV-2. Herein, SARS-CoV-2 infection in mouse models with wild-type and beta strain (B.1.351) viruses is investigated, and it is found that bone marrow-derived macrophages (BMMs) can be efficiently infected in vivo. Single-cell RNA sequencing (scRNA-Seq) analyses of infected BMMs identify distinct clusters of susceptible macrophages, including those related to osteoblast differentiation. Interestingly, SARS-CoV-2 entry on BMMs is dependent on the expression of neuropilin-1 (NRP1) rather than the widely recognized receptor angiotensin-converting enzyme 2 (ACE2). The loss of NRP1 expression during BMM-to-osteoclast differentiation or NRP1 neutralization and knockdown can significantly inhibit SARS-CoV-2 infection in BMMs. Importantly, it is found that authentic SARS-CoV-2 infection impedes BMM-to-osteoclast differentiation. Collectively, this study provides evidence for NRP1-mediated SARS-CoV-2 infection in BMMs and establishes a potential link between disturbed osteoclast differentiation and disordered skeleton metabolism in COVID-19 patients.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Humans , Macrophages/metabolism , Mice , Neuropilin-1/genetics , Osteoclasts/metabolism
19.
J Nanobiotechnology ; 20(1): 26, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34991600

ABSTRACT

Bone is one of the most sophisticated and dynamic tissues in the human body, and is characterized by its remarkable potential for regeneration. In most cases, bone has the capacity to be restored to its original form with homeostatic functionality after injury without any remaining scarring. Throughout the fascinating processes of bone regeneration, a plethora of cell lineages and signaling molecules, together with the extracellular matrix, are precisely regulated at multiple length and time scales. However, conditions, such as delayed unions (or nonunion) and critical-sized bone defects, represent thorny challenges for orthopedic surgeons. During recent decades, a variety of novel biomaterials have been designed to mimic the organic and inorganic structure of the bone microenvironment, which have tremendously promoted and accelerated bone healing throughout different stages of bone regeneration. Advances in tissue engineering endowed bone scaffolds with phenomenal osteoconductivity, osteoinductivity, vascularization and neurotization effects as well as alluring properties, such as antibacterial effects. According to the dimensional structure and functional mechanism, these biomaterials are categorized as zero-dimensional, one-dimensional, two-dimensional, three-dimensional, and four-dimensional biomaterials. In this review, we comprehensively summarized the astounding advances in emerging biomaterials for bone regeneration by categorizing them as zero-dimensional to four-dimensional biomaterials, which were further elucidated by typical examples. Hopefully, this review will provide some inspiration for the future design of biomaterials for bone tissue engineering.


Subject(s)
Biocompatible Materials , Bone Regeneration , Nanostructures , Tissue Engineering/methods , Animals , Humans , Mice , Tissue Scaffolds
20.
Adv Mater ; 33(31): e2100150, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34146359

ABSTRACT

Effective antitumor therapeutics with distinctive bactericidal and osteogenic properties are in high demand for comprehensive osteosarcoma treatment. Here, a "scaffold engineering" strategy that integrates highly active single-atomic iron catalysts (FeSAC) into a 3D printed bioactive glass (BG) scaffold is reported. Based on the atomically dispersed iron species within the catalysts, the engineered FeSAC displays prominent Fenton catalytic activity to generate toxic hydroxyl radicals (•OH) in response to the microenvironment specific to osteosarcoma. In addition, the constructed FeSAC-BG scaffold can serve as a sophisticated biomaterial platform for efficient osteosarcoma ablation, with concomitant bacterial sterilization via localized hyperthermia-reinforced nanocatalytic therapeutics. The destruction of the osteosarcoma, as well as the bacterial foci, can be achieved, further preventing susceptible chronic osteomyelitis during osteogenesis. In particular, the engineered FeSAC-BG scaffold is identified with advances in accelerated osteoconduction and osteoinduction, ultimately contributing to the sophisticated therapeutics and management of osteosarcoma. This work broadens the biomedical potential of single-atom catalysts and offers a comprehensive clinically feasible strategy for overall osteosarcoma therapeutics, bacterial inhibition, and tissue regeneration.


Subject(s)
Bone Regeneration , Osteosarcoma , Iron , Osteogenesis , Printing, Three-Dimensional , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...