Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Adv Biol (Weinh) ; 8(2): e2300453, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37957539

ABSTRACT

Accumulating evidence indicates that cellular premature senescence of the glomerulus, including endothelial cells, mesangial cells, and podocytes leads to diabetic nephropathy (DN), and DN is regarded as a clinical model of premature senescence. However, the role of cellular senescence-associated genes in the glomerulus in DN progression remains unclear. Therefore, this work aims to identify and validate potential cellular aging-related genes in the glomerulus in DN to provide novel clues for DN treatment based on anti-aging. The microarray GSE96804 dataset, including 41 diabetic glomeruli and 20 control glomeruli, is retrieved from the Gene Expression Omnibus (GEO) database and cellular senescence-related genes (CSRGs) are obtained from the GeneCards database and literature reports. Subsequently, PPI, GO, and KEGG enrichment are analyzed by screening the intersection between differentially expressed genes (DEGs) and CSRGs. scRNA-seq dataset GSE127235 is used to verify core genes expression in glomerulocytes of mice. Finally, db/db mice are utilized to validate the hub gene expression in the glomeruli, and high glucose-induced mesangial cells are used to confirm key gene expression. This study reveals that FOS and ZFP36 may play an anti-aging role in DN to ameliorate cell intracellular premature aging in mesangial cells of glomeruli.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Mice , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/prevention & control , Multiomics , Endothelial Cells/metabolism , Kidney Glomerulus/metabolism , Mice, Inbred Strains , Cellular Senescence/genetics , Diabetes Mellitus/metabolism
2.
Anticancer Res ; 43(12): 5509-5522, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38030193

ABSTRACT

BACKGROUND/AIM: It is generally accepted that low-dose metronomic (LDM) chemotherapy mostly exerts its antitumor effects by inhibiting tumor angiogenesis. However, there is some evidence that LDM chemotherapy subsequently promotes tumor angiogenesis under certain regimens in animal models. The mechanisms responsible for these contradictory results are unclear. MATERIALS AND METHODS: Cisplatin (CDDP) was intraperitoneally administered to tumor-bearing mice at doses of 0.05-3 mg/kg every other day. The effects of LDM chemotherapy with CDDP on tumor growth and angiogenesis were observed. To determine the involved mechanisms, we analyzed the expression of vascular basement membrane proteins, transcription of angiogenesis-related genes in tumor tissues, and mobilization of proangiogenic bone marrow-derived cells (BMDCs) in circulating blood. RESULTS: The mean tumor weight with the 3 mg/kg q.o.d. regimen CDDP was significantly lower (by 57.3%) in the CDDP than in the control group. However, the tumor weight was 52.1% higher for the 0.19 mg/kg q.o.d. regimen in the CDDP group, which could be antagonized using 30 mg/kg all-trans retinoic acid. For the 0.19 mg/kg q.o.d., more tumor vascular structures were observed in the CDDP than in the control group (47.9±5.0 vs. 22.3±0.8, p<0.001). The mobilization of VEGFR2+ BMDCs and the mRNA expression of the proangiogenic genes MMP9, VEGFR1, VEGFR2 and VE-cadherin were increased in the 0.19 mg/kg regimen. CONCLUSION: These results indicate that metronomic CDDP promoted tumor angiogenesis and tumor growth via increased mobilization of proangiogenic BMDCs at certain low doses. This implies a potential therapeutic risk from an inappropriate LDM chemotherapy dosage and suggests that optimizing the LDM chemotherapy regimen is urgently needed.


Subject(s)
Cisplatin , Neoplasms , Animals , Mice , Bone Marrow/pathology , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Endothelial Cells
3.
Biomolecules ; 13(5)2023 04 27.
Article in English | MEDLINE | ID: mdl-37238624

ABSTRACT

The neuron loss caused by the progressive damage to the nervous system is proposed to be the main pathogenesis of neurodegenerative diseases. Ependyma is a layer of ciliated ependymal cells that participates in the formation of the brain-cerebrospinal fluid barrier (BCB). It functions to promotes the circulation of cerebrospinal fluid (CSF) and the material exchange between CSF and brain interstitial fluid. Radiation-induced brain injury (RIBI) shows obvious impairments of the blood-brain barrier (BBB). In the neuroinflammatory processes after acute brain injury, a large amount of complement proteins and infiltrated immune cells are circulated in the CSF to resist brain damage and promote substance exchange through the BCB. However, as the protective barrier lining the brain ventricles, the ependyma is extremely vulnerable to cytotoxic and cytolytic immune responses. When the ependyma is damaged, the integrity of BCB is destroyed, and the CSF flow and material exchange is affected, leading to brain microenvironment imbalance, which plays a vital role in the pathogenesis of neurodegenerative diseases. Epidermal growth factor (EGF) and other neurotrophic factors promote the differentiation and maturation of ependymal cells to maintain the integrity of the ependyma and the activity of ependymal cilia, and may have therapeutic potential in restoring the homeostasis of the brain microenvironment after RIBI or during the pathogenesis of neurodegenerative diseases.


Subject(s)
Brain Injuries , Neurodegenerative Diseases , Humans , Ependyma/metabolism , Ependyma/pathology , Nerve Growth Factors/metabolism , Neurodegenerative Diseases/metabolism , Brain/metabolism , Brain Injuries/metabolism
4.
IUBMB Life ; 75(6): 530-547, 2023 06.
Article in English | MEDLINE | ID: mdl-36629313

ABSTRACT

Development of the hippocampus is critical for its normal maturation. However, there is no systematic study on the effects of low-dose (≤2 Gy) neonatal X-ray exposure on different cells at different developmental stages of the mouse hippocampus. The present study demonstrated that irradiation with 2 Gy at postnatal day (PD) 3 in mice induced anxiety and impairment of spatial learning and memory in adult mice. Neuroinflammatory cells were observed in the dentate gyrus (DG) and CA3 areas of the hippocampus at PD3 + 1. X-ray irradiation impaired neuronal complexity and neurogenesis. However, the number of astrocytes and microglia in the hippocampus was increased the first day after irradiation, and then decreased 21 days later. The protein expression levels of NF-κB, C/EBP homologous protein (CHOP), and γH2 A histone family member X (γH2 AX) increased from 7 to 21 days after irradiation, or till 90 days after irradiation for IL-1ß, whereas those of hippocampal sirtuin1 (SIRT1) decreased after 21 days of irradiation at PD3. These results suggest that neonatal X-ray irradiation-induced neuroinflammation impaired neuroplasticity and neurogenesis in the hippocampus, leading to the anxiety and spatial memory disorder during adulthood. The mechanisms involved in the induction of developmental neurotoxicity following low-dose irradiation may involve the inflammation-mediated signaling pathway IL-1ß/ SIRT1/CHOP.


Subject(s)
Hippocampus , Sirtuin 1 , Mice , Animals , X-Rays , Hippocampus/physiology , Neurogenesis , Neurons , Mice, Inbred C57BL
5.
Biomedicines ; 10(12)2022 Nov 23.
Article in English | MEDLINE | ID: mdl-36551767

ABSTRACT

OBJECTIVE: To investigate microRNA (miRNA) expression profiles before and after pilocarpine-induced status epilepticus (SE) in the cornu ammonis (CA) and dentated gyrus (DG) areas of the mouse hippocampus, and to predict the downstream proteins and related pathways based on bioinformatic analysis. METHODS: An epileptic mouse model was established using a pilocarpine injection. Brain tissues from the CA and DG were collected separately for miRNA analysis. The miRNAs were extracted using a kit, and the expression profiles were generated using the SurePrint G3 Mouse miRNA microarray and validated. The intersecting genes of TargetScan and miRanda were selected to predict the target genes of each miRNA. For gene ontology (GO) studies, the parent-child-intersection (pci) method was used for enrichment analysis, and Benjamini-Hochberg was used for multiple test correction. The Kyoto Encyclopedia of Genes and Genomes (KEGG) was used to detect disease-related pathways among the large list of miRNA-targeted genes. All analyses mentioned above were performed at the time points of control, days 3, 14, and 60 post-SE. RESULTS: Control versus days 3, 14, and 60 post-SE: in the CA area, a total of 131 miRNAs were differentially expressed; 53, 49, and 26 miRNAs were upregulated and 54, 10, and 22 were downregulated, respectively. In the DG area, a total of 171 miRNAs were differentially expressed; furthermore, 36, 32, and 28 miRNAs were upregulated and 78, 58, and 44 were downregulated, respectively. Of these, 92 changed in both the CA and DG, 39 only in the CA, and 79 only in the DG area. The differentially expressed miRNAs target 11-1630 genes. Most of these proteins have multiple functions in epileptogenesis. There were 15 common pathways related to altered miRNAs: nine different pathways in the CA and seven in the DG area. CONCLUSIONS: Stage- and subfield-associated hippocampal miRNA expression patterns are closely related to epileptogenesis, although the detailed mechanisms need to be explored in the future.

6.
Life Sci ; 310: 121089, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36270423

ABSTRACT

AIMS: There is emerging evidence that antineoplastic agents and the cytotoxic effects on tumor tissues attenuate the benefits of chemotherapy due to tumor microenvironment changes. Nevertheless, the actual relationship between chemotherapy and recurrent tumors in which the genotypes differ from the original tumor after chemotherapy is unclear. MATERIALS AND METHODS: Bone marrow transplantation, flow cytometer, immune inhibition and immunofluorescence will be utilized to investigate the effect of antineoplastic agents on bone-marrow-derived cells (BMDCs) release and recruitment, and to explore the pathways and mechanisms of antineoplastic agents in promoting tumor growth. KEY FINDINGS: Tumor growth and angiogenesis were significantly enhanced in the mouse model after treatment with antineoplastic agents such as cyclophosphamide, 5-fluorouracil, or cisplatin, along with large increases in proangiogenic vascular endothelial growth factor receptor-2 (VEGFR2+), ß3+, CD11b+Gr-1+, and VEGFR2+ß3+ BMDCs in circulating blood. BMDC recruitment and VEGFR2 and ß3 mRNA transcription in tumor tissues were also enhanced by antineoplastic agents. Antineoplastic-agent-treated BMDCs markedly augmented tumor and endothelial cell proliferation, and ß3 mRNA transcription in endothelial cells (ECs). SIGNIFICANCE: The results suggested that antineoplastic-agent treatment augmented the tumor microenvironment by mobilizing proangiogenic BMDCs, enhancing BMDC recruitment and angiogenesis, and increasing BMDC-mediated tumor and EC functions. These results led to tumor growth and angiogenesis aggravation. It is paramount to elucidate the potential mechanism by which the cellular and molecular effects triggered by the antineoplastic agents attenuate the effects of cancer therapy, and thereafter to explore possible methods for improving tumor treatment efficacy.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Mice , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/metabolism , Bone Marrow Cells/metabolism , Endothelial Cells/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , RNA, Messenger/metabolism , Tumor Microenvironment , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Int J Mol Sci ; 23(15)2022 Jul 27.
Article in English | MEDLINE | ID: mdl-35955439

ABSTRACT

Radiation-induced brain injury (RIBI) after radiotherapy has become an increasingly important factor affecting the prognosis of patients with head and neck tumor. With the delivery of high doses of radiation to brain tissue, microglia rapidly transit to a pro-inflammatory phenotype, upregulate phagocytic machinery, and reduce the release of neurotrophic factors. Persistently activated microglia mediate the progression of chronic neuroinflammation, which may inhibit brain neurogenesis leading to the occurrence of neurocognitive disorders at the advanced stage of RIBI. Fully understanding the microglial pathophysiology and cellular and molecular mechanisms after irradiation may facilitate the development of novel therapy by targeting microglia to prevent RIBI and subsequent neurological and neuropsychiatric disorders.


Subject(s)
Brain Injuries , Radiation Injuries , Brain/pathology , Brain Injuries/pathology , Humans , Microglia/pathology , Neurogenesis/radiation effects , Radiation Injuries/etiology , Radiation Injuries/pathology
8.
Pharmacology ; 107(1-2): 1-13, 2022.
Article in English | MEDLINE | ID: mdl-34915505

ABSTRACT

BACKGROUND: The pathobiology of diabetes and associated complications has been widely researched in various countries, but effective prevention and treatment methods are still insufficient. Diabetes is a metabolic disorder of carbohydrates, fats, and proteins caused by an absence of insulin or insulin resistance, which mediates an increase of oxidative stress, release of inflammatory factors, and macro- or micro-circulation dysfunctions, ultimately developing into diverse complications. SUMMARY: In the last decade through pathogenesis research, epigenetics has been found to affect metabolic diseases. Particularly, DNA methylation, histone acetylation, and miRNAs promote or inhibit diabetes and complications by regulating the expression of related factors. Curcumin has a wide range of beneficial pharmacological activities, including anti-inflammatory, anti-oxidation, anticancer, anti-diabetes, anti-rheumatism, and increased immunity. Key Messages: In this review, we discuss the effects of curcumin and analogs on diabetes and associated complications through epigenetics, and we summarize the preclinical and clinical researches for curcumin and its analogs in terms of management of diabetes and associated complications, which may provide an insight into the development of targeted therapy of endocrine diseases.


Subject(s)
Curcumin/pharmacology , Curcumin/therapeutic use , Diabetes Complications/drug therapy , Diabetes Mellitus/drug therapy , Epigenesis, Genetic/drug effects , Acetylation/drug effects , Animals , Curcumin/analogs & derivatives , DNA Methylation/drug effects , Diabetes Complications/genetics , Diabetes Mellitus/genetics , Humans , MicroRNAs/drug effects
9.
Dose Response ; 19(4): 15593258211057768, 2021.
Article in English | MEDLINE | ID: mdl-34887716

ABSTRACT

Background: Brain exposure to ionizing radiation during the radiotherapy of brain tumor or metastasis of peripheral cancer cells to the brain has resulted in cognitive dysfunction by reducing neurogenesis in hippocampus. The water extract of Lycium barbarum berry (Lyc), containing water-soluble Lycium barbarum polysaccharides and flavonoids, can protect the neuronal injury by reducing oxidative stress and suppressing neuroinflammation. Reseach Design: To demonstrate the long-term radioprotective effect of Lyc, we evaluated the neurobehavioral alterations and the numbers of NeuN, calbindin (CB), and parvalbumin (PV) immunopositive hippocampal neurons in BALB/c mice after acute 5.5 Gy radiation with/without oral administration of Lyc at the dosage of 10 g/kg daily for 4 weeks. Results: The results showed that Lyc could improve irradiation-induced animal weight loss, depressive behaviors, spatial memory impairment, and hippocampal neuron loss. Immunohistochemistry study demonstrated that the loss of NeuN-immunopositive neuron in the hilus of the dentate gyrus, CB-immunopositive neuron in CA1 strata radiatum, lacunosum moleculare and oriens, and PV-positive neuron in CA1 stratum pyramidum and stratum granulosum of the dentate gyrus after irradiation were significantly improved by Lyc treatment. Conclusion: The neuroprotective effect of Lyc on those hippocampal neurons may benefit the configuration of learning related neuronal networks and then improve radiation induced neurobehavioral changes such as cognitive impairment and depression. It suggests that Lycium barbarum berry may be an alternative food supplement to prevent radiation-induced neuron loss and neuropsychological disorders.

10.
Cells ; 10(12)2021 12 17.
Article in English | MEDLINE | ID: mdl-34944078

ABSTRACT

Population aging is occurring rapidly worldwide, challenging the global economy and healthcare services. Brain aging is a significant contributor to various age-related neurological and neuropsychological disorders, including Alzheimer's disease and Parkinson's disease. Several extrinsic factors, such as exposure to ionizing radiation, can accelerate senescence. Multiple human and animal studies have reported that exposure to ionizing radiation can have varied effects on organ aging and lead to the prolongation or shortening of life span depending on the radiation dose or dose rate. This paper reviews the effects of radiation on the aging of different types of brain cells, including neurons, microglia, astrocytes, and cerebral endothelial cells. Further, the relevant molecular mechanisms are discussed. Overall, this review highlights how radiation-induced senescence in different cell types may lead to brain aging, which could result in the development of various neurological and neuropsychological disorders. Therefore, treatment targeting radiation-induced oxidative stress and neuroinflammation may prevent radiation-induced brain aging and the neurological and neuropsychological disorders it may cause.


Subject(s)
Brain/pathology , Cellular Senescence/radiation effects , Radiation, Ionizing , Animals , Autophagy/radiation effects , Humans , Mitochondria/pathology , Mitochondria/radiation effects , Oxidative Stress/radiation effects
11.
Cells ; 10(9)2021 09 18.
Article in English | MEDLINE | ID: mdl-34572124

ABSTRACT

Early life radiation exposure causes abnormal brain development, leading to adult depression. However, few studies have been conducted to explore pre- or post-natal irradiation-induced depression-related neuropathological changes. Relevant molecular mechanisms are also poorly understood. We induced adult depression by irradiation of mice at postnatal day 3 (P3) to reveal hippocampal neuropathological changes and investigate their molecular mechanism, focusing on MicroRNA (miR) and its target mRNA and protein. P3 mice were irradiated by γ-rays with 5Gy, and euthanized at 1, 7 and 120 days after irradiation. A behavioral test was conducted before the animals were euthanized at 120 days after irradiation. The animal brains were used for different studies including immunohistochemistry, CAP-miRSeq, Real-Time Quantitative Reverse Transcription PCR (qRT-PCR) and western blotting. The interaction of miR-34a-5p and its target T-cell intracytoplasmic antigen-1 (Tia1) was confirmed by luciferase reporter assay. Overexpression of Tia1 in a neural stem cell (NSC) model was used to further validate findings from the mouse model. Irradiation with 5 Gy at P3 induced depression in adult mice. Animal hippocampal pathological changes included hypoplasia of the infrapyramidal blade of the stratum granulosum, aberrant and impaired cell division, and neurogenesis in the dentate gyrus. At the molecular level, upregulation of miR-34a-5p and downregulation of Tia1 mRNA were observed in both animal and neural stem cell models. The luciferase reporter assay and gene transfection studies further confirmed a direct interaction between miR-43a-5p and Tia1. Our results indicate that the early life γ-radiation-activated miR-43a-5p/Tia1 pathway is involved in the pathogenesis of adult depression. This novel finding may provide a new therapeutic target by inhibiting the miR-43a-5p/Tia1 pathway to prevent radiation-induced pathogenesis of depression.


Subject(s)
Dentate Gyrus/pathology , Depression/pathology , Gamma Rays/adverse effects , Gene Expression Regulation, Neoplastic/radiation effects , MicroRNAs/genetics , Neurogenesis , T-Cell Intracellular Antigen-1/metabolism , Animals , Apoptosis , Cell Proliferation , Dentate Gyrus/radiation effects , Depression/etiology , Depression/metabolism , Mice , Mice, Inbred BALB C , T-Cell Intracellular Antigen-1/genetics
12.
Neurochem Res ; 46(6): 1487-1501, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33710536

ABSTRACT

Activation of dopamine (DA) neurons is essential for the transition from sleep to wakefulness and maintenance of awakening, and sufficient to accelerate the emergence from general anesthesia in animals. Dopamine receptors (DR) are involve in arousal mediation. In the present study, we showed that the olfactory tubercle (OT) was active during emergence from isoflurane anesthesia, local injection of dopamine D1 receptor (D1R) agonist chloro-APB (1 mg/mL) and D2 receptor (D2R) agonist quinpirole (1 mg/mL) into OT enhanced behavioural and cortical arousal from isoflurane anesthesia, while D1R antagonist SCH-23390 (1 mg/mL) and D2R antagonist raclopride (2.5 mg/mL) prolonged recovery time. Optogenetic activation of DAergic terminals in OT also promoted behavioural and cortical arousal from isoflurane anesthesia. However, neither D1R/D2R agonists nor D1R/D2R antagonists microinjection had influences on the induction of isoflurane anesthesia. Optogenetic stimulation on DAergic terminals in OT also had no impact on the anesthesia induction. Our results indicated that DA signals in OT accelerated emergence from isoflurane anesthesia. Furthermore, the induction of general anesthesia, different from the emergence process, was not mediated by the OT DAergic pathways.


Subject(s)
Anesthetics, Inhalation/pharmacology , Arousal/physiology , Isoflurane/pharmacology , Olfactory Tubercle/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Animals , Arousal/drug effects , Benzazepines/pharmacology , Dopamine Agonists/pharmacology , Dopamine D2 Receptor Antagonists/pharmacology , Male , Mice, Inbred C57BL , Quinpirole/pharmacology , Raclopride/pharmacology , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D2/agonists
13.
Seizure ; 84: 122-128, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33348235

ABSTRACT

The astroglial network connected through gap junctions assembling from connexins physiologically balances the concentrations of ions and neurotransmitters around neurons. Astrocytic dysfunction has been associated with many neurological disorders including epilepsy. Dissociated gap junctions result in the increased activity of connexin hemichannels which triggers brain pathophysiological changes. Previous studies in patients and animal models of epilepsy indicate that the reduced gap junction coupling from assembled connexin hemichannels in the astrocytes may play an important role in epileptogenesis. This abnormal cell-to-cell communication is now emerging as an important feature of brain pathologies and being considered as a novel therapeutic target for controlling epileptogenesis. In particular, candidate drugs with ability of inhibition of connexin hemichannel activity and enhancement of gap junction formation in astrocytes should be explored to prevent epileptogenesis and control epilepsy.


Subject(s)
Astrocytes , Connexins , Animals , Cell Communication , Gap Junctions , Humans , Neurons
14.
Molecules ; 25(23)2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33287417

ABSTRACT

Patients receiving brain radiotherapy may suffer acute or chronic side effects. Ionizing radiation induces the production of intracellular reactive oxygen species and pro-inflammatory cytokines in the central nervous system, leading to brain damage. Complementary Chinese herbal medicine therapy may reduce radiotherapy-induced side effects. Flavonoids are a class of natural products which can be extracted from Chinese herbal medicine and have been shown to have neuroprotective and radioprotective properties. Flavonoids are effective antioxidants and can also inhibit regulatory enzymes or transcription factors important for controlling inflammatory mediators, affect oxidative stress through interaction with DNA and enhance genomic stability. In this paper, radiation-induced brain damage and the relevant molecular mechanism were summarized. The radio-neuro-protective effect of flavonoids, i.e., antioxidant, anti-inflammatory and maintaining genomic stability, were then reviewed. We concluded that flavonoids treatment may be a promising complementary therapy to prevent radiotherapy-induced brain pathophysiological changes and cognitive impairment.


Subject(s)
Brain/drug effects , Brain/radiation effects , Flavonoids/pharmacology , Flavonoids/therapeutic use , Radiation Injuries/drug therapy , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Animals , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Humans , Radiation, Ionizing
15.
Cells ; 8(10)2019 09 30.
Article in English | MEDLINE | ID: mdl-31575017

ABSTRACT

Long non-coding RNAs (lncRNAs) are versatile regulators of gene expression and play crucial roles in diverse biological processes. Epithelial-mesenchymal transition (EMT) is a cellular program that drives plasticity during embryogenesis, wound healing, and malignant progression. Increasing evidence shows that lncRNAs orchestrate multiple cellular processes by modulating EMT in diverse cell types. Dysregulated lncRNAs that can impact epithelial plasticity by affecting different EMT markers and target genes have been identified. However, our understanding of the landscape of lncRNAs important in EMT is far from complete. Here, we summarize recent findings on the mechanisms and roles of lncRNAs in EMT and elaborate on how lncRNAs can modulate EMT by interacting with RNA, DNA, or proteins in epigenetic, transcriptional, and post-transcriptional regulation. This review also highlights significant EMT pathways that may be altered by diverse lncRNAs, thereby suggesting their therapeutic potential.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/physiology , RNA, Long Noncoding/physiology , Humans
16.
Mol Med Rep ; 19(4): 3148-3158, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816516

ABSTRACT

Brain ischemia, including cerebral ischemia and cerebrovascular ischemia, leads to poor oxygen supply or cerebral hypoxia, and causes brain tissue death or cerebral infarction/ischemic stroke. The troxerutin and cerebroprotein hydrolysate injection (TCHI), is widely applied in China to improve blood supply in ischemic brain tissues and to enhance neuroprotective effects in clinical practice. However, the benefits and detailed underlying mechanism elaborating the effectiveness of TCHI in cerebrovascular diseases require further investigation. Therefore, in the present study, experimental in vivo and in vitro models were employed to investigate the potential mechanisms of TCHI on cerebral ischemic injury. The results demonstrated that TCHI increased the lactate dehydrogenase levels in the brain homogenate and conversely decreased lactic acid levels. TCHI was further observed to significantly increase superoxide dismutase activity and decrease malondialdehyde levels in ischemic brain tissues. In addition, TCHI significantly induced vascular maturation processes, including proliferation, adhesion, migration and tube formation in cultured human umbilical vein endothelial cells. Additionally, TCHI significantly stimulated microvessel formation in the rat aortic ring and chick chorioallantoic membrane assays. Taken together, these results provided strong evidence that TCHI stimulated angiogenesis at multiple steps, and indicated that TCHI attenuated cerebral ischemic damage through the amelioration of oxidative stress and promotion of angiogenesis.


Subject(s)
Anticoagulants/pharmacology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Hydroxyethylrutoside/analogs & derivatives , Neovascularization, Pathologic/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Biomarkers , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Cell Adhesion , Cell Movement , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Hydroxyethylrutoside/pharmacology , Male , Rats , Reactive Oxygen Species/metabolism
17.
Biomolecules ; 10(1)2019 12 31.
Article in English | MEDLINE | ID: mdl-31906046

ABSTRACT

Long non-coding RNAs (lncRNAs) play multifaceted roles in modulating gene expression under both physiological and pathological processes. The dysregulation of lncRNAs has been increasingly linked with many human diseases, including a plethora of cancers. Mounting evidence indicates that lncRNAs are aberrantly expressed in hepatocellular carcinoma (HCC) and can regulate HCC progression, as well as metastasis. In this review, we summarize the recent findings on the expanding roles of lncRNAs in modulating various functions of HCC, and elaborate on how can lncRNAs impact HCC metastasis and progression via interacting with chromatin, RNA, and proteins at the epigenetic, transcriptional, and post-transcriptional levels. This mini-review also highlights the current advances regarding the signaling pathways of lncRNAs in HCC metastasis and sheds light on the possible application of lncRNAs for the prevention and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplasm Metastasis/genetics , RNA, Long Noncoding/genetics , Carcinoma, Hepatocellular/metabolism , Epigenesis, Genetic/genetics , Humans , Liver Neoplasms/metabolism , RNA, Long Noncoding/metabolism
18.
Int J Mol Sci ; 19(9)2018 Sep 12.
Article in English | MEDLINE | ID: mdl-30213136

ABSTRACT

As an important second messenger, the calcium ion (Ca2+) plays a vital role in normal brain function and in the pathophysiological process of different neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), and epilepsy. Ca2+ takes part in the regulation of neuronal excitability, and the imbalance of intracellular Ca2+ is a trigger factor for the occurrence of epilepsy. Several anti-epileptic drugs target voltage-dependent calcium channels (VDCCs). Intracellular Ca2+ levels are mainly controlled by VDCCs located in the plasma membrane, the calcium-binding proteins (CBPs) inside the cytoplasm, calcium channels located on the intracellular calcium store (particular the endoplasmic reticulum/sarcoplasmic reticulum), and the Ca2+-pumps located in the plasma membrane and intracellular calcium store. So far, while many studies have established the relationship between calcium control factors and epilepsy, the mechanism of various Ca2+ regulatory factors in epileptogenesis is still unknown. In this paper, we reviewed the function, distribution, and alteration of VDCCs and CBPs in the central nervous system in the pathological process of epilepsy. The interaction of VDCCs with CBPs in the pathological process of epilepsy was also summarized. We hope this review can provide some clues for better understanding the mechanism of epileptogenesis, and for the development of new anti-epileptic drugs targeting on VDCCs and CBPs.


Subject(s)
Calcium Channels/metabolism , Calcium-Binding Proteins/metabolism , Epilepsy/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Humans
19.
Oncol Rep ; 40(4): 2147-2156, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30106454

ABSTRACT

Estradiol (E2) is a prime culprit for enhancing the progression of female hormone­related cancers. Bone marrow­derived cells (BMDCs) have been found to play a pivotal role in tumor growth. Estrogen receptors (ERs) are also found on certain subtypes of BMDCs, in addition to endothelial cells (ECs) and certain tumor cells. However, the role of BMDCs in E2­induced tumor biology is still unclear. Thus, the effects of E2 on ER­negative 4T1 breast cancer growth, the mobilization and recruitment of BMDCs, and interactions among BMDCs, ECs, and 4T1 cells were investigated. The results showed that E2 potentiated 4T1 tumor growth and angiogenesis in mice subjected to sham operation, ovariectomy (OVX), or OVX and E2 replacement treatment. E2 supplementation in mice with OVX upregulated the transcription of stromal cell­derived factor­1 (SDF­1) mRNA in tumor tissues and enhanced the recruitment of BMDCs into tumor tissues in vivo. E2 deficiency significantly decreased proangiogenic CXCR4+, ß3+, Sca­1+ and CXCR4+ß3+ BMDCs circulating in the peripheral blood. Cell­based system analyses showed that E2 augmented the transcription of ß3 mRNA in ECs, increased the adhesion of BMDCs to ECs. In addition, E2 enhanced the BMDC­induced EC proliferation and migration, the BMDC­induced 4T1 proliferation and the 4T1­stimulated EC proliferation in addition to enhancing the proliferation of tumor cells and the migration of ECs in vitro. Therefore, E2 enhanced the growth of breast tumors by stimulating tumor cells and ECs directly, as well as by increasing proangiogenic BMDC mobilization and recruitment leading to augmentation of the tumor and EC functions indirectly by cell proliferation assay. These findings reveal a separate mechanism via which E2 promotes the growth of female hormone­dependent tumors, which may be useful in explorations of new therapies for related cancers.


Subject(s)
Bone Marrow/pathology , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Proliferation , Endothelium, Vascular/pathology , Estrogens/toxicity , Neovascularization, Pathologic/pathology , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Bone Marrow/drug effects , Breast Neoplasms/chemically induced , Cell Adhesion , Cell Movement , Culture Media, Conditioned/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/chemically induced , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Epilepsy Res ; 126: 45-52, 2016 10.
Article in English | MEDLINE | ID: mdl-27423018

ABSTRACT

Chemokine and receptor systems play important roles in different animal models of status epileptics and epileptogenesis. Here, we identified protein and gene expression of chemokine receptor 6 in the hippocampus of Swiss mice with immunohistochemistry and RT-PCR respectively. Immunohistochemical study showed that CCR6 immunopositive product was localized in different subtypes of hippocampal interneurons, in apical dendrites of pyramidal neurons of CA1 area and other laminas of the hippocampus. Strongly stained CCR6 immunopositive product was found in calbindin, calretinin, parvalbumin immunopositive interneurons in the stratum oriens of CA1 area. During pilocarpine induced status epilepticus, a transient down-regulation of neuronal CCR6 in the stratum oriens of CA1 was demonstrated at 2h during status epilepticus. The present study provides evidence that CCR6 may be involved in normal neuronal activity in the hippocampus and play an important role in maintenance of the status epilepticus.


Subject(s)
GABAergic Neurons/metabolism , Hippocampus/metabolism , Interneurons/metabolism , Pyramidal Cells/metabolism , Receptors, CCR6/metabolism , Status Epilepticus/metabolism , Animals , Calbindin 2/metabolism , Calbindins/metabolism , Dendrites/metabolism , Dendrites/pathology , Disease Models, Animal , Female , GABAergic Neurons/pathology , Gene Expression Regulation , Hippocampus/pathology , Immunohistochemistry , Interneurons/pathology , Mice , Parvalbumins/metabolism , Pilocarpine , Polymerase Chain Reaction , Pyramidal Cells/pathology , Status Epilepticus/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...