Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1344637, 2024.
Article in English | MEDLINE | ID: mdl-38962013

ABSTRACT

Disulfidptosis, a regulated form of cell death, has been recently reported in cancers characterized by high SLC7A11 expression, including invasive breast carcinoma, lung adenocarcinoma, and hepatocellular carcinoma. However, its role in colon adenocarcinoma (COAD) has been infrequently discussed. In this study, we developed and validated a prognostic model based on 20 disulfidptosis-related genes (DRGs) using LASSO and Cox regression analyses. The robustness and practicality of this model were assessed via a nomogram. Subsequent correlation and enrichment analysis revealed a relationship between the risk score, several critical cancer-related biological processes, immune cell infiltration, and the expression of oncogenes and cell senescence-related genes. POU4F1, a significant component of our model, might function as an oncogene due to its upregulation in COAD tumors and its positive correlation with oncogene expression. In vitro assays demonstrated that POU4F1 knockdown noticeably decreased cell proliferation and migration but increased cell senescence in COAD cells. We further investigated the regulatory role of the DRG in disulfidptosis by culturing cells in a glucose-deprived medium. In summary, our research revealed and confirmed a DRG-based risk prediction model for COAD patients and verified the role of POU4F1 in promoting cell proliferation, migration, and disulfidptosis.


Subject(s)
Adenocarcinoma , Biomarkers, Tumor , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/diagnosis , Prognosis , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Biomarkers, Tumor/genetics , Female , Cell Line, Tumor , Male , Cell Proliferation/genetics , Gene Expression Profiling , Transcriptome , Nomograms , Octamer Transcription Factor-3/genetics , Cell Movement/genetics
2.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 214-219, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814210

ABSTRACT

This study explored the role of circular RNA circ_0006168 in the progression of hepatocellular carcinoma (HCC) and its interaction with microRNA-125b. The expression of circ_0006168 was examined in 42 pairs of HCC tumor and adjacent tissue specimens using quantitative polymerase chain reaction (qPCR). Elevated circ_0006168 expression in HCC tissues was significantly associated with advanced pathological staging and lower overall survival rates. Lentivirus-mediated circ_0006168 knockdown in HCC cell lines (Hep3B and Huh7) demonstrated a notable reduction in cell proliferation and an increase in apoptosis. MicroRNA-125b expression exhibited a marked reduction in HCC tissues, negatively correlating with circ_0006168 levels. Luciferase reporting assays indicated that circ_0006168 was a direct target of microRNA-125b. Additionally, cell recovery experiments suggested a reciprocal regulation between circ_0006168 and microRNA-125b, contributing to the accelerated malignant progression of HCC. The study underscored the significantly increased expression of circ_0006168 in both HCC tissues and cell lines, highlighting its association with advanced pathological stages and poor prognosis in HCC patients. Furthermore, circ_0006168 appeared to play a pivotal role in elevating the proliferation rate of HCC cells through its modulation of microRNA-125b. These findings contribute to a deeper understanding of the molecular mechanisms underlying HCC development and may offer potential therapeutic targets for intervention.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular , Cell Proliferation , Gene Expression Regulation, Neoplastic , Liver Neoplasms , MicroRNAs , RNA, Circular , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , RNA, Circular/genetics , RNA, Circular/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , Male , Female , Middle Aged , Apoptosis/genetics , Prognosis , Base Sequence , Disease Progression
3.
Cells ; 12(22)2023 11 10.
Article in English | MEDLINE | ID: mdl-37998339

ABSTRACT

E. coli is a ubiquitous pathogen that is responsible for over one million fatalities worldwide on an annual basis. In animals, E. coli can cause a variety of diseases, including mastitis in dairy cattle, which represents a potential public health hazard. However, the pathophysiology of E. coli remains unclear. We found that E. coli could induce global upregulation of m6A methylation and cause serious apoptosis in bovine mammary epithelial cells (MAC-T cells). Furthermore, numerous m6A-modified lncRNAs were identified through MeRIP-seq. Interestingly, we found that the expression of LOC4191 with hypomethylation increased in MAC-T cells upon E. coli-induced apoptosis. Knocking down LOC4191 promoted E. coli-induced apoptosis and ROS levels through the caspase 3-PARP pathway. Meanwhile, knocking down ALKBH5 resulted in the promotion of apoptosis through upregulated ROS and arrested the cell cycle in MAC-T cells. ALKBH5 silencing accelerated LOC4191 decay by upregulating its m6A modification level, and the process was recognized by hnRNP A1. Therefore, this indicates that ALKBH5 stabilizes m6A-modified LOC4191 to suppress E. coli-induced apoptosis. This report discusses an initial investigation into the mechanism of m6A-modified lncRNA in cells under E. coli-induced apoptosis and provides novel insights into infectious diseases.


