Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Affiliation country
Publication year range
1.
J Immunol ; 208(3): 618-632, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35022270

ABSTRACT

Sepsis is an acute life-threatening disorder associated with multiorgan dysfunction that remains the leading cause of death in intensive care units. As sepsis progresses, it causes prolonged immunosuppression, which results in sustained mortality, morbidity, and susceptibility to secondary infections. Using a mouse model of sepsis, we found that the long noncoding RNA HOTAIRM1 (HOXA transcript antisense RNA myeloid-specific 1) was highly expressed in mice during the late phase of sepsis. The upregulation of HOTAIRM1 was induced by Notch/Hes1 activation and, moreover, was critical for the formation of an immunosuppressive microenvironment. HOTAIRM1 induced T cell exhaustion by increasing the percentage of PD-1+ T cells and regulatory T cells, accompanied by elevated PD-L1. Blockade of either Notch/Hes1 signaling or HOTAIRM1 inhibited T cell exhaustion in late sepsis, having alleviated lung injury and improved survival of mice. Further mechanistic studies identified HOXA1 as a key transcription factor targeted by HOTAIRM1 to regulate PD-L1 expression in lung alveolar epithelial cells. These results implicated that the Notch/Hes1/HOTAIRM1/HOXA1/PD-L1 axis was critical for sepsis-induced immunosuppression and could be a potential target for sepsis therapies.


Subject(s)
Immune Tolerance/immunology , MicroRNAs/genetics , Sepsis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Sepsis/microbiology , Transcription Factor HES-1/metabolism , Transcription Factors/metabolism
2.
Dig Dis Sci ; 69(4): 1214-1227, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38376789

ABSTRACT

BACKGROUND: HSK3486 (ciprofol), a new candidate drug similar to propofol, exerts sedative and hypnotic effects through gamma-aminobutyric acid type A receptors; however, its potential role in colorectal cancer is currently unknown. AIMS: This study aimed to evaluate the effects of HSK3486 on colorectal cancer cell proliferation. METHODS: Imaging was performed to detect reactive oxygen species and mitochondrial membrane potential. Western blotting was used to determine the expression of target signals. The HSK3486 molecular mechanism was investigated through ATPase inhibitory factor 1 knockdown and xenograft model experiments to assess mitochondrial function in colorectal cancer cells. RESULTS: Cell Counting Kit-8 and Annexin V/propidium iodide double staining assays showed that HSK3486 inhibited colorectal cancer cell proliferation in a concentration-dependent manner. In addition, HSK3486 treatment increased the expression of B-cell lymphoma-2-associated X, cleaved caspase 3, and cleaved poly (ADP-ribose) polymerase, whereas myeloid cell leukemia-1 and B-cell lymphoma 2 expression decreased. HSK3486 promoted mitochondrial dysfunction by inducing ATPase inhibitor factor 1 expression. Furthermore, HSK3486 promoted oxidative stress, as shown by the increase in reactive oxygen species and lactate dehydrogenase levels, along with a decrease in mitochondrial membrane potential and ATP levels. ATPase inhibitor factor 1 small interfering RNA pretreatment dramatically increased the mitochondrial membrane potential and tumor size in a xenograft model following exposure to HSK3486. CONCLUSION: Collectively, our findings revealed that HSK3486 induces oxidative stress, resulting in colorectal cancer cell apoptosis, making it a potential candidate therapeutic strategy for colorectal cancer.


Subject(s)
Apoptosis , Colorectal Neoplasms , Humans , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/pharmacology , Adenosine Triphosphatases/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Membrane Potential, Mitochondrial , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , ATPase Inhibitory Protein/drug effects
3.
Acta Biochim Biophys Sin (Shanghai) ; 56(6): 866-878, 2024 06 25.
Article in English | MEDLINE | ID: mdl-38606479

ABSTRACT

Approximately 20% of colorectal cancer (CRC) patients are first diagnosed with metastatic colorectal cancer (mCRC) because they develop symptoms at an advanced stage. Despite advancements in treatment, patients with metastatic disease still experience inferior survival rates. Our objective is to investigate the association between long noncoding RNAs (lncRNAs) and prognosis and to explore their role in mCRC. In this study, we find that elevated expression of PCAT6 is independently linked to unfavourable survival outcomes in The Cancer Genome Atlas (TCGA) data, and this finding is further confirmed in CRC samples obtained from Fudan University Shanghai Cancer Center. Cell lines and xenograft mouse models are used to examine the impact of PCAT6 on tumor metastasis. Knockdown of PCAT6 is observed to impede the metastatic phenotype of CRC, as evidenced by functional assays, demonstrating the suppression of epithelial-mesenchymal transition (EMT) and stemness. Our findings show the significance of PCAT6 in mCRC and its potential use as a prognostic biomarker.


Subject(s)
Colorectal Neoplasms , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells , RNA, Long Noncoding , Animals , Female , Humans , Male , Mice , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Untranslated/genetics
4.
J Cell Mol Med ; 25(18): 9038-9050, 2021 09.
Article in English | MEDLINE | ID: mdl-34405537

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of death worldwide, and hence, there is a need to elucidate the molecular mechanisms contributing to the progression of CRC. In this study, we aimed at assessing the role of long non-coding RNA NBPF4 on the tumorigenesis of CRC. Silencing or overexpression experiments were performed on HCT116 and SW260 in vitro models. BALB/c athymic female nude mice aged 5-6 weeks were used as in vivo models. To assess the relationship between NBPF4 and its regulatory RNA pull-down assay, RNA immunoprecipitation, luciferase activity, Western blotting and qRT-PCR were employed. Initially, we identified that NBPF4 was downregulated in CRC tissues and cell lines. Furthermore, we observed that NBPF4 decreased tumorigenesis in both in vitro and in vivo models. Additionally, we identified that ETFA was highly expressed in CRCs and was negatively associated with NBPF4. Subsequently, we identified that EZH2, a transcriptional factor, activated ETFA by enhancing the methylation of its promoter, and EZH2 was also highly regulated in CRCs. Using COAD and READ databases, we confirmed that EZH2 and ETFA were positively correlated. Furthermore, we identified NBPF4 and EZH2 were targets for ZFP36, which bound and positively regulated NBPF4. This prevented NBPF4 from binding to its negative regulator miR-17-3p. Our results demonstrated that NBPF4 downregulated EZH2 and stabilized itself by binding to ZFP36, thus escaping from inhibition by miR-17-3p, which allowed mitigation of CRC through inhibition of ETFA.


Subject(s)
Colorectal Neoplasms/metabolism , Electron-Transferring Flavoproteins/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude
5.
Mol Med ; 27(1): 74, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34238215

ABSTRACT

BACKGROUND: Diabetic nephropathy (DN) is currently the leading cause of end-stage renal disease globally. The endothelial-to-mesenchymal transition (EndMT) of glomerular endothelial cells has been reported to play a crucial role in DN. As a specific form of epithelial-to-mesenchymal transition, EndMT and epithelial-to-mesenchymal transition may exhibit mutual modulators. Profilin 2 (PFN2) has been reported to participate in epithelial-to-mesenchymal transition. Moreover, ETS proto-oncogene 1 (ets1) and lysine methyltransferase 5A (KMT5A) have been reported to contribute to high glucose-mediated endothelial injury and epithelial-to-mesenchymal transition. In this study, we hypothesize ets1 associates with KMT5A to modulate PFN2 transcription, thus participating in high glucose-mediated EndMT in glomerular endothelial cells. METHODS: Immunohistochemistry (IHC) was performed to detect protein levels in the kidney tissues and/or aorta tissues of human subjects and rats. Western blot, qPCR and immunofluorescence were performed using human umbilical vein endothelial cells (HUVECs). Chromatin immunoprecipitation (ChIP) assays and dual luciferase assays were performed to assess transcriptional activity. The difference between the groups was compared by two-tailed unpaired t-tests or one-way ANOVAs. RESULTS: Our data indicated that vimentin, αSMA, S100A4 and PFN2 levels were increased, and CD31 levels were reduced in glomerular endothelial cells of DN patients and rats. Our cell experiments showed that high glucose induced EndMT by augmenting PFN2 expression in HUVECs. Moreover, high glucose increased ets1 expression. si-ets1 suppressed high glucose-induced PFN2 levels and EndMT. ets1 overexpression-mediated EndMT was reversed by si-PFN2. Furthermore, ets1 was determined to associate with KMT5A. High glucose attenuated KMT5A levels and histone H4 lysine 20 methylation (H4K20me1), one of the downstream targets of KMT5A. KMT5A upregulation suppressed high glucose-induced PFN2 levels and EndMT. sh-KMT5A-mediated EndMT was counteracted by si-PFN2. Furthermore, H4K20me1 and ets1 occupied the PFN2 promoter region. sh-KMT5A cooperated with ets1 overexpression to activate PFN2 promoter activity. Our in vivo study demonstrated that KMT5A was reduced, while ets1 was augmented, in glomerular endothelial cells of DN patients and rats. CONCLUSIONS: The present study indicated that ets1 cooperated with KMT5A to transcribe PFN2, thus contributing to hyperglycemia-induced EndMT in the glomerular endothelial cells of DN patients and rats. Trial registration ChiCTR, ChiCTR2000029425. 2020/1/31, http://www.chictr.org.cn/showproj.aspx?proj=48548.


Subject(s)
Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Epithelial-Mesenchymal Transition/genetics , Glucose/metabolism , Histone-Lysine N-Methyltransferase/genetics , Profilins/genetics , Proto-Oncogene Protein c-ets-1/genetics , Aged , Animals , Binding Sites , Biomarkers , Diabetic Nephropathies/pathology , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression , Gene Expression Profiling , Histone-Lysine N-Methyltransferase/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Male , Middle Aged , Models, Biological , Profilins/metabolism , Protein Binding , Proto-Oncogene Protein c-ets-1/metabolism , Rats
6.
Liver Int ; 39(1): 147-157, 2019 01.
Article in English | MEDLINE | ID: mdl-30178471

ABSTRACT

BACKGROUND & AIMS: Resistance to EGFR-targeted therapy is a major obstacle in hepatocellular carcinoma (HCC) treatment, but its underlying mechanism remains unclear. Autophagy plays a vital role in antitumour treatment. Our previous study suggested that p57 is associated with autophagy and cisplatin resistance. The present study aimed to investigate whether p57 can enhance the sensitivity of HCC cells to Erlotinib (Er)/Cetuximab(C-225) and further explore the potential mechanisms of Er/C-225 resistance. METHODS: HCC cells were transfected with pIRES2-EGFP-p57 and pIRES2-EGFP-nc, accompanied by Er/C-225 treatment. Cell viability was detected by an Annexin apoptosis kit and MTT assay. Xenograft experiments were performed to study the function of p57 in the treatment of Er/C-225 in vivo. The level of autophagy was determined by analysis of the appearance of autophagic vacuoles. Western blotting was used to investigate the potential pathways involved. RESULTS: Up-regulation of p57 decreased the level of Er/C-225-induced autophagy and enhanced the decrease in Er/C-225-induced cell viability. P57 overexpression combined with CQ treatment further enhanced the therapeutic efficiency of Er/C-225. The xenograft experiment verified that p57 up-regulation sensitizes HCC cells to Er/C-225. Moreover, a mechanistic investigation demonstrated that the up-regulation of p57 resulted in a decrease of LC3B-II and beclin-1, and an increase in p-PI3K, p-AKT and p-mTOR protein expressions. CONCLUSIONS: Through activating the PI3K/AKT/mTOR signalling pathway, p57 can reverse Er/C-225-induced autophagy, and thereby increase the therapeutic efficiency of Er/C-225 treatment. Given these results, p57 up-regulation may be applicable as a therapeutic strategy to improve EGFR-targeted therapy in HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy , Carcinoma, Hepatocellular/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , ErbB Receptors/antagonists & inhibitors , Liver Neoplasms/genetics , Animals , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation , Cetuximab/pharmacology , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Liver Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Cancer Sci ; 108(10): 1985-1995, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28771881

ABSTRACT

Aberrant expression of microRNAs (miRs) has been shown to play a critical role in the pathogenesis and progression of tumors. microRNA-219-5p (miR-219-5p) has been reported to be abnormally expressed in some types of human tumors. However, the mechanism between miR-219-5p and colorectal cancer (CRC) metastasis remains unclear. In the present study, miR-219-5p was found to be downregulated in CRC tissue compared with matched normal tissue. Through luciferase reporter assay, we demonstrated lymphoid enhancer-binding factor 1 (LEF1) as a direct target of miR-219-5p. Overexpression of miR-219-5p could inhibit motility, migration and invasion of CRC cells, and inhibit epithelial-mesenchymal transition (EMT). Furthermore, silencing LEF1 phenocopied this metastasis-suppressive function. The recovery experiment showed that re-expression of LEF1 rescued this suppressive effect on tumor metastasis and reversed the expression of EMT markers caused by miR-219-5p. Additionally, we demonstrated that miR-219-5p exerted this tumor-suppressive function by blocking activation of the AKT and ERK pathways. Finally, a nude mice experiment showed that miR-219-5p reduced the lung metastasis ability of CRC cells. Taken together, our findings indicate that miR-219-5p inhibits metastasis and EMT of CRC by targeting LEF1 and suppressing the AKT and ERK pathways, which may provide a new antitumor strategy to delay CRC metastasis.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Lung Neoplasms/secondary , Lymphoid Enhancer-Binding Factor 1/genetics , MicroRNAs/genetics , 3' Untranslated Regions , Animals , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Lung Neoplasms/genetics , MAP Kinase Signaling System , Male , Mice , Mice, Nude , Neoplasm Transplantation
8.
Biomarkers ; 21(3): 272-5, 2016.
Article in English | MEDLINE | ID: mdl-26848614

ABSTRACT

BACKGROUND: Interleukin-27 (IL-27) has been recognized as a pleiotropic cytokine with both pro- and anti-inflammatory properties. PATIENTS AND METHODS: A case-control study was conducted to investigate the possible associations of IL-27 gene polymorphisms with susceptibility to cervical cancer and clinical outcome. RESULTS: Our results suggested that the IL-27 2905T/G was significantly associated with a decreased risk of cervical cancer. Further analysis showed IL-27 2905T/G genotypes were associated with advanced tumor stages of cervical cancer patients. More interestingly, the IL-27 2905T/G genotypes were statistically significantly associated with the survival in cervical cancer patients. CONCLUSION: Our results showed that the IL-27 2905T/G genotypes were associated with decreased the susceptibility and development of cervical cancer in Chinese Han population.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Interleukins/genetics , Uterine Cervical Neoplasms/genetics , Adult , Aged , Alleles , Female , Genotype , Humans , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors , Uterine Cervical Neoplasms/pathology
9.
Tumour Biol ; 35(5): 4107-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24375196

ABSTRACT

Genetic polymorphisms in the Fas/Fas ligand (FasL) gene were proposed to be associated with susceptibility to cervical cancer, but previous studies reported controversial findings. We performed a meta-analysis to assess the associations between Fas/FasL polymorphisms and susceptibility to cervical cancer. We carried out a literature search in PubMed and Embase databases for studies on the associations between Fas/FasL polymorphisms and susceptibility to cervical cancer. The associations were assessed by odds ratio (OR) together with its 95% confidence intervals (CIs). Eleven individual studies with a total of 6,919 subjects were finally included into the meta-analysis. Overall, there was no association between Fas 1377G > A polymorphism and susceptibility to cervical cancer (A vs. G: OR = 0.99, 95% CI 0.88-1.12, P = 0.91; AA vs. GG: OR = 1.00, 95% CI 0.76-1.32, P = 0.99; AA/GA vs. GG: OR = 0.95, 95% CI 0.81-1.12, P = 0.54; AA vs. GG/GA: OR = 1.11, 95% CI 0.85-1.43, P = 0.45). In addition, there was also no association between FasL 844 T > C polymorphism and susceptibility to cervical cancer (C vs. T: OR = 1.12, 95% CI 0.91-1.36, P = 0.28; CC vs. TT: OR = 1.17, 95% CI 0.90-1.51, P = 0.24; CC/TC vs. TT: OR = 1.13, 95% CI 0.92-1.39, P = 0.24; CC vs. TT/TC: OR = 1.11, 95% CI 0.83-1.50, P = 0.47). In subgroup analysis by ethnicity, there were also no associations between Fas/FasL polymorphisms and susceptibility to cervical cancer in Asians and Africans. In conclusion, Fas 1377G > A polymorphism and FasL 844 T > C polymorphism are both not associated with susceptibility to cervical cancer.


Subject(s)
Fas Ligand Protein/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic , Uterine Cervical Neoplasms/genetics , fas Receptor/genetics , Female , Humans , Risk , Uterine Cervical Neoplasms/etiology
10.
Tumour Biol ; 35(12): 11701-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25293518

ABSTRACT

Epidermal growth factor receptor (EGFR) inhibitor treatment is a strategy for cancer therapy. However, innate and acquired resistance is a major obstacle of the efficacy. Autophagy is a self-digesting process in cells, which is considered to be associated with anti-cancer drug resistance. The activation of EGFR can regulate autophagy through multiple signal pathways. EGFR inhibitors can induce autophagy, but the specific function of the induction of autophagy by EGFR inhibitors remains biphasic. On the one hand, autophagy induced by EGFR inhibitors acts as a cytoprotective response in cancer cells, and autophagy inhibitors can enhance the cytotoxic effects of EGFR inhibitors. On the other hand, a high level of autophagy after treatment of EGFR inhibitors can also result in autophagic cell death lacking features of apoptosis, and the combination of EGFR inhibitors with an autophagy inducer might be beneficial. Thus, autophagy regulation represents a promising approach for improving the efficacy of EGFR inhibitors in the treatment of cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Animals , ErbB Receptors/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
11.
World J Surg Oncol ; 11: 5, 2013 Jan 13.
Article in English | MEDLINE | ID: mdl-23311997

ABSTRACT

BACKGROUND: Gastric cancer is a common and highly lethal malignancy in the world, but its pathogenesis remains elusive. In this study, we focus on the biological functions of CDK-associated Cullin1 (CAC1), a novel gene of the cullin family, in gastric cancer, which may help us to further understand the origin of this malignancy. METHODS: The AGS and MGC803 gastric cancer cell lines and the GES-1 gastric mucosa cell line were selected for study. At first, CAC1 expressions of those cell lines were examined by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) and western blot examinations, then CAC1 small interfering RNA (CAC1-siRNA) were designed and transfected into the AGS cell line with a relatively high level of CAC1. Once CAC1 was silenced, a series of biological characteristics of AGS cells such as cell proliferation, cell cycle, apoptosis, and expressions of apoptosis-related genes (P53, BCL2 and BAX) were determined by MTT, flow cytometry, qRT-PCR and western blot, respectively. RESULTS: CAC1 expression of AGS or MGC803 was much higher than that of GES-1. After CAC1 expression was effectively depressed by RNA interference in AGS cells, significant cell growth inhibition occurred. Furthermore, the proportion of cells treated with CAC1-siRNA increased in the G1 phase and decreased in the S phase, indicative of G1 cell cycle arrest. More importantly, the proportions of early/late apoptosis in AGS cells were enhanced with cis-diaminedichloroplatinum (cisplatin, CDDP) treatment, but to a higher extent with cisplatin plus CAC1-siRNA. Interestingly, BCL2 mRNA copies showed about a 30% decrease in the cisplatin group, but dropped by around 60% in the cisplatin plus CAC1-siRNA group. Conversely, the P53 mRNA expressions obtained nearly a two-fold increase in the cisplatin group, in addition to a five-fold increase in the cisplatin plus CAC1-siRNA group, and the BAX mRNA levels had almost a two- and four-fold augmentation, respectively. Meanwhile, P53, BAX and BCL2 showed the same alteration patterns in western blot examinations. CONCLUSIONS: CAC1 can promote cell proliferation in the AGS gastric cancer cell line. Moreover, it can prevent AGS cells from experiencing cisplatin-induced apoptosis via modulating expressions of P53, BCL2 and BAX.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cullin Proteins/physiology , Gastric Mucosa/drug effects , Stomach Neoplasms/pathology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Cycle/drug effects , Cells, Cultured , Cullin Proteins/antagonists & inhibitors , Flow Cytometry , Gastric Mucosa/metabolism , Humans , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism
12.
Biosensors (Basel) ; 13(6)2023 Jun 04.
Article in English | MEDLINE | ID: mdl-37366983

ABSTRACT

Non-enzymatic sensors with the capability of long-term stability and low cost are promising in glucose monitoring applications. Boronic acid (BA) derivatives offer a reversible and covalent binding mechanism for glucose recognition, which enables continuous glucose monitoring and responsive insulin release. To improve selectivity to glucose, a diboronic acid (DBA) structure design has been explored and has become a hot research topic for real-time glucose sensing in recent decades. This paper reviews the glucose recognition mechanism of boronic acids and discusses different glucose sensing strategies based on DBA-derivatives-based sensors reported in the past 10 years. The tunable pKa, electron-withdrawing properties, and modifiable group of phenylboronic acids were explored to develop various sensing strategies, including optical, electrochemical, and other methods. However, compared to the numerous monoboronic acid molecules and methods developed for glucose monitoring, the diversity of DBA molecules and applied sensing strategies remains limited. The challenges and opportunities are also highlighted for the future of glucose sensing strategies, which need to consider practicability, advanced medical equipment fitment, patient compliance, as well as better selectivity and tolerance to interferences.


Subject(s)
Biosensing Techniques , Blood Glucose Self-Monitoring , Humans , Blood Glucose Self-Monitoring/methods , Blood Glucose , Biosensing Techniques/methods , Glucose
13.
Cancer Res ; 83(21): 3529-3543, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37602826

ABSTRACT

As a safe, feasible, and inexpensive dietary intervention, fasting-mimicking diet (FMD) exhibits excellent antitumor efficacy by regulating metabolism and boosting antitumor immunity. A better understanding of the specific mechanisms underlying the immunoregulatory functions of FMD could help improve and expand the clinical application of FMD-mediated immunotherapeutic strategies. In this study, we aimed to elucidate the role of metabolic reprogramming induced by FMD in activation of antitumor immunity against colorectal cancer. Single-cell RNA sequencing analysis of intratumoral immune cells revealed that tumor-infiltrating IgA+ B cells were significantly reduced by FMD treatment, leading to the activation of antitumor immunity and tumor regression in murine colorectal cancer models. Mechanistically, FMD delayed tumor growth by repressing B-cell class switching to IgA. Therefore, FMD-induced reduction of IgA+ B cells overcame the suppression of CD8+ T cells. The immunoregulatory and antitumor effects of FMD intervention were reversed by IgA+ B-cell transfer. Moreover, FMD boosted fatty acid oxidation (FAO) to trigger RUNX3 acetylation, thus inactivating Cα gene transcription and IgA class switching. IgA+ B-cell expansion was also impeded in patients placed on FMD, while B-cell expression of carnitine palmitoyl transferase 1A (CPT1A), the rate-limiting enzyme of FAO, was increased. Furthermore, CPT1A expression was negatively correlated with both IgA+ B cells and IgA secretion within colorectal cancer. Together, these results highlight that FMD holds great promise for treating colorectal cancer. Furthermore, the degree of IgA+ B cell infiltration and FAO-associated metabolic status are potential biomarkers for evaluating FMD efficacy. SIGNIFICANCE: Metabolic reprogramming of B cells induced by fasting-mimicking diet suppresses IgA class switching and production to activate antitumor immunity and inhibit tumor growth. See related commentary by Bush and Perry, p. 3493.


Subject(s)
Colorectal Neoplasms , Fasting , Humans , Animals , Mice , Fasting/physiology , Diet , Biomarkers , Colorectal Neoplasms/genetics , Immunoglobulin A
14.
Oncol Res ; 20(2-3): 113-21, 2012.
Article in English | MEDLINE | ID: mdl-23193917

ABSTRACT

Hepatocellular carcinoma (HCC) is a complex and heterogeneous tumor with several genomic alterations and ranks as the third highest cause of cancer-related death globally. The unresectable or metastatic HCC has very poor prognosis. Although multikinase inhibitor sorafenib can increase the survival of patients with advanced HCC, it is becoming apparent that combination therapies are critical to overcome the complex genomic aberrations in HCC. PTEN, as one of the most commonly inactivated genes in HCC, exerts a wide range of antitumor effects. In this study, we found that PTEN was downregulated in HCC tissues, especially in those tissues with extrahepatic metastasis. And negative PTEN expression cases showed increased proliferation activity. Overexpression of PTEN significantly reduced the proliferation of tumor cells HepG2. In addition, HepG2 cells transfected with PTEN were more sensitive to sorafenib in terms of its ability to inhibit proliferation and to induce apoptosis. Moreover, overexpression of PTEN decreased phosphorylation of MEK, a key downstream effector of RAF/MEK/ERK cascade, and levels of cyclin D1, antiapoptotic Bcl-2, and VEGF. These observations indicated that combination therapies with sorafenib and PTEN overexpression have potential to further improve therapeutic options for HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Drug Resistance/drug effects , PTEN Phosphohydrolase/metabolism , Pyridines/pharmacology , Apoptosis/drug effects , Blotting, Western , Carcinoma, Hepatocellular/mortality , Case-Control Studies , Flow Cytometry , Humans , Immunoenzyme Techniques , Liver/cytology , Liver/drug effects , Liver/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Prognosis , Sorafenib , Survival Rate , Tumor Cells, Cultured
15.
BMC Gastroenterol ; 12: 53, 2012 May 28.
Article in English | MEDLINE | ID: mdl-22639890

ABSTRACT

BACKGROUND: Colon cancer is one of the most aggressive human malignancies, with a very poor prognosis. Although it has been suggested that different isoforms of the lymphoid enhancer factor (LEF-1) have opposing biological activities, the biological outcome of aberrant LEF-1 activation in colon cancer is still unclear. The aim of this study was to evaluate the effect of the different LEF-1 phenotypes on the growth of colon carcinoma cell lines. A deeper understanding of these processes might improve the targeted therapies for colon cancer by regulating the expression of LEF-1. METHODS: The role of different isoforms of LEF-1 on the growth of human colon carcinoma cell lines (SW480 and HT-29) was studied using various in vitro and in vivo assays. In vitro proliferation, migration, adhesion and apoptosis of the cells stably transfected of different isoforms of LEF-1 were monitored by MTT assay, carboxyfluorescein diacetate-succinimidyl ester staining, annexin V staining, ECM adhesion assay and transwell assay, respectively. In nude mice, the formation of neovasculature in the tumors formed by our constructed cells was measured by immunohistochemistry. All the data were analyzed using a t test, and data were treated as significant when p < 0.05. RESULTS: Overexpression of truncated LEF-1 (LEF-1-ΔL) in the colon cell lines, SW480 and HT29, inhibited their growth significantly in vitro and in vivo, but the full-length LEF-1 (LEF-1-FL) promoted the proliferation of HT29. Inactivation of Wnt signaling by LEF-1-ΔL reduced the expression of CXCR4 in colon cell lines, which may lead to a decrease in activities such as migration, adhesion and survival. In nude mice, the formation of neovasculature as well as an increase in tumor volume were inhibited by the short isoform of LEF-1. LEF-1-FL, however, caused an increase in all these parameters compared with controls. CONCLUSIONS: These findings suggest that LEF-1 might play an important role in colon carcinogenesis by acting as a regulator. Enhanced expression of LEF-1-FL, which occurs frequently in colon cancer, may be a new target for clinical therapy.


Subject(s)
Cell Proliferation , Colonic Neoplasms/pathology , Colonic Neoplasms/physiopathology , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/physiology , Phenotype , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , HT29 Cells , Humans , In Vitro Techniques , Mice , Mice, Nude , Protein Isoforms , Receptors, CXCR4/physiology , Transplantation, Heterologous , Wnt Proteins/physiology
16.
Int J Colorectal Dis ; 27(3): 287-98, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22065111

ABSTRACT

PURPOSE: Pleiotrophin (PTN) is an important developmental secretory cytokine expressed in many types of cancer and involved in angiogenesis and tumor growth; however, the significance of PTN expression in colorectal cancer (CRC) has not been established. METHODS: Immunohistochemistry, western blot, and enzyme-linked immunosorbent assay were used to detect PTN expression in CRC patients. The relationship between PTN expression and clinicopathological characteristics and survival time was statistically analyzed, and the relationship between PTN and vascular endothelial growth factor (VEGF) in tumor angiogenesis was further analyzed. RESULTS: Of CRC tissues, 74.70% (62/83) stained positive, with a strong positive ratio of 60.24% (50/83). The expression of PTN in CRC tissues was much higher than in normal colorectal tissues. PTN serum levels in CRC patients (mean = 254.59 ± 261.76 pg/ml) were significantly higher than those of normal volunteers (mean = 115.23 ± 79.53 pg/ml; p < 0.001). PTN expression was related to CRC differentiation and TNM staging. High level of PTN is a predictor of a poor prognosis and high expression of PTN is accompanied by high expression of VEGF in CRC patients. Investigation of the relationship between PTN and VEGF revealed that PTN, through the PTN/RPTPß/ζ signaling pathway, increased tyrosine phosphorylation of ß-catenin, leading to an increase in VEGF. CONCLUSIONS: Our study identifies PTN as an essential growth factor for CRC. PTN promotes VEGF expression and cooperates with VEGF in promoting CRC angiogenesis. PTN could serve as a prognostic factor for this cancer. Considering that PTN shows very limited expression in normal tissue, it may represent an attractive new target for CRC therapy.


Subject(s)
Adenocarcinoma/metabolism , Carrier Proteins/metabolism , Colonic Neoplasms/metabolism , Cytokines/metabolism , Rectal Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Analysis of Variance , Caco-2 Cells , Carrier Proteins/blood , Carrier Proteins/pharmacology , Child , Colon/metabolism , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Cytokines/blood , Cytokines/pharmacology , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neovascularization, Pathologic/metabolism , Phosphorylation/drug effects , Rectal Neoplasms/blood supply , Rectal Neoplasms/pathology , Rectum/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/drug effects , Young Adult , beta Catenin/drug effects , beta Catenin/metabolism
17.
Front Oncol ; 12: 943423, 2022.
Article in English | MEDLINE | ID: mdl-35965545

ABSTRACT

Background: The correlation between high white blood cell (WBC) count and poor prognosis has been identified in various types of cancer; however, the clinical significance and immune context of WBC count in colorectal cancer remains unclear. Methods: Between February 2009 and November 2014, 7,433 patients at the Shanghai Cancer Center who had undergone elective surgery for colorectal cancer were enrolled in this retrospective cohort study. Patients were divided into two groups: low and high preoperative WBC groups. Propensity score matching was used to address the differences in baseline characteristics. The Kaplan-Meier method and Cox regression analysis were used to identify independent prognostic factors in colorectal cancer patients. Tumor-infiltrating immune cells in the high and low preoperative WBC groups were compared using immunohistochemical staining. Results: Of the 7,433 patients who underwent colorectal cancer surgery and were available for analysis, 5,750 were included in the low preoperative WBC group, and 1,683 were included in the high preoperative WBC group. After propensity score matching, 1,553 patients were included in each group. Kaplan-Meier survival curves showed that a high preoperative WBC count was associated with a decreased overall survival (P = 0.002) and disease-free survival (P = 0.003), and that preoperative WBC count was an independent risk factor for overall survival (hazard ratio, 1.234; 95% confidence interval, 1.068-1.426; P = 0.004) and disease-free survival (hazard ratio, 1.210; 95% confidence interval, 1.047-1.397, P = 0.01). Compared to the low preoperative WBC group, the high preoperative WBC group exhibited higher expression of regulatory T cells (P = 0.0034), CD68+ macrophages (P = 0.0071), and CD66b+ neutrophils (P = 0.0041); increased expression of programmed cell death protein 1 (P = 0.005) and programmed cell death ligand 1 (P = 0.0019); and lower expression of CD8+ T cells (P = 0.0057) in colorectal cancer patients. Conclusions: Our research indicates that a high preoperative WBC count is a prognostic indicator in colorectal cancer patients and is associated with an immunosuppressive tumor microenvironment, which could aid in future risk stratification.

18.
Cell Death Discov ; 8(1): 375, 2022 Aug 27.
Article in English | MEDLINE | ID: mdl-36030287

ABSTRACT

Neutrophil extracellular traps (NETs) assist pathogen clearance, while excessive NETs formation is associated with exacerbated inflammatory responses and tissue injury in acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Autophagy is generally considered to be a protective process, but autophagy dysfunction is harmful. Whether and how NETs affect autophagic flux during sepsis-induced ALI are currently unknown. Here, we confirmed that the level of NETs was increased in ARDS patients and mice models, which led to impairment of autophagic flux and deterioration of the disease. Mechanistically, NETs activated METTL3 mediated m6A methylation of Sirt1 mRNA in alveolar epithelial cells, resulting in abnormal autophagy. These findings provide new insights into how NETs contribute to the development of sepsis-associated ALI/ARDS.

19.
Front Immunol ; 13: 950782, 2022.
Article in English | MEDLINE | ID: mdl-36081499

ABSTRACT

Colorectal cancer (CRC) is one of the most fatal cancers of the digestive system. Although cancer stem cells and metabolic reprogramming have an important effect on tumor progression and drug resistance, their combined effect on CRC prognosis remains unclear. Therefore, we generated a 21-gene mRNA stemness index-related metabolic risk score model, which was examined in The Cancer Genome Atlas and Gene Expression Omnibus databases (1323 patients) and validated using the Zhongshan Hospital cohort (200 patients). The high-risk group showed more immune infiltrations; higher levels of immunosuppressive checkpoints, such as CD274, tumor mutation burden, and resistance to chemotherapeutics; potentially better response to immune therapy; worse prognosis; and advanced stage of tumor node metastasis than the low-risk group. The combination of risk score and clinical characteristics was effective in predicting overall survival. Zhongshan cohort validated that high-risk score group correlated with malignant progression, worse prognosis, inferior adjuvant chemotherapy responsiveness of CRC, and shaped an immunoevasive contexture. This tool may provide a more accurate risk stratification in CRC and screening of patients with CRC responsive to immunotherapy.


Subject(s)
Colorectal Neoplasms , Machine Learning , Chemotherapy, Adjuvant , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Prognosis , Risk Factors
20.
Carcinogenesis ; 32(12): 1897-904, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22002319

ABSTRACT

p57 is a multifunctional protein involved in the regulation of tumor formation and development; however, the biological role of p57 in the pathogenesis of hepatocellular carcinoma (HCC) is poorly understood. To explore the role of p57 in the development of HCC, we examined p57 messenger RNA (mRNA) and protein levels in HCC tissues and adjacent non-cancerous tissues by immunohistochemistry, real-time polymerase chain reaction and western blot analysis. Moreover, we generated stable p57 knockdown HCC cell lines to investigate the impact of p57 downregulation on the growth and invasion of HCC in vitro and in vivo. Our results showed that p57 mRNA and protein levels were significantly decreased in human HCC tissues. In addition, this reduction in p57 expression was associated with increased tumor size, more advanced TNM stages, the presence of capsule invasion and extrahepatic metastasis and decreased overall survival time. In human HCC cell lines, p57 downregulation increased the expression of cyclin D1 and CDK2 and enhanced the activities of CDK4/cyclin D1 and CDK2/cyclin E complexes, resulting in increased cellular proliferation and growth of xenografts. Furthermore, p57 downregulation accelerated the invasion of HCC cells in vitro and in vivo by controlling the activity of LIMK1. In conclusion, the downregulation of p57 accelerates the growth and invasion of HCC, indicating that p57 is an important tumor suppressor in HCC. Based on these findings, p57 may be a potential target for HCC prevention and therapy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p57/physiology , Down-Regulation , Liver Neoplasms/pathology , Neoplasm Invasiveness , Adult , Aged , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Middle Aged , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL