Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 34
Filtrer
Plus de filtres











Gamme d'année
1.
Neurochem Res ; 49(10): 2957-2971, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39088164

RÉSUMÉ

Depression and anxiety disorders are prevalent stress-related neuropsychiatric disorders and involve multiple molecular changes and dysfunctions across various brain regions. However, the specific and shared pathophysiological mechanisms occurring in these regions remain unclear. Previous research used a rat model of chronic mild stress (CMS) to segregate and identify depression-susceptible, anxiety-susceptible, and insusceptible groups; then the proteomes of six distinct brain regions (the hippocampus, prefrontal cortex, hypothalamus, pituitary, olfactory bulb, and striatum) were separately and quantitatively analyzed. To gain a comprehensive and systematic understanding of the molecular abnormalities, this study aimed to investigate and compare differential proteomics data from the six regions. Differentially expressed proteins (DEPs) were identified in between specific regions and across all regions and subjected to a series of bioinformatics analyses. Regional comparisons showed that stress-induced proteomic changes and corresponding gene ontology and pathway enrichments were largely distinct, attributable to differences in cell populations, protein compositions, and brain functions of these areas. Additionally, a notable degree of overlap in the significantly enriched terms was identified, potentially suggesting strong connections in the enrichment across different regions. Furthermore, intra-regional and inter-regional protein-protein interaction networks and drug-target-DEP networks were constructed. Integrated analysis of the three association networks in the six regions, along with the DisGeNET database, identified ten DEPs as potential targets for anti-depression/anxiety drugs. Collectively, these findings revealed commonalities and differences across different brain regions at the protein level induced by CMS, and identified several novel protein targets for the development of new therapeutics for depression and anxiety.


Sujet(s)
Anxiolytiques , Encéphale , Protéome , Rat Sprague-Dawley , Stress psychologique , Animaux , Stress psychologique/métabolisme , Stress psychologique/traitement médicamenteux , Protéome/métabolisme , Mâle , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Anxiolytiques/pharmacologie , Anxiolytiques/usage thérapeutique , Anxiété/métabolisme , Anxiété/traitement médicamenteux , Dépression/métabolisme , Dépression/traitement médicamenteux , Cartes d'interactions protéiques , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Rats , Protéomique
2.
Med Oncol ; 41(7): 174, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38869721

RÉSUMÉ

Cervical cancer (CC), one of the most aggressive tumors in women, has high risk rates of recurrence and metastasis. It is essential to study the key genes and proteins involved in CC development. IRTKS, a member of the IRSp53 family, has been reported as a tumor promoter in gastric and breast cancers. However, the biological role of IRTKS in CC is still unclear. The purpose of this study was to explore the biological function of IRTKS in CC cells in vitro and the effect of IRTKS on tumorigenesis in vivo. Siha and Hela cells were treated with si-RNA and plasmids. Cell proliferation and growth were detected by CCK8, colony formation assay and nude mouse tumorigenicity assay, respectively. Transwell assay was used to analyze cell migration and invasion. The expression of epithelial-mesenchymal transition (EMT)-related proteins was determined by western blot. IRTKS was highly expressed in CC. IRTKS contributed to the proliferation of CC cells in vitro and in vivo. Furthermore, IRTKS facilitated the migration and invasion of CC cells and modulated EMT. IRTKS plays a crucial role in CC tumorigenesis, suggesting it may be a potential key gene for new therapeutic strategies in CC.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Transition épithélio-mésenchymateuse , Souris nude , Tumeurs du col de l'utérus , Animaux , Femelle , Humains , Souris , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Évolution de la maladie , Transition épithélio-mésenchymateuse/génétique , Cellules HeLa , Souris de lignée BALB C , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/métabolisme
3.
Amino Acids ; 55(2): 263-273, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-36539546

RÉSUMÉ

Comprehensive knowledge of the intracellular protein interactions of cell-surface receptors will greatly advance our comprehension of the underlying trafficking mechanisms. Hence, development of effective and high-throughput approaches is highly desired. In this work, we presented a strategy aiming to tailor toward the analysis of intracellular protein interactome of cell-surface receptors. We used α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors subunit GluA1 as an example to illustrate the methodological application. To capture intracellular proteins that interact with GluA1, after surface biotinylation of the prepared hippocampal neurons and slices, the non-biotinylated protein components as intracellular protein-enriched fraction were unconventionally applied for the following co-immunoprecipitation. The co-immuno-precipitated proteins were then analyzed through mass spectrometry-based proteomics and bioinformatics platforms. The detailed localizations indicated that intracellular proteins accounted for up to 93.7 and 90.3% of the analyzed proteins in the neurons and slices, respectively, suggesting that our protein preparation was highly effective to characterize intracellular interactome of GluA1. Further, we systematically revealed the protein functional profile of GluA1 intracellular interactome, thereby providing complete overview and better comprehension of diverse intracellular biological processes correlated with the complex GluA1 trafficking. All experimental results demonstrated that our methodology would be applicable and useful for intracellular interaction proteomics of general cell-surface receptors.


Sujet(s)
Neurones , Protéomique , Neurones/métabolisme , Hippocampe/métabolisme , Récepteurs de surface cellulaire
4.
Neuroscience ; 503: 58-68, 2022 11 01.
Article de Anglais | MEDLINE | ID: mdl-36041587

RÉSUMÉ

Recent studies have demonstrated that Camk2b expression is modified in neuropsychiatric illnesses and potentially affects synaptic plasticity. However, the molecular events arising from Camk2b dysregulation are not fully elucidated and need to be comprehensively explored. In the present study, we first induced over-expression and under-expression of Camk2b in cultured rat hippocampal neurons through transfection with lentivirus plasmids. Then isobaric tag for relative and absolute quantitation (iTRAQ)-based quantitative proteomics followed by bioinformatics analyses were carried out to explore the impacts of Camk2b dysexpression on the proteome of the neurons. Compared with the respective controls, a total of 270 proteins in the Camk2b-overexpression group and 209 proteins in the Camk2b-underexpression group were experienced a divergence in expression. Gene ontology and pathway analyses indicated that Camk2b overexpression and under-expression respectively induced two different change profiles of protein expressions and functions, reflecting the potential differences in cellular processes and biological events. Through cross comparison, several candidate target proteins regulated directly by Camk2b were revealed. Further network and immunoblot analyses demonstrated that Mapk3 could be an important linker and Camk2b-Mapk3 might serve as a new potential pathway affecting the expression of synaptic proteins in hippocampal neurons. Collectively, the present results offer a new comprehension of the regulatory molecular mechanism of Camk2b and thereby increase our understanding of Camk2b-mediated synaptogenesis in synaptic plasticity.


Sujet(s)
Hippocampe , Protéome , Animaux , Rats , Protéome/métabolisme , Hippocampe/métabolisme , Protéomique/méthodes , Neurones/métabolisme , Plasticité neuronale
5.
Cell Biol Int ; 46(6): 907-921, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35165984

RÉSUMÉ

Wilms' tumor (WT) is the most common pediatric renal malignancy. PDGFRß belongs to the type III receptor tyrosine kinase family and is known to be involved in tumor metastasis and angiogenesis. Here, we studied the effect and underlying mechanism of PDGFRß on WT G401 cells. Transwell assay and wound-healing assay were used to detect the effect of PDGFRß on G401 cells invasion and migration. Western blot and immunofluorescence were used to detect the expression of EMT-related genes. The expression of PI3K/AKT/mTOR pathway proteins was detected by Western blot. The relationship between PDGFRß and aerobic glycolysis was studied by assessing the expression of glycolysis-related enzymes detected by qRT-PCR and Western blot. The activity of HK, PK, and LDH was detected by corresponding enzyme activity kits. The concentration of lactic acid and glucose was detected by Lactic Acid Assay Kit and Glucose Assay Kit-glucose oxidase method separately. To investigate the mechanism of PDGFRß in the development of WT, the changes of glucose and lactic acid were analyzed after blocking PI3K pathway, aerobic glycolysis, or PDGFRß. The key enzyme was screened by Western blot and glucose metabolism experiment after HK2, PKM2, and PDK1 were inhibited. The results showed that PDGFRß promoted the EMT process by modulating aerobic glycolysis through PI3K/AKT/mTOR pathway in which PKM2 plays a key role. Therefore, our study of the mechanism of PDGFRß in G401 cells provides a new target for the treatment of WT.


Sujet(s)
Tumeurs du rein , Tumeur de Wilms , Bécaplermine/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Enfant , Transition épithélio-mésenchymateuse , Glucose , Glycolyse , Humains , Acide lactique , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Récepteur au PDGF bêta/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Tumeur de Wilms/métabolisme
6.
Ann Clin Lab Sci ; 52(1): 101-108, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-35181623

RÉSUMÉ

OBJECTIVE: To investigate the effect of dichloroacetate (DCA) on Wilms' tumor (WT) G401 cells. METHODS: CCK-8 assay was used to detect the influence of DCA on G401 cells viability and 10 mmol/L DCA was selected for subsequent experiments. The expression of glycolysis-related enzymes, such as hexokinase 2 (HK2), pyruvate kinase M2 (PKM2), lactic acid dehydrogenase A (LDHA), pyruvate dehydrogenase kinase 1 (PDK1), and pyruvate dehydrogenase (PDH), were detected by qRT-PCR and western blot. The extracellular lactic acid and glucose concentrations were measured by the lactic acid assay kit and glucose oxidase method kit respectively. Flow cytometry was used to detect the effect of DCA on G401 cells apoptosis. The invasion and migration ability of G401 cells were detected by Transwell assay and wound-healing assay. RESULTS: The results showed that DCA reduced glycolysis-related enzymes expression, inhibited lactic acid production, and glucose consumption. DCA also suppressed cells growth, induced cells apoptosis and inhibited cells invasion and migration. CONCLUSION: Inhibition of aerobic glycolysis by DCA can reduce the viability of G401 cells, promote cells apoptosis and inhibit cells invasion and migration. Therefore, aerobic glycolysis may be a potential therapeutic target for Wilms' tumor.


Sujet(s)
Tumeurs du rein , Tumeur de Wilms , Apoptose , Lignée cellulaire tumorale , Prolifération cellulaire , Glycolyse , Humains
7.
Front Genet ; 12: 751999, 2021.
Article de Anglais | MEDLINE | ID: mdl-34603401

RÉSUMÉ

Chronic stress as one of the most significant risk factor can trigger overactivity of hypothalamic-pituitary-adrenal (HPA) axis in depression as well as anxiety. Yet, the shared and unique neurobiological underpinnings underlying the pituitary abnormality in these two disorders have not been made clear. We previously have established depression-susceptible, anxiety-susceptible and insusceptible groups using a valid chronic mild stress (CMS) model. In this work, the possible protein expression changes in the rat pituitary of these three groups were continuously investigated through the use of the comparative quantitative proteomics and bioinformatics approaches. The pituitary-proteome analysis identified totally 197 differential proteins as a CMS response. These deregulated proteins were involved in diverse biological functions and significant pathways potentially connected with the three different behavioral phenotypes, likely serving as new investigative protein targets. Afterwards, parallel reaction monitoring-based independent analysis found out that expression alterations in Oxct1, Sec24c, Ppp1cb, Dock1, and Coq3; Lama1, Glb1, Gapdh, Sccpdh, and Renbp; Sephs1, Nup188, Spp1, Prodh1, and Srm were specifically linked to depression-susceptible, anxiety-susceptible and insusceptible groups, respectively, suggesting that the same CMS had different impacts on the pituitary protein regulatory system. Collectively, the current proteomics research elucidated an important molecular basis and furnished new valuable insights into neurochemical commonalities and specificities of the pituitary dysfunctional mechanisms in HPA axis underlying vulnerability and resistance to stress-induced anxiety or depression.

8.
Front Mol Biosci ; 8: 730473, 2021.
Article de Anglais | MEDLINE | ID: mdl-34676246

RÉSUMÉ

Chronic stress is a key factor for the onset of anxiety and depression disorders. However, the stress-induced common and unique molecular basis of the two psychiatric disorders is not fully known and still needs to be explored. Previously, we employed a chronic mild stress (CMS) procedure to induce a rat model including depression-susceptible (Dep-Sus), anxiety-susceptible (Anx-Sus), and insusceptible (Insus) cohorts. In this work, we continuously analyze the striatal proteomes of the three stressed cohorts by the use of comparative proteomics and bioinformatics approaches. Through isobaric tags for relative and absolute quantitation (iTRAQ)-based analysis, 386 abnormally expressed proteins in total were identified. These deregulated proteins are involved in various biological functions and significant pathways that are potentially connected with resistance and susceptibility to CMS-caused anxious- or depressive-like behaviors and, hence, could act as suggestive protein targets. A further parallel reaction monitoring-based independent investigation shows that alterations in Pak5, Dgkg, Scn4b, Rb1cc1, and Acin1; Ggps1, Fntb, Nudt19, Ufd1, and Ndufab1; and Dnajb12, Hbb2, Ap2s1, Ip6k1, and Stk4 were specifically connected with Dep-Sus, Anx-Sus, or Insus groups, respectively, potentially indicating that identical CMS treatment results in the different changes in the striatal protein regulations. Overall, our current proteomics study of the striatum provides an important molecular foundation and comprehensive insights into common and specific deregulations correlated with pathophysiological mechanisms that underlie resistance and susceptibility to chronic stress-induced anxiety or depression.

9.
Neuroscience ; 473: 29-43, 2021 10 01.
Article de Anglais | MEDLINE | ID: mdl-34425157

RÉSUMÉ

Chronic stress causes the abnormality of olfactory bulb (OB) in both anxiety and depression, however, the unique and common neurobiological underpinnings are still poorly understood. Previously, we built the three groups by chronic mild stress (CMS), depression-susceptible (Dep-Sus): with depression-like behavior, anxiety-susceptible (Anx-Sus): with anxiety-like behavior and insusceptible (Insus): without depression- and anxiety-like behaviors. To continuously explore the protein expression changes in these three groups, comparative quantitative proteomics analysis was conducted on the rat OB as crucial part of the olfactory system. Next, bioinformatics analyses were implemented whereas protein expressions were independently analyzed by parallel reaction monitoring (PRM) or Western blot (WB). The OB-proteome analysis identified totally 133 differentially expressed proteins as a CMS response. These deregulated proteins were involved in multiple functions and significant pathways potentially correlated with phenotypes of maladaptive behavior of depression or anxiety as well as adaptive behavior, and hence might act as potential candidate protein targets. The subsequent PRM-based or WB-based analyses showed that changes in Nefl, Mtmr7 and Tk2; Prkaca, Coa3, Cox6c2, Lamc1 and Tubal3; and Pabpn1, Nme3, Sos1 and Lum were uniquely associated with Dep-Sus, Anx-Sus, and Insus groups, respectively. These phenotype-specific deregulated proteins were primarily involved in multiple metabolic and signaling pathways, suggesting that the identical CMS differently impacted the olfactory protein regulation system and biological processes. To sum up, our present data as a useful proteomics underpinning provided the common and distinct molecular insights into the biochemical understanding of OB dysfunction underlying susceptibility and resiliency to chronic-stress-induced anxiety or depression.


Sujet(s)
Dépression , Bulbe olfactif , Animaux , Anxiété , Troubles anxieux , Modèles animaux de maladie humaine , Protéomique , Rats
10.
Amino Acids ; 53(9): 1339-1350, 2021 Sep.
Article de Anglais | MEDLINE | ID: mdl-34363538

RÉSUMÉ

Mounting studies have demonstrated that RAB3GAP1 expression is modified in brain diseases with multiple neurobiological functions and processes and acts as a potentially significant target. However, the cellular and molecular events arising from RAB3GAP1 dysexpression are still incompletely understood. In this work, underexpression and overexpression of RAB3GAP1 were first induced into cultured mouse cortical neurons by transfection with lentivirus plasmids. Then we globally explored the effects of RAB3GAP1 dysexpression on the proteome of the neurons through the use of isobaric tag for relative and absolute quantitation (iTRAQ)-based quantitative proteomics with bioinformatics. A total of 364 proteins in the RAB3GAP1-underexpression group and 314 proteins in the RAB3GAP1-overexpression group were identified to be differentially expressed. Subsequent bioinformatics analysis indicated that the proteome functional expression profiles induced by RAB3GAP1 underexpression and overexpression were different, suggesting the potential differences in biological processes and cellular effects. Subsequent intergroup cross-comparison revealed some candidate target proteins regulated directly by RAB3GAP1. Further parallel reaction monitoring (PRM) analysis illustrated that Sub1, Ssrp1, and Top1 proteins might serve as new potentially important linkers in the RAB3GAP1-mediated autophagy pathway in the cortical neurons. Collectively, the current proteomics data furnished new valuable insights to better understand the regulatory molecular mechanism of neuronal RAB3GAP1.


Sujet(s)
Cortex cérébral/métabolisme , Neurones/métabolisme , Protéome/métabolisme , Protéomique/méthodes , Protéines G rab3/métabolisme , Animaux , Biologie informatique/méthodes , Souris , Protéome/analyse , Protéines G rab3/antagonistes et inhibiteurs , Protéines G rab3/génétique
11.
Neurobiol Stress ; 15: 100347, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34113696

RÉSUMÉ

Despite studies providing insight into the neurobiology of chronic stress, depression and anxiety, long noncoding RNA (lncRNA)-mediated mechanisms underlying the common and distinct pathophysiology of these stress-induced disorders remain nonconclusive. In a previous study, we used the chronic mild stress paradigm to separate depression-susceptible, anxiety-susceptible and insusceptible rat subpopulations. In the current study, lncRNA and messenger RNA (mRNA) expression was comparatively profiled in the hippocampus of the three stress groups using microarray technology. Groupwise comparisons identified distinct sets of lncRNAs and mRNAs associated with the three different behavioral phenotypes of the stressed rats. To investigate the regulatory roles of the dysregulated lncRNAs upon mRNA expression, correlations between the differential lncRNAs and mRNAs were first analyzed by combined use of weighted gene coexpression network analysis and ceRNA theory-based methods. Subsequent functional analysis of strongly correlated mRNAs indicated that the dysregulated lncRNAs were involved in various biological pathways and processes to specifically induce rat susceptibility or resiliency to depression or anxiety. Further intersectional analysis of phenotype-associated and drug-associated lncRNA-mRNA networks and subnetworks assisted in identifying 16 hub lncRNAs as potential targets of anti-depression/anxiety drugs. Collectively, our study established the molecular basis for understanding the similarities and differences in pathophysiological mechanisms underlying stress-induced depression or anxiety and stress resiliency, revealing several important lncRNAs that represent potentially new therapeutic drug targets for depression and anxiety disorders.

12.
Front Mol Neurosci ; 14: 633398, 2021.
Article de Anglais | MEDLINE | ID: mdl-33737865

RÉSUMÉ

Chronic stress as a known risk factor leads to hyperactivity of the hypothalamus-pituitary-adrenal (HPA) axis in both depression and anxiety. However, the stress-induced dysfunction of the HPA axis in these disorders especially the common and unique molecular dysregulations have not been well-explored. Previously, we utilized a chronic mild stress (CMS) paradigm to segregate and gain depression-susceptible, anxiety-susceptible, and insusceptible groups. In this study, we continue to examine the possible protein expression alterations of the hypothalamus as the center of the HPA axis in these three groups by using a proteomic approach. Though isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative analysis, a total of 593 dysregulated proteins were identified. These were potentially associated with vulnerability and adaptability of CMS-caused depression or anxiety and therefore might become novel investigative protein targets. Further independent analysis using parallel reaction monitoring (PRM) indicated that 5, 7, and 21 dysregulated proteins were specifically associated with depression-susceptible, anxiety-susceptible, and insusceptible groups, respectively, suggesting that the same CMS differently affected the regulation system of the rat hypothalamic proteome. In summary, the current proteomic research on the hypothalamus provided insights into the specific and common molecular basis for the HPA dysfunction mechanisms that underlie resiliency and vulnerability to stress-induced depression or anxiety.

13.
Transl Psychiatry ; 11(1): 143, 2021 02 24.
Article de Anglais | MEDLINE | ID: mdl-33627638

RÉSUMÉ

Chronic stress is a significant risk factor for depression as well as anxiety disorders. Yet, the stress-induced specific and common molecular dysregulations of these disorders have not been fully understood. Previously, we constructed a chronic mild stress (CMS) rat model to separate and obtain depression-susceptible, anxiety-susceptible, and insusceptible groups. In this study, the prefrontal cortical proteomes of the three stressed groups were comparatively profiled utilizing isobaric tags for relative and absolute quantitation (iTRAQ)-coupled tandem mass spectrometry approach. A total of 212 protein dysregulations were identified, potentially correlating to susceptibility or resilience to CMS-induced depression or anxiety, and thus might serve as potential protein targets for further investigation. In addition, independent analysis by parallel reaction monitoring identified changes in Gfap, Rhog, Gnai2, Ppp1r1b, and Uqcrh; Tubb6, Urod, Cul1, Spred1, and Gpcpd1; Acadl, Ppp1r1a, Grm2, Mtor, Lsm8, Cplx2, and Tsta3 that were distinctly correlated to depression-susceptible, anxiety-susceptible, or insusceptible groups, respectively. This suggested that identical CMS had different effects on the protein regulation system of the rat prefrontal cortex. Collectively, the present proteomics study of the prefrontal cortex established a significant molecular basis and offered new insights into the specificity and commonality of pathophysiologic mechanisms underlying susceptibility and resiliency to stress-induced depression or anxiety.


Sujet(s)
Dépression , Protéomique , Animaux , Anxiété , Troubles anxieux , Cortex préfrontal , Rats , Stress psychologique
14.
FEBS Open Bio ; 10(9): 1748-1757, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32580247

RÉSUMÉ

Osteosarcoma (OS) is the most common malignant bone tumor primarily influencing children and adults. Approximately one-fifth of patients have micrometastasis in the lungs when OS is diagnosed. Platelet-derived growth factor receptor (PDGFR) beta (PDGFRß) is a subtype of PDGFR. PDGFRß has been noted to be highly expressed in OS cell lines and patient specimens, and is associated with metastasis and poor prognosis of OS. However, mechanistic insights into the exact role of PDGFRß in OS pathogenesis and development are still lacking. Here we assessed the effects of PDGFRß on invasive and migratory abilities, such as the epithelial-mesenchymal transition and phosphatidylinositol 3-kinase (PI3K), Akt and mammalian target of rapamycin (mTOR) pathways in HOS cells. Depleting PDGFRß resulted in reduced migration of HOS cells in the small interfering RNA duplexes specific for the PDGFRß group compared with the mock and scramble-treated groups in Transwell invasion assays. Using wound-healing assays, we demonstrate the rate of wound healing in the PDGF-BB-stimulated group was higher compared with the mock-treated group. Western blot showed that down-regulation of PDGFRß decreased the expression of stromal phenotype markers and phosphorylation pathway proteins (PI3K, AKT and mTOR), but the epithelial phenotype marker was increased in HOS cells. Treating HOS cells with PDGF-BB revealed a treatment time-dependent increase of phosphorylated, but not total, PI3K, AKT and mTOR. Taken together, we suggest that PDGFRß plays an important role in OS invasion, migration and epithelial-mesenchymal transition by influencing the PI3K, Akt and mTOR pathways, hence highlighting PDGFRß as a potential therapeutic target for OS.


Sujet(s)
Tumeurs osseuses/métabolisme , Régulation négative , Ostéosarcome/métabolisme , Récepteur au PDGF bêta/métabolisme , Tumeurs osseuses/anatomopathologie , Mouvement cellulaire , Transition épithélio-mésenchymateuse , Humains , Ostéosarcome/anatomopathologie , Récepteur au PDGF bêta/génétique , Cellules cancéreuses en culture
15.
Gene ; 689: 11-17, 2019 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-30553996

RÉSUMÉ

Most cancer cells predominantly produce their energy through a high rate of glycolysis in the presence of abundant oxygen. Glycolysis has become a target of anticancer strategies. Previous researches showed that glucose transporter 1 (GLUT1) inhibitor is effective as anticancer agents. This study assessed the effects of the selective GLUT1 inhibitor WZB117 on regulation of neuroblastoma (NB) cell line SH-SY5Y viability, cell cycle and glycolysis in vitro. SH-SY5Y cells were grown and treated with WZB117 for up to 72 h and then subjected to cell viability, qRT-PCR, Western blot and flow cytometry analysis. Level of ATP and LDH was also analyzed. The result showed that WZB117 treatment reduced tumor cells viability, downregulated level of GLUT1 protein. Moreover, WZB117 treatment arrested tumor cells at the G0-G1 phase of the cell cycle, induced tumor cells to undergo necrosis instead of apoptosis. In addition, WZB117 treatment downregulated the levels of intracellular ATP, LDH and glycolytic enzymes. Thus, WZB117-induced GLUT1 inhibition suppressed tumor cell growth, induced cell cycle arrest and reduced glycolysis metabolites in NB cells in vitro. This study suggested that GLUT1 can be used as a potential therapeutic target for NB.


Sujet(s)
Transporteur de glucose de type 1/antagonistes et inhibiteurs , Hydroxybenzoates/pharmacologie , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Transporteur de glucose de type 1/génétique , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/génétique , Humains , Neuroblastome/génétique
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 34(11): 994-999, 2018 Nov.
Article de Chinois | MEDLINE | ID: mdl-30591108

RÉSUMÉ

Objective To study the effect of inhibiting the expression of glucose transporter 1 (GLUT1) in neuroblastoma cells on the proliferation, invasion and migration of tumor cells. Methods The specific small molecule inhibitor WZB117 was used to inhibit the expression of GLUT1 in neuroblastoma cells. The expression of GLUT1 mRNA was detected by real-time quantitative PCR; the expression of GLUT1 protein was detected by Western blotting; the ability of cell proliferation was detected by CCK-8 assay; the ability of cell invasion and migration were detected by TranswellTM invasion and migration assay, respectively. Results After treated with WZB117, the expression level of GLUT1 mRNA increased, while the protein expression level decreased in the neuroblastoma cells. Cell proliferation was inhibited, and the ability of cell invasion and migration were reduced. Conclusion Inhibiting the expression of GLUT1 in neuroblastoma cells might attenuate the malignant biological behaviors of tumor cells.


Sujet(s)
Mouvement cellulaire , Transporteur de glucose de type 1/métabolisme , Neuroblastome/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , Humains , Invasion tumorale , ARN messager
17.
Asian Pac J Cancer Prev ; 16(2): 571-8, 2015.
Article de Anglais | MEDLINE | ID: mdl-25684489

RÉSUMÉ

Prostate cancer is the most common cancer in men. In this study, we investigated immune responses of cytotoxic T lymphocytes (CTLs) against TRAMP-C2 prostate cancer cells after activation by dendritic cells (DCs) loaded with TRAMP-C2 freeze-thaw antigen and/or PEP-3 peptide in vitro. Bone marrow-derived DC from the bone marrow of the C57BL/6 were induced to mature by using the cytokine of rhGM-CSF and rhIL-4, and loaded with either the freeze-thaw antigen or PEP-3 peptide or both of them. Maturation of DCs was detected by flow cytometry. The killing efficiency of the CTLs on TRAMP-C2 cells were detected by flow cytometry, CCK8, colony formation, transwell migration, and wound-healing assay. The levels of the IFN-γ, TNF-ß and IL-12 were measured by enzyme-linked immunosorbent assay (ELISA). Compared with the unloaded DCs, the loaded DCs had significantly increased expression of several phenotypes related to DC maturation. CTLs activated by DCs loaded with freeze-thaw antigen and PEP-3 peptide had more evident cytotoxicity against TRAMP-C2 cells in vitro. The secretion levels of IFN-γ, TNF-ß and IL-12, secreted by DCs loaded with antigen and PEP-3 and interaction with T cells, were higher than in the other groups. Our results suggest that the CTLs activated by DCs loaded with TRAMP-C2 freeze-thaw antigen and PEP-3 peptide exert a remarkable killing efficiency against TRAMP-C2 cells in vitro.


Sujet(s)
Antigènes néoplasiques/immunologie , Cellules dendritiques/immunologie , Congélation , Peptides/immunologie , Tumeurs de la prostate/immunologie , Lymphocytes T cytotoxiques/immunologie , Animaux , Antigènes néoplasiques/métabolisme , Apoptose , Technique de Western , Mouvement cellulaire , Prolifération cellulaire , Cellules cultivées , Cytokines/métabolisme , Cellules dendritiques/métabolisme , Cellules dendritiques/anatomopathologie , Cytométrie en flux , Humains , Mâle , Souris , Souris de lignée C57BL , Peptides/métabolisme , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Lymphocytes T cytotoxiques/métabolisme , Lymphocytes T cytotoxiques/anatomopathologie , Cicatrisation de plaie , Tests d'activité antitumorale sur modèle de xénogreffe
18.
J Cancer Res Clin Oncol ; 140(8): 1331-41, 2014 Aug.
Article de Anglais | MEDLINE | ID: mdl-24816726

RÉSUMÉ

OBJECT: Trop2 plays an important role in proliferation and invasion of tumors. Extensive research has shown that the expression level of Trop2 is closely related to the progress of cervical diseases. This study was to explore the effects of Trop2 on cell proliferation and apoptosis in cervical cancer. METHODS: Trop2 was knocked down by shRNA in CaSki cells. The expression level of mRNA and protein was detected by real-time PCR and western blot, respectively. Cell proliferation was determined by CCK-8 and clone formation assay; apoptosis was measured by flow cytometry; cell cycle and apoptosis-related proteins cyclinD1, P53, bcl-2, bax, caspase 3, 8 and 9 were analyzed as well to investigate possible mechanism. RESULTS: Trop2 expression was effectively repressed in CaSki cells by Trop2 shRNA, which resulted in inhibition of proliferation and colony formation, whereas apoptosis rate was significantly increased. Furthermore, in Trop2 knockdown CaSki cells, the expression of cyclinD1 and bcl-2 was significantly down-regulated, while that of P53 and bax was up-regulated accompanied by increased activities of caspase 9 and 3 but not caspase 8. CONCLUSIONS: Trop2 is important in proliferation and apoptosis regulation in CaSki cells, which may become a novel target for cervical cancer treatment.


Sujet(s)
Antigènes néoplasiques/génétique , Apoptose , Molécules d'adhérence cellulaire/génétique , Prolifération cellulaire , Antigènes néoplasiques/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Caspases/métabolisme , Molécules d'adhérence cellulaire/métabolisme , Lignée cellulaire tumorale , Femelle , Expression des gènes , Techniques de knock-down de gènes , Humains , ARN messager/génétique , ARN messager/métabolisme , Petit ARN interférent/génétique , Tumeurs du col de l'utérus
19.
Asian Pac J Cancer Prev ; 15(6): 2447-51, 2014.
Article de Anglais | MEDLINE | ID: mdl-24761845

RÉSUMÉ

Human papillomavirus (HPV) is the primary etiologic agent of cervical cancer. Consideration of safety and non human leukocyte antigen restriction, protein vaccine has become the most likely form of HPV therapeutic vaccine, although none have so far been reported as effective. Since tumor cells consistently express the two proteins E6 and E7, most therapeutic vaccines target one or both of them. In this study, we fabricated DC vaccines by transducing replication-defective recombinant adenoviruses expressing E6/E7 fusion gene of HPV-16, to investigate the lethal effects of specific cytotoxic T lymphocytes (CTL) against CaSki cells in vitro. Mouse immature dendritic cells (DC) were generated from bone marrow, and transfected with pAd-E6/E7 to prepare a DC vaccine and to induce specific CTL. The surface expression of CD40, CD68, MHC II and CD11c was assessed by flow cytometry (FCM), and the lethal effects of CTL against CaSki cells were determined by DAPI, FCM and CCK-8 methods. Immature mouse DC was successfully transfected by pAd-E6/E7 in vitro, and the transfecting efficiency was 40%-50%. A DC vaccine was successfully prepared and was used to induce specific CTL. Experimental results showed that the percentage of apoptosis and killing rate of CaSki cells were significantly increased by coculturing with the specific CTL (p <0.05). These results illustrated that a DC vaccine modified by HPV-16 E6/E7 gene can induce apoptosis of CaSki cells by inducing CTL, which may be used as a new strategy for biological treatment of cervical cancer.


Sujet(s)
Cytotoxicité immunologique/immunologie , Cellules dendritiques/immunologie , Protéines des oncogènes viraux/génétique , Protéines E7 de papillomavirus/génétique , Infections à papillomavirus/prévention et contrôle , Vaccins contre les papillomavirus/administration et posologie , Protéines de répression/génétique , Tumeurs du col de l'utérus/prévention et contrôle , Adenoviridae/génétique , Animaux , Apoptose , Cellules dendritiques/virologie , Femelle , Cytométrie en flux , Antigènes d'histocompatibilité de classe I/immunologie , Papillomavirus humain de type 16/génétique , Humains , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Infections à papillomavirus/immunologie , Infections à papillomavirus/virologie , Cellules cancéreuses en culture , Tumeurs du col de l'utérus/immunologie , Tumeurs du col de l'utérus/virologie
20.
Mol Cell Biochem ; 392(1-2): 95-107, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-24633962

RÉSUMÉ

MCPH1, initially identified as an hTERT repressor, has recently been implicated in mediating DNA damage response and maintaining chromosome integrity. This study is to investigate its potential role in the onset of cervical cancer. In the study, decreased expression of MCPH1 was observed in 19 of 31 cases (61.3%) at mRNA level and 44 of 63 cases (69.8%) at protein level of cervical tumor tissues compared with the paired nontumor tissues. Reduced MCPH1 protein expression was significantly associated with high-tumor grade (1 vs. 3 P = 0.013; 2 vs. 3 P = 0.047). In addition to inhibit SiHa cell migration and invasion, the overexpression of MCPH1 inhibited cervical cancer cells growth through inducing S phase arrest and mitochondrial apoptosis. Further analysis demonstrated cyclinA2/CDK2, CDC25C-cyclinB/CDC2, and p53/p21 pathways were involved in the MCPH1 overexpression-induced S phase arrest. Moreover, the overexpression of MCPH1 activated mitochondrial apoptosis through regulating several apoptosis-related proteins such as p53, Bcl-2, Bax, cytochrome c, caspase-3, and PARP-1. Our findings indicate that downregulated MCPH1 correlates with tumor progression in cervical cancer, and MCPH1 has an important role in regulating cell growth through regulating the cell cycle and apoptosis. Thus, it may be a crucial tumor suppressor gene and a novel candidate therapeutic target for cervical cancer.


Sujet(s)
Caspase-3/métabolisme , Protéines du cycle cellulaire/physiologie , Division cellulaire/physiologie , Cytochromes c/métabolisme , Protéines de tissu nerveux/physiologie , Tumeurs du col de l'utérus/anatomopathologie , Séquence nucléotidique , Lignée cellulaire , Protéines du cytosquelette , Amorces ADN , Femelle , Humains , Protéines de tissu nerveux/métabolisme , Réaction de polymérisation en chaine en temps réel , Transduction du signal , Tumeurs du col de l'utérus/enzymologie , Tumeurs du col de l'utérus/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE