1.Effect of retinoic acid on delayed encephalopathy after acute carbon monoxide poisoning: Role of the lncRNA SNHG15/LINGO-1/BDNF/TrkB axis.
Fangling HUANG ; Su'e WANG ; Zhengrong PENG ; Xu HUANG ; Sufen BAI
Journal of Central South University(Medical Sciences) 2025;50(6):955-969
OBJECTIVES:
The neurotoxicity of carbon monoxide (CO) to the central nervous system is a key pathogenesis of delayed encephalopathy after acute carbon monoxide poisoning (DEACMP). Our previous study found that retinoic acid (RA) can suppress the neurotoxic effects of CO. This study further explores, in vivo and in vitro, the molecular mechanisms by which RA alleviates CO-induced central nervous system damage.
METHODS:
A cytotoxic model was established using the mouse hippocampal neuronal cell line HT22 and primary oligodendrocytes exposed to CO, and a DEACMP animal model was established in adult Kunming mice. Cell viability and apoptosis of hippocampal neurons and oligodendrocytes were assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and Annexin V/propidium iodide (PI) double staining. The transcriptional and protein expression of each gene was detected using real-time fluorescence quantitative PCR (RT-qPCR) and Western blotting. Long noncoding RNA (lncRNA) SNHG15 and LINGO-1 were knocked down or overexpressed to observe changes in neurons and oligodendrocytes. In DEACMP mice, SNHG15 or LINGO-1 were knocked down to assess changes in central nervous tissue and downstream protein expression.
RESULTS:
RA at 10 and 20 μmol/L significantly reversed CO-induced apoptosis of hippocampal neurons and oligodendrocytes, downregulation of SNHG15 and LINGO-1, and upregulation of brain-derived neurotrophic factor (BDNF) and tyrosine kinase receptor B (TrkB) (all P<0.05). Overexpression of SNHG15 or LINGO-1 weakened the protective effect of RA against CO-induced cytotoxicity (all P<0.05). Knockdown of SNHG15 or LINGO-1 alleviated CO-induced apoptosis of hippocampal neurons and oligodendrocytes and upregulated BDNF and TrkB expression levels (all P<0.05). Experiments in DEACMP model mice showed that knockdown of SNHG15 or LINGO-1 mitigated central nervous system injury in DEACMP (all P<0.05).
CONCLUSIONS
RA alleviates CO-induced apoptosis of hippocampal neurons and oligodendrocytes, thereby reducing central nervous system injury and exerting neuroprotective effects. LncRNA SNHG15 and LINGO-1 are key molecules mediating RA-induced inhibition of neuronal apoptosis and are associated with the BDNF/TrkB pathway. These findings provide a theoretical framework for optimizing the clinical treatment of DEACMP and lay an experimental foundation for elucidating its molecular mechanisms.
Animals
;
RNA, Long Noncoding/physiology*
;
Brain-Derived Neurotrophic Factor/genetics*
;
Carbon Monoxide Poisoning/complications*
;
Mice
;
Tretinoin/pharmacology*
;
Nerve Tissue Proteins/metabolism*
;
Membrane Proteins/metabolism*
;
Apoptosis/drug effects*
;
Hippocampus/cytology*
;
Receptor, trkB/metabolism*
;
Neurons/drug effects*
;
Male
;
Brain Diseases/etiology*
;
Oligodendroglia/drug effects*
;
Signal Transduction
;
Cell Line
2.The Medial Prefrontal Cortex-Basolateral Amygdala Circuit Mediates Anxiety in Shank3 InsG3680 Knock-in Mice.
Jiabin FENG ; Xiaojun WANG ; Meidie PAN ; Chen-Xi LI ; Zhe ZHANG ; Meng SUN ; Tailin LIAO ; Ziyi WANG ; Jianhong LUO ; Lei SHI ; Yu-Jing CHEN ; Hai-Feng LI ; Junyu XU
Neuroscience Bulletin 2025;41(1):77-92
Anxiety disorder is a major symptom of autism spectrum disorder (ASD) with a comorbidity rate of ~40%. However, the neural mechanisms of the emergence of anxiety in ASD remain unclear. In our study, we found that hyperactivity of basolateral amygdala (BLA) pyramidal neurons (PNs) in Shank3 InsG3680 knock-in (InsG3680+/+) mice is involved in the development of anxiety. Electrophysiological results also showed increased excitatory input and decreased inhibitory input in BLA PNs. Chemogenetic inhibition of the excitability of PNs in the BLA rescued the anxiety phenotype of InsG3680+/+ mice. Further study found that the diminished control of the BLA by medial prefrontal cortex (mPFC) and optogenetic activation of the mPFC-BLA pathway also had a rescue effect, which increased the feedforward inhibition of the BLA. Taken together, our results suggest that hyperactivity of the BLA and alteration of the mPFC-BLA circuitry are involved in anxiety in InsG3680+/+ mice.
Animals
;
Prefrontal Cortex/metabolism*
;
Basolateral Nuclear Complex/metabolism*
;
Mice
;
Anxiety/metabolism*
;
Nerve Tissue Proteins/genetics*
;
Male
;
Gene Knock-In Techniques
;
Pyramidal Cells/physiology*
;
Mice, Transgenic
;
Neural Pathways/physiopathology*
;
Mice, Inbred C57BL
;
Microfilament Proteins
3.Reprogramming miR-146b-snphb Signaling Activates Axonal Mitochondrial Transport in the Zebrafish M-cell and Facilitates Axon Regeneration After Injury.
Xin-Liang WANG ; Zong-Yi WANG ; Xing-Han CHEN ; Yuan CAI ; Bing HU
Neuroscience Bulletin 2025;41(4):633-648
Acute mitochondrial damage and the energy crisis following axonal injury highlight mitochondrial transport as an important target for axonal regeneration. Syntaphilin (Snph), known for its potent mitochondrial anchoring action, has emerged as a significant inhibitor of both mitochondrial transport and axonal regeneration. Therefore, investigating the molecular mechanisms that influence the expression levels of the snph gene can provide a viable strategy to regulate mitochondrial trafficking and enhance axonal regeneration. Here, we reveal the inhibitory effect of microRNA-146b (miR-146b) on the expression of the homologous zebrafish gene syntaphilin b (snphb). Through CRISPR/Cas9 and single-cell electroporation, we elucidated the positive regulatory effect of the miR-146b-snphb axis on Mauthner cell (M-cell) axon regeneration at the global and single-cell levels. Through escape response tests, we show that miR-146b-snphb signaling positively regulates functional recovery after M-cell axon injury. In addition, continuous dynamic imaging in vivo showed that reprogramming miR-146b significantly promotes axonal mitochondrial trafficking in the pre-injury and early stages of regeneration. Our study reveals an intrinsic axonal regeneration regulatory axis that promotes axonal regeneration by reprogramming mitochondrial transport and anchoring. This regulation involves noncoding RNA, and mitochondria-associated genes may provide a potential opportunity for the repair of central nervous system injury.
Animals
;
Zebrafish
;
MicroRNAs/genetics*
;
Nerve Regeneration/physiology*
;
Mitochondria/metabolism*
;
Zebrafish Proteins/genetics*
;
Axons/metabolism*
;
Signal Transduction/physiology*
;
Axonal Transport/physiology*
;
Nerve Tissue Proteins/genetics*
4.Connexin43 Modulates X-Ray-Induced Pyroptosis in Human Umbilical Vein Endothelial Cells.
Chen LI ; Mei TIAN ; Qiao GOU ; Yong Rui JIA ; Xu SU
Biomedical and Environmental Sciences 2019;32(3):177-188
OBJECTIVE:
Pyroptosis is an inflammatory form of programmed cell death. This phenomenon has been recently reported to play an important role in radiation-induced normal tissue injury. Connexin43 (Cx43) is a gap junction protein that regulates cell growth and apoptosis. In this study, we investigated the effect of Cx43 on X-ray-induced pyroptosis in the human umbilical vein endothelial cells (HUVECs).
METHODS:
HUVECs, Cx43 overexpression, and Cx43 knockdown strains were irradiated with 10 Gy. Proteins were detected using western blot analysis. Cell pyroptosis was evaluated using the fluorescence-labeled inhibitor of caspase assay (FLICA) and propidium iodide staining through flow cytometry and confocal microscopy. Cell morphology and cytotoxicity were detected by scanning electron microscopy and lactate dehydrogenase release assay, respectively.
RESULTS:
Irradiation with 10 Gy X-ray induced pyroptosis in the HUVECs and reduced Cx43 expression. The pyroptosis in the HUVECs was significantly attenuated by overexpression of Cx43 as it decreased the level of active caspase-1. However, interference of Cx43 expression with siRNA significantly promoted pyroptosis by increasing the active caspase-1 level. Pannexin1 (Panx1), a gap junction protein regulates pyroptosis, and its cleaved form is used to evaluate channel opening and active state. The level of cleaved Panx1 in the HUVECs and Cx43 knockdown strains increased in the presence of X-ray, but decreased in the Cx43 overexpression strains. Furthermore, interference of Panx1 with siRNA alleviated the upregulation of pyroptosis caused by Cx43 knockdown.
CONCLUSION
Results suggest that single high-dose X-ray irradiation induces pyroptosis in the HUVECs. In addition, Cx43 regulates pyroptosis directly by activating caspase-1 or indirectly by cleaving Panx1.
Caspase 1
;
genetics
;
metabolism
;
Connexin 43
;
genetics
;
metabolism
;
Connexins
;
genetics
;
metabolism
;
Gene Expression Regulation
;
radiation effects
;
Human Umbilical Vein Endothelial Cells
;
physiology
;
radiation effects
;
Humans
;
Nerve Tissue Proteins
;
genetics
;
metabolism
;
Pyroptosis
;
X-Rays
;
adverse effects
5.Mutant Huntingtin Causes a Selective Decrease in the Expression of Synaptic Vesicle Protein 2C.
Chaohua PENG ; Gaochun ZHU ; Xiangqian LIU ; He LI
Neuroscience Bulletin 2018;34(5):747-758
Huntington's disease (HD) is a neurodegenerative disease caused by a polyglutamine expansion in the huntingtin (Htt) protein. Mutant Htt causes synaptic transmission dysfunctions by interfering in the expression of synaptic proteins, leading to early HD symptoms. Synaptic vesicle proteins 2 (SV2s), a family of synaptic vesicle proteins including 3 members, SV2A, SV2B, and SV2C, plays important roles in synaptic physiology. Here, we investigated whether the expression of SV2s is affected by mutant Htt in the brains of HD transgenic (TG) mice and Neuro2a mouse neuroblastoma cells (N2a cells) expressing mutant Htt. Western blot analysis showed that the protein levels of SV2A and SV2B were not significantly changed in the brains of HD TG mice expressing mutant Htt with 82 glutamine repeats. However, in the TG mouse brain there was a dramatic decrease in the protein level of SV2C, which has a restricted distribution pattern in regions particularly vulnerable in HD. Immunostaining revealed that the immunoreactivity of SV2C was progressively weakened in the basal ganglia and hippocampus of TG mice. RT-PCR demonstrated that the mRNA level of SV2C progressively declined in the TG mouse brain without detectable changes in the mRNA levels of SV2A and SV2B, indicating that mutant Htt selectively inhibits the transcriptional expression of SV2C. Furthermore, we found that only SV2C expression was progressively inhibited in N2a cells expressing a mutant Htt containing 120 glutamine repeats. These findings suggest that the synaptic dysfunction in HD results from the mutant Htt-mediated inhibition of SV2C transcriptional expression. These data also imply that the restricted distribution and decreased expression of SV2C contribute to the brain region-selective pathology of HD.
Aging
;
metabolism
;
Animals
;
Brain
;
metabolism
;
pathology
;
Cell Line, Tumor
;
Gene Expression
;
physiology
;
Huntingtin Protein
;
genetics
;
metabolism
;
Membrane Glycoproteins
;
metabolism
;
Mice
;
Mice, Transgenic
;
Mutation
;
Nerve Tissue Proteins
;
metabolism
;
RNA, Messenger
;
metabolism
;
Transcription, Genetic
;
physiology
6.Inhibition of KLF7-Targeting MicroRNA 146b Promotes Sciatic Nerve Regeneration.
Wen-Yuan LI ; Wei-Ting ZHANG ; Yong-Xia CHENG ; Yan-Cui LIU ; Feng-Guo ZHAI ; Ping SUN ; Hui-Ting LI ; Ling-Xiao DENG ; Xiao-Feng ZHU ; Ying WANG
Neuroscience Bulletin 2018;34(3):419-437
A previous study has indicated that Krüppel-like factor 7 (KLF7), a transcription factor that stimulates Schwann cell (SC) proliferation and axonal regeneration after peripheral nerve injury, is a promising therapeutic transcription factor in nerve injury. We aimed to identify whether inhibition of microRNA-146b (miR-146b) affected SC proliferation, migration, and myelinated axon regeneration following sciatic nerve injury by regulating its direct target KLF7. SCs were transfected with miRNA lentivirus, miRNA inhibitor lentivirus, or KLF7 siRNA lentivirus in vitro. The expression of miR146b and KLF7, as well as SC proliferation and migration, were subsequently evaluated. In vivo, an acellular nerve allograft (ANA) followed by injection of GFP control vector or a lentiviral vector encoding an miR-146b inhibitor was used to assess the repair potential in a model of sciatic nerve gap. miR-146b directly targeted KLF7 by binding to the 3'-UTR, suppressing KLF7. Up-regulation of miR-146b and KLF7 knockdown significantly reduced the proliferation and migration of SCs, whereas silencing miR-146b resulted in increased proliferation and migration. KLF7 protein was localized in SCs in which miR-146b was expressed in vivo. Similarly, 4 weeks after the ANA, anti-miR-146b increased KLF7 and its target gene nerve growth factor cascade, promoting axonal outgrowth. Closer analysis revealed improved nerve conduction and sciatic function index score, and enhanced expression of neurofilaments, P0 (anti-peripheral myelin), and myelinated axon regeneration. Our findings provide new insight into the regulation of KLF7 by miR-146b during peripheral nerve regeneration and suggest a potential therapeutic strategy for peripheral nerve injury.
Animals
;
Cell Movement
;
genetics
;
Cell Proliferation
;
genetics
;
Disease Models, Animal
;
Female
;
Ganglia, Spinal
;
cytology
;
Gene Expression Regulation
;
genetics
;
physiology
;
HEK293 Cells
;
Humans
;
Kruppel-Like Transcription Factors
;
genetics
;
metabolism
;
Male
;
MicroRNAs
;
genetics
;
metabolism
;
Motor Endplate
;
genetics
;
Myelin P0 Protein
;
metabolism
;
Nerve Regeneration
;
genetics
;
physiology
;
Nerve Tissue Proteins
;
metabolism
;
RNA, Small Interfering
;
genetics
;
metabolism
;
Rats
;
Rats, Sprague-Dawley
;
Rats, Wistar
;
Sciatic Neuropathy
;
metabolism
;
surgery
;
therapy
7.Recapitulating cortical development with organoid culture in vitro and modeling abnormal spindle-like (ASPM related primary) microcephaly disease.
Rui LI ; Le SUN ; Ai FANG ; Peng LI ; Qian WU ; Xiaoqun WANG
Protein & Cell 2017;8(11):823-833
The development of a cerebral organoid culture in vitro offers an opportunity to generate human brain-like organs to investigate mechanisms of human disease that are specific to the neurogenesis of radial glial (RG) and outer radial glial (oRG) cells in the ventricular zone (VZ) and subventricular zone (SVZ) of the developing neocortex. Modeling neuronal progenitors and the organization that produces mature subcortical neuron subtypes during early stages of development is essential for studying human brain developmental diseases. Several previous efforts have shown to grow neural organoid in culture dishes successfully, however we demonstrate a new paradigm that recapitulates neocortical development process with VZ, OSVZ formation and the lamination organization of cortical layer structure. In addition, using patient-specific induced pluripotent stem cells (iPSCs) with dysfunction of the Aspm gene from a primary microcephaly patient, we demonstrate neurogenesis defects result in defective neuronal activity in patient organoids, suggesting a new strategy to study human developmental diseases in central nerve system.
Action Potentials
;
physiology
;
Biomarkers
;
metabolism
;
Cell Culture Techniques
;
Embryoid Bodies
;
cytology
;
metabolism
;
Gene Expression
;
Humans
;
Induced Pluripotent Stem Cells
;
cytology
;
metabolism
;
Lateral Ventricles
;
cytology
;
growth & development
;
metabolism
;
Microcephaly
;
genetics
;
metabolism
;
pathology
;
Models, Biological
;
Mutation
;
Neocortex
;
cytology
;
growth & development
;
metabolism
;
Nerve Tissue Proteins
;
deficiency
;
genetics
;
Neurogenesis
;
genetics
;
Neurons
;
cytology
;
metabolism
;
Organoids
;
cytology
;
metabolism
;
PAX6 Transcription Factor
;
genetics
;
metabolism
;
Patch-Clamp Techniques
;
SOXB1 Transcription Factors
;
genetics
;
metabolism
;
Zonula Occludens-1 Protein
;
genetics
;
metabolism
8.Restoration of Brain Acid Soluble Protein 1 Inhibits Proliferation and Migration of Thyroid Cancer Cells.
Run-Sheng GUO ; Yue YU ; Jun CHEN ; Yue-Yu CHEN ; Na SHEN ; Ming QIU
Chinese Medical Journal 2016;129(12):1439-1446
BACKGROUNDBrain acid soluble protein 1 (BASP1) is identified as a novel potential tumor suppressor in several cancers. However, its role in thyroid cancer has not been investigated yet. In the present study, the antitumor activities of BASP1 against the growth and migration of thyroid cancer cells were evaluated.
METHODSBASP1 expression in thyroid cancer tissues and normal tissues were examined by immunohistochemical staining and the association between its expression and prognosis was analyzed. pcDNA-BASP1 carrying full length of BASP1 cDNA was constructed to restore the expression of BASP1 in thyroid cancer cell lines (BHT-101 and KMH-2). The cell proliferation in vitro and in vivo was evaluated by WST-1 assay and xenograft tumor models, respectively. Cell cycle distribution after transfection was analyzed using flow cytometry. Cell apoptosis after transfection was examined by annexin V/propidium iodide assay. The migration was examined using transwell assay.
RESULTSBASP1 expression was abundant in normal tissues while it is significantly decreased in cancer tissues (P = 0.000). pcDNA-BASP1 restored the expression of BASP1 and significantly inhibited the growth of BHT-101 and KMH-2 cells as well as xenograft tumors in nude mice (P = 0.000). pcDNA-BASP1 induced G1 arrest and apoptosis in BHT-101 and KMH-2 cells. In addition, pcDNA-BASP1 significantly inhibited the cell migration.
CONCLUSIONSDownregulation of BASP1 expression may play a role in the tumorigenesis of thyroid cancer. Restoration of BASP1 expression exerted extensive antitumor activities against growth and migration of thyroid cancer cells, which suggested that BASP1 gene might act as a potential therapeutic agent for the treatment of thyroid cancer.
Aged ; Animals ; Apoptosis ; genetics ; physiology ; Calmodulin-Binding Proteins ; genetics ; metabolism ; Cell Cycle ; genetics ; physiology ; Cell Line, Tumor ; Cell Movement ; genetics ; physiology ; Cell Proliferation ; genetics ; physiology ; Cytoskeletal Proteins ; genetics ; metabolism ; Female ; Gene Expression Regulation, Neoplastic ; genetics ; physiology ; Humans ; Male ; Membrane Proteins ; genetics ; metabolism ; Mice ; Mice, Nude ; Middle Aged ; Nerve Tissue Proteins ; genetics ; metabolism ; Repressor Proteins ; genetics ; metabolism ; Thyroid Neoplasms ; metabolism ; pathology ; Xenograft Model Antitumor Assays
9.Molecular Mechanism of Action of hnRNP K and RTN3 in the Replication of Enterovirus 71.
Li LI ; Haiyan ZHONG ; Mao FAN ; Liyue KUI ; Huiying LI ; Jianying ZHANG
Chinese Journal of Virology 2015;31(2):197-200
Enterovirus 71 (EV71) is a neurotropic pathogen that can induce hand, foot and mouth disease in children. There is an appreciable mortality rate after EV71 infections. The mechanism of action of EV71 replication is not known. Recent work has identified some of cell factors of the host that participate in the synthesis of the RNA and proteins of EV71 (e.g., hnRNP K, reticulon 3 (RTN 3)). In that work, researchers used a competitive assay to show that hnRNP K can interact with EV71 5' UTR, which is required for efficient synthesis of viral RNA. Using a yeast two-hybrid system, other researchers demonstrated that RTN 3 interacts with the N-terminal domain of EV71 2C, which is crucial for replication of viral RNA. Here, we discuss recent work focusing on the molecular mechanisms of hnRNP K and RTN 3 in the synthesis of the RNA and proteins of EV71.
Animals
;
Carrier Proteins
;
genetics
;
metabolism
;
Enterovirus A, Human
;
genetics
;
physiology
;
Enterovirus Infections
;
genetics
;
metabolism
;
virology
;
Heterogeneous-Nuclear Ribonucleoprotein K
;
Host-Pathogen Interactions
;
Humans
;
Membrane Proteins
;
genetics
;
metabolism
;
Nerve Tissue Proteins
;
genetics
;
metabolism
;
Ribonucleoproteins
;
genetics
;
metabolism
;
Viral Proteins
;
genetics
;
metabolism
;
Virus Replication
10.USP33, a new player in lung cancer, mediates Slit-Robo signaling.
Pushuai WEN ; Ruirui KONG ; Jianghong LIU ; Li ZHU ; Xiaoping CHEN ; Xiaofei LI ; Yongzhan NIE ; Kaichun WU ; Jane Y WU
Protein & Cell 2014;5(9):704-713
Ubiquitin specific protease 33 (USP33) is a multifunctional protein regulating diverse cellular processes. The expression and role of USP33 in lung cancer remain unexplored. In this study, we show that USP33 is down-regulated in multiple cohorts of lung cancer patients and that low expression of USP33 is associated with poor prognosis. USP33 mediates Slit-Robo signaling in lung cancer cell migration. Downregulation of USP33 reduces the protein stability of Robo1 in lung cancer cells, providing a previously unknown mechanism for USP33 function in mediating Slit activity in lung cancer cells. Taken together, USP33 is a new player in lung cancer that regulates Slit-Robo signaling. Our data suggest that USP33 may be a candidate tumor suppressor for lung cancer with potential as a prognostic marker.
Blotting, Western
;
Cell Line, Tumor
;
Cell Movement
;
genetics
;
physiology
;
Cohort Studies
;
Down-Regulation
;
Female
;
Gene Expression Regulation, Neoplastic
;
HEK293 Cells
;
Humans
;
Immunohistochemistry
;
Intercellular Signaling Peptides and Proteins
;
metabolism
;
Kaplan-Meier Estimate
;
Lung Neoplasms
;
genetics
;
metabolism
;
pathology
;
Male
;
Middle Aged
;
Nerve Tissue Proteins
;
metabolism
;
Prognosis
;
RNA Interference
;
Receptors, Immunologic
;
metabolism
;
Reverse Transcriptase Polymerase Chain Reaction
;
Signal Transduction
;
genetics
;
physiology
;
Ubiquitin Thiolesterase
;
genetics
;
metabolism

Result Analysis
Print
Save
E-mail