Subject(s)
Apoptosis , Escherichia coli , Female , Animals , Cattle , Escherichia coli/metabolism , Reactive Oxygen Species/metabolism , Apoptosis/genetics , DNA Methylation
4.
Acta Biochim Pol ; 70(4): 799-806, 2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37788370

ABSTRACT

Multidrug resistance severely limits the efficacy of ovarian cancer (OC) treatment. Recent studies have revealed the carcinogenic role of LINC00707 RNA. However, the role of LINC00707 in the development of multidrug resistance in OC has not been clarified. Therefore, the aim of this study was to investigate the relationship between LINC00707 and multidrug resistance in OC, which can facilitate the development of new therapeutic agents for effectively addressing this issue. The RNA expression of LINC00707, miR-382-5p and leucine-rich repeat kinase 2 (LRRK2) in SKOV3 (a human OC cell line) cells was detected by qRT-PCR. The effects of LINC00707 on the proliferation and viability of SKOV3 cells were determined by MTT assay and colony formation assay. The interaction of LINC00707, miR-382-5p, and LRRK2 was bioinformatically predicted and verified with dual-luciferase reporter assay. In addition, the effect of LINC00707 on drug resistance in SKOV3 cells through targeting the miR-382-5p/LRRK2 axis was explored. The expression of LINC00707 and LRRK2 was significantly increased in SKOV3 cells, while miR-382-5p expression was significantly decreased. The results of bioinformatic prediction and colony formation assay demonstrated that LINC00707 could regulate LRRK2 expression in SKOV3 cells by targeting miR-382-5p. Additionally, knockdown of LINC00707 markedly increased expression of miR-382-5p and decreased that of LRRK2, increased cell proliferation and viability, as well as sensitivity to chemotherapeutic agents in SKOV3 cells. Notably, these manifestations were more obvious with simultaneous knockdown of LINC00707 and miR-382-5p compared with knockdown of LINC00707 alone. LINC00707 is overexpressed in SKOV3 cells and promotes SKOV3 cell proliferation and resistance to chemotherapeutic drugs via targeting the miR-382-5p/LRRK2 axis.


Subject(s)
MicroRNAs , Ovarian Neoplasms , RNA, Long Noncoding , Humans , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Cell Proliferation/genetics , Drug Resistance, Multiple/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics
5.
Oxid Med Cell Longev ; 2022: 9744771, 2022.
Article in English | MEDLINE | ID: mdl-36578520

ABSTRACT

N6-Methyladenosine (m6A) is the most abundant epigenetic RNA modification in eukaryotes, regulating RNA metabolism (export, stability, translation, and decay) in cells through changes in the activity of writers, erasers, and readers and ultimately affecting human life or disease processes. Inflammation is a response to infection and injury in various diseases and has therefore attracted significant attention. Currently, extensive evidence indicates that m6A plays an essential role in inflammation. In this review, we focus on the mechanisms of m6A in inflammatory autoimmune diseases, metabolic disorder, cardio-cerebrovascular diseases, cancer, and pathogen-induced inflammation, as well as its possible role as targets for clinical diagnosis and treatment.


Subject(s)
Neoplasms , RNA , Humans , RNA/metabolism , Neoplasms/metabolism , Adenosine , Epigenesis, Genetic
6.
Front Vet Sci ; 9: 846700, 2022.
Article in English | MEDLINE | ID: mdl-35464378

ABSTRACT

Mastitis is one of the most common and significant infectious diseases in dairy cattle and is responsible for significant financial losses for the dairy industry globally. An important pathogen of bovine mastitis, Mycoplasma bovis (M. bovis) has a high infection rate, requires a long course of treatment, and is difficult to cure. Bovine mammary epithelial cells (BMECs) are the first line of defense of the mammary gland, and their natural immune system plays a critical role in resisting M. bovis infection. This study aimed to explore and demonstrate the regularity of Toll-like receptors (TLRs) activation during M. bovis infection and their function during M. bovis mastitis. An in vitro model of M. bovis-induced mastitis showed that the expression of IL-6, IL-8, and TNF-α increased significantly following infection. M. bovis infection also upregulated the expression of TLR1/2/6 on the cell membrane and TLR3/9 in the cytoplasm. There is a crosstalk effect between TLR1-TLR2 and TLR2-TLR6. Furthermore, M. bovis infection was found to activate the TLR1/2/6/9/MyD88/NF-κB and TLR3/TRIF/IRF signal transduction pathways, which in turn activate inflammatory factors. These findings lay the theoretical foundation for understanding the pathogenesis of M. bovis, permitting the development of effective measures for preventing and controlling M. bovis mastitis.

7.
Front Immunol ; 13: 873330, 2022.
Article in English | MEDLINE | ID: mdl-35444650

ABSTRACT

Mastitis is a common disease that hinders the development of dairy industry and animal husbandry. It leads to the abuse of antibiotics and the emergence of super drug-resistant bacteria, and poses a great threat to human food health and safety. Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) are the most common pathogens of mastitis in dairy cows and usually cause subclinical or clinical mastitis. CircRNAs and N6-methyladenosine (m6A) play important roles in immunological diseases. However, the mechanisms by which m6A modifies circRNA in bovine mammary epithelial cells remain poorly understood. The aim of our study was to investigate m6A-modified circRNAs in bovine mammary epithelial cells (MAC-T cells) injured by S. aureus and E. coli. The profile of m6A-modified circRNA showed a total of 1,599 m6A peaks within 1,035 circRNAs in the control group, 35 peaks within 32 circRNAs in the S. aureus group, and 1,016 peaks within 728 circRNAs in the E. coli group. Compared with the control group, 67 peaks within 63 circRNAs were significantly different in the S. aureus group, and 192 peaks within 137 circRNAs were significantly different in the E. coli group. Furthermore, we found the source genes of these differentially m6A-modified circRNAs in the S. aureus and E. coli groups with similar functions according to GO and KEGG analyses, which were mainly associated with cell injury, such as inflammation, apoptosis, and autophagy. CircRNA-miRNA-mRNA interaction networks predicted the potential circRNA regulation mechanism in S. aureus- and E. coli-induced cell injury. We found that the mRNAs in the networks, such as BCL2, MIF, and TNFAIP8L2, greatly participated in the MAPK, WNT, and inflammation pathways. This is the first report on m6A-modified circRNA regulation of cells under S. aureus and E. coli treatment, and sheds new light on potential mechanisms and targets from the perspective of epigenetic modification in mastitis and other inflammatory diseases.


Subject(s)
Escherichia coli Infections , Mastitis , Staphylococcal Infections , Adenosine/analogs & derivatives , Animals , Cattle , Epithelial Cells , Escherichia coli , Female , Humans , Inflammation/metabolism , RNA, Circular/genetics , RNA, Messenger/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus
8.
Oxid Med Cell Longev ; 2022: 1704172, 2022.
Article in English | MEDLINE | ID: mdl-35251466

ABSTRACT

The symptoms of mastitis caused by Staphylococcus aureus (S. aureus) in dairy cows are not obvious and difficult to identify, resulting in major economic losses. N6-Methyladenosine (m6A) modification has been reported to be closely associated with the occurrence of many diseases. However, only a few reports have described the role of m6A modification in S. aureus-induced mastitis. In this study, after 24 h of treatment with inactivated S. aureus, MAC-T cells (an immortalized bovine mammary epithelial cell line) showed increased expression levels of the inflammatory factors IL-1ß, IL-6, TNF-α, and reactive oxygen species. We found that the mRNA levels of METLL3, METLL14, WTAP, and ALKBH5 were also upregulated. Methylated RNA immunoprecipitation sequencing analysis revealed that 133 genes were m6A hypermethylated, and 711 genes were m6A hypomethylated. Biological functional analysis revealed that the differential m6A methylated genes were mainly related to oxidative stress, lipid metabolism, inflammatory response, and so on. In the present study, we also identified 62 genes with significant changes in m6A modification and mRNA expression levels. These findings elucidated the m6A modification spectrum induced by S. aureus in MAC-T cells and provide the basis for subsequent m6A research on mastitis.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/microbiology , Hot Temperature , Mammary Glands, Animal/cytology , Mastitis/metabolism , Microbial Viability , Signal Transduction/genetics , Staphylococcal Infections/metabolism , Staphylococcus aureus , Adenosine/analogs & derivatives , Animals , Cattle , Cell Line, Transformed , Cytokines/metabolism , Female , Mastitis/genetics , Mastitis/microbiology , Methylation , RNA/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Up-Regulation/genetics
9.
Biol Trace Elem Res ; 200(4): 1750-1762, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34185276

ABSTRACT

Mastitis caused by Staphylococcus aureus infection not only causes serious economic losses, but also affects human health. Se plays an important role in body immunity. However, the mechanisms by which Se regulates mastitis induced by S. aureus are still principally unknown. The purpose of this study is to investigate whether Se can inhibit mastitis induced by S. aureus through regulation of MerTK. Sixty BALB/c female mice were fed low, normal, or high Se concentrations for 7 weeks and then randomly divided into six groups (Se-Low Control group (LSN), Se-Normal Control group (NSN), Se-High Control group (HSN), Se-Low S. aureus group (LSS), Se-Normal S. aureus group (NSS), Se-High S. aureus group (HSS)). The regulation of Se on MerTK was detected via histopathological staining, western blot analysis, enzyme-linked immunosorbent assay, and qRT-PCR. With increased selenium concentrations, the levels of IL-1ß, IL-6, and TNF-α decreased, while the phosphorylation levels of MerTK, PI3K, AKT, and mTOR increased. Therefore, this study showed that Se could alleviate S. aureus mastitis by activating MerTK and PI3K/AKT/mTOR pathway.


Subject(s)
Mastitis , Selenium , Staphylococcal Infections , Animals , Female , Humans , Inflammation/drug therapy , Inflammation/metabolism , Mastitis/metabolism , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Selenium/metabolism , Selenium/pharmacology , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , TOR Serine-Threonine Kinases , c-Mer Tyrosine Kinase
10.
Int J Mol Sci ; 22(18)2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34575880

ABSTRACT

Escherichia coli and Staphylococcus aureus are two common pathogenic microorganisms that cause mastitis in dairy cows. They can cause clinical mastitis and subclinical mastitis. In recent studies, lncRNAs have been found to play an important role in the immune responses triggered by microbial inducers. However, the actions of lncRNAs in bovine mastitis remain unclear. The purpose of this study was to investigate the effects of bovine mammary epithelial cell injuries induced by treatment with E. coli and S. aureus, and to explore the lncRNA profile on cell injuries. The lncRNA transcriptome analysis showed a total of 2597 lncRNAs. There were 2234 lncRNAs differentially expressed in the E. coli group and 2334 in the S. aureus group. Moreover, we found that the E. coli and S. aureus groups of maternal genes targeted signaling pathways with similar functions according to KEGG and GO analyses. Two lncRNA-miRNA-mRNA interaction networks were constructed in order to predict the potential molecular mechanisms of regulation in the cell injuries. We believe that this is the first report demonstrating the dysregulation of lncRNAs in cells upon E. coli and S. aureus infections, suggesting that they have the potential to become important diagnostic markers and to provide novel insights into controlling and preventing mastitis.


Subject(s)
Escherichia coli Infections/genetics , Escherichia coli , Mastitis, Bovine/etiology , Mastitis, Bovine/pathology , RNA, Long Noncoding/genetics , Staphylococcal Infections/genetics , Staphylococcus aureus , Animals , Cattle , Computational Biology/methods , Epithelial Cells/metabolism , Escherichia coli/physiology , Escherichia coli Infections/microbiology , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Genome-Wide Association Study/methods , Host-Pathogen Interactions/genetics , MicroRNAs/genetics , Models, Biological , RNA Interference , RNA, Messenger/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology
11.
Int J Mol Sci ; 22(12)2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34200743

ABSTRACT

Mastitis is a common disease in dairy cows that is mostly caused by E. coli, and it brings massive losses to the dairy industry. N6-Methyladenosine (m6A), a methylation at the N6 position of RNA adenine, is a type of modification strongly associated with many diseases. However, the role of m6A in mastitis has not been investigated. In this study, we used MeRIP-seq to sequence the RNA of bovine mammary epithelial cells treated with inactivated E. coli for 24 h. In this in vitro infection model, there were 16,691 m6A peaks within 7066 mRNA transcripts in the Con group and 10,029 peaks within 4891 transcripts in the E. coli group. Compared with the Con group, 474 mRNAs were hypermethylated and 2101 mRNAs were hypomethylated in the E. coli group. Biological function analyses revealed differential m6A-modified genes mainly enriched in the MAPK, NF-κB, and TGF-ß signaling pathways. In order to explore the relationship between m6A and mRNA expression, combined MeRIP-seq and mRNA-seq analyses revealed 212 genes with concomitant changes in the mRNA expression and m6A modification. This study is the first to present a map of RNA m6A modification in mastitis treated with E. coli, providing a basis for future research.


Subject(s)
Adenosine/analogs & derivatives , DNA Methylation , Epithelial Cells/metabolism , Escherichia coli Infections/veterinary , Gene Expression Regulation , Mammary Glands, Animal/metabolism , Mastitis, Bovine/genetics , Adenosine/chemistry , Animals , Cattle , Epithelial Cells/immunology , Epithelial Cells/microbiology , Escherichia coli/isolation & purification , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Female , Gene Expression Profiling , Mammary Glands, Animal/immunology , Mammary Glands, Animal/microbiology , Mastitis, Bovine/immunology , Mastitis, Bovine/microbiology
12.
Int Immunopharmacol ; 86: 106697, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32585608

ABSTRACT

Mastitis is one of the most common diseases among dairy cows. There is still much debate worldwide as to whether antibiotic therapy should be given to dairy cows, or if natural products should be taken as a substitute for antibacterial therapy. As the antibiotic treatment leads to the bacterial resistance and drug residue in milk, introducing natural products for mastitis is becoming a trend. This study investigates the mechanisms of the protective effects of the natural product gambogic acid (GA) in lipopolysaccharide (LPS)-induced mastitis. For in vitro treatments, it was found that GA reduced IL-6, TNF-α, and IL-1ß levels by inhibiting the phosphorylation of proteins in the nuclear factor κB (NF-κB) and the mitogen-activated protein kinase (MAPK) pathway. GA also maintained a stable membrane mitochondrial potential and inhibited the overproduction of reactive oxygen species, which protected the cells from apoptosis. On the other hand, in vivo treatments with GA were found to reduce pathological symptoms markedly, and protected the blood-milk barrier from damage induced by LPS. The results demonstrate that GA plays a vital role in suppressing inflammation, alleviating the apoptosis effect, and protecting the blood-milk barrier in mastitis induced by LPS. Thus, these results suggest that the natural product GA plays a potential role in mastitis treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Mammary Glands, Animal/drug effects , Mastitis/drug therapy , Xanthones/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Female , Inflammation/drug therapy , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Mammary Glands, Animal/immunology , Mammary Glands, Animal/pathology , Mammary Glands, Animal/ultrastructure , Mastitis/chemically induced , Mastitis/immunology , Mastitis/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , NF-kappa B p50 Subunit/metabolism , Phosphorylation/drug effects , Pregnancy , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Xanthones/therapeutic use
13.
Onco Targets Ther ; 13: 1471-1480, 2020.
Article in English | MEDLINE | ID: mdl-32110045

ABSTRACT

BACKGROUND: Pancreatic cancer (PC) is a highly lethal malignancy worldwide. Our previous study indicated that overexpression of USP34 could promote tumor growth in PC cells. Therefore, this study aimed to further investigate the role of USP34 during the tumorigenesis of PC. METHODS: The level of USP34 in PANC-1 and MiaPaCa-2 cells transfected with USP34-shRNAs was detected by RT-qPCR. Moreover, transwell migration and Annexin V/PI analysis were conducted to detect cell migration and apoptosis, respectively. RESULTS: In this study, downregulation of USP34 markedly inhibited proliferation and migration, and induced apoptosis in PANC-1 cells. Moreover, silencing of USP34 obviously downregulated the levels of PRR11 and p-p38 in PANC-1 cells. An in vivo study in nude mice bearing PANC-1 cell xenografts confirmed these results. CONCLUSION: Downregulation of USP34 could inhibit proliferation and migration in PANC-1 cells via inhibiting PRR11, and inactivating p38 MAPK signaling. Therefore, USP34 might be a potential therapeutic target for the treatment of PC.

14.
Biol Pharm Bull ; 42(4): 573-579, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30686807

ABSTRACT

Pancreatic cancer is known to be a fatal disease, which is difficult to be diagnosed in its early stages. Ubiquitin-Specific Protease 34 (USP34) are closely related to human cancers in the development and progression. However, there are rarely studies about the role of USP34 in pancreatic cancer. Thus, we aimed to investigate the effect of USP34 in human pancreatic cancer. Short-hairpin RNA targeting USP34 (USP34-shRNA) and USP34 overexpression lentivirus were used in the current study. The level of USP34 in human pancreatic cancer (PANC-1) cells were then analyzed by quantitative (q)RT-PCR. In addition, Western blotting was used to examine phosphorylated (p)-AKT, p-protein kinase C (PKC) and p-extracellular signal-regulated kinase (ERK) protein levels. CCK-8 assay, flow cytometry, and migration assay were used to detect cell proliferation, apoptosis and migration, respectively in vitro. According to the result of qRT-PCR and Western blotting, USP34-shRNA1 significantly downregulated USP34 gene level in PANC-1 cell. Subsequently, Western blotting assay indicated that USP34 silencing significantly down-regulated the expression of p-AKT and p-PKC in cells. On the other hand, USP34 overexpressing remarkably up-regulated the expression of p-AKT and p-PKC in cells. In addition, USP34 overexpression promoted PANC-1 cell proliferation and migration via up-regulating the proteins of p-AKT and p-PKC. Moreover, USP34 overexpression reversed AKT inhibitor and PKC inhibitor induced PACN-1 cell apoptosis. Our results indicated USP34 regulated h PANC-1 cell survival via AKT and PKC pathways, and which played a pro-survival role in human pancreatic cancer. Therefore, we suggested USP34 could be a potential therapeutic target for pancreatic cancer.


Subject(s)
Gene Expression Regulation, Enzymologic , Pancreatic Neoplasms/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitin-Specific Proteases/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Gene Knockdown Techniques , Humans , Lentivirus/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Plasmids , RNA, Small Interfering/genetics , Signal Transduction
15.
J Adv Model Earth Syst ; 11(10): 3305-3320, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31894191

ABSTRACT

Increasing vapor pressure deficit (VPD) increases atmospheric demand for water. While increased evapotranspiration (ET) in response to increased atmospheric demand seems intuitive, plants are capable of reducing ET in response to increased VPD by closing their stomata. We examine which effect dominates the response to increasing VPD: atmospheric demand and increases in ET or plant response (stomata closure) and decreases in ET. We use Penman-Monteith, combined with semiempirical optimal stomatal regulation theory and underlying water use efficiency, to develop a theoretical framework for assessing ET response to VPD. The theory suggests that depending on the environment and plant characteristics, ET response to increasing VPD can vary from strongly decreasing to increasing, highlighting the diversity of plant water regulation strategies. The ET response varies due to (1) climate, with tropical and temperate climates more likely to exhibit a positive ET response to increasing VPD than boreal and arctic climates; (2) photosynthesis strategy, with C3 plants more likely to exhibit a positive ET response than C4 plants; and (3) plant type, with crops more likely to exhibit a positive ET response, and shrubs and gymniosperm trees more likely to exhibit a negative ET response. These results, derived from previous literature connecting plant parameters to plant and climate characteristics, highlight the utility of our simplified framework for understanding complex land-atmosphere systems in terms of idealized scenarios in which ET responds to VPD only. This response is otherwise challenging to assess in an environment where many processes coevolve together.

SELECTION OF CITATIONS
SEARCH DETAIL
...