Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Affiliation country
Publication year range
1.
J Bone Miner Metab ; 40(6): 940-950, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36350408

ABSTRACT

INTRODUCTION: The bone lacunar-canalicular system (LCS) is an important microstructural basis for signaling and material transport in bone tissue, guaranteeing normal physiological processes in tissues. Spaceflight astronauts and elderly osteoporosis are related to its function, so it is necessary to reveal the mass transfer laws in bone microstructure under different gravity fields to provide insight for effective clinical treatment. MATERIALS AND METHODS: Using the natural LCS structure of bovine tibial cortical bone as the object, the mass transfer experiments on cortical bone were conducted by using sodium fluorescein tracer through different frequency pulsating pressure provided by dynamic perfusion loading device and different high G environments provided by high-speed centrifuge to analyze the mass transfer laws under different gravity fields and different pulsating pressures. RESULTS: The fluorescence intensity of lacunae within the osteon was lower the farther away from the Haversian canal. As the gravity field magnitude increased, the fluorescence intensity within each lacuna enhanced, and the more distant the lacunae from the Haversian canal, the greater the fluorescence intensity enhancement. High-frequency pulsating pressure simulated high-intensity exercise in humans can improve mass transfer efficiency in the LCS. CONCLUSION: High-intensity exercise may greatly increase solute molecules, nutrients, and signaling molecules in osteocytes and improve the activity of osteocytes. Hypergravity can enhance the transport of solute molecules, nutrients, and signaling molecules in the LCS, especially promoting mass transfer to deep layer lacunae. Conversely, mass transfer to deep layer lacunae may be inhibited under microgravity, causing bone loss and ultimately leading to osteoporosis.


Subject(s)
Haversian System , Osteoporosis , Humans , Animals , Cattle , Aged , Osteocytes , Tibia , Cortical Bone
2.
J Mater Sci Mater Med ; 32(4): 31, 2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33751254

ABSTRACT

No effective treatment has been established for nerve dysfunction caused by spinal cord injury (SCI). Orderly axonal growth at the site of spinal cord transection and creation of an appropriate biological microenvironment are important for functional recovery. To axially guiding axonal growth, designing a collagen/silk fibroin scaffold fabricated with 3D printing technology (3D-C/SF) emulated the corticospinal tract. The normal collagen/silk fibroin scaffold with freeze-drying technology (C/SF) or 3D-C/SF scaffold were implanted into rats with completely transected SCI to evaluate its effect on nerve repair during an 8-week observation period. Electrophysiological analysis and locomotor performance showed that the 3D-C/SF implants contributed to significant improvements in the neurogolical function of rats compared to C/SF group. By magnetic resonance imaging, 3D-C/SF implants promoted a striking degree of axonal regeneration and connection between the proximal and distal SCI sites. Compared with C/SF group, rats with 3D-C/SF scaffold exhibited fewer lesions and disordered structures in histological analysis and more GAP43-positive profiles at the lesion site. The above results indicated that the corticospinal tract structure of 3D printing collagen/silk fibroin scaffold improved axonal regeneration and promoted orderly connections within the neural network, which could provided a promising and innovative approach for tissue repair after SCI.


Subject(s)
Collagen/chemistry , Fibroins/chemistry , Printing, Three-Dimensional , Spinal Cord Injuries/therapy , Tissue Scaffolds/chemistry , Animals , Axons/pathology , Calorimetry, Differential Scanning , Compressive Strength , Electrophysiology , Female , Magnetic Resonance Imaging , Movement , Nerve Net , Nerve Regeneration , Rats , Rats, Sprague-Dawley , Recovery of Function , Spectroscopy, Fourier Transform Infrared , Stress, Mechanical , X-Ray Diffraction
3.
Mol Cancer ; 19(1): 153, 2020 Oct 30.
Article in English | MEDLINE | ID: mdl-33126881

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

4.
Med Sci Monit ; 26: e920855, 2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32150531

ABSTRACT

BACKGROUND Transplantation of exosomes derived from mesenchymal stem cells (MSCs-Exo) can improve the recovery of neurological function in rats after traumatic brain injury (TBI). We tested a new hypothesis that BDNF-mediated MSCs-Exo could effectively promote functional recovery and neurogenesis of rats after TBI. MATERIAL AND METHOD BMSCs of rats were extracted by whole bone marrow culture, BDNF was added to BMSCs for intervention, supernatant was collected, and exosomes were separated and purified by hypercentrifugation. Exosomes were identified by WB, TEM and particle size analysis and subsequently used in cell and animal experiments. We investigated the recovery of sensorimotor function and spatial learning ability, inflammation inhibition and neuron regeneration in rats after TBI. RESULTS Compared with group MSCs-Exo, group BDNF-mediated MSCs-Exo showed better effects in promoting the recovery of sensorimotor function and spatial learning ability. BDNF-mediated MSCs-Exo successfully inhibited inflammation and promoted neuronal regeneration in vivo and in vitro. We further analyzed miRNA in BDNF-mediated MSCs-Exo and MSCs-Exo, and found that the expression of miR-216a-5p in BDNF-mediated MSCs-Exo was significantly higher than that in MSCs-Exo by qRT-PCR. Rescue experiment indicated that miR-216a-5p has a similar function to BDNF-mediated MSCs-Exo. CONCLUSIONS In conclusion, we found that BDNF-mediated MSCs-Exo can better promote neurogenesis and inhibit apoptosis than MSCs-Exo in rats after TBI, and the mechanism may be related to the high expression of miR-216a-5p.


Subject(s)
Brain Injuries, Traumatic/therapy , Brain-Derived Neurotrophic Factor/metabolism , Exosomes/transplantation , Mesenchymal Stem Cells/cytology , MicroRNAs/metabolism , Animals , Apoptosis , Brain/physiopathology , Brain Injuries, Traumatic/physiopathology , Cell Line, Tumor , Cell Movement/physiology , Culture Media/metabolism , Disease Models, Animal , Exosomes/metabolism , Humans , Male , Mesenchymal Stem Cells/metabolism , Nerve Regeneration/physiology , Neurogenesis/physiology , Neurons , Primary Cell Culture/methods , Rats
5.
Mol Cancer ; 17(1): 166, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30470262

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) have been found to play critical roles in the development and progression of various cancers. However, little is known about the effects of the circular RNA network on glioblastoma multiforme (GBM). METHODS: A microarray was used to screen circRNA expression in GBM. Quantitative real-time PCR was used to detect the expression of circMMP9. GBM cells were transfected with a circMMP9 overexpression vector or siRNA, and cell proliferation, migration and invasion, as well as tumorigenesis in nude mice, were assessed to examine the effect of circMMP9 in GBM. Biotin-coupled miRNA capture, fluorescence in situ hybridization and luciferase reporter assays were conducted to confirm the relationship between circMMP9 and miR-124. RESULTS: In this study, we screened differentially expressed circRNAs and identified circMMP9 in GBM. We found that circMMP9 acted as an oncogene, was upregulated in GBM and promoted the proliferation, migration and invasion abilities of GBM cells. Next, we verified that circMMP9 served as a sponge that directly targeted miR-124; circMMP9 accelerated GBM cell proliferation, migration and invasion by targeting miR-124. Furthermore, we found that cyclin-dependent kinase 4 (CDK4) and aurora kinase A (AURKA) were involved in circMMP9/miR-124 axis-induced GBM tumorigenesis. Finally, we found that eukaryotic initiation factor 4A3 (eIF4A3), which binds to the MMP9 mRNA transcript, induced circMMP9 cyclization and increased circMMP9 expression in GBM. CONCLUSIONS: Our findings indicate that eIF4A3-induced circMMP9 is an important underlying mechanism in GBM cell proliferation, invasion and metastasis through modulation of the miR-124 signaling pathway, which could provide pivotal potential therapeutic targets for the treatment of GBM.


Subject(s)
Brain Neoplasms/genetics , Carcinogenesis/genetics , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A/genetics , Glioblastoma/genetics , Matrix Metalloproteinase 9/genetics , MicroRNAs/genetics , RNA/genetics , Animals , Aurora Kinase A/genetics , Brain Neoplasms/pathology , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Heterografts , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , MicroRNAs/metabolism , RNA/biosynthesis , RNA, Circular , Up-Regulation
6.
Cell Physiol Biochem ; 47(2): 617-629, 2018.
Article in English | MEDLINE | ID: mdl-29794460

ABSTRACT

BACKGROUND/AIMS: Current therapies for spinal cord injury (SCI) have limited efficacy, and identifying a therapeutic target is a pressing need. Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) plays an important role in regulating calcium homeostasis, which has been shown to inhibit apoptosis. Exendin-4 has been shown to inhibit the apoptosis of nerve cells in SCI, which can also improve SERCA2 expression. In this study, we sought to determine whether exendin-4 plays a protective role in a rat model of SCI via SERCA2. METHODS: To investigate the effects of exendin-4 on SCI, a rat model of SCI was induced by a modified version of Allen's method. Spinal cord tissue sections from rats and western blot analysis were used to examine SERCA2 expression after treatment with the long-acting glucagon-like peptide 1 receptor exendin-4 or the SERCA2 antagonist 5(6)-carboxyfluorescein diacetate N-succinimidyl ester (CE). Locomotor function was evaluated using the Basso Beattie Bresnahan locomotor rating scale and slanting board test. RESULTS: Cell apoptosis was increased with CE treatment and decreased with exendin-4 treatment. Upregulation of SERCA2 in female rats with SCI resulted in an improvement of motor function scores and histological changes. CONCLUSION: These findings suggest that exendin-4 plays a protective role in a rat model of SCI through SERCA2 via inhibition of apoptosis. Existing drugs targeting SERCA2 may be an effective therapeutic strategy for the treatment of SCI.


Subject(s)
Peptides/pharmacology , Protective Agents/pharmacology , Recovery of Function/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Venoms/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Exenatide , Locomotion/drug effects , Microscopy, Fluorescence , PC12 Cells , Peptides/therapeutic use , Protective Agents/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Signal Transduction/drug effects , Spinal Cord Injuries/prevention & control , Venoms/therapeutic use , bcl-2-Associated X Protein/metabolism
7.
Cell Physiol Biochem ; 41(4): 1313-1324, 2017.
Article in English | MEDLINE | ID: mdl-28278500

ABSTRACT

BACKGROUND/AIMS: Aspirin has been demonstrated to possess potent chemopreventive and anticancer effects on prostate cancer. However, the more detailed molecular mechanisms of aspirin to suppress prostate cancer cell invasion have not been clearly elucidated. METHODS: Transwell assays were performed to evaluate the effects of aspirin on cell invasion. Matrix metalloproteinases (MMPs) and serine proteinases activities in cell media were examined by gelatin zymography and ELISA. In addition, inhibitor of κB (IκB) kinase-ß (IKK-ß) phosphorylation and IKK-ß kinase activity were measured to assess the effects of aspirin on IKK-ß activation. RESULTS: We found that aspirin suppressed the invasion and attachment in human prostate cancer cells. Aspirin treatment significantly resulted in reduction of matrix metalloproteinase-9 (MMP-9) and upregulation of tissue inhibitors of metalloproteinase-1 (TIMP-1) activity, which are the proteolytic enzymes contributing to the degradation of extracellular matrix and basement membrane in cell invasion and metastasis. Our data further showed that aspirin was able to inhibit both urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor-1 (PAI-1) expression in the cells. In addition, aspirin treatment caused a strong decrease in nuclear factor-kappa B (NF-κB) activation, inhibitor of κB (IκB)-α phosphorylation together with translocation of NF-κB p65 to nucleus and IκB kinase (IKK)- ß activation. Moreover, the inhibitory effects of aspirin on cell invasion were reversed by IKK-ß overexpression, while the IKK inhibitor sensitizes the anti-invasive effect of aspirin in prostate cancer cells. CONCLUSION: The present research concluded that aspirin suppressed prostate cancer cell invasion by reducing MMP-9 activity and uPA expression through decreasing of IKK-ß-mediated NF-κB activation, indicating that the ability of aspirin to inhibit cell invasion might be useful in the chemoprevention of metastatic prostate cancer.


Subject(s)
Aspirin/pharmacology , I-kappa B Kinase/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Cell Line, Tumor , Humans , Male , Neoplasm Invasiveness , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transcription Factor RelA/metabolism
8.
Anal Chem ; 86(20): 10021-4, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-24517078

ABSTRACT

In this work, a novel enzyme-linked immunosorbent assay (ELISA) with a low limit of detection and high sensitivity was developed using atom transfer radical polymer (ATRP)-modified gold nanoparticles (AuNPs). Clear signal amplification was achieved by introducing an abundance of horseradish peroxidase (HRP) to the AuNPs, because of the ATRP modification. This result suggested that the new ELISA was able to detect antigens in complex mixtures, and the limit of detection (LOD) was lower than that of conventional ELISA by a factor of 81. The new ELISA strategy greatly decreased the LOD during analysis and exhibited excellent reproducibility, stability, and feasibility. Therefore, it is a promising technique with many potential applications in biochemistry and medical science research.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Gold/chemistry , Metal Nanoparticles/chemistry , Polymers/chemistry , Horseradish Peroxidase/chemistry , Microscopy, Electron, Transmission
9.
Neural Regen Res ; 19(9): 1940-1946, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38227519

ABSTRACT

Controlling intracranial pressure, nerve cell regeneration, and microenvironment regulation are the key issues in reducing mortality and disability in acute brain injury. There is currently a lack of effective treatment methods. Hibernation has the characteristics of low temperature, low metabolism, and hibernation rhythm, as well as protective effects on the nervous, cardiovascular, and motor systems. Artificial hibernation technology is a new technology that can effectively treat acute brain injury by altering the body's metabolism, lowering the body's core temperature, and allowing the body to enter a state similar to hibernation. This review introduces artificial hibernation technology, including mild hypothermia treatment technology, central nervous system regulation technology, and artificial hibernation-inducer technology. Upon summarizing the relevant research on artificial hibernation technology in acute brain injury, the research results show that artificial hibernation technology has neuroprotective, anti-inflammatory, and oxidative stress-resistance effects, indicating that it has therapeutic significance in acute brain injury. Furthermore, artificial hibernation technology can alleviate the damage of ischemic stroke, traumatic brain injury, cerebral hemorrhage, cerebral infarction, and other diseases, providing new strategies for treating acute brain injury. However, artificial hibernation technology is currently in its infancy and has some complications, such as electrolyte imbalance and coagulation disorders, which limit its use. Further research is needed for its clinical application.

10.
Cell Death Dis ; 14(2): 80, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36737428

ABSTRACT

Increased glycolysis is one of the key metabolic hallmarks of cancer cells. However, the roles of lncRNAs in energy metabolism and cancer metastasis remain unclear. Here, the expression of TMEM105 associated with glycolysis was dramatically elevated from normal to breast cancer to breast cancer liver metastasis tissues, and the survival analysis revealed that high TMEM105 expression was related to poor survival, especially in patients with liver metastasis. Moreover, TMEM105 facilitated the glycolysis of breast cancer cells and induced cell invasion and breast cancer liver metastasis (BCLM). Mechanistically, TMEM105 regulated LDHA expression by sponging miR-1208, which further promoted cell glycolysis and BCLM. Importantly, glycolytic production of lactate enhanced TMEM105 expression in breast cancer cells by activating the SHH-MAZ signaling pathway. These findings suggested that the lactate-responsive TMEM105 acted as a miRNA sponge, inducing BCLM via a glycolysis-mediated positive feedback loop, which might be a rational target for the treatment of BCLM patients.


Subject(s)
Breast Neoplasms , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Female , Humans , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5/metabolism , Lactates , Liver Neoplasms/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
11.
Neural Regen Res ; 18(9): 1999-2004, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36926725

ABSTRACT

Animal experiments have shown that injectable collagen scaffold with human umbilical cord-derived mesenchymal stem cells can promote recovery from spinal cord injury. To investigate whether injectable collagen scaffold with human umbilical cord-derived mesenchymal stem cells can be used to treat spontaneous intracerebral hemorrhage, this non-randomized phase I clinical trial recruited patients who met the inclusion criteria and did not meet the exclusion criteria of spontaneous intracerebral hemorrhage treated in the Characteristic Medical Center of Chinese People's Armed Police Force from May 2016 to December 2020. Patients were divided into three groups according to the clinical situation and patient benefit: control (n = 18), human umbilical cord-derived mesenchymal stem cells (n = 4), and combination (n = 8). The control group did not receive any transplantation. The human umbilical cord-derived mesenchymal stem cells group received human umbilical cord-derived mesenchymal stem cell transplantation. The combination group received injectable collagen scaffold with human umbilical cord-derived mesenchymal stem cells. Patients who received injectable collagen scaffold with human umbilical cord-derived mesenchymal stem cells had more remarkable improvements in activities of daily living and cognitive function and smaller foci of intracerebral hemorrhage-related encephalomalacia. Severe adverse events associated with cell transplantation were not observed. Injectable collagen scaffold with human umbilical cord-derived mesenchymal stem cells appears to have great potential treating spontaneous intracerebral hemorrhage.

12.
Neural Regen Res ; 18(9): 1990-1998, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36926724

ABSTRACT

There are various clinical treatments for traumatic brain injury, including surgery, drug therapy, and rehabilitation therapy; however, the therapeutic effects are limited. Scaffolds combined with exosomes represent a promising but challenging method for improving the repair of traumatic brain injury. In this study, we determined the ability of a novel 3D-printed collagen/chitosan scaffold loaded with exosomes derived from neural stem cells pretreated with insulin-like growth factor-1 (3D-CC-INExos) to improve traumatic brain injury repair and functional recovery after traumatic brain injury in rats. Composite scaffolds comprising collagen, chitosan, and exosomes derived from neural stem cells pretreated with insulin-like growth factor-1 (INExos) continuously released exosomes for 2 weeks. Transplantation of 3D-CC-INExos scaffolds significantly improved motor and cognitive functions in a rat traumatic brain injury model, as assessed by the Morris water maze test and modified neurological severity scores. In addition, immunofluorescence staining and transmission electron microscopy showed that 3D-CC-INExos implantation significantly improved the recovery of damaged nerve tissue in the injured area. In conclusion, this study suggests that transplanted 3D-CC-INExos scaffolds might provide a potential strategy for the treatment of traumatic brain injury and lay a solid foundation for clinical translation.

13.
Regen Biomater ; 9: rbac043, 2022.
Article in English | MEDLINE | ID: mdl-35855109

ABSTRACT

The secretome secreted by stem cells and bioactive material has emerged as a promising therapeutic choice for traumatic brain injury (TBI). We aimed to determine the effect of 3D-printed collagen/chitosan/secretome derived from human umbilical cord blood mesenchymal stem cells scaffolds (3D-CC-ST) on the injured tissue regeneration process. 3D-CC-ST was performed using 3D printing technology at a low temperature (-20°C), and the physical properties and degeneration rate were measured. The utilization of low temperature contributed to a higher cytocompatibility of fabricating porous 3D architectures that provide a homogeneous distribution of cells. Immediately after the establishment of the canine TBI model, 3D-CC-ST and 3D-CC (3D-printed collagen/chitosan scaffolds) were implanted into the cavity of TBI. Following implantation of scaffolds, neurological examination and motor evoked potential detection were performed to analyze locomotor function recovery. Histological and immunofluorescence staining were performed to evaluate neuro-regeneration. The group treated with 3D-CC-ST had good performance of behavior functions. Implanting 3D-CC-ST significantly reduced the cavity area, facilitated the regeneration of nerve fibers and vessel reconstruction, and promoted endogenous neuronal differentiation and synapse formation after TBI. The implantation of 3D-CC-ST also markedly reduced cell apoptosis and regulated the level of systemic inflammatory factors after TBI.

14.
Brain Res Bull ; 181: 65-76, 2022 04.
Article in English | MEDLINE | ID: mdl-35093467

ABSTRACT

Although the anti-inflammatory properties of developmental endothelial locus-1 (DEL-1) are well known, few studies have examined the role of DEL-1 in spinal cord injury (SCI). Here, the protective effect of DEL-1 on SCI was investigated using hypoxia/recovery (H/R) injury of astrocytes and a mouse SCI model. The effects of DEL-1 overexpression/silencing on primary astrocytes were assessed by flow cytometry, immunofluorescence, and western blotting. Female Sprague-Dawley rats were intrathecally injected with recombinant adeno-associated virus (AAV) at T10, and DEL-1 was permanently expressed. Protein levels in the spinal cord, functional testing, and electrophysiology, pathology, and immunofluorescence were all measured after treatment. DEL-1 overexpression significantly increased the expression of SIRT1/SERCA2At the same time, inflammation, endoplasmic reticulum stress, and apoptosis were all significantly inhibited, the motor function of SCI rats was noticeably restored, and the myelin sheath of the injured site was more complete. Furthermore, after DEL-1 silencing SIRT1/SERCA2 expression decreased, while inflammation, endoplasmic reticulum stress, and apoptotic responses increased significantly. DEL-1 treatment, however, did not increase SERCA2 expression after SIRT1 silencing. These findings demonstrate that DEL-1 protects against SCI via SIRT1/SERCA2 signaling, promoting spinal neural recovery.


Subject(s)
Apoptosis/physiology , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/metabolism , Endoplasmic Reticulum Stress/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sirtuin 1/metabolism , Spinal Cord Injuries/metabolism , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
15.
J Orthop Surg (Hong Kong) ; 29(2): 23094990211012293, 2021.
Article in English | MEDLINE | ID: mdl-34060363

ABSTRACT

BACKGROUND: Due to endogenous neuronal deficiency and glial scar formation, spinal cord injury (SCI) often leads to irreversible neurological loss. Accumulating evidence has shown that a suitable scaffold has important value for promoting nerve regeneration after SCI. Collagen/heparin sulfate scaffold (CHSS) has shown effect for guiding axonal regeneration and decreasing glial scar deposition after SCI. The current research aimed to evaluate the utility of the CHSSs adsorbed with mesenchymal stem cells (MSCs) on nerve regeneration, and functional recovery after acute complete SCI. METHODS: CHSSs were prepared, and evaluated for biocompatibility. The CHSSs adsorbed with MSCs were transplanted into these canines with complete SCI. RESULTS: We observed that MSCs had good biocompatibility with CHSSs. In complete transverse SCI models, the implantation of CHSS co-cultured with MSCs exhibited significant improvement in locomotion, motor evoked potential, magnetic resonance imaging, diffusion tensor imaging, and urodynamic parameters. Meanwhile, nerve fibers were markedly improved in the CHSS adsorbed with MSCs group. Moreover, we observed that the implantation of CHSS combined with MSCs modulated inflammatory cytokine levels. CONCLUSIONS: The results preliminarily demonstrated that the transplantation of MSCs on a CHSS could improve the recovery of motor function after SCI. Thus, implanting the MSCs-laden CHSS is a promising combinatorial therapy for treatment in acute SCI.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries , Tissue Scaffolds , Animals , Collagen , Diffusion Tensor Imaging , Dogs , Feasibility Studies , Heparin , Mesenchymal Stem Cell Transplantation/veterinary , Spinal Cord Injuries/therapy , Spinal Cord Injuries/veterinary , Sulfates
16.
Comput Intell Neurosci ; 2021: 5579888, 2021.
Article in English | MEDLINE | ID: mdl-34306053

ABSTRACT

Episodic memory allows a person to recall and mentally reexperience specific episodes from one's personal past. Studies of episodic memory are of great significance for the diagnosis and the exploration of the mechanism of memory generation. Most of the current studies focus on certain brain regions and pay less attention to the interrelationship between multiple brain regions. To explore the interrelationship in the brain network, we use an open fMRI dataset to construct the brain functional connectivity and effective connectivity network. We establish a binary directed network of the memory when it is reactivated. The binary directed network shows that the occipital lobe and parietal lobe have the most causal connections. The number of edges starting from the superior parietal lobule is the highest, with 49 edges, and 31 of which are connected to the occipital cortex. This means that the interaction between the superior parietal lobule and the occipital lobe plays the most important role in episodic memory, and the superior parietal lobule plays a more causal role in causality. In addition, memory regions such as the precuneus and fusiform also have some edges. The results show that the posterior parietal cortex plays an important role of hub node in the episodic memory network. From the brain network model, more information can be obtained, which is conducive to exploring the brain's changing pattern in the whole memory process.


Subject(s)
Memory, Episodic , Brain/diagnostic imaging , Brain Mapping , Humans , Magnetic Resonance Imaging , Mental Recall , Parietal Lobe/diagnostic imaging
17.
Mol Ther Nucleic Acids ; 24: 310-324, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-33850635

ABSTRACT

Hypoxia induces a series of cellular adaptive responses that enable promotion of inflammation and cancer development. Hypoxia-inducible factor-1α (HIF-1α) is involved in the hypoxia response and cancer promotion, and it accumulates in hypoxia and is degraded under normoxic conditions. Here we identify prostate cancer associated transcript-1 (PCAT-1) as a hypoxia-inducible long non-coding RNA (lncRNA) that regulates HIF-1α stability, crucial for cancer progression. Extensive analyses of clinical data indicate that PCAT-1 is elevated in breast cancer patients and is associated with pathological grade, tumor size, and poor clinical outcomes. Through gain- and loss-of-function experiments, we find that PCAT-1 promotes hypoxia-associated breast cancer progression including growth, migration, invasion, colony formation, and metabolic regulation. Mechanistically, PCAT-1 directly interacts with the receptor of activated protein C kinase-1 (RACK1) protein and prevents RACK1 from binding to HIF-1α, thus protecting HIF-1α from RACK1-induced oxygen-independent degradation. These findings provide new insight into lncRNA-mediated mechanisms for HIF-1α stability and suggest a novel role of PCAT-1 as a potential therapeutic target for breast cancer.

18.
Rev Assoc Med Bras (1992) ; 67(9): 1342-1348, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34816932

ABSTRACT

OBJECTIVE: This study aimed to assess the effect of the collagen/silk fibroin scaffolds seeded with human umbilical cord-mesenchymal stem cells on functional recovery after acute complete spinal cord injury. METHODS: The fibroin and collagen were mixed (mass ratio, 3:7), and the composite scaffolds were produced. Forty rats were randomly divided into the Sham group (without spinal cord injury), spinal cord injury group (spinal cord transection without any implantation), collagen/silk fibroin scaffolds group (spinal cord transection with implantation of the collagen/silk fibroin scaffolds), and collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group (spinal cord transection with the implantation of the collagen/silk fibroin scaffolds co-cultured with human umbilical cord-mesenchymal stem cells). Motor evoked potential, Basso-Beattie-Bresnahan scale, modified Bielschowsky's silver staining, and immunofluorescence staining were performed. RESULTS: The BBB scores in the collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group were significantly higher than those in the spinal cord injury and collagen/silk fibroin scaffolds groups (p<0.05 or p<0.01). The amplitude and latency were markedly improved in the collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group compared with the spinal cord injury and collagen/silk fibroin scaffolds groups (p<0.05 or p<0.01). Meanwhile, compared to the spinal cord injury and collagen/silk fibroin scaffolds groups, more neurofilament positive nerve fiber ensheathed by myelin basic protein positive structure at the injury site were observed in the collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group (p<0.01, p<0.05). The results of Bielschowsky's silver staining indicated more nerve fibers was observed at the lesion site in the collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group compared with the spinal cord injury and collagen/silk fibroin scaffolds groups (p<0.01, p< 0.05). CONCLUSION: The results demonstrated that the transplantation of human umbilical cord-mesenchymal stem cells on a collagen/silk fibroin scaffolds could promote nerve regeneration, and recovery of neurological function after acute spinal cord injury.


Subject(s)
Fibroins , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries , Animals , Collagen , Humans , Rats , Recovery of Function , Spinal Cord , Tissue Scaffolds , Umbilical Cord
19.
Biomater Sci ; 9(3): 745-764, 2021 Feb 07.
Article in English | MEDLINE | ID: mdl-33200759

ABSTRACT

The objective of this study was to evaluate the therapy effects of a novel biological scaffold containing heparin, collagen and vascular endothelial growth factor (VEGF) in treating traumatic brain injury (TBI). In our research, a functional composite scaffold constituted by collagen, heparin and vascular endothelial growth factor was used to stimulate angiogenesis and improve nerve-tissue regeneration in a rat model of TBI. The composite scaffold possessed excellent mechanical properties and good porosity, and could effectively control the release rate of VEGF. Motor and cognitive functions such as motor evoked potential, Morris water maze test and modified neurological severity score were evidently improved after the scaffold was grafted onto the injury site in the rat TBI model. There was clearly improved restoration of damaged nerve tissue at the injured site. Furthermore, brain edema and inflammatory reactions were significantly alleviated. Newly formed neurons with associated synaptic structures, nerve fibers, myelin sheaths and functional angiogenesis with intact endothelium at the injury site were observed. In conclusion, our data revealed that the collagen/heparin scaffold combined with VEGF could create excellent microenvironment stimuli for damaged nerve-tissue regeneration, providing a potential strategy for treating TBI.


Subject(s)
Brain Injuries, Traumatic , Vascular Endothelial Growth Factor A , Animals , Brain Injuries, Traumatic/drug therapy , Collagen , Heparin , Rats , Recovery of Function , Tissue Scaffolds
20.
Theranostics ; 11(2): 768-788, 2021.
Article in English | MEDLINE | ID: mdl-33391504

ABSTRACT

Rationale: The combination of medical and tissue engineering in neural regeneration studies is a promising field. Collagen, silk fibroin and seed cells are suitable options and have been widely used in the repair of spinal cord injury. In this study, we aimed to determine whether the implantation of a complex fabricated with collagen/silk fibroin (SF) and the human umbilical cord mesenchymal stem cells (hUCMSCs) can promote cerebral cortex repair and motor functional recovery in a canine model of traumatic brain injury (TBI). Methods: A porous scaffold was fabricated with cross-linked collagen and SF. Its physical properties and degeneration rate were measured. The scaffolds were co-cultured with hUCMSCs after which an implantable complex was formed. After complex implantation to a canine model of TBI, the motor evoked potential (MEP) and magnetic resonance imaging (MRI) were used to evaluate the integrity of the cerebral cortex. The neurologic score, motion capture, surface electromyography (sEMG), and vertical ground reaction force (vGRF) were measured in the analysis of motor functions. In vitro analysis of inflammation levels was performed by Elisa while immunohistochemistry was used in track the fate of hUCMSCs. In situ hybridization, transmission electron microscope, and immunofluorescence were used to assess neural and vascular regeneration. Results: Favorable physical properties, suitable degradation rate, and biocompatibility were observed in the collagen/SF scaffolds. The group with complex implantation exhibited the best cerebral cortex integrity and motor functions. The implantation also led to the regeneration of more blood vessels and nerve fibers, less glial fibers, and inflammatory factors. Conclusion: Implantation of this complex enhanced therapy in traumatic brain injury (TBI) through structural repair and functional recovery. These effects exhibit the translational prospects for the clinical application of this complex.


Subject(s)
Brain Injuries, Traumatic/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Motor Activity , Nerve Regeneration , Neural Pathways , Recovery of Function , Animals , Brain Injuries, Traumatic/pathology , Collagen/chemistry , Dogs , Fibroins/chemistry , Male , Tissue Engineering , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL