Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Placenta ; 149: 18-28, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490094

ABSTRACT

INTRODUCTION: Gestational trophoblastic disease (GTD) encompasses a spectrum of rare pre-malignant and malignant entities originating from trophoblastic tissue, including partial hydatidiform mole, complete hydatidiform mole and choriocarcinoma. ß-galactoside α2,6 sialyltransferase 1 (ST6Gal1), the primary sialyltransferase responsible for the addition of α2,6 sialic acids, is strongly associated with the occurrence and development of several tumor types. However, the role of ST6Gal1/α2,6 -sialylation of trophoblast cells in GTD is still not well understood. METHODS: The expression of ST6Gal1 was investigated in GTD and human immortalized trophoblastic HTR-8/SVneo cells and human gestational choriocarcinoma JAR cells. We evaluated the effect of ST6Gal1 on proliferation and stemness of trophoblastic cells. We also examined the effect of internal miR-199a-5p on ST6Gal1 expression. The role of ST6Gal1 in regulating α2,6-sialylated integrin ß1 and its significance in the activation of integrin ß1/focal adhesion kinase (FAK) signaling pathway were also explored. RESULTS: ST6Gal1 was observed to be highly expressed in GTD. Overexpression of ST6Gal1 promoted the proliferation and stemness of HTR-8/SVneo cells, whereas knockdown of ST6Gal1 suppressed the viability and stemness of JAR cells. MiR-199a-5p targeted and inhibited the expression of ST6Gal1 in trophoblastic cells. In addition, we revealed integrin ß1 was highly α2,6-sialylated in JAR cells. Inhibition of ST6Gal1 reduced α2,6-sialylation on integrin ß1 and suppressed the integrin ß1/FAK pathway in JAR cells, thereby affecting its biological functions. DISCUSSION: This study demonstrated that ST6Gal1 plays important roles in promoting proliferation and stemness through the integrin ß1 signaling pathway in GTD. Therefore, ST6Gal1 may have a potential role in the occurrence and development of GTD.


Subject(s)
Choriocarcinoma , Gestational Trophoblastic Disease , Integrin beta1 , MicroRNAs , Female , Humans , Pregnancy , Cell Proliferation , Choriocarcinoma/pathology , Integrin beta1/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism
2.
J Cereb Blood Flow Metab ; 44(7): 1128-1144, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38230663

ABSTRACT

The neural cell adhesion molecule (NCAM) promotes neural development and regeneration. Whether NCAM mimetic peptides could synergize with bone marrow mesenchymal stem cells (BMSCs) in stroke treatment deserves investigation. We found that the NCAM mimetic peptide P2 promoted BMSC proliferation, migration, and neurotrophic factor expression, protected neurons from oxygen-glucose deprivation through ERK and PI3K/AKT activation and anti-apoptotic mechanisms in vitro. Following middle cerebral artery occlusion (MCAO) in rats, P2 alone or in combination with BMSCs inhibited neuronal apoptosis and induced the phosphorylation of ERK and AKT. P2 combined with BMSCs enhanced neurotrophic factor expression and BMSC proliferation in the ischemic boundary zone. Moreover, combined P2 and BMSC therapy induced translocation of nuclear factor erythroid 2-related factor, upregulated heme oxygenase-1 expression, reduced infarct volume, and increased functional recovery as compared to monotreatments. Treatment with LY294002 (PI3K inhibitor) and PD98059 (ERK inhibitor) decreased the neuroprotective effects of combined P2 and BMSC therapy in MCAO rats. Collectively, P2 is neuroprotective while P2 and BMSCs work synergistically to improve functional outcomes after ischemic stroke, which may be attributed to mechanisms involving enhanced BMSC proliferation and neurotrophic factor release, anti-apoptosis, and PI3K/AKT and ERK pathways activation.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Neural Cell Adhesion Molecules , Rats, Sprague-Dawley , Recovery of Function , Stroke , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Male , Neural Cell Adhesion Molecules/metabolism , Stroke/therapy , Stroke/metabolism , Recovery of Function/drug effects , Recovery of Function/physiology , Mesenchymal Stem Cell Transplantation/methods , Rats , Infarction, Middle Cerebral Artery/therapy , Infarction, Middle Cerebral Artery/metabolism , Cell Proliferation/drug effects , Apoptosis/drug effects , Peptides/pharmacology , Peptides/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cells, Cultured
3.
Cell Death Dis ; 14(10): 651, 2023 10 06.
Article in English | MEDLINE | ID: mdl-37798282

ABSTRACT

Embryo implantation into the uterus is the gateway for successful pregnancy. Proper migration and invasion of embryonic trophoblast cells are the key for embryo implantation, and dysfunction causes pregnancy failure. Protein glycosylation plays crucial roles in reproduction. However, it remains unclear whether the glycosylation of trophoblasts is involved in trophoblast migration and invasion processes during embryo implantation failure. By Lectin array, we discovered the decreased α1,3-fucosylation, especially difucosylated Lewis Y (LeY) glycan, in the villus tissues of miscarriage patients when compared with normal pregnancy women. Downregulating LeY biosynthesis by silencing the key enzyme fucosyltransferase IV (FUT4) inhibited migration and invasion ability of trophoblast cells. Using proteomics and translatomics, the specific LeY scaffolding glycoprotein of mesoderm-specific transcript (MEST) with glycosylation site at Asn163 was identified, and its expression enhanced migration and invasion ability of trophoblast cells. The results also provided novel evidence showing that decreased LeY modification on MEST hampered the binding of MEST with translation factor eIF4E2, and inhibited implantation-related gene translation initiation, which caused pregnancy failure. The α1,3-fucosylation of MEST by FUT4 may serve as a new biomarker for evaluating the functional state of pregnancy, and a target for infertility treatment.


Subject(s)
Embryo Implantation , Trophoblasts , Pregnancy , Humans , Female , Glycosylation , Trophoblasts/metabolism , Epithelial Cells/metabolism , Biomarkers/metabolism , Fucosyltransferases/genetics , Fucosyltransferases/metabolism
5.
Biol Reprod ; 108(4): 553-563, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36723873

ABSTRACT

Trophoblast cells are critical to placental angiogenesis in the first trimester of pregnancy. Dysfunction of trophoblast leads to defective vascular remodeling and impaired angiogenesis, which is believed as the major cause of placental insufficiency and pregnancy failure. Protein O-fucosyltransferase 1 (poFUT1) is mainly responsible for O-fucosylated glycan biosynthesis on glycoproteins, and poFUT1 deficiency causes embryonic lethality in mice. However, the expression and function of poFUT1 in trophoblast-mediated human placental vessel formation remain unclear. In the current study, we showed that fewer blood vessels were observed in the villi and decidua of miscarriage patients than in normal pregnancy women. The expression of poFUT1 was decreased in the trophoblast cells of miscarriage patients compared with normal pregnancy women. Employing HTR/SVneo cells and an in vivo chorioallantoic membrane assay, we demonstrated that poFUT1 promoted the proliferation, migration ability, and angiogenesis potential of trophoblast cells. The results also indicated that poFUT1 upregulated O-fucosylation on uPA, facilitated the binding of uPA and uPAR, activated the RhoA signaling pathway, and further enhanced the angiogenic capacity of trophoblast cells. Our study provides new evidence for a relationship between poFUT1/O-fucosylation and placental angiogenesis. These findings may provide potential diagnostic biomarkers and targeted therapies for miscarriage patients.


Subject(s)
Abortion, Spontaneous , Fucosyltransferases , Placenta , Animals , Female , Humans , Mice , Pregnancy , Cell Movement , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Placenta/metabolism , Pregnancy Trimester, First , Signal Transduction , Trophoblasts/metabolism
6.
J Cereb Blood Flow Metab ; 43(6): 882-892, 2023 06.
Article in English | MEDLINE | ID: mdl-36651130

ABSTRACT

The pathogenesis of cerebral atrophy (CA) is not clear. Previous studies show a high incidence of preterm CA in hemodialysis patients. This study aims to investigate the factors influencing CA and to derive a CA prediction nomogram in maintenance-hemodialysis patients. First, brain volumes of hemodialysis patients (≤55 years) were compared against age- and sex-matched healthy controls, and differences were revealed in bilateral insular cisterns width, maximum cerebral sulci width, Evans index, ventricular-brain ratio, frontal atrophy index, and temporal lobe ratio. Then, the patients were divided equally into "no or mild" or "severe" CA groups. Potential factors influencing CA were screened. Kt/V (urea removal index) and hemoglobin levels negatively correlated with CA degree, and were used to establish a nomogram within randomly assigned training and validation patient groups. The areas under the receiver operating characteristic curves (AUROC) for training and validation groups were 0.703 and 0.744, respectively. When potassium and calcium were added to the nomogram, the AUROC for training/validation group increased to 0.748/0.806. The nomogram had optimal AUROC for training (0.759) and validation (0.804) groups when albumin was also included. Hemodialysis patients showed reduced anterior brain volumes and the nomogram established herein may have predictive value for developing CA.


Subject(s)
Renal Dialysis , Urea , Infant, Newborn , Humans , Atrophy , Hemoglobins , Retrospective Studies
7.
Pathol Res Pract ; 241: 154257, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36470043

ABSTRACT

Both clear cell odontogenic carcinoma (CCOC) and sclerosing odontogenic carcinoma (SOC) are rare odontogenic malignancies. Here, we report a case of maxillary CCOC whose clinical and histologic features resembled those of SOC. Radiologically, the tumor presented as an ill-defined, expansile radiolucency with local bone destruction. Histologically, the tumor was comprised of thin cords or strands of odontogenic epithelium permeating through a sclerosed fibrous stroma with occasional clear cell foci. It damaged the cortical plates and invaded the adjacent soft tissue. Immunohistochemical expression of Pancytokeratin, Cytokeratin 19, p63, Cytokeratin 5/6, and Cytokeratin 14, as well as focal expression of Cytokeratin 7, demonstrated the epithelial nature of the tumor. Alcian Blue Periodic acid Schiff staining revealed a lack of intracellular mucin. Fluorescence in situ hybridization analysis revealed Ewing sarcoma RNA binding protein 1 and activating transcription factor 1 gene translocation, further confirming the diagnosis of CCOC. Lastly, we contextualized the genetic analysis of our case to that of CCOC in the literature.


Subject(s)
Carcinoma , Mouth Neoplasms , Odontogenic Tumors , Humans , In Situ Hybridization, Fluorescence , Odontogenic Tumors/diagnosis , Odontogenic Tumors/genetics , Odontogenic Tumors/pathology , RNA-Binding Protein EWS/genetics , Oncogene Proteins, Fusion/genetics
8.
Cells ; 11(19)2022 10 06.
Article in English | MEDLINE | ID: mdl-36231102

ABSTRACT

Peritoneal metastasis is the main cause of poor prognoses and high mortality in ovarian cancer patients. Abnormal protein glycosylation modification is associated with cancer malignancy. Elevated α1,3-mannosyltransferase 3 (ALG3), which catalyzes the α1,3-mannosylation of glycoproteins, has been found in some malignant tumors. However, the pathological significance of ALG3 and its regulatory mechanism in ovarian cancer metastasis is unclear. The results showed that the level of ALG3/α1,3-mannosylation was higher in human ovarian cancer tissues compared with normal ovarian tissues, as measured by Lectin chip, Western blot and Lectin blot analyses, as well as ovarian tissue microarray analysis. ALG3 was also correlated with the poor prognosis of ovarian cancer patients, according to survival analysis. The downregulation of ALG3 decreased the proliferation, stemness and peritoneal metastasis of ovarian cancer cells. The increase in urokinase plasminogen activator receptor (uPAR) α1,3-mannosylation catalyzed by ALG3 enhanced urokinase plasminogen activator (uPA)/uPAR activation and the interaction of uPAR with a disintegrin and metalloproteinase 8 (ADAM8), which promoted ovarian cancer peritoneal metastasis via the ADAM8/Ras/ERK pathway. Furthermore, decreased ALG3 suppressed ascites formation and the peritoneal metastasis of ovarian cancer cells in mice. This study highlights ALG3 as a potential diagnostic biomarker and prospective therapeutic target for ovarian cancer.


Subject(s)
Ovarian Neoplasms , Peritoneal Neoplasms , ADAM Proteins/metabolism , Animals , Antigens, CD , Biomarkers/metabolism , Carcinoma, Ovarian Epithelial , Disintegrins/metabolism , Female , Glycosylation , Humans , Lectins/metabolism , Mannosyltransferases/metabolism , Membrane Proteins/metabolism , Mice , Ovarian Neoplasms/pathology , Receptors, Urokinase Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism
9.
Clin Neuropathol ; 41(6): 253-262, 2022.
Article in English | MEDLINE | ID: mdl-35652545

ABSTRACT

Primary histiocytic sarcoma of the central nervous system is a rare lymphohematopoietic tumor originating from histiocytes. Here we report such a case with somatic NF2 mutation. Based on imaging studies, a 24-year-old woman presented with a homogeneously enhancing lesion in the right parietal lobe region and without other organ involvement. Histological analysis showed that the pleomorphic tumor cells were loosely arranged, and the neoplastic cells are characterized by abundant eosinophilic cytoplasm, highly atypical nuclei, and prominent nucleoli. The lesional cells were immunoreactive with antibodies against -CD68KP1, CD163 focally, lysozyme, and BRAF V600E. NGS-based genetic profiling revealed a pathogenic somatic NF2 (p.R196*) mutation. Additionally, BRAF (p.V600E), PDGFRA (p.V561D), BRCA1 (p.H437Q, VUS), and BRCA2 (p.E2343A, VUS) mutations were detected. However, the tumor did not respond to apatinib and anlotinib treatment, and the patient died 10 months after the initial diagnosis.


Subject(s)
Central Nervous System Neoplasms , Histiocytic Sarcoma , Female , Humans , Young Adult , Central Nervous System/pathology , Central Nervous System Neoplasms/genetics , Histiocytic Sarcoma/genetics , Histiocytic Sarcoma/pathology , Mutation , Proto-Oncogene Proteins B-raf
10.
Pathol Res Pract ; 236: 153984, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35753135

ABSTRACT

Follicular thyroid carcinoma (FTC) is one of the most common malignant tumors of the endocrine system. Recent studies have shown that voltage-gated sodium channels (VGSCs) affect the proliferation, migration, and invasion of tumor cells. However, the expression and functions of VGSCs, and the molecular pathways activated by VGSCs in FTC cells remain unclear. Our studies revealed that the expression of Nav1.6, encoded by SCN8A, was the predominantly upregulated subtype of VGSCs in FTC tissues. Knockdown of Nav1.6 significantly inhibited the proliferation, epithelial-mesenchymal transition and invasiveness of FTC cells. Using gene set enrichment analysis and Kyoto Encyclopedia of Genes and Genomics, SCN8A was predicted to be related to the JAK-STAT signaling pathway. Hence, we targeted the JAK-STAT pathway and demonstrated that Nav1.6 enhanced FTC cell proliferation, epithelial-mesenchymal transition, and invasion by phosphorylating JAK2 to activate STAT3. Furthermore, downregulating the expression of Nav1.6 improve the susceptibility of FTC cells to ubenimex in vitro. These results suggest Nav1.6 accelerates FTC progression through JAK/STAT signaling and may be a potential target for FTC therapy.


Subject(s)
Adenocarcinoma, Follicular , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Thyroid Neoplasms , Adenocarcinoma, Follicular/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
11.
Br J Cancer ; 126(12): 1795-1805, 2022 06.
Article in English | MEDLINE | ID: mdl-35217799

ABSTRACT

BACKGROUND: The lack of non-invasive methods for detection of early micro-metastasis is a major cause of the poor prognosis of non-small cell lung cancer (NSCLC) brain metastasis (BM) patients. Herein, we aimed to identify circulating biomarkers based on proteomics for the early diagnosis and monitoring of patients with NSCLC BM. METHODS: Upregulated proteins were detected by secretory proteomics in the animal-derived high brain metastatic lung cancer cell line. A well-designed study composed of three independent cohorts was then performed to verify these blood-based protein biomarkers: the serum discovery and verification cohorts (n = 80; n = 459), and the tissue verification cohort (n = 76). Logistic regression was used to develop a diagnostic biomarker panel. Model validation cohort (n = 160) was used to verify the stability of the constructed predictive model. Changes in serum Cathepsin F (CTSF) levels of patients were tracked to monitor the treatment response. Progression-free survival (PFS) and overall survival (OS) were analysed to assess their prognostic relevance. RESULTS: CTSF and Fibulin-1 (FBLN1) levels were specifically upregulated in sera and tissues of patients with NSCLC BM compared with NSCLC without BM and primary brain tumour. The combined diagnostic performance of CTSF and FBLN1 was superior to their individual ones. CTSF serum changes were found to reflect the therapeutic response of patients with NSCLC BM and the trends of progression were detected earlier than the magnetic resonance imaging changes. Elevated expression of CTSF in NSCLC BM tissues was associated with poor PFS, and was found to be an independent prognostic factor. CONCLUSIONS: We report a novel blood-based biomarker panel for early diagnosis, monitoring of therapeutic response, and prognostic evaluation of patients with NSCLC BM.


Subject(s)
Brain Neoplasms , Calcium-Binding Proteins , Carcinoma, Non-Small-Cell Lung , Cathepsin F , Lung Neoplasms , Animals , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Brain Neoplasms/blood , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Calcium-Binding Proteins/blood , Calcium-Binding Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cathepsin F/blood , Cathepsin F/metabolism , Humans , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Prognosis , Up-Regulation
12.
Stem Cell Rev Rep ; 18(2): 585-594, 2022 02.
Article in English | MEDLINE | ID: mdl-34449012

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs) have been shown to promote stroke recovery, however, the underlying mechanisms are not well understood. In this study naïve rats were intravenously injected with syngeneic BMSCs to screen for potential differences in brain metabolite spectrum versus vehicle-treated controls by capillary electrophoresis-mass spectrometry. A total of 65 metabolites were significantly changed after BMSC treatment. Among them, 5-oxoproline, an intermediate in the biosynthesis of the endogenous glutathione (GSH), was increased. To confirm the obtained results and investigate the metabolic pathways, BMSCs were injected into rats 24 h after middle cerebral artery occlusion (MCAO). Rats receiving vehicle solution and sham-operated animals served as controls. High performance liquid chromatography, reverse transcription-quantitative polymerase chain reaction, and Western blotting revealed that intravenous BMSC application increased the levels of 5-oxoproline and GSH in MCAO rats, as well as the expression of key enzymes involved in GSH synthesis including, gamma-glutamylcyclotransferase and gamma-glutamylcysteine ligase. Subsequent clinical investigation confirmed that acute ischemic stroke patients had higher plasma 5-oxoproline and GSH levels than age- and sex-matched non-stroke controls. The optimal cutoff value for 5-oxoproline diagnosing acute ischemic stroke (≤ 7d) was 3.127 µg/mL (sensitivity, 63.4 %; specificity, 81.2 %) determined by receiver characteristic operator curve. The area under the curve was 0.782 (95 % confidence interval: 0.718-0.845). Our findings indicate that BMSCs play a protective role in ischemic stroke through upregulation of GSH and 5-oxoproline is a potential biomarker for acute ischemic stroke. Ischemic stroke causes oxidative stress and induction of endogenous, glutathione-dependent anti-oxidative mechanisms. 5-oxoproline, an important metabolite in glutathione biosynthesis, could serve as a biomarker of acute ischemic stroke. Moreover, intravenous bone marrow mesenchymal stem cell (BMSC) treatment after experimental stroke upregulates the expression of key enzymes involved in glutathione synthesis, which results in better antioxidative defense and improved stroke outcome.


Subject(s)
Ischemic Stroke , Mesenchymal Stem Cells , Stroke , Animals , Bone Marrow Cells/metabolism , Glutathione/metabolism , Glutathione/pharmacology , Glutathione/therapeutic use , Humans , Infarction, Middle Cerebral Artery/metabolism , Mesenchymal Stem Cells/metabolism , Pyrrolidonecarboxylic Acid/metabolism , Pyrrolidonecarboxylic Acid/pharmacology , Pyrrolidonecarboxylic Acid/therapeutic use , Rats , Stroke/metabolism , Stroke/therapy , Up-Regulation
14.
Exp Cell Res ; 393(2): 112095, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32442537

ABSTRACT

Aberrant protein glycosylation is involved in many diseases including cancer. This study investigated the role of fucosyltransferase VII (FUT7) in the progression of follicular thyroid carcinoma (FTC). FUT7 expression was found to be upregulated in FTC compared to paracancerous thyroid tissue, and in FTC with T2 stage of TMN classification compared to FTC with T1 stage. FUT7 overexpression promoted cell proliferation, epithelial-mesenchymal transition (EMT), and the migration and invasion of primary FTC cell line FTC-133. Consistently, FUT7 knock-down inhibited cell proliferation, EMT, as well as the migration and invasion of the metastatic FTC cell line FTC-238. Mechanistic investigation revealed that FUT7 catalyzed the α1,3-fucosylation of epidermal growth factor receptor (EGFR) in FTC cells. The extent of glycan α1,3-fucosylation on EGFR was positively correlated with the activation of EGFR in the presence/absence of epidermal growth factor (EGF) treatment. Furthermore, FUT7 was shown to enhance EGF-induced progression of FTC cells through mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathways. These findings provide a new perspective on FUT7 that may be a novel diagnostic and therapeutic target of FTC.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Fucosyltransferases/metabolism , Adenocarcinoma, Follicular/drug therapy , Adenocarcinoma, Follicular/pathology , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Epidermal Growth Factor/metabolism , ErbB Receptors/drug effects , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Up-Regulation/drug effects
15.
J Cell Physiol ; 235(2): 1076-1089, 2020 02.
Article in English | MEDLINE | ID: mdl-31276203

ABSTRACT

Glycosylation alters the molecular and functional features of glycoproteins, which is closely related with many physiological processes and diseases. During "window of implantation", uterine endometrium transforms into a receptive status to accept the embryo, thereby establishing successful embryo implantation. In this article, we aimed at investigating the role of N-glycosylation, a major modification type of glycoproteins, in the process of endometrial receptivity establishment. Results found that human uterine endometrial tissues at mid-secretory phase exhibited Lectin PHA-E+L (recognizes the branched N-glycans) positive N-glycans as measured by the Lectin fluorescent staining analysis. By utilizing in vitro implantation model, we found that de-N-glycosylation of human endometrial Ishikawa and RL95-2 cells by tunicamycin (inhibitor of N-glycosylation) and peptide-N-glycosidase F (PNGase F) impaired their receptive ability to human trophoblastic JAR cells. Meanwhile, N-glycosylation of integrin αvß3 and leukemia inhibitory factor receptor (LIFR) are found to play key roles in regulating the ECM-dependent FAK/Paxillin and LIF-induced STAT3 signaling pathways, respectively, thus affecting the receptive potentials of endometrial cells. Furthermore, in vivo experiments and primary mouse endometrial cells-embryos coculture model further verified that N-glycosylation of mouse endometrial cells contributed to the successful implantation. Our results provide new evidence to show that N-glycosylation of uterine endometrium is essential for maintaining the receptive functions, which gives a better understanding of the glycobiology of implantation.


Subject(s)
Embryo Implantation/physiology , Endometrium/cytology , Animals , Cell Line , Female , Glycosylation , Humans , Mice , Models, Biological , Polysaccharides/metabolism , Pregnancy , Trophoblasts/physiology
16.
J Cell Biol ; 219(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31816056

ABSTRACT

The development of cerebral cortex requires spatially and temporally orchestrated proliferation, migration, and differentiation of neural progenitor cells (NPCs). The molecular mechanisms underlying cortical development are, however, not fully understood. The neural cell adhesion molecule (NCAM) has been suggested to play a role in corticogenesis. Here we show that NCAM is dynamically expressed in the developing cortex. NCAM expression in NPCs is highest in the neurogenic period and declines during the gliogenic period. In mice bearing an NPC-specific NCAM deletion, proliferation of NPCs is reduced, and production of cortical neurons is delayed, while formation of cortical glia is advanced. Mechanistically, NCAM enhances actin polymerization in NPCs by interacting with actin-associated protein profilin2. NCAM-dependent regulation of NPCs is blocked by mutations in the profilin2 binding site. Thus, NCAM plays an essential role in NPC proliferation and fate decision during cortical development by regulating profilin2-dependent actin polymerization.


Subject(s)
CD56 Antigen/physiology , Cell Differentiation , Cerebral Cortex/cytology , Neural Stem Cells/cytology , Neurogenesis , Neurons/cytology , Profilins/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cerebral Cortex/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/metabolism , Profilins/genetics
17.
Front Oncol ; 9: 1258, 2019.
Article in English | MEDLINE | ID: mdl-31799200

ABSTRACT

Cancer cells adopt glycolysis to facilitate the generation of biosynthetic substrates demanded by cell proliferation and growth, and to adapt to stress conditions such as excessive reactive oxygen species (ROS) accumulation. TIGAR (TP53-induced glycolysis and apoptosis regulator) is a fructose-2,6-bisphosphatase that is regulated by p53. TIGAR functions to inhibit glycolysis and promote antioxidative activities, which assists the generation of NADPH to maintain the levels of GSH and thus reduces intracellular ROS. However, the functions of TIGAR in gastric cancer (GC) remain unclear. TIGAR expression levels were detected by immunoblotting and immunohistochemistry in gastric cancer samples, along with four established cell lines of GC. The functions of TIGAR were determined by utilizing shRNA-mediated knockdown experiments. The NADPH/NADP+ ratio, ROS, mitochondrial ATP production, and phosphorus oxygen ratios were determined in TIGAR-depleted cells. Xenograft experiment was conducted with BALB/c nude mice. TIGAR was up-regulated compared with corresponding non-cancerous tissues in primary GCs. TIGAR knockdown significantly reduced cell proliferation and increased apoptosis. TIGAR protected cancer cells from oxidative stress-caused damages, but also glycolysis defects. TIGAR also increased the production of NADPH in gastric cancer cells. TIGAR knockdown led to increased ROS production, elevated mitochondrial ATP production, and phosphorus oxygen ratios. The prognosis of high TIGAR expression patients was significantly poorer than those with low TIGAR expression. Taken together, TIGAR exhibits oncogenic features in GC, which can be evaluated as a target for intervention in the treatment of GC.

18.
Brain ; 142(11): 3456-3472, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31529023

ABSTRACT

The immunological barrier currently precludes the clinical utilization of allogeneic stem cells. Although glial-restricted progenitors have become attractive candidates to treat a wide variety of neurological diseases, their survival in immunocompetent recipients is limited. In this study, we adopted a short-term, systemically applicable co-stimulation blockade-based strategy using CTLA4-Ig and anti-CD154 antibodies to modulate T-cell activation in the context of allogeneic glial-restricted progenitor transplantation. We found that co-stimulation blockade successfully prevented rejection of allogeneic glial-restricted progenitors from immunocompetent mouse brains. The long-term engrafted glial-restricted progenitors myelinated dysmyelinated adult mouse brains within one month. Furthermore, we identified a set of plasma miRNAs whose levels specifically correlated to the dynamic changes of immunoreactivity and as such could serve as biomarkers for graft rejection or tolerance. We put forward a successful strategy to induce alloantigen-specific hyporesponsiveness towards stem cells in the CNS, which will foster effective therapeutic application of allogeneic stem cells.


Subject(s)
Immune Tolerance , Microglia/immunology , Microglia/transplantation , Myelin Sheath , Neural Stem Cells/immunology , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Adoptive Transfer , Allografts , Animals , Cytokines/biosynthesis , Graft Rejection , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , T-Lymphocytes/immunology , Transplantation, Homologous
19.
EBioMedicine ; 44: 563-573, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31201143

ABSTRACT

BACKGROUND: Endometrial stromal cell decidualization is critical for embryo implantation. Dysfunctional decidualization leads to implantation failure, miscarriage and even pregnancy associated disorders in subsequent pregnancy trimesters. Protein glycosylation is involved in many physiological and pathological processes. Protein O-fucosyltransferase 1 (poFUT1) is the key enzyme for the O-fucosylation of proteins. However, the role and mechanism of poFUT1 in human endometrial stromal cell decidualization remain elusive. METHODS: We employed immunohistochemistry to detect the level of poFUT1 in the uterine endometrium from those of the proliferative phase, secretory phase, early pregnancy women and miscarriage patients. Using human endometrial stromal cells (hESCs) and a mouse model, the underlying mechanisms of poFUT1 in decidualization was investigated. FINDINGS: The level of poFUT1 was increased in the stromal cells of the secretory phase relative to those in the proliferative phase of the menstrual cycle, and decreased in the stromal cells of miscarriage patients compared to women with healthy early pregnancies. Furthermore, we found that poFUT1 promoted hESCs decidualization. The results also demonstrated that poFUT1 increased O-fucosylation on Notch1 in hESCs, which activated Notch1 signaling pathway. Activated Notch1 (NICD), as a specific trans-factor of PRL and IGFBP1 promoters, enhanced PRL and IGFBP1 transcriptional activity, thus inducing hESCs decidualization. INTERPRETATION: Level of poFUT1 is lower in the uterine endometrium from miscarriage patients than early pregnancy women. poFUT1 is critical in endometrial decidualization by controlling the O-fucosylation on Notch1. Our findings provide a new mechanism perspective on poFUT1 in uterine decidualization that may be a useful diagnostic and therapeutic target for miscarriage. FUND: National Natural Science Foundation of China (31770857, 31670810 and 31870794). Liaoning Provincial Program for Top Discipline of Basic Medical Sciences.


Subject(s)
Decidua/physiology , Endometrium/physiology , Fucosyltransferases/metabolism , Receptor, Notch1/metabolism , Adult , Animals , Cell Line , Cell Proliferation , Female , Fucosyltransferases/genetics , Gene Expression Regulation , Glycosylation , Humans , Immunohistochemistry , Menstrual Cycle , Mice , Models, Biological , Pregnancy , Stromal Cells/metabolism
20.
Placenta ; 75: 45-53, 2019 01.
Article in English | MEDLINE | ID: mdl-30712666

ABSTRACT

INTRODUCTION: Trophoblast proliferation and invasion are essential for embryo implantation and placentation. Protein glycosylation is one of the most common and vital post-translational modifications, regulates protein physical and biochemical properties. FUT8 is the only known fucosyltransferase responsible for catalyzing α1,6-fucosylation in mammals, and α1,6-fucosylated glycoproteins are found to participate in various physiopathological processes. However, whether FUT8/α1,6-fucosylation modulates the functions of trophoblastic cells remains elusive. METHODS: FUT8 in human placenta villi during 6-8 gestational weeks and trophoblastic cells were detected by Western blot and immunofluorescent staining. α1,6-fucosylation in tissues or cells were measured by Lectin LCA (Lens culinaris) fluorescent staining and Lectin blot. FUT8 expression was down-regulated by siRNA transfection in JAR and JEG-3 cells, and cell viability, motility and invasiveness ability were detected by the functional experiments. α1,6-fucosylation of insulin-like growth factor receptor (IGF-1R) was examined by immunoprecipitation, and the amount of phosphorylated IGF-1R was detected in FUT8 down-regulated JAR cells. RESULTS: Human placenta villi and trophoblastic cells expressed FUT8/α1,6-fucosylation. Knockdown FUT8 by siRNA transfection suppressed the proliferation, epithelial-mesenchymal transition, migration and invasion of JAR and JEG-3 cells. Furthermore, we found that FUT8 modified the α1,6-fucosylation of IGF-1R, and regulated IGF-1 dependent activation of IGF-1R, MAPK and PI3K/Akt signaling pathways in JAR cells. CONCLUSIONS: Our results implicate a critical role for FUT8 in maintaining the normal functions of trophoblastic cells, suggesting manipulating FUT8 may be an effective approach in pregnancy.


Subject(s)
Fucosyltransferases/physiology , Trophoblasts/physiology , Cell Line , Cell Proliferation , Humans , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...