Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biol Reprod ; 108(4): 553-563, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36723873

RESUMEN

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.


Asunto(s)
Aborto Espontáneo , Fucosiltransferasas , Placenta , Animales , Femenino , Humanos , Ratones , Embarazo , Movimiento Celular , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Placenta/metabolismo , Primer Trimestre del Embarazo , Transducción de Señal , Trofoblastos/metabolismo
2.
Br J Cancer ; 126(12): 1795-1805, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35217799

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas , Proteínas de Unión al Calcio , Carcinoma de Pulmón de Células no Pequeñas , Catepsina F , Neoplasias Pulmonares , Animales , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Proteínas de Unión al Calcio/sangre , Proteínas de Unión al Calcio/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Catepsina F/sangre , Catepsina F/metabolismo , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Pronóstico , Regulación hacia Arriba
3.
Clin Neuropathol ; 41(6): 253-262, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35652545

RESUMEN

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.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Sarcoma Histiocítico , Femenino , Humanos , Adulto Joven , Sistema Nervioso Central/patología , Neoplasias del Sistema Nervioso Central/genética , Sarcoma Histiocítico/genética , Sarcoma Histiocítico/patología , Mutación , Proteínas Proto-Oncogénicas B-raf
4.
Exp Cell Res ; 393(2): 112095, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32442537

RESUMEN

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.


Asunto(s)
Adenocarcinoma Folicular/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Fucosiltransferasas/metabolismo , Adenocarcinoma Folicular/tratamiento farmacológico , Adenocarcinoma Folicular/patología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Regulación hacia Arriba/efectos de los fármacos
5.
J Cell Physiol ; 235(2): 1076-1089, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31276203

RESUMEN

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.


Asunto(s)
Implantación del Embrión/fisiología , Endometrio/citología , Animales , Línea Celular , Femenino , Glicosilación , Humanos , Ratones , Modelos Biológicos , Polisacáridos/metabolismo , Embarazo , Trofoblastos/fisiología
6.
Brain ; 142(11): 3456-3472, 2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31529023

RESUMEN

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.


Asunto(s)
Tolerancia Inmunológica , Microglía/inmunología , Microglía/trasplante , Vaina de Mielina , Células-Madre Neurales/inmunología , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Traslado Adoptivo , Aloinjertos , Animales , Citocinas/biosíntesis , Rechazo de Injerto , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Linfocitos T/inmunología , Trasplante Homólogo
7.
J Cutan Pathol ; 2018 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-29943394

RESUMEN

BACKGROUND: Endocrine mucin-producing sweat gland carcinoma (EMPSGC) is a rare, low-grade adnexal neoplasm that most commonly involves the eyelid. Analogous to solid papillary carcinoma of the breast, it probably represents a precursor lesion to mucinous carcinoma. Here, we describe 11 cases of EMPSGC with molecular analysis. METHODS: We performed a retrospective search of the Johns Hopkins Medical Institute pathology database and identified 11 cases of EMPSGC. Immunohistochemistry was performed for chromogranin, synaptophysin, neuron specific enolase, estrogen receptor (ER), epithelial membrane antigen (EMA), cytokeratin 7 (CK7), and cytokeratin 20 (CK20). Array comparative genomic hybridization (aCGH) and BRAFV600E pyrosequencing were performed on two and three cases, respectively. RESULTS: We observed a strong female predilection (73% females, 8/11 cases) with an average age of 66 years (range, 56-83 years). EMPSGCs were associated with adjacent benign sweat gland cysts (3/11), atypical intraductal proliferation (1/11), and mucinous carcinoma (1/11). Immunohistochemically, all tumors expressed at least one neuroendocrine marker, ER, EMA, and CK7, and were negative for CK20. aCGH demonstrated a 6p11.2 to 6q16.1 deletion (1/2 cases). All cases were negative for BRAFV600E mutation (3/3 cases). CONCLUSION: This series provides further histopathologic support that EMPSGC represents a multistage progression to mucinous carcinoma. Additional studies are needed to understand its molecular mechanisms.

8.
J Cereb Blood Flow Metab ; 44(7): 1128-1144, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38230663

RESUMEN

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.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Moléculas de Adhesión de Célula Nerviosa , Péptidos , Recuperación de la Función , Accidente Cerebrovascular , Animales , Masculino , Ratas , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Infarto de la Arteria Cerebral Media/terapia , Infarto de la Arteria Cerebral Media/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Péptidos/farmacología , Péptidos/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Accidente Cerebrovascular/terapia , Accidente Cerebrovascular/metabolismo
9.
Placenta ; 149: 18-28, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38490094

RESUMEN

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.


Asunto(s)
Coriocarcinoma , Enfermedad Trofoblástica Gestacional , Integrina beta1 , MicroARNs , Femenino , Humanos , Embarazo , Proliferación Celular , Coriocarcinoma/patología , Integrina beta1/metabolismo , Sialiltransferasas/genética , Sialiltransferasas/metabolismo
10.
Pathol Res Pract ; 241: 154257, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36470043

RESUMEN

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.


Asunto(s)
Carcinoma , Neoplasias de la Boca , Tumores Odontogénicos , Humanos , Hibridación Fluorescente in Situ , Tumores Odontogénicos/diagnóstico , Tumores Odontogénicos/genética , Tumores Odontogénicos/patología , Proteína EWS de Unión a ARN/genética , Proteínas de Fusión Oncogénica/genética
11.
Cell Death Dis ; 14(10): 651, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37798282

RESUMEN

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.


Asunto(s)
Implantación del Embrión , Trofoblastos , Embarazo , Humanos , Femenino , Glicosilación , Trofoblastos/metabolismo , Células Epiteliales/metabolismo , Biomarcadores/metabolismo , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo
12.
J Cereb Blood Flow Metab ; 43(6): 882-892, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36651130

RESUMEN

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.


Asunto(s)
Diálisis Renal , Urea , Recién Nacido , Humanos , Atrofia , Hemoglobinas , Estudios Retrospectivos
13.
Cells ; 11(19)2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36231102

RESUMEN

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.


Asunto(s)
Neoplasias Ováricas , Neoplasias Peritoneales , Proteínas ADAM/metabolismo , Animales , Antígenos CD , Biomarcadores/metabolismo , Carcinoma Epitelial de Ovario , Desintegrinas/metabolismo , Femenino , Glicosilación , Humanos , Lectinas/metabolismo , Manosiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Neoplasias Ováricas/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
14.
Pathol Res Pract ; 236: 153984, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35753135

RESUMEN

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.


Asunto(s)
Adenocarcinoma Folicular , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neoplasias de la Tiroides , Adenocarcinoma Folicular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Humanos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología
15.
Stem Cell Rev Rep ; 18(2): 585-594, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34449012

RESUMEN

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.


Asunto(s)
Accidente Cerebrovascular Isquémico , Células Madre Mesenquimatosas , Accidente Cerebrovascular , Animales , Células de la Médula Ósea/metabolismo , Glutatión/metabolismo , Glutatión/farmacología , Glutatión/uso terapéutico , Humanos , Infarto de la Arteria Cerebral Media/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ácido Pirrolidona Carboxílico/metabolismo , Ácido Pirrolidona Carboxílico/farmacología , Ácido Pirrolidona Carboxílico/uso terapéutico , Ratas , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/terapia , Regulación hacia Arriba
16.
J Cell Biol ; 219(1)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31816056

RESUMEN

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.


Asunto(s)
Antígeno CD56/fisiología , Diferenciación Celular , Corteza Cerebral/citología , Células-Madre Neurales/citología , Neurogénesis , Neuronas/citología , Profilinas/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Corteza Cerebral/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/metabolismo , Profilinas/genética
17.
Mol Cell Biochem ; 331(1-2): 1-7, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19449091

RESUMEN

LeY oligosaccharide is stage specifically expressed by the embryo and uterine endometrium, and it plays important roles in embryo implantation. In addition to participating in the recognition and adhesion on fetal-maternal interface, LeY potentially regulates the expression of some implantation-related factors. However, it remains elusive whether it can mediate the involved signaling pathway. In this study, agarose-LeY beads were used to mimic the embryos, and the effects of LeY oligosaccharide on DAG/PKC signaling pathway was studied in human endometrial epithelial cells. Results showed that LeY could significantly trigger the activation of cPKCalpha and cPKCbeta2, and their translocation from the cytosol to the plasma membrane. The cellular DAG content was also upregulated, and the activation of PLCgamma1 was promoted. On the contrary, DAG/PKC signaling pathway was significantly inhibited when anti-LeY antibody was used after confirmation of LeY expression in human endometrial epithelial cells by immunohistochemistry and flow cytometry. These results suggest that LeY oligosaccharide acts as a signal molecule to modulate DAG/PKC signaling pathway.


Asunto(s)
Diglicéridos/metabolismo , Endometrio/citología , Endometrio/enzimología , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal , Regulación hacia Arriba , Línea Celular , Células Epiteliales/citología , Células Epiteliales/enzimología , Femenino , Humanos , Oligosacáridos/metabolismo , Fosfolipasa C gamma/metabolismo , Fosforilación , Transporte de Proteínas
18.
EBioMedicine ; 44: 563-573, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31201143

RESUMEN

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.


Asunto(s)
Decidua/fisiología , Endometrio/fisiología , Fucosiltransferasas/metabolismo , Receptor Notch1/metabolismo , Adulto , Animales , Línea Celular , Proliferación Celular , Femenino , Fucosiltransferasas/genética , Regulación de la Expresión Génica , Glicosilación , Humanos , Inmunohistoquímica , Ciclo Menstrual , Ratones , Modelos Biológicos , Embarazo , Células del Estroma/metabolismo
19.
Placenta ; 75: 45-53, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30712666

RESUMEN

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.


Asunto(s)
Fucosiltransferasas/fisiología , Trofoblastos/fisiología , Línea Celular , Proliferación Celular , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal
20.
Front Oncol ; 9: 1258, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31799200

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA