Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Sci ; 115(4): 1224-1240, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38403332

ABSTRACT

The transcription factor forkhead box protein O1 (FoxO1) is closely related to the occurrence and development of ovarian cancer (OC), however its role and molecular mechanisms remain unclear. Herein, we found that FoxO1 was highly expressed in clinical samples of OC patients and was significantly correlated with poor prognosis. FoxO1 knockdown inhibited the proliferation of OC cells in vitro and in vivo. ChIP-seq combined with GEPIA2 and Kaplan-Meier database analysis showed that structural maintenance of chromosome 4 (SMC4) is a downstream target of FoxO1, and FoxO1 promotes SMC4 transcription by binding to its -1400/-1390 bp promoter. The high expression of SMC4 significantly blocked the tumor inhibition effect of FoxO1 knockdown. Furtherly, FoxO1 increased SMC4 mRNA abundance by transcriptionally activating methyltransferase-like 14 (METTL14) and increasing SMC4 m6A methylation on its coding sequence region. The Cancer Genome Atlas dataset analysis confirmed a significant positive correlation between FoxO1, SMC4, and METTL14 expression in OC. In summary, this study revealed the molecular mechanisms of FoxO1 regulating SMC4 and established a clinical link between the expression of FoxO1/METTL14/SMC4 in the occurrence of OC, thus providing a potential diagnostic target and therapeutic strategy.


Subject(s)
Chromosomes, Human, Pair 4 , Ovarian Neoplasms , Female , Humans , Adenosine Triphosphatases/genetics , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human, Pair 4/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Kaplan-Meier Estimate , Methyltransferases/genetics , Ovarian Neoplasms/pathology
2.
J Transl Med ; 22(1): 58, 2024 01 14.
Article in English | MEDLINE | ID: mdl-38221609

ABSTRACT

BACKGROUND: Chimeric antigen receptor CAR-T cell therapies have ushered in a new era of treatment for specific blood cancers, offering unparalleled efficacy in cases of treatment resistance or relapse. However, the emergence of cytokine release syndrome (CRS) as a side effect poses a challenge to the widespread application of CAR-T cell therapies. Melatonin, a natural hormone produced by the pineal gland known for its antioxidant and anti-inflammatory properties, has been explored for its potential immunomodulatory effects. Despite this, its specific role in mitigating CAR-T cell-induced CRS remains poorly understood. METHODS: In this study, our aim was to investigate the potential of melatonin as an immunomodulatory agent in the context of CD19-targeting CAR-T cell therapy and its impact on associated side effects. Using a mouse model, we evaluated the effects of melatonin on CAR-T cell-induced CRS and overall survival. Additionally, we assessed whether melatonin administration had any detrimental effects on the antitumor efficacy and persistence of CD19 CAR-T cells. RESULTS: Our findings demonstrate that melatonin effectively mitigated the severity of CAR-T cell-induced CRS in the mouse model, leading to improved overall survival outcomes. Remarkably, melatonin administration did not compromise the antitumor effectiveness or persistence of CD19 CAR-T cells, indicating its compatibility with therapeutic goals. These results suggest melatonin's potential as an immunomodulatory compound to alleviate CRS without compromising the therapeutic benefits of CAR-T cell therapy. CONCLUSION: The study's outcomes shed light on melatonin's promise as a valuable addition to the existing treatment protocols for CAR-T cell therapies. By attenuating CAR-T cell-induced CRS while preserving the therapeutic impact of CAR-T cells, melatonin offers a potential strategy for optimizing and refining the safety and efficacy profile of CAR-T cell therapy. This research contributes to the evolving understanding of how to harness immunomodulatory agents to enhance the clinical application of innovative cancer treatments.


Subject(s)
Cytokine Release Syndrome , Immunotherapy, Adoptive , Melatonin , Antigens, CD19 , Cell- and Tissue-Based Therapy , Cytokine Release Syndrome/therapy , Immunologic Factors/pharmacology , Immunotherapy, Adoptive/adverse effects , Melatonin/pharmacology , Neoplasm Recurrence, Local , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Animals , Mice
3.
FASEB J ; 36(6): e22362, 2022 06.
Article in English | MEDLINE | ID: mdl-35608902

ABSTRACT

Malignant tumors display profound changes in cellular metabolism, yet how these altered metabolites affect the development and growth of tumors is not fully understood. Here, we used metabolomics to analyze the metabolic profile differences in ovarian cancer and found that citric acid (CA) is the most significantly downregulated metabolite. Recently, CA has been reported to inhibit the growth of a variety of tumor cells, but whether it is involved in pyroptosis of ovarian cancer and its potential molecular mechanisms still remains to be further investigated. Here, we demonstrated that CA inhibits the growth of ovarian cancer cells in a dose-dependent manner. RNA-seq analysis revealed that CA significantly promoted the expression of thioredoxin interacting protein (TXNIP) and caspase-4 (CASP4). Morphologic examination by transmission electron microscopy indicated that CA-treated ovarian cancer cells exhibited typical pyroptosis characteristics. Further mechanistic analyses showed that CA facilitates pyroptosis via the CASP4/TXNIP-NLRP3-Gesdermin-d (GSDMD) pathway in ovarian cancer. This study elucidated that CA induces ovarian cancer cell death through classical and non-classical pyroptosis pathways, which may be beneficial as an ovarian cancer therapy.


Subject(s)
Ovarian Neoplasms , Pyroptosis , Carcinoma, Ovarian Epithelial , Carrier Proteins , Caspase 1/metabolism , Citric Acid , Female , Humans , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins
4.
Soft Matter ; 19(43): 8423-8433, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37877309

ABSTRACT

To exploit the chemical asymmetry of diblock copolymer chains on the design of high-performance switch sensors, we propose an analytically tractable model system which contains an adsorption-responsive diblock copolymer in an otherwise inert brush, and study its phase transitions by using both analytical theory and self-consistent field calculations. The copolymer chain is chemically asymmetric in the sense that the two blocks assume different adsorption strengths, which is characterized by the defined adsorption ratio. We found that the conformation states, the number of stable phases, and transition types are mainly controlled by the length of each block and the adsorption ratio. In particular, when the length of the ungrafted block is longer than the brush chains, and the adsorption ratio is smaller than a critical value, the copolymer chain shows three thermodynamically stable states, and undergoes two unsynchronous transitions, where the two blocks respond to the adsorption in a different manner, when the adsorption changes from weak to sufficiently strong. For this kind of three-state transition, the transition point, transition barrier, and transition width are evaluated by using the self-consistent field method, and their scaling relationship with respect to the system parameters is extracted, which matches reasonably well with the predictions from the analytical theory. The self-consistent field calculations also indicate that the conformational transitions involved in the three-state transition process are sharp with a low energy barrier, and interestingly, barrier-free transitions are observed. Our finding shows that the three-state transitions not only specify a region where high performance unsynchronous switch sensors can be exploited, but may also provide a useful model understanding the unsynchronous biological processes.

5.
Mol Biol Rep ; 50(11): 9229-9237, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37805662

ABSTRACT

BACKGROUND: Precise quantification of grafted human cells in preclinical animal models such as non-human primates, rodents and rabbits is needed for the evaluations of the safety and efficacy of cell therapy. Quantitative PCR (qPCR) as a swift, sensitive and powerful assay is suitable for human cell quantification. However, it is a formidable challenge due to that the genome of non-human primates share more than 95% of similarity as human. METHODS: In the present study, we developed a probe-based quantitative PCR (qPCR) assay for the quantification of human cells in preclinical animal models via targeting human specific DNA in the intron of BRCA1 (termed BRCA1-qPCR). The 5' and 3' end of BRCA1-qPCR probe was conjugated with FAM and non-fluorescent quencher-minor groove binder (NFQ-MGB), respectively. 1 µg of genomic DNA from human and preclinical animal models including rhesus monkeys, cynomolgus monkeys, New Zealand white rabbits, SD rats, C57BL/6 and BALB/c mice were used for determining the specificity and sensitivity of the BRCA1-qPCR assay. A calibration curve was generated by BRCA1-qPCR analysis of linearized plasmid containing targeted human specific DNA in BRCA1. The BRCA1-qPCR assay was validated by analysis of 0.003%, 0.03% and 0.3% of human leukocytes mixed within murine leukocytes. RESULTS: The BRCA1-qPCR assay detected human DNA rather than DNA from tested species. The amplification efficiency of the BRCA1-qPCR assay was 95.4% and the linearity of the calibration curve was R2 = 0.9997. The BRCA1-qPCR assay detected as low as 5 copies of human specific DNA and is efficient to specially amplify 30 pg human DNA in the presence of 1 µg of genomic DNA from tested species, respectively. The BRCA1-qPCR assay was able to quantify as low as 0.003% of human cells within murine leukocytes. CONCLUSION: The BRCA1-qPCR assay is efficient for the quantification of human cells in preclinical animal models.


Subject(s)
DNA , Primates , Humans , Animals , Rats , Mice , Rabbits , Introns , Mice, Inbred C57BL , Rats, Sprague-Dawley , Polymerase Chain Reaction , Models, Animal , BRCA1 Protein/genetics
6.
Cytometry A ; 101(2): 150-158, 2022 02.
Article in English | MEDLINE | ID: mdl-34173319

ABSTRACT

Human basophils are terminally differentiated granulocytes that are least abundant in the peripheral blood but play important roles in allergic diseases. Studies on human basophils are limited by the high cost on the isolation of human basophils by magnetic-activated cell sorting (MACS) for negative depletion of non-basophils, followed by CD123-based positive selection of basophils. Moreover, such CD123-based purification of basophils may be limited by blocking of the binding of IL-3/anti-CD123 to the surface CD123. Here we identified SSClow CD4- CD127- HLA-DR- CRTH2high as unique markers for the identification of human basophils through stringent flow cytometric analysis of leukocytes from buffy coat. We established an efficient and cost-effective method for isolating human basophils from buffy coat based on positive magnetic selection of CRTH2+ cells followed by flow cytometric sorting of SSClow CD4- CD127- HLA-DR- CRTH2high cells. Approximately 1 to 1.5 million basophils were isolated from one buffy coat with a purity of >97%. Basophils purified by this method were viable and efficiently responded to key regulators of basophils including IL-3 and anti-IgE. This method can be used for purifying human basophils for subsequent functional studies.


Subject(s)
Basophils , Interleukin-3 Receptor alpha Subunit , Cost-Benefit Analysis , HLA-DR Antigens , Humans , Interleukin-3/metabolism , Interleukin-3 Receptor alpha Subunit/metabolism
7.
Int J Mol Sci ; 23(20)2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36293287

ABSTRACT

Phosphatidyl ethanolamine-binding proteins (PEBPs) are involved in regulating flowering time and various developmental processes. Functions and expression patterns in cultivated peanuts (Arachis hypogaea L.) remain unknown. In this study, 33 PEBP genes in cultivated peanuts were identified and divided into four subgroups: FT, TFL, MFT and FT-like. Gene structure analysis showed that orthologs from A and B genomes in cultivated peanuts had highly similar structures, but some orthologous genes have subgenomic dominance. Gene collinearity and phylogenetic analysis explain that some PEBP genes play key roles in evolution. Cis-element analysis revealed that PEBP genes are mainly regulated by hormones, light signals and stress-related pathways. Multiple PEPB genes had different expression patterns between early and late-flowering genotypes. Further detection of its response to temperature and photoperiod revealed that PEBPs ArahyM2THPA, ArahyEM6VH3, Arahy4GAQ4U, ArahyIZ8FG5, ArahyG6F3P2, ArahyLUT2QN, ArahyDYRS20 and ArahyBBG51B were the key genes controlling the flowering response to different flowering time genotypes, photoperiods and temperature. This study laid the foundation for the functional study of the PEBP gene in cultivated peanuts and the adaptation of peanuts to different environments.


Subject(s)
Arachis , Gene Expression Regulation, Plant , Arachis/genetics , Arachis/metabolism , Phylogeny , Flowers/metabolism , Plant Proteins/metabolism , Genomics , Hormones/metabolism , Ethanolamines/metabolism
8.
Immunology ; 164(2): 292-304, 2021 10.
Article in English | MEDLINE | ID: mdl-33999409

ABSTRACT

Allergic diseases are caused by dysregulated Th2 immune responses involving multiple effector cells including basophils. Short chain fatty acids (SCFAs), mainly acetate, propionate and butyrate, exert immunomodulatory functions via activation of its receptors GPR41 and GPR43, and inhibition of the histone deacetylases (HDACs) activity. In allergic diseases, SCFAs suppress the activity of mast cells, eosinophils and type 2 innate lymphoid cells (ILC2) but enhance the function of Th2 cells. Here, we aimed to elucidate the function of SCFAs on human basophils. Human basophils were purified from healthy donors by flow cytometric sorting. The surface proteins, apoptosis and degranulation of basophils were analyzed by flow cytometric analysis. The mRNA expression was assayed using real-time PCR. Interleukin 4 (IL-4) and IL-13 were measured by ELISA. Histone acetylation was examined by western blot. GPR41 was expressed by basophils and was enhanced by IL-3. Acetate induced intracellular calcium influx in basophils which was suppressed by blocking GPR41. Propionate and butyrate, but not acetate, induced the expression of CD69 and IL-13. In addition, propionate and butyrate enhanced IgE-mediated basophil degranulation but inhibited basophil survival and IL-4 secretion. Propionate and butyrate induced histone acetylation of basophils and suppression of HDACs activity mimicked the effects of propionate and butyrate on human basophils. Our findings demonstrate that propionate and butyrate may play a complex role in regulating basophil apoptosis, activation and degranulation via inhibiting HDACs activity. The in vivo effects of SCFAs on the regulation of basophil-associated allergic diseases need to be further explored.


Subject(s)
Apoptosis/drug effects , Basophils/drug effects , Butyrates/pharmacology , Histones/metabolism , Interleukin-13/metabolism , Propionates/pharmacology , Apoptosis/immunology , Basophils/immunology , Basophils/metabolism , Cells, Cultured , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/metabolism , Fatty Acids, Volatile , Gene Expression/drug effects , Gene Expression/immunology , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunity, Innate/drug effects , Immunity, Innate/immunology , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
9.
Mov Disord ; 36(8): 1911-1918, 2021 08.
Article in English | MEDLINE | ID: mdl-33876851

ABSTRACT

BACKGROUND: The accumulation of α-synuclein (α-Syn) aggregates that leads to the onset of Parkinson's disease (PD) has been postulated to begin in the gastrointestinal tract. The normal human appendix contains pathogenic forms of α-Syn, and appendectomy has been reported to affect the incidence of PD. OBJECTIVE: This study investigated appendix abnormality in patients with PD. METHODS: We assessed appendix morphology in 100 patients with PD and 50 control subjects by multislice spiral computed tomography. We analyzed the clinical characteristics of patients with PD with diseased appendices, which was confirmed in seven patients by histopathological analysis. RESULTS: Chronic appendicitis-like lesions were detected in 53% of patients with PD, but these were not associated with the duration of motor symptoms. Appendicitis-like lesions, impaired olfaction, and rapid eye movement sleep behavior disorder were risk factors for PD. The following clinical symptoms could be used to identify patients with PD with appendicitis-like lesions: first motor symptoms were bradykinesia/rigidity, onset of motor symptoms in the central axis or left limb, prodromal constipation, high ratio of Unified Parkinson's Disease Rating Scale Part III score to symptom duration, low Montreal Cognitive Assessment score, and high Epworth Sleepiness Scale score. The seven patients with PD who were diagnosed with chronic appendicitis underwent appendectomy, and histopathological analysis revealed structural changes associated with chronic appendicitis and α-Syn aggregation. CONCLUSIONS: These results indicate an association between chronic appendicitis-like lesions and PD, and suggest that α-Syn accumulation in the diseased appendix occurs in PD. © 2021 International Parkinson and Movement Disorder Society.


Subject(s)
Appendix , Parkinson Disease , REM Sleep Behavior Disorder , Appendectomy , Humans , Parkinson Disease/complications , Parkinson Disease/diagnostic imaging , alpha-Synuclein
10.
Int J Med Sci ; 18(14): 3125-3139, 2021.
Article in English | MEDLINE | ID: mdl-34400883

ABSTRACT

Background: Regulating the polarization of macrophages to antitumor M1 macrophages is a promising strategy for overcoming the immunosuppression of the tumor microenvironment for cancer therapy. Ferumoxytol (FMT) can not only serve as a drug deliver agent but also exerts anti-tumor activity. ß-glucan has immuno-modulating properties to prevent tumor growth. Thus, a nanocomposite of FMT surface-coated with ß-glucan (FMT-ß-glucan) was prepared to explore its effect on tumor suppression. Methods: Male B16F10 melanoma mouse model was established to explore the antitumor effect of FMT-ß-glucan. The viability and apoptotic rates of B16F10 cells were detected by cell counting kit-8 and Annexin-V/PI experiments. The levels of M1 markers were quantified by quantitative reverse transcription-polymerase chain reaction and enzyme linked immunosorbent assay. Phagocytic activity and intracellular reactive oxygen species (ROS) in macrophages were evaluated by the neutral red uptake assay and flow cytometry, respectively. Small interfering RNA (siRNA) transfection was applied to knock down the Dectin-1 gene in RAW 264.7 cells. Results: FMT-ß-glucan suppressed tumor growth to a greater extent and induced higher infiltration of M1 macrophages than the combination of FMT and ß-glucan (FMT+ß-glucan) in vivo. In vitro, supernatant from FMT-ß-glucan-treated RAW 264.7 cells led to lower cell viability and induced more apoptosis of B16F10 cells than that from the FMT+ß-glucan group. Moreover, FMT-ß-glucan boosted the expression of M1 type markers, and increased phagocytic activity and ROS in RAW 264.7 cells. Further research indicated that FMT-ß-glucan treatment promoted the level of Dectin-1 on the surface of RAW 264.7 cells and that knockdown of Dectin-1 abrogated the phosphorylation levels of several components in MAPK and NF-κB signaling. Conclusion: The nanocomposite FMT-ß-glucan suppressed melanoma growth by inducing the M1 macrophage-activated tumor microenvironment.


Subject(s)
Ferrosoferric Oxide/pharmacology , Lectins, C-Type/agonists , Melanoma/drug therapy , Skin Neoplasms/drug therapy , beta-Glucans/pharmacology , Animals , Disease Models, Animal , Ferrosoferric Oxide/chemistry , Ferrosoferric Oxide/therapeutic use , Gene Knockdown Techniques , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Melanoma/immunology , Melanoma/pathology , Mice , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , beta-Glucans/chemistry , beta-Glucans/therapeutic use
11.
Environ Toxicol ; 36(3): 308-319, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33035382

ABSTRACT

Prostate cancer (PCa) has become the second leading cause of cancer-related mortality in males worldwide. Although the long noncoding RNA DLX6-AS1 has been recognized to be an oncogene in multiple cancers, the biological function and regulatory mechanism of DLX6-AS1 in prostate cancer are still obscure. In the present study, we observed that DLX6-AS1 was significantly upregulated in PCa tissues and cells. Knockdown of DLX6-AS1 inhibited PCa progression by suppressing cell proliferation and accelerating cell apoptosis. Molecular mechanism exploration indicated that DLX6-AS1 acted as a sponge for miR-497-5p and synuclein gamma (SNCG) was a downstream target gene of miR-497-5p. In addition, there was a negative correlation between DLX6-AS1 and miR-497-5p in PCa tissues. Rescue assays showed that SNCG overexpression could partially recover DLX6-AS1 knockdown-mediated inhibition of progression in PCa. Furthermore, xenograft tumor model was established to determine the role of DLX6-AS1 in PCa tumor growth and the results suggested that DLX6-AS1 could facilitate tumor growth by regulating SNCG in vivo. In conclusion, our study investigated the biological function and underlying mechanism of DLX6-AS1 in PCa and validated that DLX6-AS1 functioned as an oncogene through miR-497-5p/SNCG axis.


Subject(s)
Cell Proliferation/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Apoptosis , Disease Progression , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Neoplasm Proteins/genetics , RNA, Long Noncoding/genetics , Up-Regulation , gamma-Synuclein/genetics , gamma-Synuclein/metabolism
12.
Mol Cancer ; 19(1): 140, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32907590

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

13.
Cytokine ; 127: 154934, 2020 03.
Article in English | MEDLINE | ID: mdl-31783259

ABSTRACT

Basophils are important effector cells in allergic disorders and anti-parasitic immune response. A number of activators including interleukin 3 (IL-3) and IgE have been identified in the regulation of human basophils expressing mediators such as histamine and leukotriene C4 (LTC4) and cytokines, including IL-4 and IL-13. Human basophils express high levels of IL-2 receptors. However, the function of the IL-2 pathway in basophils remains unknown. Here, we identified that IL-2 induced the activation of human basophils in vitro to express a variety of inflammatory cytokines and chemokines including IL-5, IL-13, GM-CSF and CCL-17. This effect by IL-2 is confirmed by an upstream regulator analysis using Ingenuity pathway analysis. Of note, one of the top regulated cytokines, IL-5, was for the first time identified to be induced by IL-2 in human basophils rather than IL-3 or anti-IgE. Immunofluorescence analysis of skin specimens from bullous pemphigoid and eczema revealed that infiltrating basophils in skin lesions widely expressed IL-5 and GM-CSF. Together, our findings reveal IL-2 as a novel regulator of human basophils. This adds a new layer to support the importance of basophils in allergic disorders.


Subject(s)
Basophils/drug effects , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Interleukin-2/pharmacology , Interleukin-5/genetics , Apoptosis/drug effects , Apoptosis/genetics , Basophils/immunology , Basophils/metabolism , Cells, Cultured , Dermatitis, Atopic/genetics , Dermatitis, Atopic/metabolism , Eczema/genetics , Eczema/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-5/metabolism , Pemphigoid, Bullous/genetics , Pemphigoid, Bullous/metabolism , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology
14.
Andrologia ; 52(10): e13750, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32668495

ABSTRACT

Prostate cancer (PCa) is known as one of the most common cancers in men all over the world. Previous studies have identified that the pro-inflammatory mediator interleukin-17F (IL-17F) aggravates the progression of several diseases. However, whether IL-17F plays a role in PCa is still lack of enough exploration. In this study, IL-17F expression was strikingly upregulated in PCa tissues. Treatment of IL-17F promoted cell viability at a dose-dependent manner. Further, functional assays were implemented by treatment of 100 ng/ml of IL-17F. Cell viability, proliferation, migration, invasion and stemness were promoted by 100 ng/ml of IL-17F. IL-17F increased the expression of p-PI3K and p-AKT in PCa cells, indicating that IL-17F might activate the PI3K/AKT signalling pathway in PCa cells. LY294002 (the inhibitor of the PI3K/AKT signalling pathway) could reverse the facilitating effects of IL-17F treatment on PCa cell viability, proliferation, migration, invasion and stemness. Taken together, current study revealed that IL-17F facilitated PCa cell malignant phenotypes via activation of the PI3K/AKT signalling pathway, offering a potential therapeutic target for PCa.


Subject(s)
Phosphatidylinositol 3-Kinases , Prostatic Neoplasms , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Interleukin-17 , Male , Phenotype , Proto-Oncogene Proteins c-akt
15.
Int J Mol Sci ; 21(6)2020 Mar 11.
Article in English | MEDLINE | ID: mdl-32168930

ABSTRACT

Plants tolerate cold stress by regulating gene networks controlling cellular and physiological traits to modify growth and development. Transcription factor (TF)-directed regulation of transcription within these gene networks is key to eliciting appropriate responses. Identifying TFs related to cold tolerance contributes to cold-tolerant crop breeding. In this study, a comparative transcriptome analysis was carried out to investigate global gene expression of entire TFs in two peanut varieties with different cold-tolerant abilities. A total of 87 TF families including 2328 TF genes were identified. Among them, 445 TF genes were significantly differentially expressed in two peanut varieties under cold stress. The TF families represented by the largest numbers of differentially expressed members were bHLH (basic helix-loop-helix protein), C2H2 (Cys2/His2 zinc finger protein), ERF (ethylene-responsive factor), MYB (v-myb avian myeloblastosis viral oncogene homolog), NAC (NAM, ATAF1/2, CUC2) and WRKY TFs. Phylogenetic evolutionary analysis, temporal expression profiling, protein-protein interaction (PPI) network, and functional enrichment of differentially expressed TFs revealed the importance of plant hormone signal transduction and plant-pathogen interaction pathways and their possible mechanism in peanut cold tolerance. This study contributes to a better understanding of the complex mechanism of TFs in response to cold stress in peanut and provides valuable resources for the investigation of evolutionary history and biological functions of peanut TFs genes involved in cold tolerance.


Subject(s)
Arachis/growth & development , Data Mining/methods , Gene Expression Profiling/methods , Transcription Factors/genetics , Arachis/genetics , Cold-Shock Response , Evolution, Molecular , Gene Expression Regulation, Plant , Phylogeny , Plant Breeding , Plant Proteins/genetics , Protein Interaction Maps
16.
J Cell Physiol ; 234(5): 7051-7061, 2019 05.
Article in English | MEDLINE | ID: mdl-30387134

ABSTRACT

Type I interferon (IFN) signaling in neoplastic cells has a chemo-sensitizing effect in cancer therapy. Toll-like receptor 3 (TLR3) activation promotes IFN-ß production, which induces apoptosis and impairs proliferation in some cancer cells. Herein, we tested whether the TLR3 agonist polyinosinic: polycytidylic acid (poly I:C) can improve chemotherapeutic efficacy in paclitaxel (PTX) resistant cell lines. Human colon cancer cell lines HCT116, SW620, HCT-8 (sensitive to PTX), and HCT-8/PTX (resistant to PTX) were treated with poly I:C and the cell viability was measured. Results showed that poly I:C specifically impaired the cell viability of HCT-8/PTX by simultaneously promoting cell apoptosis and inhibiting cell proliferation. In addition, when TLR3 was overexpressed in HCT-8/PTX cells, we found that TLR3 contributed to the production of IFN-ß that reduced cell viability, and poly I:C preferentially activated the TLR3-UNC93B1 signaling pathway to mediate this effect. Moreover, cotreatment of poly I:C and PTX acted synergistically to induce cell apoptosis of HCT-8/PTX via upregulating the expression of TLR3 and its molecular chaperone UNC93B1, assisting in the secretion of IFN-ß. Notably, a combination of poly I:C and PTX synergistically inhibited the PTX-resistant tumor growth in vivo without side effects. In conclusion, our studies demonstrate that poly I:C reinforces the potency of cytotoxic chemotherapeutics in PTX-resistant cell line through the TLR3-UNC93B1-IFN-ß signaling pathway, which supplies a novel mechanism of poly I:C for the chemotherapy sensitizing effect in a PTX-resistant tumor.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Interferon-beta/metabolism , Membrane Transport Proteins/metabolism , Paclitaxel/pharmacology , Poly I-C/pharmacology , Toll-Like Receptor 3/agonists , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Interferon-beta/genetics , Membrane Transport Proteins/genetics , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
Mol Cancer ; 18(1): 25, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30777076

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) are a class of non-coding RNAs with a loop structure, but its functions remain largely unknown. Growing evidence has revealed that circRNAs play a striking role as functional RNAs in the progression of malignant disease. However, the precise role of circRNAs in gastric cancer (GC) remains unclear. METHODS: CircRNAs were determined by human circRNA array analysis and quantitative reverse transcription polymerase reaction. Luciferase reporter, RNA pull down, and fluorescence in situ hybridization assays were employed to test the interaction between circPSMC3 and miR-296-5p. Ectopic over-expression and siRNA-mediated knockdown of circPSMC3, proliferation, migration and invasion in vitro, and in vivo experiment of metastasis were used to evaluate the function of circPSMC3. RESULTS: CircPSMC3 rather than liner PSMC3 mRNA was down-regulated in GC tissues, corresponding plasmas from GC patients as well as GC cell lines compared to normal controls. Lower circPSMC3 expression in GC patients was correlated with higher TNM stage and shorter overall survival. Over-expression of circPSMC3 and miR-296-5p inhibitor could inhibit the tumorigenesis of gastric cancer cells in vivo and vitro whereas co-transfection of circPSMC3 and miRNA-296-5p could counteract this effect. Importantly, we demonstrated that circPSMC3 could act as a sponge of miR-296-5p to regulate the expression of Phosphatase and Tensin Homolog (PTEN), and further suppress the tumorigenesis of gastric cancer cells. CONCLUSION: Our study reveals that circPSMC3 can serve as a novel potential circulating biomarker for detection of GC. CircPSMC3 participates in progression of gastric cancer by sponging miRNA-296-5p with PTEN, providing a new insight into the treatment of gastric cancer.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Proteasome Endopeptidase Complex/genetics , RNA/genetics , Stomach Neoplasms/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Heterografts , Humans , Lymphatic Metastasis , Male , Mice , MicroRNAs/metabolism , Middle Aged , Neoplasm Staging , PTEN Phosphohydrolase/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA/antagonists & inhibitors , RNA/metabolism , RNA, Circular , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Analysis
18.
Biochem Biophys Res Commun ; 515(2): 345-351, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31153637

ABSTRACT

Prostate cancer (PCa) is a major malignancy in men. Hitherto that date, surgical or chemical castration is the standard treatment to PCa. Nevertheless, there are still many patients with initial treatment progress to metastatic castration-resistant prostate cancer (mCRPC). There are many effective chemotherapeutic drugs for mCRPC, but the tumors will be resistant to these chemotherapeutic drugs, which is an urgent problem to be solved. Specifically, tumor therapy resistance driven by the pathologically active host stroma has gradually becoming the spotlight of oncotherapy in recent years. The exosome-derived miR-27a plays an important role in PCa cell chemoresistance. However, the functions of miR-27a on PCa developing chemoresistance remain unknown. In the present study, we aimed to construct potential regulatory networks of exosomal miR-27a in PCa chemoresistance. The expression of miR-27a was significantly increased by treatment with cisplatin, doxorubicin (DOX) and docetaxel in PCa tissues. We next co-cultured PCa cells (PC3 cells) with primary prostate fibroblasts (PSC27 cells) to explore the mechanisms of tumor therapy resistance. Further studies delineate that exosome-derived miR-27a produced by PSC-27 cells improved chemoresistance by restraining the expression of P53 gene. Our studies provide a new direction for exploring the effects of PCa tumor microenvironment of chemoresistance.


Subject(s)
Genes, p53 , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cells, Cultured , Cisplatin/pharmacology , Coculture Techniques , Docetaxel/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Exosomes/genetics , Exosomes/metabolism , Fibroblasts/metabolism , Gene Expression/drug effects , Humans , Male , Prostate/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
19.
Microb Cell Fact ; 18(1): 55, 2019 Mar 18.
Article in English | MEDLINE | ID: mdl-30885243

ABSTRACT

Naturally occurring carotenoids have been isolated and used as colorants, antioxidants, nutrients, etc. in many fields. There is an ever-growing demand for carotenoids production. To comfort this, microbial production of carotenoids is an attractive alternative to current extraction from natural sources. This review summarizes the biosynthetic pathway of carotenoids and progresses in metabolic engineering of various microorganisms for carotenoid production. The advances in synthetic pathway and systems biology lead to many versatile engineering tools available to manipulate microorganisms. In this context, challenges and possible directions are also discussed to provide an insight of microbial engineering for improved production of carotenoids in the future.


Subject(s)
Bacterial Physiological Phenomena , Carotenoids/biosynthesis , Carotenoids/genetics , Metabolic Engineering/methods , Microorganisms, Genetically-Modified/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL