Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Cell Commun Signal ; 22(1): 101, 2024 02 08.
Article in English | MEDLINE | ID: mdl-38326896

ABSTRACT

BACKGROUND: Our previous studies demonstrated that 1-Pyrroline-5-carboxylate (P5C) released by prostate cancer cells inhibits T cell proliferation and function by increasing SHP1 expression. We designed this study to further explore the influence of P5C on T cell metabolism, and produced an antibody for targeting P5C to restore the functions of T cells. METHOD: We co-immunoprecipated SHP1 from T cells and analyzed the proteins that were bound to it using liquid chromatography mass spectrometry (LC/MS-MS). The influence of P5C on T cells metabolism was also detected by LC/MS-MS. Seahorse XF96 analyzer was further used to identify the effect of P5C on T cells glycolysis. We subsequently designed and produced an antibody for targeting P5C by monoclonal technique and verified its effectiveness to restore the function of T cells in vitro and in vivo. RESULT: PKM2 and LDHB bind SHP1 in T cells, and P5C could increase the levels of p-PKM2 while having no effect on the levels of PKM2 and LDHB. We further found that P5C influences T cell energy metabolism and carbohydrate metabolism. P5C also inhibits the activity of PKM2 and decreases the content of intracellular lactic acid while increasing the activity of LDH. Using seahorse XF96 analyzer, we confirmed that P5C remarkably inhibits glycolysis in T cells. We produced an antibody for targeting P5C by monoclonal technique and verified that the antibody could oppose the influence of P5C to restore the process of glycolysis and function in T cells. Meanwhile, the antibody also inhibits the growth of prostate tumors in an animal model. CONCLUSION: Our study revealed that P5C inhibits the process of glycolysis in T cells by targeting SHP1/PKM2/LDHB complexes. Moreover, it is important that the antibody for targeting P5C could restore the function of T cells and inhibit the growth of prostate tumors.


Subject(s)
Prostatic Neoplasms , Pyrroles , T-Lymphocytes , Humans , Male , Animals , Prostate , Tumor Microenvironment , Cell Proliferation , Glycolysis , Cell Line, Tumor
2.
J Clin Lab Anal ; 36(11): e24662, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36181310

ABSTRACT

BACKGROUND: GRAP2 is an adaptor protein involved in leukocyte signal activation; however, the prognostic value of GRAP2 and its correlation with immune infiltration in lung adenocarcinoma (LUAD) are unclear. METHODS: Original data were downloaded from the TCGA database and Gene Expression Omnibus (GEO) database. GRAP2 expression was analyzed with the TCGA and TIMER databases. We evaluated the influence of GRAP2 on clinical prognosis using the Kaplan-Meier plotter, GEO, and GEPIA database. The TIMER and TISIDB databases were used to investigate correlations between GRAP2 expression and cancer immune characteristics. Finally, we confirmed the expression of GRAP2 in LUAD by immunohistochemistry staining. RESULTS: The transcription levels of GRAP2 were significantly lower in several human cancer types, including LUAD, than in adjacent normal tissues. Immunohistochemistry staining confirmed that LUAD tumor tissues had lower GRAP2 protein expression levels than adjacent normal tissues. GRAP2 downregulation was associated with poorer overall survival, pathologic stage, T stage, N stage, and primary therapy outcome in LUAD. Mechanistically, we found a hub gene set that included a total of 91 genes coexpressed with GRAP2, which were closely related to the immune response in LUAD. The expression levels of GRAP2 were positively correlated with the infiltration levels of multiple immune cells and the cumulative survival time of a few immune cells. GRAP2 expression was found to be positively correlated with that of multiple immune markers, chemokines, chemokine receptors, and MHC molecules in LUAD. CONCLUSIONS: GRAP2 can be used as a biomarker for assessing prognosis and immune infiltration levels in LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Prognosis , Lung Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Adenocarcinoma of Lung/pathology , Biomarkers
3.
Dig Dis Sci ; 61(8): 2315-2327, 2016 08.
Article in English | MEDLINE | ID: mdl-27156077

ABSTRACT

BACKGROUND: The high mortality rate of hepatocellular carcinoma (HCC) is partly due to a lack of good diagnostic markers and treatment strategies. Recently, several microRNA (miRNA) profiling studies were conducted with HCC; however, their inconsistency means that their diagnostic or therapeutic value is debatable. AIMS: This study aims to systematically evaluate the consistency of miRNAs from multiple independent studies. METHODS: A systematic analysis of miRNAs from eligible publications was conducted, followed by real-time PCRs. The targets of highly consistent miRNAs were collected using online programs, followed by enrichment analyses for gene ontology terms and Kyoto encyclopedia of genes and genomes pathways. RESULTS: In total, 241 differentially expressed miRNAs were reported in 13 HCC profiling studies, of which 137 were upregulated and 104 downregulated. Among consistently upregulated miRNAs (cutoff > fourfold), miRNA-222, miRNA-21, miRNA-221, miRNA-210, and miRNA-224 were found increased in 8, 6, 6, 5, and 5 different studies, respectively. Among 137 downregulated miRNAs, miRNA-195, miRNA-199a, miRNA-125b, and miRNA-99a were reported in 8, 8, 5, and 5 studies, respectively. These results were confirmed by real-time PCR. Enrichment analyses demonstrated that programmed cell death and proliferation play important roles during the interplay of miRNA with HCC. CONCLUSIONS: miRNAs most consistently related to HCC are oncomirs miRNA-221/222 and tumor suppressors miRNA-199a/195.


Subject(s)
Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Cell Proliferation/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Humans
4.
Am J Pathol ; 182(5): 1617-28, 2013 May.
Article in English | MEDLINE | ID: mdl-23499375

ABSTRACT

Inflammatory bowel diseases are characterized by epithelial barrier disruption and alterations in immune regulation. Ste20-like proline/alanine-rich kinase (SPAK) plays a role in intestinal inflammation, but the underlying mechanisms need to be defined. Herein, SPAK knockout (KO) C57BL/6 mice exhibited significant increases in intestinal transepithelial resistance, a marked decrease in paracellular permeability to fluorescence isothiocyanate-dextran, and altered apical side tight junction sodium ion selectivity, compared with wild-type mice. Furthermore, the expression of junction protein, claudin-2, decreased. In contrast, expressions of occludin, E-cadherin, ß-catenin, and claudin-5 increased significantly, whereas no obvious change of claudin-1, claudin-4, zonula occludens protein 1, and zonula occludens protein 2 expressions was observed. In murine models of colitis induced by dextran sulfate sodium and trinitrobenzene sulfuric acid, KO mice were more tolerant than wild-type mice, as demonstrated by colonoscopy features, histological characteristics, and myeloperoxidase activities. Consistent with these findings, KO mice showed increased IL-10 levels and decreased proinflammatory cytokine secretion, ameliorated bacterial translocation on treatment with dextran sulfate sodium, and regulation of with no lysine (WNK) kinase activity. Together, these features may reduce epithelial permeability. In conclusion, SPAK deficiency increases intestinal innate immune homeostasis, which is important for control or attenuation of pathological responses in inflammatory bowel diseases.


Subject(s)
Gene Knockout Techniques , Inflammation/enzymology , Inflammation/prevention & control , Intestines/enzymology , Intestines/pathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Animals , Bacteria/metabolism , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate , Epithelial Cells/pathology , Inflammation/pathology , Lysine/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Tight Junction Proteins/metabolism
5.
J Cutan Pathol ; 41(8): 630-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24995964

ABSTRACT

BACKGROUND: Kaposi sarcoma (KS), caused by the infection of Kaposi sarcoma-associated herpesvirus (KSHV), is a disease manifested mainly by dark purple skin and mouth nodules. Cancer care studies showed that co-infection of KSHV and human immunodeficiency virus (HIV) was able to increase the patients' survival, but the underlying mechanisms are still elusive. METHODS: To understand the mechanism underlying the prolonged survival in KSHV-HIV co-infected patients, we performed microarray analysis on RNA extracted from biopsies from KS tumors and adjacent healthy tissues in four KS patients. Subsequently, we performed hierarchical clustering, gene ontology (GO) and ingenuity pathway analysis. We then characterized the roles of tight junction protein claudin-2 in the endothelial barrier function. RESULTS: Three hundred and forty-three differentially expressed genes were identified, of which 246 genes exhibited significantly increased expression in the tumor compared to the adjacent healthy tissue and 97 genes showed downregulated expression, including claudin-2. Knockdown of claudin-2 in cultured endothelial cells enhances barrier function by altering the charge selectivity, but not the size selectivity. CONCLUSION: Claudin-2 expression is decreased in KS tumors from patients co-infected with KSHV and HIV. Decreased claudin-2 enhances endothelial barrier function and may play a role in the prolonged survival of patients with KSHV and HIV co-infection.


Subject(s)
Capillary Permeability/genetics , Claudins/biosynthesis , Endothelial Cells/metabolism , Herpesvirus 8, Human , Sarcoma, Kaposi/genetics , Blotting, Western , Cluster Analysis , Coinfection , Down-Regulation , Endothelial Cells/pathology , HIV Infections/complications , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/pathology , Transcriptome
6.
Cell Immunol ; 281(1): 44-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23435349

ABSTRACT

Extracellular acidosis is involved in various pathological situations of central nervous system and the effects are largely mediated by acid sensing ion channels (ASICs). However, it remains unclear whether extracellular acidosis affects immune cells. Macrophages are immune cells that play important role in immune reactions. In this study we investigated the impact of extracellular acidosis on the function of bone marrow derived macrophages (BMMs). The results showed that extracellular acidosis upregulated the endocytosis, surface molecular expression and interleukin-10 secretion of BMMs, in which the expression of ASIC1 and ASIC3 was detected. Notably, extracellular acidosis stimulated endocytosis and upregulation of surface molecules expression in BMMs could be abolished by amiloride, a blocker of ASICs, and nonsteroid anti-inflammatory drugs. Our findings provide new insight into the role of extracellular acidosis in the regulation of immune function and suggest ASICs as new targets for the modulation of immune response.


Subject(s)
Acidosis/metabolism , Bone Marrow Cells/metabolism , Endocytosis/immunology , Macrophages/metabolism , Acid Sensing Ion Channel Blockers/pharmacology , Acid Sensing Ion Channels/drug effects , Acid Sensing Ion Channels/metabolism , Amiloride/pharmacology , Animals , Cells, Cultured , Female , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL
7.
J Immunol ; 187(3): 1496-505, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21705622

ABSTRACT

Inflammatory bowel disease, mainly Crohn's disease and ulcerative colitis, are characterized by epithelial barrier disruption and altered immune regulation. Colonic Ste20-like proline/alanine-rich kinase (SPAK) plays a role in intestinal inflammation, but its underlying mechanisms need to be defined. Both SPAK-transfected Caco2-BBE cells and villin-SPAK transgenic (TG) FVB/6 mice exhibited loss of intestinal barrier function. Further studies demonstrated that SPAK significantly increased paracellular intestinal permeability to FITC-dextran. In vivo studies using the mouse models of colitis induced by dextran sulfate sodium (DSS) and trinitrobenzene sulfonic acid showed that TG FVB/6 mice were more susceptible to DSS and trinitrobenzene sulfonic acid treatment than wild-type FVB/6 mice, as demonstrated by clinical and histological characteristics and enzymatic activities. Consistent with this notion, we found that SPAK increased intestinal epithelial permeability, which likely facilitated the production of inflammatory cytokines in vitro and in vivo, aggravated bacterial translocation in TG mice under DSS treatment, and consequently established a context favorable for the triggering of intestinal inflammation cascades. In conclusion, overexpression of SPAK inhibits maintenance of intestinal mucosal innate immune homeostasis, which makes regulation of SPAK important to attenuate pathological responses in inflammatory bowel disease.


Subject(s)
Genetic Predisposition to Disease , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/genetics , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Adaptive Immunity/genetics , Animals , Caco-2 Cells , Cells, Cultured , Disease Models, Animal , Humans , Immunity, Innate/genetics , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , Permeability , Protein Serine-Threonine Kinases/physiology
8.
Front Oncol ; 13: 1207081, 2023.
Article in English | MEDLINE | ID: mdl-37746262

ABSTRACT

Background: 2',5'-oligoadenylate synthetase 1 (OAS1), has been reported as a tumor driver gene in breast carcinoma and pancreatic carcinoma. However, the role of OAS1 in most tumors has not been reported. Methods: The original data of 35 tumor types were down load from the TCGA (The Cancer Genome Atlas) database and Human Protein Atlas (HPA) database. TIMER2, Kmplot, UALCAN, and TISIDB tools were used to investigate the expression and function of OAS1, and the role of OAS1 in prognosis, diagnostic value, and immune characteristics of pan-cancer. LUAD and PRAD cell lines, A549, H1975, PC-3 and C4-2 were utilized to perform cell function tests. Results: OAS1 expression was up-regulated in 12 tumor types and down-regulated in 2 tumor types. High OAS1 expression was correlated with poor prognosis in 6 tumor types, while high OAS1 expression was correlated with good prognosis in 2 tumor types. OAS1 was correlated with molecular subtypes in 8 tumor types and immune subtypes in 12 tumor types. OAS1 was positively associated with the expression of numerous immune checkpoint genes and tumor mutational burden (TMB). OAS1 had potential diagnostic value in 15 tumor types. Silence of OAS1 significantly inhibited the cell proliferation ability, and promoted G2/M cell cycle arrest of LUAD and PRAD cells. Meanwhile, silence of OAS1 enhanced cisplatin-induced apoptosis of LUAD and PRAD cells, but weakened cell migration. Conclusion: This pan-cancer study suggests that OAS1can be used as a molecular biomarker for prognosis in pan-cancer and may play an important role in tumor immune response.

9.
Heliyon ; 9(11): e22090, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38027636

ABSTRACT

Radiation resistance has always been one of the main obstacles to tumor radiotherapy. Therefore, understanding the mechanisms underlying radiotherapy resistance is a focus of research. In this study, we induced two radiation-resistant cell lines to mimic the radiation resistance of NSCLC and investigated the mechanisms of radiotherapy resistance. Cell radiosensitivity was analyzed by single-cell gel electrophoresis, colony formation and tumor sphere formation assays. A wound healing assay was used to analyze cell migration. Western blotting and siRNA were used to identify the potential mechanism. In animal model experiments, xenograft tumors were used to verify the difference between radiotherapy-resistant and nonresistant NSCLC models after radiotherapy. Our results showed that NSCLC radiation-resistant cells exhibited more radioresistance and migratory abilities under low-dose irradiation. The expression of LIMK2 and p-CFL1 were upregulated in NSCLC radiation-resistant cells. Knockdown of LIMK2 significantly enhanced the radiosensitivity of NSCLC-resistant cells. In vivo, low-dose radiotherapy suppressed tumor growth, induced apoptosis and upregulated the expression of LIMK2 in xenograft tumors. However, radiotherapy had little effect on the NSCLC radiation resistance model. In conclusion, NSCLC radiation-resistant cells exhibit more radioresistance and migratory ability under low-dose irradiation. Strikingly, knockdown of LIMK2 enhanced the radiosensitivity of NSCLC-resistant cells.

10.
J Biol Chem ; 286(35): 31003-31013, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21757725

ABSTRACT

The oligopeptide transporter PepT1 expressed in inflamed colonic epithelial cells transports small bacterial peptides, such as muramyl dipeptide (MDP) and l-Ala-γ-D-Glu-meso-diaminopimelic acid (Tri-DAP) into cells. The innate immune system uses various proteins to sense pathogen-associated molecular patterns. Nucleotide-binding oligomerization domain (NOD)-like receptors of which there are more than 20 related family members are present in the cytosol and recognize intracellular ligands. NOD proteins mediate NF-κB activation via receptor-interacting serine/threonine-protein kinase 2 (RICK or RIPK). The specific ligands for some NOD-like receptors have been identified. NOD type 1 (NOD1) is activated by peptides that contain a diaminophilic acid, such as the PepT1 substrate Tri-DAP. In other words, PepT1 transport activity plays an important role in controlling intracellular loading of ligands for NOD1 in turn determining the activation level of downstream inflammatory pathways. However, no direct interaction between Tri-DAP and NOD1 has been identified. In the present work, surface plasmon resonance and atomic force microscopy experiments showed direct binding between NOD1 and Tri-DAP with a K(d) value of 34.5 µM. In contrast, no significant binding was evident between muramyl dipeptide and NOD1. Furthermore, leucine-rich region (LRR)-truncated NOD1 did not interact with Tri-DAP, indicating that Tri-DAP interacts with the LRR domain of NOD1. Next, we examined binding between RICK and NOD1 proteins and found that such binding was significant with a K(d) value of 4.13 µM. However, NOD1/RICK binding was of higher affinity (K(d) of 3.26 µM) when NOD1 was prebound to Tri-DAP. Furthermore, RICK phosphorylation activity was increased when NOD was prebound to Tri-DAP. In conclusion, we have shown that Tri-DAP interacts directly with the LRR domain of NOD1 and consequently increases RICK/NOD1 association and RICK phosphorylation activity.


Subject(s)
Diaminopimelic Acid/analogs & derivatives , Leucine/chemistry , Nod1 Signaling Adaptor Protein/chemistry , Oligopeptides/chemistry , Biophysics/methods , Caco-2 Cells , Diaminopimelic Acid/chemistry , Diaminopimelic Acid/metabolism , Humans , Immunity, Innate , Inflammation , Microscopy, Atomic Force/methods , Nucleotides/chemistry , Oligopeptides/metabolism , Peptides/chemistry , Phosphorylation , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Lab Invest ; 92(8): 1203-12, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22641098

ABSTRACT

The transmembrane glycoprotein CD98 regulates integrin signaling that in turn controls cell proliferation and survival. CD98 expression is upregulated in various carcinomas, including colorectal cancer. Recently, by generating gain- and loss-of-function mouse models featuring genetic manipulation of CD98 expression specifically in intestinal epithelial cells (IECs), we have explored the crucial role of CD98 in the regulation of intestinal homeostasis and inflammation-associated tumorigenesis. In the present study, we investigated the contribution of CD98 to intestinal tumorigenesis in Apc(Min/+) mice and the underlying mechanism of action. Mice featuring IEC-specific CD98 overexpression (Tg animals) were crossed with Apc(Min/+) mice, and the characteristics of intestinal adenoma formation were assessed. Compared with Apc(Min/+) mice, Tg/Apc(Min/+) animals exhibited increases in both intestinal tumor incidence and tumor size; these parameters correlated with enhanced proliferation and decreased apoptosis of IECs. IEC-specific CD98 overexpression resulted in increased synthesis of the oncogenic proteins c-myc and cyclin-D1 in Apc(Min/+) mice, independently of the Wnt-APC-ß-catenin pathway, suggesting the implication of CD98 overexpression-mediated Erk activation. IEC-specific CD98 overexpression enhanced the production of proinflammatory cytokines and chemokines that are crucial for tumorigenesis. We validated our results in mice exhibiting IEC-specific CD98 downregulation (CD98(flox/+)VillinCre animals). IEC-specific CD98 downregulation efficiently attenuated tumor incidence and growth in Apc(Min/+) mice. The reduction of intestinal tumorigenesis upon IEC-specific CD98 downregulation was caused by the attenuation of IEC proliferation and cytokine/chemokine production. In conclusion, we show that CD98 exerts an oncogenic activity in terms of intestinal tumorigenesis, via an ability to regulate tumor growth and survival.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Fusion Regulatory Protein-1/biosynthesis , Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Apoptosis/physiology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Histocytochemistry , Intestinal Mucosa/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , MAP Kinase Signaling System , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism
12.
Am J Physiol Gastrointest Liver Physiol ; 302(5): G484-92, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22194420

ABSTRACT

Intestinal inflammation is characterized by epithelial disruption, leading to loss of barrier function and the recruitment of immune cells, including neutrophils. Although the mechanisms are not yet completely understood, interactions between environmental and immunological factors are thought to be critical in the initiation and progression of intestinal inflammation. In recent years, it has become apparent that the di/tripeptide transporter PepT1 may play an important role in the pathogenesis of such inflammation. In healthy individuals, PepT1 is primarily expressed in the small intestine and transports di/tripeptides for metabolic purposes. However, during chronic inflammation such as that associated with inflammatory bowel disease, PepT1 expression is upregulated in the colon, wherein the protein is normally expressed either minimally or not at all. Several recent studies have shown that PepT1 binds to and transports various bacterial di/tripeptides into colon cells, leading to activation of downstream proinflammatory responses via peptide interactions with innate immune receptors. In the present review, we examine the relationship between colonic PepT1-mediated peptide transport in the colon and activation of innate immune responses during disease. It is important to understand the mechanisms of PepT1 action during chronic intestinal inflammation to develop future therapies addressing inappropriate immune activation in the colon.


Subject(s)
Gastroenteritis/etiology , Inflammatory Bowel Diseases/etiology , Symporters/physiology , Animals , Colorectal Neoplasms/physiopathology , Gastroenteritis/drug therapy , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/physiopathology , Nod Signaling Adaptor Proteins/physiology , Oligopeptides/metabolism , Peptide Transporter 1 , Symporters/genetics
13.
Am J Physiol Gastrointest Liver Physiol ; 302(11): G1282-91, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22499850

ABSTRACT

The transmembrane glycoprotein CD98 is known to be involved in intestinal inflammation. In the present study, we found that CD98 overexpression in intestinal epithelial cells does not normally affect the expression of colonic (epithelial and immune cell) microRNAs (miRNAs), small noncoding RNAs that posttranscriptionally regulate a wide variety of biological processes. However, upon dextran sulfate sodium (DSS) treatment, the expression of several colonic miRNAs, but not miRNAs from other tissues such as liver and spleen, were differentially regulated in mice overexpressing CD98 in epithelial cells compared with wild-type (WT) animals. For example, the level of colonic miRNA 132 was not affected by DSS treatment in WT animals but was upregulated in mice overexpressing CD98 in intestinal epithelial cells. Other colonic miRNAs, including colonic miRNA 23a and 23b, were downregulated in WT animals after DSS treatment but not in colonic epithelial cell CD98-overexpressing mice. Interestingly, the expression of potential miRNA target genes affected intestinal epithelial cells that overexpress CD98 and cell types that did not overexpress CD98 but were in close proximity to CD98-overexpressing intestinal epithelial cells. Taken together, these observations show that the combination of an inflammatory context and intestinal epithelial cell expression of CD98 affects the regulation of miRNA expression in colonic epithelial and immune cells. This is new evidence that protein expression modulates miRNA expression and suggests the existence of regulatory crosstalk between proteins and miRNAs in diseases such as colitis.


Subject(s)
Colitis/metabolism , Colon/metabolism , Fusion Regulatory Protein-1/biosynthesis , Intestinal Mucosa/metabolism , MicroRNAs/metabolism , Animals , Colitis/genetics , Epithelial Cells/metabolism , Inflammation , Mice , MicroRNAs/genetics
14.
Gastroenterology ; 141(4): 1334-45, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21762661

ABSTRACT

BACKGROUND & AIMS: The human di/tripeptide transporter human intestinal H-coupled oligonucleotide transporter (hPepT1) is abnormally expressed in colons of patients with inflammatory bowel disease, although its exact role in pathogenesis is unclear. We investigated the contribution of PepT1 to intestinal inflammation in mouse models of colitis and the involvement of the nucleotide-binding oligomerization domain 2 (NOD2) signaling pathway in the pathogenic activity of colonic epithelial hPepT1. METHODS: Transgenic mice were generated in which hPepT1 expression was regulated by the ß-actin or villin promoters; colitis was induced using 2,4,6-trinitrobenzene sulfonic acid (TNBS) or dextran sodium sulfate (DSS) and the inflammatory responses were assessed. The effects of NOD2 deletion in the hPepT1 transgenic mice also was studied to determine the involvement of the PepT1-NOD2 signaling pathway. RESULTS: TNBS and DSS induced more severe levels of inflammation in ß-actin-hPepT1 transgenic mice than wild-type littermates. Intestinal epithelial cell-specific hPepT1 overexpression in villin-hPepT1 transgenic mice increased the severity of inflammation induced by DSS, but not TNBS. Bone marrow transplantation studies showed that hPepT1 expression in intestinal epithelial cells and immune cells has an important role in the proinflammatory response. Antibiotics abolished the effect of hPepT1 overexpression on the inflammatory response in DSS-induced colitis in ß-actin-hPepT1 and villin-hPepT1 transgenic mice, indicating that commensal bacteria are required to aggravate intestinal inflammation. Nod2-/-, ß-actin-hPepT1 transgenic/Nod2-/-, and villin-hPepT1 transgenic/Nod2-/- littermates had similar levels of susceptibility to DSS-induced colitis, indicating that hPepT1 overexpression increased intestinal inflammation in a NOD2-dependent manner. CONCLUSIONS: The PepT1-NOD2 signaling pathway is involved in aggravation of DSS-induced colitis in mice.


Subject(s)
Colitis/metabolism , Colon/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction , Symporters/metabolism , Actins/genetics , Animals , Anti-Bacterial Agents/pharmacology , Bone Marrow Transplantation , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Colitis/prevention & control , Colon/drug effects , Colon/immunology , Colon/microbiology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Peptide Transporter 1 , Promoter Regions, Genetic , Severity of Illness Index , Signal Transduction/drug effects , Symporters/genetics , Time Factors , Trinitrobenzenesulfonic Acid
15.
Materials (Basel) ; 15(8)2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35454531

ABSTRACT

Electrodeposition has attracted tremendous interest in functional coatings due to its advantages of high efficiency, inexpensiveness and ease of implementation. In this work, nickel graphene oxide (Ni-GO), nickel silicon carbide (Ni-SiC) and nickel graphene oxide/silicon carbide (Ni-GO/SiC) composite coatings were electrodeposited on the 2218 aluminum alloy (2218AlA) substrate. The microstructure, microhardness, bonding strength and tribological behaviors of the composite coatings were carried out. According to the results obtained, the composite coatings were dense and compact, with no visible defects and microcracks, and well bonded to 2218AlA substrate. The microhardness of composite coatings was significantly increased compared to that of the 2218AlA substrate. The microhardness of Ni-SiC composite coating was the highest, reaching 3.14 times that of the 2218AlA substrate. The friction response time, friction coefficient and wear rate of the composite coatings were obviously lower. For the Ni-GO composite coating, the average friction coefficient is the smallest at 45.35% of the 2218AlA substrate, while the wear rate is the smallest at 46.97% of the 2218AlA substrate. However, the comprehensive tribological performances of the Ni-GO/SiC composite coating were superior. The abrasive and adhesive wear were the main wear mechanisms of composite coatings, but the degree of damage was different.

16.
ACS Omega ; 7(21): 17921-17928, 2022 May 31.
Article in English | MEDLINE | ID: mdl-35664597

ABSTRACT

To improve the hydrophobicity of precious hardwood, a facile sanding pretreatment and alkyl ketene dimer (AKD) modification were performed. After the AKD modification, the wood was highly hydrophobic, and the contact angle was 143°. The increased hydrophobicity could be attributed to the ester bond formed between the wood hydroxyl groups and AKD, which was confirmed by attenuated total reflectance-Fourier transform infrared spectroscopy. Sanding pretreatment could further greatly increase the wood hydrophobicity and render it superhydrophobic, not only in a cross-section but also in the tangential and radial sections. The changed wood surface roughness could be responsible for the increased hydrophobicity, which was confirmed by characterization with a scanning electron microscope (SEM) and a three-dimensional optical microscope. Apart from the improved hydrophobicity, the AKD-modified wood exhibited excellent water, acid, and toluene resistance. After a 12 h immersion, the contact angle did not change significantly, and the acid immersion contributed to an improvement in the hydrophobicity of the wood. Furthermore, the resultant AKD-modified wood exhibited an excellent self-cleaning effect.

17.
ACS Omega ; 7(33): 29369-29379, 2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36033710

ABSTRACT

An efficient and durable flame-retardant coating was constructed on wood via a layer-by-layer (LBL) self-assembly approach by using a chitosan (CS), graphene oxide (GO), and ammonium polyphosphate (APP) ternary flame-retardant system. Both scanning electron microscopy (SEM) characterization and Fourier transform infrared spectroscopy (FT-IR) analysis indicated that CS-GO and APP polyelectrolytes were successfully deposited on wood, and the deposition amount was increased with the numbers of the LBLs. Thermogravimetric analysis revealed that the CS-GO-APP coating could decrease the initial and maximum thermal decomposition temperature of the coated wood while increase the char residue significantly, which may be attributed to the earlier degradation of CS and APP and effective heat barrier of the incorporated GO, thus increasing the thermal stability of the modified wood. The limited oxygen index (LOI) and cone calorimeter analysis results of the pristine and coated wood indicated that the fire resistance was significantly improved after CS-GO-APP modification; when 15 BLs were deposited on the wood, the LOI was increased from pristine 22 to 42, while the heat release rate and total heat release decreased from pristine 105.50 kW/m2 and 62.43 MJ/m2 to 57.51 kW/m2 and 34.31 MJ/m2, respectively. What is more, the 24 h immersion experiments and abrasion tests proved the excellent durability of the deposited coating. Furthermore, the SEM images of the char residues after flaming test proved that the CS-GO-APP assembly coating could promote the char layer formation on the wood surface and block the heat and flame spread, thus protecting the wood from fire attacking.

18.
J Biol Chem ; 285(2): 1479-89, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19892711

ABSTRACT

The transmembrane glycoprotein CD98 regulates multiple cellular functions, including extracellular signaling, epithelial cell adhesion/polarity, amino acid transport, and cell-cell interactions. MicroRNAs post-transcriptionally regulate gene expression, thereby functioning as modulators of numerous cellular processes, such as cell differentiation, proliferation, and apoptosis. Here, we investigated if microRNAs regulate CD98 expression during intestinal epithelial cell differentiation and inflammation. We found that microRNA-7 repressed CD98 expression in Caco2-BBE cells by directly targeting the 3'-untranslated region of human CD98 mRNA. Expression of CD98 was decreased, whereas that of microRNA-7 was increased in well-differentiated Caco2-BBE cells compared with undifferentiated cells. Undifferentiated crypt cells isolated from mouse jejunum showed higher CD98 levels and lower levels of mmu-microRNA-706, a murine original microRNA candidate for CD98, than well-differentiated villus cells. Importantly, microRNA-7 decreased Caco2-BBE cell attachment on laminin-1, and CD98 overexpression recovered this inhibition, suggesting that microRNA-7 modulates epithelial cell adhesion to extracellular matrix, which in turn could affect proliferation and differentiation during the migration of enterocytes across the crypt-villus axis, by regulating CD98 expression. In a pathological context, the pro-inflammatory cytokine interleukin 1-beta increased CD98 expression in Caco2-BBE cells by decreasing microRNA-7 levels. Consistent with the in vitro findings, microRNA-7 levels were decreased in actively inflamed Crohn disease colonic tissues, where CD98 expression was up-regulated, compared with normal tissues. Together, these results reveal a novel mechanism underlying regulation of CD98 expression during patho-physiological states. This study raises microRNAs as a promising target for therapeutic modulations of CD98 expression in intestinal inflammatory disorders.


Subject(s)
Cell Differentiation , Fusion Regulatory Protein-1/biosynthesis , Gene Expression Regulation , Intestinal Mucosa/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Animals , Caco-2 Cells , Cell Adhesion/genetics , Cell Communication/genetics , Cell Polarity/genetics , Crohn Disease/genetics , Crohn Disease/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fusion Regulatory Protein-1/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Jejunum/metabolism , Laminin/genetics , Laminin/metabolism , Male , Mice , MicroRNAs/genetics
19.
J Biol Chem ; 285(24): 18184-90, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20388705

ABSTRACT

We have reported that epithelial adenosine 2B receptor (A(2B)AR) mRNA and protein are up-regulated in colitis, which we demonstrated to be regulated by tumor necrosis factor alpha (TNF-alpha). Here, we examined the mechanism that governs A(2B)AR expression during colitis. A 1.4-kb sequence of the A(2B)AR promoter was cloned into the pFRL7 luciferase vector. Anti-microRNA (miRNA) was custom-synthesized based on specific miRNA binding sites. The binding of miRNA to the 3'-untranslated region (UTR) of A(2B)AR mRNA was examined by cloning this 3'-UTR downstream of the luciferase gene in pMIR-REPORT. In T84 cells, TNF-alpha induced a 35-fold increase in A(2B)AR mRNA but did not increase promoter activity in luciferase assays. By nuclear run-on assay, no increase in A(2B)AR mRNA following TNF-alpha treatment was observed. Four putative miRNA target sites (miR27a, miR27b, miR128a, miR128b) in the 3'-UTR of the A(2B)AR mRNA were identified in T84 cells and mouse colon. Pretreatment of cells with TNF-alpha reduced the levels of miR27b and miR128a by 60%. Over expression of pre-miR27b and pre-miR128a reduced A(2B)AR levels by >60%. Blockade of miR27b increased A(2B)AR mRNA levels by 6-fold in vitro. miR27b levels declined significantly in colitis-affected tissue in mice in the presence of increased A(2B)AR mRNA. Collectively, these data demonstrate that TNF-alpha-induced A(2B)AR expression in colonic epithelial cells is post-transcriptionally regulated by miR27b and miR128a and show that miR27b influences A(2B)AR expression in murine colitis.


Subject(s)
MicroRNAs/metabolism , Receptor, Adenosine A2B/biosynthesis , Transcription, Genetic , 3' Untranslated Regions , Animals , Cell Nucleus/metabolism , Colitis/metabolism , Cyclic AMP/metabolism , Humans , Mice , Mice, Inbred C57BL , RNA Processing, Post-Transcriptional , Tumor Necrosis Factor-alpha/metabolism
20.
Article in English | MEDLINE | ID: mdl-21030610

ABSTRACT

MicroRNAs (miRNAs), which are noncoding RNAs that posttranscriptionally inhibit expression of target genes, have recently emerged as important regulators of many cellular functions such as cell differentiation. The epithelial di/tripeptide membrane transporter PepT1 is expressed in highly differentiated cells (the villous tip) but not in undifferentiated cells (the crypt) of the small intestine. Here, we investigated the regulation of PepT1 expression by miRNAs and its functional consequences. We observed a reverse correlation between the expression levels of PepT1 and mature miRNA-92b (miR-92b) during the differentiation of intestinal epithelial Caco2-BBE cells, suggesting a miR-92b-mediated regulation of PepT1 expression. We demonstrate that miR-92b suppressed PepT1 expression at both mRNA and protein levels, with subsequent reduced PepT1 transport activity, in Caco2-BBE cells by directly targeting the PepT1 3'-untranslated region. In addition, miR-92b suppresses bacterial peptide-induced proinflammatory responses in intestinal epithelial cells by inhibiting PepT1 expression. Altogether, our study provides for the first time evidence for the regulation of PepT1 expression at a posttranscriptional level by miRNAs in intestinal epithelial cells during pathophysiological states.


Subject(s)
Intestinal Mucosa/metabolism , MicroRNAs/physiology , Symporters/biosynthesis , Caco-2 Cells , Cell Differentiation , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Down-Regulation , Humans , Inflammation/chemically induced , Inflammation/prevention & control , Intestinal Mucosa/cytology , Oligopeptides/pharmacology , Peptide Transporter 1
SELECTION OF CITATIONS
SEARCH DETAIL