Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
ACS Biomater Sci Eng ; 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38557027

ABSTRACT

The intricate electrophysiological functions and anatomical structures of spinal cord tissue render the establishment of in vitro models for spinal cord-related diseases highly challenging. Currently, both in vivo and in vitro models for spinal cord-related diseases are still underdeveloped, complicating the exploration and development of effective therapeutic drugs or strategies. Organoids cultured from human induced pluripotent stem cells (hiPSCs) hold promise as suitable in vitro models for spinal cord-related diseases. However, the cultivation of spinal cord organoids predominantly relies on Matrigel, a matrix derived from murine sarcoma tissue. Tissue-specific extracellular matrices are key drivers of complex organ development, thus underscoring the urgent need to research safer and more physiologically relevant organoid culture materials. Herein, we have prepared a rat decellularized brain extracellular matrix hydrogel (DBECMH), which supports the formation of hiPSC-derived spinal cord organoids. Compared with Matrigel, organoids cultured in DBECMH exhibited higher expression levels of markers from multiple compartments of the natural spinal cord, facilitating the development and maturation of spinal cord organoid tissues. Our study suggests that DBECMH holds potential to replace Matrigel as the standard culture medium for human spinal cord organoids, thereby advancing the development of spinal cord organoid culture protocols and their application in in vitro modeling of spinal cord-related diseases.

2.
ACS Biomater Sci Eng ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38593429

ABSTRACT

Spinal cord organoids are of significant value in the research of spinal cord-related diseases by simulating disease states, thereby facilitating the development of novel therapies. However, the complexity of spinal cord structure and physiological functions, along with the lack of human-derived inducing components, presents challenges in the in vitro construction of human spinal cord organoids. Here, we introduce a novel human decellularized placenta-derived extracellular matrix hydrogel (DPECMH) and, combined with a new induction protocol, successfully construct human spinal cord organoids. The human placenta-sourced decellularized extracellular matrix (dECM), verified through hematoxylin and eosin staining, DNA quantification, and immunofluorescence staining, retained essential ECM components such as elastin, fibronectin, type I collagen, laminin, and so forth. The temperature-sensitive hydrogel made from human placenta dECM demonstrated good biocompatibility and promoted the differentiation of human induced pluripotent stem cell (hiPSCs)-derived spinal cord organoids into neurons. It displayed enhanced expression of laminar markers in comparison to Matrigel and showed higher expression of laminar markers compared to Matrigel, accelerating the maturation process of spinal cord organoids and demonstrating its potential as an organoid culture substrate. DPECMH has the potential to replace Matrigel as the standard additive for human spinal cord organoids, thus advancing the development of spinal cord organoid culture protocols and their application in the in vitro modeling of spinal cord-related diseases.

3.
Biomaterials ; 299: 122161, 2023 08.
Article in English | MEDLINE | ID: mdl-37236138

ABSTRACT

We previously constructed a three-dimensional gelatin sponge (3D-GS) scaffold as a delivery vehicle for therapeutic cells and trophic factors in the treatment of spinal cord injury (SCI), and this study aimed to assess the biosafety and efficacy of the scaffold in a non-human primate SCI model. However, because it has only been tested in rodent and canine models, the biosafety and efficacy of the scaffold should ideally be assessed in a non-human primate SCI model before its use in the clinic. No adverse reactions were observed over 8 weeks following 3D-GS scaffold implantation into in a Macaca fascicularis with hemisected SCI. Scaffold implantation also did not add to neuroinflammatory or astroglial responses already present at the injured site, suggesting good biocompatibility. Notably, there was a significant reduction in α-smooth muscle actin (αSMA)-positive cells at the injury/implantation interface, leading to alleviation of fibrotic compression of the residual spinal cord tissue. The regenerating tissue in the scaffold showed numerous cells migrating into the implant secreting abundant extracellular matrix, resulting in a pro-regenerative microenvironment. Consequently, nerve fiber regeneration, myelination, vascularization, neurogenesis, and electrophysiological improvements were achieved. These results indicated that the 3D-GS scaffold had good histocompatibility and effectiveness in the structural repair of injured spinal cord tissue in a non-human primate and is suitable for use in the treatment of patients with SCI.


Subject(s)
Gelatin , Spinal Cord Injuries , Animals , Dogs , Gelatin/chemistry , Tissue Scaffolds/chemistry , Spinal Cord Injuries/therapy , Nerve Regeneration/physiology , Spinal Cord , Primates
4.
ACS Nano ; 16(1): 823-836, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35025206

ABSTRACT

The microorganism has become a promising therapeutic tool for many diseases because it is a kind of cell factory that can efficiently synthesize a variety of bioactive substances. However, the metabolic destiny of microorganisms is difficult to predict in vivo. Here, a timing bionic dormant body with programmable destiny is reported, which can predict the metabolic time and location of microorganisms in vivo and can prevent it from being damaged by the complex biological environment in vivo. Taking the complex digestive system as an example, the bionic dormant body exists in the upper digestive tract as a nonmetabolic dormant body after oral administration and will be awakened to synthesize bioactive substances about 2 h after reaching the intestine. Compared with oral microorganisms alone, the bioavailability of the biomimetic dormant body in the intestine is almost 3.5 times higher. The utilization rate of the oral bionic dormant body to synthesize drugs is 2.28 times higher than oral drugs. We demonstrated the significant efficacies of treatment using Parkinson's disease (PD) mice by dormant body capable of timed neurotransmitter production after oral delivery. The timed bionic dormant body with programmable destiny may provide an effective technology to generate advanced microbial therapies for the treatment of various diseases.


Subject(s)
Bionics , Parkinson Disease , Mice , Animals , Pharmaceutical Preparations
5.
Sci China Life Sci ; 65(5): 909-926, 2022 05.
Article in English | MEDLINE | ID: mdl-34406569

ABSTRACT

Spinal cord injury (SCI) often results in an inhibitory environment at the injury site. In our previous studies, transplantation of a scaffold combined with stem cells was proven to induce neural regeneration in animal models of complete SCI. Based on these preclinical studies, collagen scaffolds loaded with the patients' own bone marrow mononuclear cells or human umbilical cord mesenchymal stem cells were transplanted into SCI patients. Fifteen patients with acute complete SCI and 51 patients with chronic complete SCI were enrolled and followed up for 2 to 5 years. No serious adverse events related to functional scaffold transplantation were observed. Among the patients with acute SCI, five patients achieved expansion of their sensory positions and six patients recovered sensation in the bowel or bladder. Additionally, four patients regained voluntary walking ability accompanied by reconnection of neural signal transduction. Among patients with chronic SCI, 16 patients achieved expansion of their sensation level and 30 patients experienced enhanced reflexive defecation sensation or increased skin sweating below the injury site. Nearly half of the patients with chronic cervical SCI developed enhanced finger activity. These long-term follow-up results suggest that functional scaffold transplantation may represent a feasible treatment for patients with complete SCI.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries , Animals , Humans , Mesenchymal Stem Cell Transplantation/methods , Nerve Regeneration , Spinal Cord , Spinal Cord Injuries/surgery , Tissue Scaffolds
6.
Mol Med Rep ; 23(3)2021 03.
Article in English | MEDLINE | ID: mdl-33495805

ABSTRACT

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that several pairings of panels in Fig. 5, as shown on p. 5599, were strikingly similar. After having examined their original data, the authors realized that they uploaded some images incorrectly during the process of compiling this figure, and that there were duplicated data panels in this figure. However, the authors were able to consult their original data, and had access to the correct images. The revised version of Fig. 5, showing the correct data for the Akt/Control, p­Akt/Control, mTOR/0.05 µM Ouabain, HIF­1α/0.05 µM Ouabain and Akt/0.5 µM Ouabain experiments, is shown opposite. Note that the replacement of the erroneous data does not affect either the results or the conclusions reported in this paper, and all the authors agree to this Corrigendum. The authors are grateful to the Editor of Molecular Medicine Reports for granting them this opportunity to publish a Corrigendum, and apologize to the readership for any inconvenience caused. [the original article was published in Molecular Medicine Reports 17: 5595­5600, 2018; DOI: 10.3892/mmr.2018.8587].

7.
Front Neurosci ; 14: 403, 2020.
Article in English | MEDLINE | ID: mdl-32581664

ABSTRACT

Traumatic brain injury (TBI) contributes to hypocoagulopathy associated with prolonged bleeding and hemorrhagic progression. Bloodletting puncture therapy at hand twelve Jing-well points (BL-HTWP) has been applied as a first aid measure in various emergent neurological diseases, but the detailed mechanisms of the modulation between the central nervous system and systemic circulation after acute TBI in rodents remain unclear. To investigate whether BL-HTWP stimulation modulates hypocoagulable state and exerts neuroprotective effect, experimental TBI model of mice was produced by the controlled cortical impactor (CCI), and treatment with BL-HTWP was immediately made after CCI. Then, the effects of BL-HTWP on the neurological function, cerebral perfusion state, coagulable state, and cerebrovascular histopathology post-acute TBI were determined, respectively. Results showed that BL-HTWP treatment attenuated cerebral hypoperfusion and improve neurological recovery post-acute TBI. Furthermore, BL-HTWP stimulation reversed acute TBI-induced hypocoagulable state, reduced vasogenic edema and cytotoxic edema by regulating multiple hallmarks of coagulopathy in TBI. Therefore, we conclude for the first time that hypocoagulopathic state occurs after acute experimental TBI, and the neuroprotective effect of BL-HTWP relies on, at least in part, the modulation of hypocoagulable state. BL-HTWP therapy may be a promising strategy for acute severe TBI in the future.

8.
Int Immunopharmacol ; 86: 106715, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32570036

ABSTRACT

Traumatic brain injury (TBI) can be exacerbated and prolonged for months or even years by chronic inflammatory processes with long-term consequences on neurodegeneration and neurological impairment. However, there are no clear pharmacological therapies of benefit to manage neurological dysfunctions, which, relating to the molecular mechanisms underlying the behavioral deficits after TBI, have yet to be fully identified. Recently, a glucagon-like peptide 1 (GLP-1) agonist, Exendin-4, was approved not only for the treatment of type 2 diabetes mellitus, but it also played a neurotrophic role in various CNS neurological diseases. In this study, we evaluated the neuroprotective effects of Exendin-4 on neurological outcome, cerebral blood flow, neurodegeneration, and inflammatory responses by utilizing a cortical contusion impact injury (CCI) model in rats. We found that TBI rats displayed neurological impairments, neurodegeneration, reduction of cerebral blood flow, and inflammatory responses, while Exendin-4 promoted neurological, cognitive, and cerebral blood flow recovery and attenuated neural degeneration and inflammatory cytokines after TBI. Furthermore, Exendin-4 treatment significantly diminished the TBI-induced overexpression of TNFα and IL-1ß, as well as phosphorylation of p38 and ERK1/2. These data suggest a strong beneficial action of the glucagon-like peptide-1 receptor agonist Exendin-4 in improving neurological outcomes by attenuating inflammatory responses induced by traumatic brain injury, which is of therapeutic potential for TBI.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Exenatide/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Inflammation/drug therapy , Neuroprotective Agents/pharmacology , Animals , Behavior, Animal/drug effects , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Cerebrovascular Circulation/drug effects , Cognition/drug effects , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Exenatide/therapeutic use , Inflammation/etiology , Interleukin-1beta/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuroglia/drug effects , Neuroprotective Agents/therapeutic use , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Zhongguo Zhen Jiu ; 39(10): 1075-80, 2019 Oct 12.
Article in Chinese | MEDLINE | ID: mdl-31621260

ABSTRACT

OBJECTIVE: To observe the effect of bloodletting acupuncture at twelve jing-well points of hand on microcirculatory disturbance in mice with traumatic brain injury (TBI), and to explore the protective effect of bloodletting therapy on TBI. METHODS: Sixty clean adult male C57BL/6J mice were randomly divided into a sham-operation group, a model group and a treatment group, 20 mice in each group. The TBI model was established by using electronic controlled cerebral cortex impact instrument in the model group and the treatment group. The mice in the treatment group were treated with bloodletting acupuncture at bilateral "Shaoshang" (LU 11), "Shangyang" (LI 1), "Zhongchong" (PC 9), "Guanchong" (TE 1), "Shaochong" (HT 9) and "Shaoze" (SI 1) immediately after trauma. The mice in the sham-operation group only opened the bone window but did not receive the strike. The regional cerebral blood flow (rCBF) was monitored by laser speckle contrast analysis (LASCA) using a PeriCam PSI System before trauma, immediately after trauma and 1, 2, 12, 24, 48, 72 h after trauma. The brain water content was measured by wet-dry weight method 24 h after trauma. The severity of functional impairment at 2, 12, 24, 48 and 72 h after trauma was evaluated by modified neurological scale scores (mNSS). RESULTS: ① 2 h after trauma, the mNSS in the model group and treatment group were >7 points, suggesting the successful establishment of model; compared with the sham-operation group, the mNSS was increased significantly from 12 to 72 h after trauma in the model group ( all P<0.01), but the mNSS in the treatment group was significantly lower than that in the model group from 2 to 24 h after trauma (P<0.01, P<0.05). ② Compared with the sham-operation group, rCBF in the model group was decreased significantly immediately after trauma (P<0.01), and the rCBF in the model group was lower than that in the sham-operation group from 1 to 72 h after trauma ( all P<0.01); rCBF in the treatment group began to rise and was significantly higher than that in the model group 1-2 h after trauma (P<0.01); 12-48 h after trauma, the increasing of rCBF in the two groups tended to be gentle until 72 h after injury, and rCBF in the model group was decreased while that in the treatment group continued to rise and was higher than that in the model group (P<0.01). ③ 24 h after trauma, the brain water content in the model group was significantly higher than that in the sham-operation group (P<0.01), and brain water content in the treatment group was significantly lower than that in the model group (P<0.01). CONCLUSION: The bloodletting acupuncture at twelve jing-well points of hand could improve microcirculation disturbance, increase microcirculation perfusion, alleviate secondary brain edema and promote the recovery of nerve function in mice with TBI.


Subject(s)
Acupuncture Therapy , Bloodletting , Brain Injuries, Traumatic , Acupuncture Points , Animals , Brain Injuries, Traumatic/therapy , Male , Mice , Mice, Inbred C57BL , Microcirculation , Random Allocation
10.
Biomed Res Int ; 2019: 5653212, 2019.
Article in English | MEDLINE | ID: mdl-31355268

ABSTRACT

OBJECTIVE: Casein kinase 2 interacting protein-1 (CKIP-1) has exhibited multiple functions in regulating cell proliferation, apoptosis, differentiation, and cytoskeleton. CKIP-1 also plays an important role as a critical regulator in tumorigenesis. The aim of this study is to further examine the function of CKIP-1 in glioma cells. METHODS: The expression level of CKIP-1 protein was determined in gliomas tissues and cell lines by immunohistochemistry stain and western blotting while the association of CKIP-1 expression with prognosis was analyzed by Kaplan-Meier method and compared by log-rank test. CKIP-1 was overexpressed or silenced in gliomas cell lines. CCK-8, colony formation assay, and BrdU incorporation assay were used to determine cell proliferation and DNA synthesis. Cell cycle and apoptosis rate were determined with fluorescence-activated cell sorting (FACS) method. Then, expression of key members in AKT/GSK3ß/ß-catenin pathway was detected by western blot analysis. RESULTS: In the present study, we reported new evidence that CKIP-1 was reversely associated with the proliferation of glioma cells and survival in glioma patients. Additionally, the overexpressed CKIP-1 significantly inhibited glioma cell proliferation. Further experiments revealed that CKIP-1 functioned through its antiproliferative and proapoptotic activity in glioma cells. Importantly, mechanistic investigations suggested that CKIP-1 sharply suppressed the activity of AKT by inhibiting the phosphorylation, markedly downregulated the phosphorylated GSK3ß at Ser9, and promoted ß-catenin degradation. CONCLUSIONS: Overall, our results provided new insights into the clinical significance and molecular mechanism of CKIP-1 in glioma, which indicated CKIP1 might function as a therapeutic target for clinical treatment of glioma.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioma , Glycogen Synthase Kinase 3 beta/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , beta Catenin/metabolism , Adult , Cell Line, Tumor , Female , Glioma/metabolism , Glioma/mortality , Glioma/pathology , Humans , Male , Middle Aged
11.
Int J Nanomedicine ; 14: 721-732, 2019.
Article in English | MEDLINE | ID: mdl-30705588

ABSTRACT

BACKGROUND: Traumatic spinal cord injury (SCI) causes neuronal death, demyelination, axonal degeneration, inflammation, glial scar formation, and cystic cavitation resulting in interruption of neural signaling and loss of nerve function. Multifactorial targeted therapy is a promising strategy for SCI. METHODS: The anti-inflammatory peptide KAFAKLAARLYRKALARQLGVAA (KAFAK) and brain-derived neurotrophic factor (BDNF)-modified hyaluronan-methylcellulose (HAMC) hydrogel was designed for minimally invasive, localized, and sustained intrathecal protein delivery. The physical and biological characteristics of HAMC-KAFAK/BDNF hydrogel were measured in vitro. SCI model was performed in rats and HAMC-KAFAK/BDNF hydrogel was injected into the injured site of spinal cord. The neuronal regeneration effect was evaluated by inflammatory cytokine levels, behavioral test and histological analysis at 8 weeks post operation. RESULTS: HAMC-KAFAK/BDNF hydrogel showed minimally swelling property and sustained release of the KAFAK and BDNF. HAMC-KAFAK/BDNF hydrogel significantly improved the proliferation of PC12 cells in vitro without cytotoxicity. Significant recovery in both neurological function and nerve tissue morphology in SCI rats were observed in HAMC-KAFAK/BDNF group. HAMC-KAFAK/BDNF group showed significant reduction in proinflammatory cytokines expression and cystic cavitation, decreased glial scar formation, and improved neuronal survival in the rat SCI model compared to HAMC group and SCI group. CONCLUSION: The HAMC-KAFAK/BDNF hydrogel promotes functional recovery of rats with spinal cord injury by regulating inflammatory cytokine levels and improving axonal regeneration.


Subject(s)
Brain-Derived Neurotrophic Factor/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Methylcellulose/chemistry , Nerve Regeneration/drug effects , Peptides/pharmacology , Spinal Cord Injuries/physiopathology , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Female , Peptides/chemistry , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord Injuries/metabolism
12.
Neural Regen Res ; 14(6): 991-996, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30762010

ABSTRACT

An accurate and effective neurological evaluation is indispensable in the treatment and rehabilitation of traumatic brain injury. However, most of the existing evaluation methods in basic research and clinical practice are not objective or intuitive for assessing the neurological function of big animals, and are also difficult to use to qualify the extent of damage and recovery. In the present study, we established a big animal model of traumatic brain injury by impacting the cortical motor region of beagles. At 2 weeks after successful modeling, we detected neurological deficiencies in the animal model using a series of techniques, including three-dimensional motion capture, electromyogram and ground reaction force. These novel technologies may play an increasingly important role in the field of traumatic brain injury diagnosis and rehabilitation in the future. The experimental protocol was approved by the Animal Care and Use Committee of Logistics University of People's Armed Police Force (approval No. 2017-0006.2).

13.
J Neurosci Methods ; 307: 95-105, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29960029

ABSTRACT

BACKGROUND: Animal models are essential in simulating clinical diseases and facilitating relevant studies. NEW METHOD: We established a precise canine model of traumatic brain injury (TBI) based on cerebral motor cortex injury which was confirmed by neuroimaging, electrophysiology, and a series of motor function assessment methods. Twelve beagles were divided into control, sham, and model groups. The cerebral motor cortex was identified by diffusion tensor imaging (DTI), a simple marker method, and intraoperative electrophysiological measurement. Bony windows were designed by magnetic resonance imaging (MRI) scan and DTI. During the operation, canines in the control group were under general anesthesia. The canines were operated via bony window craniotomy and dura mater opening in the sham group. After opening of the bony window and dura mater, the motor cortex was impacted by a modified electronic cortical contusion impactor (eCCI) in the model group. RESULTS: Postoperative measurements revealed damage to the cerebral motor cortex and functional defects. Comparisons between preoperative and postoperative results demonstrated that the established model was successful. COMPARISON WITH EXISTING METHOD(S): Compared with conventional models, this is the first brain trauma model in large animal that was constructed based on injury to the cerebral motor cortex under the guidance of DTI, a simple marker method, and electrophysiology. CONCLUSION: The method used to establish this model can be standardized to enhance reproducibility and provide a stable and precise large animal model of TBI for clinical and basic research.


Subject(s)
Brain Injuries, Traumatic/etiology , Disease Models, Animal , Evoked Potentials, Motor/physiology , Motor Cortex/injuries , Animals , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/veterinary , Craniotomy , Dogs , Electric Stimulation , Glasgow Coma Scale , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Motor Activity/physiology , Motor Cortex/diagnostic imaging
14.
Cell Transplant ; 27(6): 907-915, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29871514

ABSTRACT

Stem cells and biomaterials transplantation hold a promising treatment for functional recovery in spinal cord injury (SCI) animal models. However, the functional recovery of complete SCI patients was still a huge challenge in clinic. Additionally, there is no clinical standard procedure available to diagnose precisely an acute patient as complete SCI. Here, two acute SCI patients, with injury at thoracic 11 (T11) and cervical 4 (C4) level respectively, were judged as complete injury by a stricter method combined with American Spinal Injury Association (ASIA) Impairment Scale, magnetic resonance imaging (MRI) and nerve electrophysiology. Collagen scaffolds, named NeuroRegen scaffolds, with human umbilical cord mesenchymal stem cells (MSCs) were transplanted into the injury site. During 1 year follow up, no obvious adverse symptoms related to the functional scaffolds implantation were found after treatment. The recovery of the sensory and motor functions was observed in the two patients. The sensory level expanded below the injury level, and the patients regained the sense function in bowel and bladder. The thoracic SCI patient could walk voluntary with the hip under the help of brace. The cervical SCI patient could raise his lower legs against the gravity in the wheelchair and shake his toes under control. The injury status of the two patients was improved from ASIA A complete injury to ASIA C incomplete injury. Furthermore, the improvement of sensory and motor functions was accompanied with the recovery of the interrupted neural conduction. These results showed that the supraspinal control of movements below the injury was regained by functional scaffolds implantation in the two patients who were judged as the complete injury with combined criteria, it suggested that functional scaffolds transplantation could serve as an effective treatment for acute complete SCI patients.


Subject(s)
Collagen/chemistry , Mesenchymal Stem Cell Transplantation/methods , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Tissue Scaffolds/chemistry , Adult , Collagen/therapeutic use , Humans , Male , Mesenchymal Stem Cells/cytology , Recovery of Function , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Treatment Outcome
15.
Mol Med Rep ; 17(4): 5595-5600, 2018 04.
Article in English | MEDLINE | ID: mdl-29436645

ABSTRACT

Glioma is one of the most malignant forms of brain tumor, and has been of persistent concern due to its high recurrence and mortality rates, and limited therapeutic options. As a cardiac glycoside, ouabain has widespread applications in congestive heart diseases due to its positive cardiac inotropic effect by inhibiting Na+/K+­ATPase. Previous studies have demonstrated that ouabain has antitumor activity in several types of human tumor, including glioma. However, the exact underlying mechanism remains to be elucidated. The purpose of present study was to elucidate the effect of ouabain on human glioma cell apoptosis and investigate the exact mechanism. U­87MG cells were treated with various concentrations of ouabain for 24 h, following which cell viability and survival rate were assessed using a 3­(4,5-dimethylthiazol-2­yl)­2,5­diphenyltetrazolium bromide assay. The dynamic changes and cell motility were observed using digital holographic microscopy. Additionally, western blot analysis and high­content screening assays were used to detect the protein expression levels of phosphorylated (p­)Akt, mammalian target of rapamycin (mTOR), p­mTOR and hypoxia­inducible factor (HIF)­1α, respectively. Compared with the control group, ouabain suppressed U­87MG cell survival, and attenuated cell motility in a dose­dependent manner (P<0.01). The downregulation of p­Akt, mTOR, p­mTOR and HIF­1α were observed following treatment with 2.5 and 25 µmol/l of ouabain. These results suggested that ouabain exerted suppressive effects on tumor cell growth and motility, leading to cell death via regulating the intracellular Akt/mTOR signaling pathway and inhibiting the expression of HIF­1α in glioma cells. The present study examined the mechanism underlying the antitumor property of ouabain, providing a novel potential therapeutic agent for glioma treatment.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ouabain/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Glioma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
16.
Neurotherapeutics ; 15(1): 216-232, 2018 01.
Article in English | MEDLINE | ID: mdl-29247448

ABSTRACT

This study aimed to investigate the effects of targeted temperature management (TTM) modulation on traumatic brain injury (TBI) and the involved mechanisms using quantitative proteomics technology. SH-SY5Y and HT-22 cells were subjected to moderate stretch injury using the cell injury controller (CIC), followed by incubation at TTM (mild hypothermia, 32°C), or normothermia (37°C). The real-time morphological changes, cell cycle phase distribution, death, and cell viability were evaluated. Moderate TBI was produced by the controlled cortical impactor (CCI), and the effects of TTM on the neurological damage, neurodegeneration, cerebrovascular histopathology, and behavioral outcome were determined in vivo. Results showed that TTM treatment prevented TBI-induced neuronal necrosis in the brain, achieved a substantial reduction in neuronal death both in vitro and in vivo, reduced cortical lesion volume and neuronal loss, attenuated cerebrovascular histopathological damage, brain edema, and improved behavioral outcome. Using an iTRAQ proteomics approach, proteins that were significantly associated with TTM in experimental TBI were identified. Importantly, changes in four candidate molecules (plasminogen [PLG], antithrombin III [AT III], fibrinogen gamma chain [FGG], transthyretin [TTR]) were verified using TBI rat brain tissues and TBI human cerebrospinal fluid (CSF) samples. This study is one of the first to investigate the neuroprotective effects of TTM on the proteome of human and experimental models of TBI, providing an overall landscape of the TBI brain proteome and a scientific foundation for further assessment of candidate molecules associated with TTM for the promotion of reparative strategies post-TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Hypothermia, Induced/methods , Proteomics , Animals , Brain Injuries, Traumatic/pathology , Cell Cycle , Cell Line, Tumor , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Cold Temperature , Hippocampus/metabolism , Hippocampus/pathology , Humans , Maze Learning , Mice , Necrosis , Neurons/metabolism , Neurons/pathology , Physical Stimulation , Rats , Rats, Sprague-Dawley
17.
Reprod Toxicol ; 74: 10-22, 2017 12.
Article in English | MEDLINE | ID: mdl-28843701

ABSTRACT

Long-term exposure to fine particulate matter (PM2.5) may cause adverse pregnancy outcomes but the mechanisms are not clear. Our research confirms that PM2.5 induced DNA damage, and inhibited cell proliferation in HTR-8/SVneo cells, presenting in a dose- and time-dependent manners. Using quantitative proteomics, the 182 and 486 differentially expressed proteins in cells treated with 120µgml-1 PM2.5 for 24 and 48h were involved in many critical biological processes, including of cell proliferation, response to DNA damage, regulation of small GTPase mediated signal transduction, and etc. Further validation indicated that PM2.5 blocked the cell cycle at the G2/M phase through activation of the ATR-Cyclin E1/Cdk6 pathway, and it reduced the migration and invasion by upregulating TIMP1 and TIMP2 expression and downregulating Collagen I expression. Our findings were consistent with the observed effects of PM2.5 on cell cycle arrest and inhibition of migration and invasion in human extravillous trophoblast.


Subject(s)
Air Pollutants/toxicity , Particulate Matter/toxicity , Trophoblasts/drug effects , Air Pollutants/analysis , Arsenic/analysis , Arsenic/toxicity , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen Type I/metabolism , DNA Damage , Humans , Metals, Heavy/analysis , Metals, Heavy/toxicity , Particulate Matter/analysis , Polycyclic Aromatic Hydrocarbons/analysis , Polycyclic Aromatic Hydrocarbons/toxicity , Proteomics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Trophoblasts/metabolism , Trophoblasts/physiology
18.
Sci Rep ; 7(1): 5818, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28724891

ABSTRACT

Necroptosis, a novel type of programmed cell death, is involved in stroke-induced ischemic brain injury. Although studies have sought to explore the mechanisms of necroptosis, its signaling pathway has not yet to be completely elucidated. Thus, we used oxygen-glucose deprivation (OGD) and middle cerebral artery occlusion (MCAO) models mimicking ischemic stroke (IS) conditions to investigate mechanisms of necroptosis. We found that OGD and MCAO induced cell death, local brain ischemia and neurological deficit, while zVAD-fmk (zVAD, an apoptotic inhibitor), GSK'872 (a receptor interacting protein kinase-3 (RIP3) inhibitor), and combined treatment alleviated cell death and ischemic brain injury. Moreover, OGD and MCAO upregulated protein expression of the triggers of necroptosis: receptor interacting protein kinase-1 (RIP1), RIP3 and mixed lineage kinase domain-like protein (MLKL). The upregulation of these proteins was inhibited by GSK'872, combination treatments and RIP3 siRNA but not zVAD treatment. Intriguingly, hypoxia-inducible factor-1 alpha (HIF-1α), an important transcriptional factor under hypoxic conditions, was upregulated by OGD and MCAO. Similar to their inhibitory effects on aforementioned proteins upregulation, GSK'872, combination treatments and RIP3 siRNA decreased HIF-1α protein level. These findings indicate that necroptosis contributes to ischemic brain injury induced by OGD and MCAO and implicate HIF-1α, RIP1, RIP3, and MLKL in necroptosis.


Subject(s)
Apoptosis , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , GTPase-Activating Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Amino Acid Chloromethyl Ketones , Animals , Brain Injuries/complications , Brain Ischemia/complications , Cell Line , Down-Regulation/drug effects , Glucose/deficiency , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Necrosis , Oxygen , RNA, Small Interfering/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
19.
Biomaterials ; 137: 73-86, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28544974

ABSTRACT

Studies have shown that endogenous neural stem cells (NSCs) activated by spinal cord injury (SCI) primarily generate astrocytes to form glial scar. The NSCs do not differentiate into neurons because of the adverse microenvironment. In this study, we defined the activation timeline of endogenous NSCs in rats with severe SCI. These injury-activated NSCs then migrated into the lesion site. Cetuximab, an EGFR signaling antagonist, significantly increased neurogenesis in the lesion site. Meanwhile, implanting cetuximab modified linear ordered collagen scaffolds (LOCS) into SCI lesion sites in dogs resulted in neuronal regeneration, including neuronal differentiation, maturation, myelination, and synapse formation. The neuronal regeneration eventually led to a significant locomotion recovery. Furthermore, LOCS implantation could also greatly decrease chondroitin sulfate proteoglycan (CSPG) deposition at the lesion site. These findings suggest that endogenous neurogenesis following acute complete SCI is achievable in species ranging from rodents to large animals via functional scaffold implantation. LOCS-based Cetuximab delivery system has a promising therapeutic effect on activating endogenous neurogenesis, reducing CSPGs deposition and improving motor function recovery.


Subject(s)
Cetuximab/chemistry , Cetuximab/pharmacology , Collagen/chemistry , ErbB Receptors/antagonists & inhibitors , Neural Stem Cells/pathology , Neurogenesis/drug effects , Spinal Cord Injuries/metabolism , Tissue Scaffolds , Acute Disease , Animals , Cell Differentiation , Chondroitin Sulfate Proteoglycans/metabolism , Dogs , Female , Humans , Nerve Regeneration/drug effects , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries/pathology
20.
Cell Transplant ; 26(5): 891-900, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28185615

ABSTRACT

Regeneration of damaged neurons and recovery of sensation and motor function after complete spinal cord injury (SCI) are challenging. We previously developed a collagen scaffold, NeuroRegen, to promote axonal growth along collagen fibers and inhibit glial scar formation after SCI. When functionalized with multiple biomolecules, this scaffold promoted neurological regeneration and functional recovery in animals with SCI. In this study, eight patients with chronic complete SCI were enrolled to examine the safety and efficacy of implanting NeuroRegen scaffold with human umbilical cord mesenchymal stem cells (hUCB-MSCs). Using intraoperative neurophysiological monitoring, we identified and surgically resected scar tissues to eliminate the inhibitory effect of glial scarring on nerve regeneration. We then implanted NeuroRegen scaffold loaded with hUCB-MSCs into the resection sites. No adverse events (infection, fever, headache, allergic reaction, shock, perioperative complications, aggravation of neurological status, or cancer) were observed during 1 year of follow-up. Primary efficacy outcomes, including expansion of sensation level and motor-evoked potential (MEP)-responsive area, increased finger activity, enhanced trunk stability, defecation sensation, and autonomic neural function recovery, were observed in some patients. Our findings suggest that combined application of NeuroRegen scaffold and hUCB-MSCs is safe and feasible for clinical therapy in patients with chronic SCI. Our study suggests that construction of a regenerative microenvironment using a scaffold-based strategy may be a possible future approach to SCI repair.


Subject(s)
Spinal Cord Injuries/therapy , Adolescent , Adult , Aged , Cells, Cultured , Female , Humans , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Microscopy, Electron, Scanning , Middle Aged , Recovery of Function/physiology , Spinal Cord Regeneration/physiology , Tissue Scaffolds , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...