Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Glia ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38899731

ABSTRACT

Spinal cord injury (SCI) can result in severe motor and sensory deficits, for which currently no effective cure exists. The pathological process underlying this injury is extremely complex and involves many cell types in the central nervous system. In this study, we have uncovered a novel function for macrophage G protein-coupled receptor kinase-interactor 1 (GIT1) in promoting remyelination and functional repair after SCI. Using GIT1flox/flox Lyz2-Cre (GIT1 CKO) mice, we identified that GIT1 deficiency in macrophages led to an increased generation of tumor necrosis factor-alpha (TNFα), reduced proportion of mature oligodendrocytes (mOLs), impaired remyelination, and compromised functional recovery in vivo. These effects in GIT1 CKO mice were reversed with the administration of soluble TNF inhibitor. Moreover, bone marrow transplantation from GIT1 CWT mice reversed adverse outcomes in GIT1 CKO mice, further indicating the role of macrophage GIT1 in modulating spinal cord injury repair. Our in vitro experiments showed that macrophage GIT1 plays a critical role in secreting TNFα and influences the differentiation of oligodendrocyte precursor cells (OPCs) after stimulation with myelin debris. Collectively, our data uncovered a new role of macrophage GIT1 in regulating the transformation of OPCs into mOLs, essential for functional remyelination after SCI, suggesting that macrophage GIT1 could be a promising treatment target of SCI.

2.
Exp Cell Res ; 411(1): 112937, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34863709

ABSTRACT

Spinal cord injury (SCI) often causes severe neurological dysfunction, and facilitating neurite elongation is particularly important in its treatment. Astrocytes (AS) play an important role in the central nervous system (CNS), and their high plasticity and versatility provide a feasible entry point for relevant research. Our purpose was to explore whether extracellular vesicles (EVs) from astrocytes (AS-EVs) and lipopolysaccharide (LPS)-preactivated astrocytes (LPAS-EVs) facilitate neurite elongation, to explore the underlying mechanism, and to verify whether these EVs promote locomotor recovery in rats. We used LPS to preactivate astrocytes and cocultured them with PC12 cells to observe neurite changes, then extracted and identified AS-EVs and LPAS-EVs and the role and mechanism of these EVs in facilitating neurite elongation was examined in vivo and vitro. We demonstrated that AS-EVs and LPAS-EVs facilitated the elongation of neurites and the recovery of rats with SCI. LPAS-EVs had a stronger effect than AS-EVs, by activating the Hippo pathway, promoting monopole spindle binding protein 1 (MOB1) expression, and reducing Yes-associated protein (YAP) levels. The data also suggest a feedback regulation between MOB1 and p-YAP/YAP. In sum, AS-EVs and LPAS-EVs can play an active role in facilitating neurite elongation by activating the Hippo pathway. These findings provide a new strategy for treating SCI and other CNS-related injuries.


Subject(s)
Astrocytes/cytology , Extracellular Vesicles/transplantation , Hippo Signaling Pathway , Neurites/physiology , Neurons/cytology , Spinal Cord Injuries/therapy , Animals , Astrocytes/metabolism , Extracellular Vesicles/metabolism , PC12 Cells , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
3.
J Nanobiotechnology ; 21(1): 452, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38012616

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) remains a significant health concern, with limited available treatment options. This condition poses significant medical, economic, and social challenges. SCI is typically categorized into primary and secondary injuries. Inflammation, oxidative stress, scar formation, and the immune microenvironment impede axon regeneration and subsequent functional restoration. Numerous studies have shown that the destruction of the blood-brain barrier (BBB) and microvessels is a crucial factor in severe secondary injury. Additionally, reactive oxygen species (ROS)-induced lipid peroxidation significantly contributes to endothelial cell death. Pericytes are essential constituents of the BBB that share the basement membrane with endothelial cells and astrocytes. They play a significant role in the establishment and maintenance of BBB. RESULTS: Immunofluorescence staining at different time points revealed a consistent correlation between pericyte coverage and angiogenesis, suggesting that pericytes promote vascular repair via paracrine signaling. Pericytes undergo alterations in cellular morphology and the transcriptome when exposed to hypoxic conditions, potentially promoting angiogenesis. We simulated an early ischemia-hypoxic environment following SCI using glucose and oxygen deprivation and BBB models. Co-culturing pericytes with endothelial cells improved barrier function compared to the control group. However, this enhancement was reduced by the exosome inhibitor, GW4869. In vivo injection of exosomes improved BBB integrity and promoted motor function recovery in mice following SCI. Subsequently, we found that pericyte-derived exosomes exhibited significant miR-210-5p expression based on sequencing analysis. Therefore, we performed a series of gain- and loss-of-function experiments in vitro. CONCLUSION: Our findings suggest that miR-210-5p regulates endothelial barrier function by inhibiting JAK1/STAT3 signaling. This process is achieved by regulating lipid peroxidation levels and improving mitochondrial function, suggesting a potential mechanism for restoration of the blood-spinal cord barrier (BSCB) after SCI.


Subject(s)
MicroRNAs , Spinal Cord Injuries , Mice , Animals , Pericytes/metabolism , Endothelial Cells/metabolism , Lipid Peroxidation , Axons , Nerve Regeneration , Spinal Cord Injuries/metabolism , Signal Transduction , MicroRNAs/genetics , MicroRNAs/metabolism
4.
J Nanobiotechnology ; 20(1): 529, 2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36514078

ABSTRACT

Neuroinflammation is an important cause of poor prognosis in patients with spinal cord injury. pyroptosis is a new type of inflammatory cell death. Treg cells has been shown to play an anti-inflammatory role in a variety of inflammatory diseases, including inflammatory bowel disease, amyotrophic lateral sclerosis, and arthritis. However, little is known about Treg cells' potential role in pyroptosis following spinal cord injury. The aim of this research was to look into the effect of Treg cells to motor function recovery, pyroptosis and the mechanism behind it after SCI. Here, we found that pyroptosis mainly occurred in microglia on the seventh day after spinal cord injury. Konckout Treg cells resulted in widely pyroptosis and poor motor recovery after SCI. In conversely, over-infiltration of Treg cell in mice by tail vein injection had beneficial effects following SCI.Treg cell-derived exosomes promote functional recovery by inhibiting microglia pyroptosis in vivo. Bioinformatic analysis revealed that miRNA-709 was significantly enriched in Treg cells and Treg cell-secreted exosomes. NKAP has been identified as a miRNA-709 target gene. Moreover, experiments confirmed that Treg cells targeted the NKAP via exosomal miR-709 to reduce microglia pyroptosis and promote motor function recovery after SCI. More importantly, The miR-709 overexpressed exosomes we constructed significantly reduced the inflammatory response and improved motor recovery after spinal cord injury. In brief, our findings indicate a possible mechanism for communication between Treg cells and microglia, which opens up a new perspective for alleviating neuroinflammation after SCI.


Subject(s)
Exosomes , MicroRNAs , Spinal Cord Injuries , Animals , Mice , Exosomes/metabolism , Microglia/metabolism , MicroRNAs/metabolism , Neuroinflammatory Diseases , Pyroptosis , Recovery of Function , Spinal Cord Injuries/metabolism , T-Lymphocytes, Regulatory/metabolism
5.
J Nanobiotechnology ; 20(1): 56, 2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35093078

ABSTRACT

Subtalar osteoarthritis (STOA) is often secondary to chronic ankle sprains, which seriously affects the quality of life of patients. Due to its etiology and pathogenesis was not studied equivocally yet, there is currently a lack of effective conservative treatments. Although they have been used for tissue repair, platelet-rich plasma-derived exosomes (PRP-Exo) have the disadvantage of low retention and short-lived therapeutic effects. This study aimed to determine whether incorporation of PRP-Exo in thermosensitive hydrogel (Gel) increased their retention in the joint and thereby playing a therapeutic role on STOA due to chronic mechanical instability established by transecting lateral ligaments (anterior talofibular ligament (ATFL)/calcaneal fibular ligament (CFL)). PRP-Exo incorporated Gel (Exo-Gel) system, composed of Poloxamer-407 and 188 mixture-based thermoresponsive hydrogel matrix in an optimal ratio, was determined by its release ability of Exo and rheology of Gel response to different temperature. The biological activity of Exo-Gel was evaluated in vitro, and the therapeutic effect of Exo-Gel on STOA was evaluated in vivo. Exo released from Exo-Gel continuously for 28 days could promote the proliferation and migration of mouse bone mesenchymal stem cells (mBMSCs) and chondrocytes, at the same time enhance the chondrogenic differentiation of mBMSCs, and inhibit inflammation-induced chondrocyte degeneration. In vivo experiments confirmed that Exo-Gel increased the local retention of Exo, inhibited the apoptosis and hypertrophy of chondrocytes, enhanced their proliferation, and potentially played the role in stem cell recruitment to delay the development of STOA. Thus, Delivery of PRP-Exo incorporated in thermosensitive Gel provides a novel approach of cell-free therapy and has therapeutic effect on STOA.


Subject(s)
Exosomes , Osteoarthritis , Platelet-Rich Plasma , Animals , Cartilage/metabolism , Exosomes/metabolism , Humans , Mice , Osteoarthritis/metabolism , Platelet-Rich Plasma/metabolism , Quality of Life
6.
Glia ; 69(7): 1782-1798, 2021 07.
Article in English | MEDLINE | ID: mdl-33694203

ABSTRACT

Reactive astrogliosis is a pathological feature of spinal cord injury (SCI). The ubiquitin-proteasome system plays a crucial role in maintaining protein homeostasis and has been widely studied in neuroscience. Little, however, is known about the underlying function of deubiquitinating enzymes in reactive astrogliosis following SCI. Here, we found that ubiquitin-specific protease 18 (USP18) was significantly upregulated in astrocytes following scratch injury, and in the injured spinal cord in mice. Knockdown of USP18 in vitro and conditional knockout of USP18 in astrocytes (USP18 CKO) in vivo significantly attenuated reactive astrogliosis. In mice, this led to widespread inflammation and poor functional recovery following SCI. In contrast, overexpression of USP18 in mice injected with adeno-associated virus (AAV)-USP18 had beneficial effects following SCI. We showed that USP18 binds, deubiquitinates, and thus, stabilizes SRY-box transcription factor 9 (SOX9), thereby regulating reactive astrogliosis. We also showed that the Hedgehog (Hh) signaling pathway induces expression of USP18 through Gli2-mediated transcriptional activation after SCI. Administration of the Hh pathway activator SAG significantly increased reactive astrogliosis, reduced lesion area and promoted functional recovery in mice following SCI. Our results demonstrate that USP18 positively regulates reactive astrogliosis by stabilizing SOX9 and identify USP18 as a promising target for the treatment of SCI.


Subject(s)
Gliosis , SOX9 Transcription Factor , Spinal Cord Injuries , Ubiquitin Thiolesterase , Animals , Astrocytes/metabolism , Deubiquitinating Enzymes/metabolism , Gliosis/pathology , Hedgehog Proteins/metabolism , Inflammation/metabolism , Mice , SOX9 Transcription Factor/metabolism , Spinal Cord Injuries/pathology
7.
J Neuroinflammation ; 18(1): 196, 2021 Sep 12.
Article in English | MEDLINE | ID: mdl-34511129

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) is a severe traumatic disease which causes high disability and mortality rates. The molecular pathological features after spinal cord injury mainly involve the inflammatory response, microglial and neuronal apoptosis, abnormal proliferation of astrocytes, and the formation of glial scars. However, the microenvironmental changes after spinal cord injury are complex, and the interactions between glial cells and nerve cells remain unclear. Small extracellular vesicles (sEVs) may play a key role in cell communication by transporting RNA, proteins, and bioactive lipids between cells. Few studies have examined the intercellular communication of astrocytes through sEVs after SCI. The inflammatory signal released from astrocytes is known to initiate microglial activation, but its effects on neurons after SCI remain to be further clarified. METHODS: Electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blotting were applied to characterize sEVs. We examined microglial activation and neuronal apoptosis mediated by astrocyte activation in an experimental model of acute spinal cord injury and in cell culture in vitro. RESULTS: Our results indicated that astrocytes activated after spinal cord injury release CCL2, act on microglia and neuronal cells through the sEV pathway, and promote neuronal apoptosis and microglial activation after binding the CCR2. Subsequently, the activated microglia release IL-1ß, which acts on neuronal cells, thereby further aggravating their apoptosis. CONCLUSION: This study elucidates that astrocytes interact with microglia and neurons through the sEV pathway after SCI, enriching the mechanism of CCL2 in neuroinflammation and spinal neurodegeneration, and providing a new theoretical basis of CCL2 as a therapeutic target for SCI.


Subject(s)
Extracellular Vesicles , Spinal Cord Injuries , Apoptosis , Astrocytes/metabolism , Chemokine CCL2/metabolism , Extracellular Vesicles/metabolism , Humans , Microglia/metabolism , Neuroinflammatory Diseases , Neurons , Spinal Cord/pathology , Spinal Cord Injuries/metabolism
8.
FASEB J ; 34(2): 2055-2074, 2020 02.
Article in English | MEDLINE | ID: mdl-31908016

ABSTRACT

In spinal cord ischemia-reperfusion (I/R) injury, large amounts of reactive oxygen species can cause mitochondrial damage. Therefore, mitophagy acts as the main mechanism for removing damaged mitochondria and protects nerve cells. This study aimed to illustrate the important role of GPCR kinase 2-interacting protein-1 (GIT1) in mitophagy in vivo and in vitro. The level of mitophagy in the neurons of Git1 knockout mice was significantly reduced after ischemia-reperfusion. However, the overexpression of adeno-associated virus with Git1 promoted mitophagy and inhibited the apoptosis of neurons. GIT1 regulated the phosphorylation of Beclin-1 in Thr119, which could promote the translocation of Parkin to the mitochondrial outer membrane. This process was independent of PTEN-induced kinase 1 (PINK1), but it could not rescue the role in the absence of PINK1. Overall, GIT1 enhanced mitophagy and protected neurons against ischemia-reperfusion injury and, hence, might serve as a new research site for the protection of ischemia-reperfusion injury.


Subject(s)
Beclin-1/metabolism , Cell Cycle Proteins/metabolism , GTPase-Activating Proteins/metabolism , Mitophagy , Reperfusion Injury , Spinal Cord Diseases , Ubiquitin-Protein Ligases/metabolism , Animals , Beclin-1/genetics , Cell Cycle Proteins/genetics , GTPase-Activating Proteins/genetics , Mice , Mice, Knockout , Protein Kinases/genetics , Protein Kinases/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Spinal Cord Diseases/genetics , Spinal Cord Diseases/metabolism , Spinal Cord Diseases/pathology , Spinal Cord Diseases/prevention & control , Ubiquitin-Protein Ligases/genetics
9.
J Pineal Res ; 71(4): e12769, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34562326

ABSTRACT

Spinal cord injury (SCI) is a devastating trauma that leads to irreversible motor and sensory dysfunction and is, so far, without effective treatment. Recently, however, nano-sized extracellular vesicles derived from preconditioned mesenchymal stem cells (MSCs) have shown great promise in treating various diseases, including SCI. In this study, we investigated whether extracellular vesicles (MEVs) derived from MSCs pretreated with melatonin (MT), which is well recognized to be useful in treating diseases, including Alzheimer's disease, non-small cell lung cancer, acute ischemia-reperfusion liver injury, chronic kidney disease, and SCI, are better able to promote functional recovery in mice after SCI than extracellular vesicles derived from MSCs without preconditioning (EVs). MEVs were found to facilitate motor behavioral recovery more than EVs and to increase microglia/macrophages polarization from M1-like to M2-like in mice. Experiments in BV2 microglia and RAW264.7 macrophages confirmed that MEVs facilitate M2-like polarization and also showed that they reduce the production of reactive oxygen species (ROS) and regulate mitochondrial function. Proteomics analysis revealed that ubiquitin-specific protease 29 (USP29) was markedly increased in MEVs, and knockdown of USP29 in MEVs (shUSP29-MEVs) abolished MEVs-mediated benefits in vitro and in vivo. We then showed that USP29 interacts with, deubiquitinates and therefore stabilizes nuclear factor-like 2 (NRF2), thereby regulating microglia/macrophages polarization. In NRF2 knockout mice, MEVs failed to promote functional recovery and M2-like microglia/macrophages polarization. We also showed that MT reduced global N6-methyladenosine (m6 A) modification and levels of the m6 A "writer" methyltransferase-like 3 (METTL3). The stability of USP29 mRNA in MSCs was enhanced by treatment with MT, but inhibited by overexpression of METTL3. This study describes a very promising extracellular vesicle-based approach for treating SCI.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Extracellular Vesicles , Lung Neoplasms , Melatonin , Mesenchymal Stem Cells , Spinal Cord Injuries , Animals , Mice , Spinal Cord Injuries/therapy , Ubiquitin-Specific Proteases
10.
BMC Musculoskelet Disord ; 22(1): 156, 2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33557804

ABSTRACT

BACKGROUND: Bilateral decompression via unilateral approach (BDUA) is an effective surgical approach for treating lumbar degenerative diseases. However, no studies of prognosis, especially the recovery of the soft tissue, have reported using BDUA in an elderly population. The aims of these research were to investigate the early efficacy of the bilateral decompression via unilateral approach versus conventional approach transforaminal lumbar interbody fusion (TLIF) for the treatment of lumbar degenerative disc disease in the patients over 65 years of age, especially in the perioperative factors and the recovery of the soft tissue. METHODS: The clinical data from 61 aging patients with lumbar degenerative disease who received surgical treatment were retrospectively analyzed. 31 cases who received the lumbar interbody fusion surgery with bilateral decompression via unilateral approach (BDUA) were compared with 30 cases who received conventional approach transforaminal lumbar interbody fusion. The radiographic parameters were measured using X-ray including lumbar lordosis angle and fusion rate. Japanese Orthopedic Association (JOA), Visual Analogue Scale (VAS) and Oswestry Disability Index (ODI) scores were used to evaluate the clinical outcomes at different time points. Fatty degeneration ratio and area of muscle/vertebral body were used to detect recovery of soft tissue. RESULTS: The BDUA approach group was found to have significantly less intraoperative blood loss(p < 0.05) and postoperative drainage(p < 0.05) compared to conventional approach transforaminal lumbar interbody fusion group. Symptoms of spinal canal stenosis and nerve compression were significantly relieved postoperatively, as compared with the preoperative state. However, the opposite side had a lower rate of fatty degeneration (9.42 ± 3.17%) comparing to decompression side (11.68 ± 3.08%) (P < 0.05) six months after surgery in the BDUA group. While there were no significant differences (P > 0.05) in two sides of conventional transforaminal lumbar interbody fusion approach group six months after surgery. CONCLUSIONS: Bilateral decompression via unilateral approach (BDUA) is able to reduce the intraoperative and postoperative body fluid loss in the elderly. The opposite side of decompression in BDUA shows less fatty degeneration in 6 months, which indicates better recovery of the soft tissue of the aging patients.


Subject(s)
Intervertebral Disc Degeneration , Spinal Fusion , Aged , Decompression , Humans , Intervertebral Disc Degeneration/diagnostic imaging , Intervertebral Disc Degeneration/surgery , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/surgery , Minimally Invasive Surgical Procedures , Retrospective Studies , Treatment Outcome
11.
Int J Neurosci ; 131(2): 170-182, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32223487

ABSTRACT

OBJECTIVE: This study aims to explore the effects of exosomes derived from G protein-coupled receptor kinase 2 interacting protein 1 (GIT1)-overexpressing bone marrow mesenchymal stem cell (GIT1-BMSC-Exos) on the treatment of traumatic spinal cord injury (SCI) in a rat model. METHODS: All the rats underwent a T10 laminectomy. A weight-drop impact was performed using a 10-g rod from a height of 12.5 mm except the sham group. Rats with SCI were distributed into three groups randomly and then treated with tail vein injection of GIT1-BMSCs-Exos, BMSCs-Exos and PBS, respectively. The effects of GIT1-Exos on glutamate (GLU)-induced apoptosis in vitro were also evaluated by TUNEL staining. RESULTS: The results showed that rats treated with GIT1-BMSCs-Exos had better functional behavioral recovery than those treated with PBS or BMSCs-Exos only. The overexpression of GIT1 in BMSCs-Exos not only restrained glial scar formation and neuroinflammation after SCI, but also attenuated apoptosis and promoted axonal regeneration in the injured lesion area. Neuronal cell death induced by GLU was controlled remarkably in vitro as well. CONCLUSION: In conclusion, our study suggested that the application of GIT1-BMSCs-Exos may provide a novel avenue for traumatic SCI treatment.


Subject(s)
Cell Cycle Proteins/metabolism , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Spinal Cord Injuries/metabolism , Animals , Cells, Cultured , Female , Membrane Glycoproteins , Rats, Sprague-Dawley , Receptors, Interleukin-1 , Recovery of Function
12.
BMC Surg ; 21(1): 170, 2021 Mar 29.
Article in English | MEDLINE | ID: mdl-33781244

ABSTRACT

BACKGROUND: For a long time, surgical difficulty is mainly evaluated based on subjective perception rather than objective indexes. Moreover, the lack of systematic research regarding the evaluation of surgical difficulty potentially has a negative effect in this field. This study was aimed to evaluate the risk factors for the surgical difficulty of anterior cervical spine surgery (ACSS). METHODS: This was a retrospective cohort study totaling 291 consecutive patients underwent ACSS from 2012.3 to 2017.8. The surgical difficulty of ACSS was defined by operation time longer than 120 min or intraoperative blood loss equal to or greater than 200 ml. Evaluation of risk factors was performed by analyzing the patient's medical records and radiological parameters such as age, sex, BMI, number of operation levels, high signal intensity of spinal cord on T2-weighted images, ossified posterior longitudinal ligament (OPLL), sagittal and coronal cervical circumference, cervical length, spinal canal occupational ratio, coagulation function index and platelet count. RESULTS: Significant differences were reported between low-difficulty and high-difficulty ACSS groups in terms of age (p = 0.017), sex (p = 0.006), number of operation levels (p < 0.001), high signal intensity (p < 0.001), OPLL (p < 0.001) and spinal canal occupational ratio (p < 0.001). Multivariate logistic regression analysis revealed that number of operation levels (OR = 5.224, 95%CI = 2.125-12.843, p < 0.001), high signal intensity of spinal cord (OR = 4.994, 95%CI = 1.636-15.245, p = 0.005), OPLL (OR = 6.358, 95%CI = 1.932-20.931, p = 0.002) and the spinal canal occupational ratio > 0.45 (OR = 3.988, 95%CI = 1.343-11.840, p = 0.013) were independently associated with surgical difficulty in ACSS. A nomogram was established and ROC curve gave a 0.906 C-index. There was a good calibration curve for difficulty estimation. CONCLUSION: This study indicated that the operational level, OPLL, high signal intensity of spinal cord, and spinal canal occupational ratio were independently associated with surgical difficulty and a predictive nomogram can be established using the identified risk factors. Optimal performance was achieved for predicting surgical difficulty of ACSS based on preoperative factors.


Subject(s)
Cervical Vertebrae/surgery , Decompression, Surgical/methods , Ossification of Posterior Longitudinal Ligament/surgery , Aged , Aged, 80 and over , Cervical Vertebrae/diagnostic imaging , Female , Humans , Incidence , Male , Middle Aged , Nomograms , Predictive Value of Tests , Retrospective Studies , Treatment Outcome
13.
J Cell Mol Med ; 24(21): 12633-12641, 2020 11.
Article in English | MEDLINE | ID: mdl-32945076

ABSTRACT

Long non-coding RNAs have been demonstrated to be important regulators of various cancers, though the precise mechanisms remain unclear. Although lincFOXF1 has been reported to act as a tumour suppressor, its function and underlying mechanisms in osteosarcoma have not yet been explored. We employed quantitative real-time polymerase chain reaction (qRT-PCR) to evaluate the expression of lincFOXF1 and GAPDH in osteosarcoma tissues and cell lines, and colony-formation, CCK8, wound-healing, and transwell assays were conducted to analyse the proliferation, migration, and invasion capacity of osteosarcoma cells. Subcellular localization analysis by fractionation and RNA immunoprecipitation assays were performed to elucidate the mechanism responsible for lincFOXF1-mediated phenotypes of osteosarcoma cells. The results revealed that lincFOXF1 expression is significantly decreased and strongly related to Enneking stage as well as metastasis in osteosarcoma patients. Further experiments showed that lincFOXF1 inhibits the migration, invasion and metastasis of cells in vitro and vivo. Mechanistic investigation demonstrated that lincFOXF1 physically binds to EZH2, a polycomb repressive complex 2 (PRC2) component, and a search for downstream targets suggested that G-protein-coupled receptor kinase-interacting protein 1 (GIT1) is involved in the lincFOXF1-mediated repression of osteosarcoma cells migration and invasion. Moreover, GIT1 expression is inversely correlated with lincFOXF1 in osteosarcoma. The present findings indicate that lincFOXF1 is involved in the progression of osteosarcoma through binding with EZH2, further regulating GIT1 expression. Our results suggest that lincFOXF1 may serve as a biomarker and therapeutic target for osteosarcoma patients.


Subject(s)
Cell Movement/genetics , Disease Progression , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA, Long Noncoding/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adult , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Down-Regulation/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Polycomb Repressive Complex 2/metabolism , Protein Binding , RNA, Long Noncoding/genetics , Young Adult
14.
Biochem Biophys Res Commun ; 525(4): 850-856, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32169280

ABSTRACT

AIM: To investigate the effect of endogenous PTH deficiency on osteoclasts during fracture healing and its mechanism. METHODS: A femoral fracture model was used to determine the role of endogenous PTH in fracture healing. Immunohistochemistry, qPCR, and Western blot were used to determine the potential functions and mechanisms of endogenous PTH. RESULT: In this study, we found that expression of RANKL and CK was lower in PTH knockout (KO) mice than in wild type (WT) mice. In vitro culture of osteoclasts showed that under the same stimulation, there was no statistical difference in the number of osteoclasts and the area of bone resorption areas in PTH WT mice and PTH KO mice. We found that a high concentration of RANKL could promote the number and activity of osteoclasts. Upon induction of osteoblasts in vitro, those from the PTH WT group expressed higher RANKL protein and mRNA than those from the PTH KO group. Lastly, we confirmed that the PI3K/AKT/STAT5 pathway promotes RANKL increase from osteoblasts. CONCLUSION: During fracture healing, endogenous PTH deficiency can affect osteoclast activity by reducing RANKL expression in osteoblasts.


Subject(s)
Fracture Healing/physiology , Osteoblasts/metabolism , Osteoclasts/metabolism , Parathyroid Hormone/metabolism , RANK Ligand/metabolism , Animals , Cell Communication , Cells, Cultured , Femoral Fractures/metabolism , Femoral Fractures/pathology , Mice, Knockout , Parathyroid Hormone/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism
15.
J Neuroinflammation ; 17(1): 62, 2020 Feb 17.
Article in English | MEDLINE | ID: mdl-32066456

ABSTRACT

BACKGROUND: A sustained inflammatory response following spinal cord injury (SCI) contributes to neuronal damage, inhibiting functional recovery. Macrophages, the major participants in the inflammatory response, transform into foamy macrophages after phagocytosing myelin debris, subsequently releasing inflammatory factors and amplifying the secondary injury. Here, we assessed the effect of macrophage scavenger receptor 1 (MSR1) in phagocytosis of myelin debris after SCI and explained its possible mechanism. METHODS: The SCI model was employed to determine the critical role of MSR1 in phagocytosis of myelin debris in vivo. The potential functions and mechanisms of MSR1 were explored using qPCR, western blotting, and immunofluorescence after treating macrophages and RAW264.7 with myelin debris in vitro. RESULTS: In this study, we found improved recovery from traumatic SCI in MSR1-knockout mice over that in MSR1 wild-type mice. Furthermore, MSR1 promoted the phagocytosis of myelin debris and the formation of foamy macrophage, leading to pro-inflammatory polarization in vitro and in vivo. Mechanistically, in the presence of myelin debris, MSR1-mediated NF-κB signaling pathway contributed to the release of inflammatory mediators and subsequently the apoptosis of neurons. CONCLUSIONS: Our study elucidates a previously unrecognized role of MSR1 in the pathophysiology of SCI and suggests that its inhibition may be a new treatment strategy for this traumatic condition.


Subject(s)
Apoptosis/physiology , Macrophages/metabolism , Neurons/metabolism , Scavenger Receptors, Class A/deficiency , Spinal Cord Injuries/metabolism , Animals , Cells, Cultured , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , RAW 264.7 Cells , Scavenger Receptors, Class A/genetics , Spinal Cord Injuries/pathology
16.
J Nanobiotechnology ; 18(1): 47, 2020 Mar 16.
Article in English | MEDLINE | ID: mdl-32178675

ABSTRACT

Transplantation of mesenchymal stem cells (MSCs) has been considered an effective therapeutic treatment for a variety of diseases including bone fracture. However, there are associated complications along with MSCs transplantation. There is evidence to show that exosomes (Exos) derived from MSCs exert a similar paracrine function. In addition, repair capabilities of MSCs decline with age. Hence, this study aims to confirm whether the Exos protective function on osteogenic differentiation and fracture healing from aged MSCs was attenuated. This information was used in order to investigate the underlying mechanism. MSCs were successfully isolated and identified from young and aged rats, and Exos were then obtained. Aged-Exos exhibited significantly attenuated effects on MSCs osteogenic differentiation in vitro and facture healing in vivo. Using miRNA array analysis, it was shown that miR-128-3p was markedly upregulated in Aged-Exos. In vitro experiments confirmed that Smad5 is a direct downstream target of miR-128-3p, and was inhibited by overexpressed miR-128-3p. A series gain- and loss- function experiment indicated that miR-128-3P serves a suppressor role in the process of fracture healing. Furthermore, effects caused by miR-128-3P mimic/inhibitor were reversed by the application of Smad5/siSmad5. Taken together, these results suggest that the therapeutic effects of MSCs-derived Exos may vary according to differential expression of miRNAs. Exosomal miR-128-3P antagomir may act as a promising therapeutic strategy for bone fracture healing, especially for the elderly.


Subject(s)
Exosomes/metabolism , Fracture Healing/physiology , Mesenchymal Stem Cell Transplantation/methods , MicroRNAs/metabolism , Osteogenesis/physiology , Smad5 Protein/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Fractures, Bone/pathology , Fractures, Bone/therapy , Male , Mesenchymal Stem Cells , Rats , Rats, Sprague-Dawley
17.
J Clin Lab Anal ; 34(7): e23273, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32157746

ABSTRACT

OBJECTIVE: Lumbar disk herniation (LDH) is a complex condition based on lumbar disk degeneration (LDD). Previous studies have shown that genetic factors are highly associated with the severity and risk for LDH. This case-control study was aimed to evaluate the association between the matrix metalloproteinase (MMP)-3 gene rs591058 C/T polymorphism and LDH risk in a southern Chinese population. METHODS: A total of 231 LDH patients and 312 healthy controls were recruited in this study. Genotyping was analyzed using a standard polymerase chain reaction and restriction fragment length polymorphism (PCR-RFLP). RESULTS: It was observed that TT genotype or T allele carriers of the MMP-3 gene rs591058 C/T polymorphism was more likely associated with an increased risk for LDH. Subgroup analyses showed the following characteristics increased the risk for LDH: female sex; cigarette smoking; and alcohol consumption. Furthermore, individuals with high whole body vibration, bending/twisting, and lifting were associated with an increased risk for LDH. CONCLUSION: Taken together, these data indicated that the MMP-3 gene rs591058 C/T polymorphism was associated with an increased risk for LDH. The MMP-3 gene rs591058 C/T polymorphism might serve as a clinical indicator and marker for LDH risk in the Chinese population.


Subject(s)
Intervertebral Disc Degeneration/genetics , Intervertebral Disc Displacement/genetics , Matrix Metalloproteinase 3/genetics , Occupational Diseases/etiology , Polymorphism, Single Nucleotide , Adult , Aged , Asian People/genetics , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Intervertebral Disc Degeneration/diagnostic imaging , Intervertebral Disc Displacement/diagnostic imaging , Male , Middle Aged , Occupational Diseases/genetics , Risk Factors
18.
J Cell Biochem ; 120(2): 1903-1915, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30367520

ABSTRACT

In this experiment, the cross-talk betweenNotch and the NF-κB signaling pathway was examined to reveal the mechanism of slowing down the type II collagen (ColII) and aggrecan degeneration affected by inflammatory cytokines. The expression levels of ColII and aggrecan in the intervertebral disc were observed through immunohistochemistry and hematoxylin-eosin staining+alcian blue staining, respectively. The expression levels of ColII, aggrecan, Runx2, and NF-κB in the nuclei of human nucleus pulposus cells (hNPCs) in each group, as well as the phosphorylation and acetylation levels of p65, were examined through Western blot analysis. The 293T cells were transfected with a plasmid containing the overexpressed relative domain of Notch1 intracellular domain (NICD1), and immunoprecipitation (IP) was performed to observe the combination of NICD1 and p65. HNPCs were transfected with a lentiviral-contained overexpression lacking the ANK region of NICD1, and IP was performed to observe the combination of NICD1 and p65. The expression of ColII and aggrecan in the intervertebral disc culture increased when γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-1-alanyl]-Sphenylglycine t-butyl ester (DAPT) was added to the disc culture medium. Western blot revealed that DAPT inhibited p65 phosphorylation and acetylation, and the p65 and p50 levels in the nucleus decreased. NICD1 was found to be combined with p65 in contrast to the reverse consequences after ANK domain deletion in hNPCs. In nucleus pulposus cells, the combination of p65 and the ANK domain of NICD1 is a critical procedure for the degeneration related to the NF-κB signaling pathway activation induced by IL-1ß and TNF-α.

19.
Biochem Biophys Res Commun ; 510(3): 479-486, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30737029

ABSTRACT

Osteosarcoma (OS) has emerged as the most common primary musculoskeletal malignant tumor which affects children and adolescents. A growing number of relevant studies have shown that many microRNAs (miRNAs) play a vital regulatory role in the etiology of various types of cancer. miR-1258 has been widely studied in various cancers, but there have been few studies of its role in OS. In this present study, miR-1258 expression was dramatically decreased in OS tissues as well as OS cell lines. In addition, decreased expression of miR-1258 was significantly associated with malignant clinical manifestations and poor clinical prognosis of patients with OS. Moreover, upregulation of miR-1258 significantly inhibited cell proliferation as well as promoting cell cycle arrest at G0/G1. AKT3 was identified as a direct target of miR-1258 by binding to its 3'-UTR, and miR-1258 was negatively correlated with AKT3 expression in clinical OS tissues. AKT3 was evidently upregulated in OS tissues and cells and upregulation of AKT3 accelerated the progression of OS. Moreover, through a series of rescue experiments, we demonstrated that AKT3 can abolish the role of miR-1258 in suppressing proliferation as well as regulating the cell cycle in OS cells. In conclusion, our results suggest that the miR-1258-AKT3 axis may be a promising prognostic marker and therapeutic target for human OS.


Subject(s)
Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Osteosarcoma/genetics , Proto-Oncogene Proteins c-akt/genetics , Adolescent , Adult , Bone Neoplasms/enzymology , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Osteosarcoma/enzymology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Proto-Oncogene Proteins c-akt/metabolism
20.
FASEB J ; : fj201800548, 2018 Jun 18.
Article in English | MEDLINE | ID: mdl-29912587

ABSTRACT

GPCR kinase 2-interacting protein-1 (GIT1) is a scaffold protein that plays an important role in cell adaptation, proliferation, migration, and differentiation; however, the role of GIT1 in the regulation of neuronal death after spinal cord injury remains obscure. Here, we demonstrate that GIT1 deficiency remarkably increased neuronal apoptosis and enhanced JNK/p38 signaling, which resulted in stronger motor deficits by ischemia-reperfusion in vivo, consistent with the finding of oxygen-glucose deprivation/reoxygenation-induced neuronal injury in vitro. After treatment with JNK and p38 inhibitors, abnormally necroptotic cell death caused by GIT1 knockdown could be partially rescued, with the recovery of neuronal viability, which was still poorer than that in control neurons. Meanwhile, overactivation of JNK/p38 after GIT1 depletion was concomitant with excessive activity of apoptosis signal-regulating kinase-1 (ASK1) that could be abolished by ASK1 silencing in HEK293T cells. Finally, GIT1 could disrupt the oligomerization of ASK1 via interaction between the synaptic localization domain that contains the coiled-coil (CC)-2 domain of GIT1 and the C-terminal CC domain of ASK1. It suppressed the autophosphorylation of ASK1 and led to decreasing activity of the ASK1/JNK/p38 pathway. These data reveal a protective role for GIT1 in neuronal damage by modulating ASK1/JNK/p38 signaling.-Chen, J., Wang, Q., Zhou, W., Zhou, Z., Tang, P.-Y., Xu, T., Liu, W., Li, L.-W., Cheng, L., Zhou, Z.-M., Fan, J., Yin, G.-Y. GPCR kinase 2-interacting protein-1 protects against ischemia-reperfusion injury of the spinal cord by modulating ASK1/JNK/p38 signaling.

SELECTION OF CITATIONS
SEARCH DETAIL