Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Neural Regen Res ; 16(2): 350-356, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32859797

ABSTRACT

Flavonoids have been reported to have therapeutic potential for spinal cord injury. Hawthorn leaves have abundant content and species of total flavonoids, and studies of the effects of the total flavonoids of hawthorn leaves on spinal cord injury have not been published in or outside China. Therefore, Sprague-Dawley rats were used to establish a spinal cord injury model by Allen's method. Rats were intraperitoneally injected with 0.2 mL of different concentrations of total flavonoids of hawthorn leaves (5, 10, and 20 mg/kg) after spinal cord injury. Injections were administered once every 6 hours, three times a day, for 14 days. After treatment with various concentrations of total flavonoids of hawthorn leaves, the Basso, Beattie, and Bresnahan scores and histological staining indicated decreases in the lesion cavity and number of apoptotic cells of the injured spinal cord tissue; the morphological arrangement of the myelin sheath and nerve cells tended to be regular; and the Nissl bodies in neurons increased. The Basso, Beattie, and Bresnahan scores of treated spinal cord injury rats were increased. Western blot assays showed that the expression levels of pro-apoptotic Bax and cleaved caspase-3 were decreased, but the expression level of the anti-apoptotic Bcl-2 protein was increased. The improvement of the above physiological indicators showed a dose-dependent relationship with the concentration of total flavonoids of hawthorn leaves. The above findings confirm that total flavonoids of hawthorn leaves can reduce apoptosis and exert neuroprotective effects to promote the recovery of the motor function of rats with spinal cord injury. This study was approved by the Ethics Committee of the Guangxi Medical University of China (approval No. 201810042) in October 2018.

2.
Curr Top Med Chem ; 20(21): 1911-1915, 2020.
Article in English | MEDLINE | ID: mdl-32493190

ABSTRACT

AIMS: A series of 8-methoxy ciprofloxacin- hydrazone/acylhydrazone hybrids were evaluated for their activity against a panel of cancer cell lines including HepG2 liver cancer cells, MCF-7, doxorubicin- resistant MCF-7 (MCF-7/DOX) breast cancer cells, DU-145 and multidrug-resistant DU145 (MDR DU-145) prostate cancer cells to seek for novel anticancer agents. BACKGROUND: Ciprofloxacin with excellent pharmacokinetic properties as well as few side effects, is one of the most common used antibacterial agents. Notably, Ciprofloxacin could induce cancer cells apoptosis, and cell cycle arrest at the S/G2 stage. The structure-activity relationship reveals that the introduction of the methoxy group into the C-8 position of the fluoroquinolone moiety has resulted in a greater binding affinity to the binding site, and 8-methoxy ciprofloxacin derivatives have proved a variety of biological activities even against drug-resistant organisms. However, to the best of our current knowledge, there are no studies that have reported the anticancer activity of 8-methoxy ciprofloxacin derivatives so far. Furthermore, many fluoroquinolone-hydrazone/acylhydrazone hybrids possess promising anticancer activity. Thus, it is rational to screen the anticancer activity of 8-methoxy ciprofloxacin derivatives. OBJECTIVE: To enrich the structure-activity relationship and provide new anticancer candidates for further investigations. METHODS: The desired 8-methoxy ciprofloxacin-hydrazone/acylhydrazone hybrids 5 and 6 were screened for their in vitro anticancer activity against liver cancer cells HepG2, breast cancer cells MCF-7, MCF7/DOX, prostate cancer cells DU-145 and MDR DU-145 by MTT assay. RESULTS: Some of 8-methoxy ciprofloxacin-hydrazone hybrids showed potential activity against HepG2, MCF-7, MCF-7/DOX, DU-145 and MDR DU-145 cancer cell lines, low cytotoxicity towards VERO cells and promising inhibitory activity on tubulin polymerization. CONCLUSION: Compounds 5d and 5f showed promising anticancer activity, low cytotoxicity, and potential tubulin polymerization inhibitory activity, were worthy of investigation. Other: The structure-activity relationship was enriched.


Subject(s)
Antineoplastic Agents/pharmacology , Ciprofloxacin/pharmacology , Hydrazones/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Ciprofloxacin/chemistry , Drug Screening Assays, Antitumor , Humans , Hydrazones/chemistry , Molecular Structure , Structure-Activity Relationship
3.
Transl Stroke Res ; 11(2): 254-266, 2020 04.
Article in English | MEDLINE | ID: mdl-31250378

ABSTRACT

Hippocampal injury and cognitive impairments are common after cardiac arrest and stroke and do not have an effective intervention despite much effort. Therefore, we developed a new approach aimed at reversing synaptic dysfunction by targeting TRPM2 channels. Cardiac arrest/cardiopulmonary resuscitation (CA/CPR) in mice was used to investigate cognitive deficits and the role of the calcium-permeable ion channel transient receptor potential-M2 (TRPM2) in ischemia-induced synaptic dysfunction. Our data indicates that absence (TRPM2-/-) or acute inhibition of TRPM2 channels with tatM2NX reduced hippocampal cell death in males only, but prevented synaptic plasticity deficits in both sexes. Remarkably, administration of tatM2NX weeks after injury reversed hippocampal plasticity and memory deficits. Finally, TRPM2-dependent activation of calcineurin-GSK3ß pathway contributes to synaptic plasticity impairments. These data suggest persistent TRPM2 activity following ischemia contributes to impairments of the surviving hippocampal network and that inhibition of TRPM2 channels at chronic time points may represent a novel strategy to improve functional recovery following cerebral ischemia that is independent of neuroprotection.


Subject(s)
Cognitive Dysfunction/physiopathology , Heart Arrest/complications , Hippocampus/physiopathology , Ischemia/complications , Neurons/physiology , TRPM Cation Channels/physiology , Animals , Calcineurin/physiology , Cardiopulmonary Resuscitation , Cognitive Dysfunction/etiology , Female , Glycogen Synthase Kinase 3 beta/physiology , Ischemia/physiopathology , Male , Mice, Knockout , Neuronal Plasticity , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics
4.
Rejuvenation Res ; 22(5): 420-430, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30632458

ABSTRACT

MicroRNAs (miRNAs) are endogenous noncoding small molecule RNAs that regulate cell proliferation, differentiation, fat metabolism, and hormone secretion. Studies have shown that miRNAs regulate the processes related to osteoporosis, including the differentiation of osteoblasts, osteoclasts, and chondrocytes, and are one of the important regulatory factors of some bone metabolic diseases. In our previous study, it has been revealed that natural compound Polygonatum sibiricum polysaccharide (PSP) can promote osteoblast formation and block osteoclastogenesis through Wnt/ß-catenin signaling pathway. This study was designed to investigate whether PSP can inhibit expression of osteoclast-related genes by Hippo signaling pathway, which was prevented by effectively blocking the expression of miR-1224. This study showed that there were 27 differentially expressed miRNAs when PSP inhibits osteoclastogenesis, the most notable of which was miR-1224. Furthermore, the study showed that PSP increased the level of Limd1, which was the target gene of miR-1224. In conclusion, these findings demonstrate that PSP suppressed osteoclastogenesis in vitro through the Hippo signaling pathway based on miR-1224. This study may aid in the development of a therapeutic approach utilizing PSP for the enhancement of bone health and prevention of osteoporosis.


Subject(s)
Macrophages/drug effects , Macrophages/metabolism , MicroRNAs/genetics , Osteoclasts/drug effects , Osteoclasts/metabolism , Polygonatum , Polysaccharides/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Gene Expression/drug effects , Hippo Signaling Pathway , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Osteoclasts/cytology , Osteogenesis/drug effects , Osteogenesis/genetics , Plants, Medicinal , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
5.
Cell Physiol Biochem ; 50(2): 512-524, 2018.
Article in English | MEDLINE | ID: mdl-30308489

ABSTRACT

BACKGROUND/AIMS: miR-136-5p participates in recovery after spinal cord injury (SCI) via an unknown mechanism. We investigated the mechanism underlying the involvement of miR-136-5p in the inflammatory response in a rat model of SCI. METHODS: Sprague-Dawley rat astrocytes were cultured in vitro to construct a reporter plasmid. Luciferase assays were used to detect the ability of miR-136-5p to target the IKKß and A20 genes. Next, recombinant lentiviral vectors were constructed, which either overexpressed miR-136-5p or inhibited its expression. The influence of miR-136-5p overexpression and miR-136-5p silencing on inflammation was observed in vivo in an SCI rat model. The expression of IL-1ß, IL-6, TNF-α, IFN-α, and related proteins (A20, IKKß, and NF-κB) was detected. RESULTS: In vitro studies showed that luciferase activity was significantly activated in the presence of the 3' untranslated region (UTR) region of the IKKß gene after stimulation of cells with miR-136-5p. However, luciferase activity was significantly inhibited in the presence of the 3'UTR region of the A20 gene. Thus, miR-136-5p may act directly on the 3'UTR regions of the IKKß and A20 genes to regulate their expression. miR-136-5p overexpression promoted the production of related cytokines and NF-κB in SCI rats and inhibited the expression of A20 protein. CONCLUSION: Overexpression of miR-136-5p promotes the generation of IL-1ß, IL-6, TNF-α, IFN-α, IKKß, and NF-κB in SCI rats but inhibits the expression of A20. Under these conditions, inflammatory cell infiltration into the rat spinal cord increases and injury is significantly aggravated. Silencing of miR-136-5p significantly reduces the protein expression results described after miR-136-5p overexpression and ameliorates the inflammatory cell infiltration and damage to the spinal cord. Therefore, miR-136-5p might be a new target for the treatment of SCI.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Kinase/metabolism , MicroRNAs/metabolism , NF-kappa B/metabolism , Spinal Cord Injuries/pathology , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Cytokines/analysis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , I-kappa B Kinase/chemistry , I-kappa B Kinase/genetics , Interleukin-1beta/analysis , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , NF-kappa B/genetics , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Spinal Cord Injuries/genetics , Spinal Cord Injuries/veterinary , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/analysis
6.
Neural Plast ; 2018: 9275239, 2018.
Article in English | MEDLINE | ID: mdl-29853851

ABSTRACT

Ischemic long-term potentiation (iLTP) is a form of synaptic plasticity that occurs in acute brain slices following oxygen-glucose deprivation. In vitro, iLTP can occlude physiological LTP (pLTP) through saturation of plasticity mechanisms. We used our murine cardiac arrest and cardiopulmonary resuscitation (CA/CPR) model to produce global brain ischemia and assess whether iLTP is induced in vivo, contributing to the functionally relevant impairment of pLTP. Adult male mice were subjected to CA/CPR, and slice electrophysiology was performed in the hippocampal CA1 region 7 or 30 days later. We observed increased miniature excitatory postsynaptic current amplitudes, suggesting a potentiation of postsynaptic AMPA receptor function after CA/CPR. We also observed increased phosphorylated GluR1 in the postsynaptic density of hippocampi after CA/CPR. These data support the in vivo induction of ischemia-induced plasticity. Application of a low-frequency stimulus (LFS) to CA1 inputs reduced excitatory postsynaptic potentials in slices from mice subjected to CA/CPR, while having no effects in sham controls. These results are consistent with a reversal, or depotentiation, of iLTP. Further, depotentiation with LFS partially restored induction of pLTP with theta burst stimulation. These data provide evidence for iLTP following in vivo ischemia, which occludes pLTP and likely contributes to network disruptions that underlie memory impairments.


Subject(s)
Brain Ischemia/physiopathology , CA1 Region, Hippocampal/physiopathology , Heart Arrest/physiopathology , Long-Term Potentiation , Neurons/physiology , Animals , Brain Ischemia/complications , Heart Arrest/complications , Long-Term Synaptic Depression , Male , Mice, Inbred C57BL , Receptors, AMPA/physiology
7.
J Cereb Blood Flow Metab ; 38(12): 2223-2235, 2018 12.
Article in English | MEDLINE | ID: mdl-29611441

ABSTRACT

Global ischemia in childhood often leads to poor neurologic outcomes, including learning and memory deficits. Using our novel model of childhood cardiac arrest/cardiopulmonary resuscitation (CA/CPR), we investigate the mechanism of ischemia-induced cognitive deficits and recovery. Memory is impaired seven days after juvenile CA/CPR and completely recovers by 30 days. Consistent with this remarkable recovery not observed in adults, hippocampal long-term potentiation (LTP) is impaired 7-14 days after CA/CPR, recovering by 30 days. This recovery is not due to the replacement of dead neurons (neurogenesis), but rather correlates with brain-derived neurotrophic factor (BDNF) expression, implicating BDNF as the molecular mechanism underlying impairment and recovery. Importantly, delayed activation of TrkB receptor signaling reverses CA/CPR-induced LTP deficits and memory impairments. These data provide two new insights (1) endogenous recovery of memory and LTP through development may contribute to improved neurological outcome in children compared to adults and (2) BDNF-enhancing drugs speed recovery from pediatric cardiac arrest during the critical school ages.


Subject(s)
Brain Ischemia/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Recovery of Function/physiology , Animals , Brain Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Signal Transduction/physiology
8.
J Cereb Blood Flow Metab ; 37(8): 3053-3064, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28168893

ABSTRACT

Cardiac arrest and cardiopulmonary resuscitation (CA/CPR) produce brain ischemia that results in cognitive and motor coordination impairments subsequent to injury of vulnerable populations of neurons, including cerebellar Purkinje neurons. To determine the effects of CA/CPR on plasticity in the cerebellum, we used whole cell recordings from Purkinje neurons to examine long-term depression (LTD) at parallel fiber (PF) synapses. Acute slices were prepared from adult male mice subjected to 8 min cardiac arrest at 1, 7, and 30 days after resuscitation. Concurrent stimulation of PF and climbing fibers (CFs) resulted in robust LTD of PF-evoked excitatory postsynaptic currents (EPSCs) in controls. LTD was absent in recordings obtained from mice subjected to CA/CPR, with no change in EPSC amplitude from baseline at any time point tested. AMPA and mGluR-mediated responses at the PF were not altered by CA/CPR. In contrast, CF-evoked NMDA currents were reduced following CA/CPR, which could account for the loss of LTD observed. A loss of GluN1 protein was observed following CA/CPR that was surprisingly not associated with changes in mRNA expression. These data demonstrate sustained impairments in synaptic plasticity in Purkinje neurons that survive the initial injury and which likely contribute to motor coordination impairments observed after CA/CPR.


Subject(s)
Cardiopulmonary Resuscitation , Excitatory Postsynaptic Potentials/physiology , Heart Arrest/physiopathology , Long-Term Synaptic Depression/physiology , Purkinje Cells , Animals , Disease Models, Animal , Heart Arrest/metabolism , Heart Arrest/pathology , Male , Mice, Inbred C57BL , Purkinje Cells/metabolism , Purkinje Cells/physiology , Receptors, Glutamate/metabolism
9.
Cell Rep ; 18(5): 1109-1117, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28147268

ABSTRACT

The Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a major mediator of physiological glutamate signaling, but its role in pathological glutamate signaling (excitotoxicity) remains less clear, with indications for both neuro-toxic and neuro-protective functions. Here, the role of CaMKII in ischemic injury is assessed utilizing our mouse model of cardiac arrest and cardiopulmonary resuscitation (CA/CPR). CaMKII inhibition (with tatCN21 or tatCN19o) at clinically relevant time points (30 min after resuscitation) greatly reduces neuronal injury. Importantly, CaMKII inhibition also works in combination with mild hypothermia, the current standard of care. The relevant drug target is specifically Ca2+-independent "autonomous" CaMKII activity generated by T286 autophosphorylation, as indicated by substantial reduction in injury in autonomy-incompetent T286A mutant mice. In addition to reducing cell death, tatCN19o also protects the surviving neurons from functional plasticity impairments and prevents behavioral learning deficits, even at extremely low doses (0.01 mg/kg), further highlighting the clinical potential of our findings.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Arrest/metabolism , Heart Arrest/physiopathology , Neuroprotection/physiology , Animals , Calcium/metabolism , Cell Death/physiology , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Phosphorylation/physiology
10.
Brain Behav Immun ; 48: 115-22, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25774010

ABSTRACT

Despite dramatic improvement in cardiopulmonary resuscitation (CPR) and other techniques for cardiac arrest (CA), the majority of survivors continue to show signs of decreased memory or executive cognitive function. Such memory impairment may be due to hippocampal CA1 neuronal death, which is delayed by several days after CA/CPR. Classical microgliosis in the CA1 region may contribute to neuronal death, yet the role of a key activation receptor Toll Like Receptor 4 (TLR4) has not been previously investigated for such neuronal death after CA/CPR. We show that (+)-naltrexone was neuroprotective after CA/CPR. TLR4 blockade was associated with decreased expression of markers for microglial/macrophage activation and T cell and B cell infiltration, as well as decreased pro-inflammatory cytokine levels. Notably, IL-10 expression was elevated in response to CA/CPR, but was not attenuated by (+)-naltrexone, suggesting that the local monocyte/microglial phenotype had shifted towards alternative activation. This was confirmed by elevated expression of Arginase-1, and decreased expression of NFκB p65 subunit. Thus, (+)-naltrexone and other TLR4 antagonists may represent a novel therapeutic strategy to alleviate the substantial burden of memory or executive cognitive function impairment after CA/CPR.


Subject(s)
Cell Death/drug effects , Heart Arrest/pathology , Hippocampus/drug effects , Naltrexone/pharmacology , Neuroprotective Agents/pharmacology , Animals , Cardiopulmonary Resuscitation , Heart Arrest/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Interleukin-10/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology
11.
J Neuroimmunol ; 274(1-2): 132-40, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25084739

ABSTRACT

Although inflammatory mechanisms have been linked to neuronal injury following global cerebral ischemia, the presence of infiltrating peripheral immune cells remains understudied. We performed flow cytometry of single cell suspensions obtained from the brains of mice at varying time points after global cerebral ischemia induced by cardiac arrest and cardiopulmonary resuscitation (CA/CPR) to characterize the influx of lymphocytes into the injured brain. We observed that CA/CPR caused a large influx of lymphocytes within 3h of resuscitation that was maintained for the 3day duration of our experiments. Using cell staining flow cytometry we observed that the large majority of infiltrating lymphocytes were CD4(+) T cells. Intracellular stains revealed a large proportion of pro-inflammatory T cells expressing either TNFα or INFγ. Importantly, the lack of functional T cells in TCRα knockout mice reduced neuronal injury following CA/CPR, implicating pro-inflammatory T cells in the progression of ischemic neuronal injury. Finally, we made the remarkable observation that the novel CD4(+)CD40(+) (Th40) population of pro-inflammatory T cells that are strongly associated with autoimmunity are present in large numbers in the injured brain. These data indicate that studies investigating the neuro-immune response after global cerebral ischemia should consider the role of infiltrating T cells in orchestrating the acute and sustained immune response.


Subject(s)
Brain Ischemia/immunology , CD4-Positive T-Lymphocytes/immunology , Cardiopulmonary Resuscitation , Heart Arrest/immunology , Heart Arrest/therapy , Animals , Brain Ischemia/pathology , CD4-Positive T-Lymphocytes/cytology , CD40 Antigens/immunology , Cell Movement/immunology , Hippocampus/immunology , Hippocampus/pathology , Immunophenotyping , Male , Mice , Mice, Inbred C57BL
12.
Exp Neurol ; 259: 10-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24512750

ABSTRACT

Cerebral ischemia caused by loss of blood supply to the brain during cardiac arrest or stroke are major causes of death and disability. Biological sex is an important factor in predicting vulnerability of the brain to an ischemic insult, with males being at higher risk for cardio-cerebrovascular events than females of the same age. However, relative incidence of stroke between the genders appears to normalize at advanced ages. Therefore, many scientists have focused on the mechanisms of sex differences in outcome following brain ischemic injury, with a particular emphasis on the role of sex steroids. The majority of studies indicate that female sex steroids, such as estrogen and progesterone, play important roles in the relative neuroprotection following cerebral ischemia observed in females. However, less is known about male sex steroids and brain damage. This review describes the state of our knowledge of androgen-related contributions to neurological injury and recovery following cerebral ischemia that occurs following stroke. Experimental studies examining the effects of castration, androgenic agonists and antagonists and aging provide valuable insights into the role of androgens in clinical outcome following cerebrovascular events.


Subject(s)
Androgens/physiology , Androgens/therapeutic use , Stroke/physiopathology , Adult , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Child , Disease Models, Animal , Female , Humans , Male , Recovery of Function , Sex Characteristics , Stroke/genetics
SELECTION OF CITATIONS
SEARCH DETAIL