Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 610
Filtrar
1.
DNA Cell Biol ; 40(11): 1418-1427, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34767735

RESUMEN

In this study, we examined the regulatory role of CCDC34 in the sorafenib sensitivity of hepatocellular carcinoma (HCC) and its functional partners. Wide-type Huh7 and Hep3B and induced sorafenib-resistant (SR) Huh7/SR and Hep3B/SR cells were used as in vitro cell models. Immunofluorescent staining and coimmunoprecipitation were performed to check protein-protein interaction. Cell Counting Kit-8 (CCK-8), terminal deoxynucleotidyl transferase-mediated nick end labeling (TUNEL), PI/Annexin V staining, and western blot analysis were performed to assess cell response to sorafenib. The results showed that CCDC34 upregulation in HCC was associated with poor survival. Huh7/SR and Hep3B/SR cells had significantly higher CCDC34 expression than the parental cell lines. RABL2A expression was significantly upregulated in SR HCC cells and interacted with CCDC34 in its GTP-bound state in Huh7/SR and Hep3B/SR cells. RABL2A depletion sensitized Huh7/SR and Hep3B/SR cells to sorafenib. RABL2A Q80L mutant (GTP-bound state locked), but not S35N mutant (GDP-bound state locked) overexpression increased sorafenib IC50 of Huh7 and Hep3B cells. CCDC34 depletion nearly abrogated the protective effects of RABL2A Q80L overexpression both in vitro and in vivo. RABL2A Q80L overexpression significantly increased the expression of p-p38 and p-JNK, the effects of which were significantly attenuated by CCDC34 depletion. In summary, we infer that the RABL2A-CCDC34 axis plays an important role in mediating p38/MAPK and JNK/MAPK signaling, thereby contributing to acquired sorafenib resistance in HCC.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Carcinoma Hepatocelular/metabolismo , Proteínas de Neoplasias/metabolismo , Sorafenib/metabolismo , Antígenos de Neoplasias/fisiología , Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , China , Resistencia a Antineoplásicos/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/fisiología , Transducción de Señal/efectos de los fármacos , Sorafenib/farmacología , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab/fisiología
2.
Mediators Inflamm ; 2021: 2255017, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34733114

RESUMEN

BACKGROUND: Apoptosis is one of the causes of immune depression in sepsis. Pyroptosis also occurs in sepsis. The toll-like receptor (TLR) 4 and receptor for advanced glycation end products (RAGE) have been shown to play important roles in apoptosis and pyroptosis. However, it is still unknown whether TLR4 inhibition decreases apoptosis in sepsis. METHODS: Stimulated peripheral blood mononuclear cells (PBMCs) with or without lipopolysaccharides (LPS) and high-mobility group box 1 (HMGB1) were cultured with or without TLR4 inhibition using monoclonal antibodies from 20 patients with sepsis. Caspase-3, caspase-8, and caspase-9 activities were measured. The expression of B cell lymphoma 2 (Bcl2) and Bcl2-associated X (Bax) was measured. The cell death of PBMCs was detected using a flow cytofluorimeter. RESULTS: After TLR4 inhibition, Bcl2 to Bax ratio elevated both in LPS and HMGB1-stimulated PBMCs. The activities of caspase-3, caspase-8, and caspase-9 did not change in LPS or HMGB1-stimulated PBMCs. The cell death of LPS and HMGB1-stimulated CD8 lymphocytes and monocytes increased after TLR4 inhibition. The cell death of CD4 lymphocytes was unchanged. CONCLUSION: The apoptosis did not decrease, while TLR4 was inhibited. After TLR4 inhibition, there was an unknown mechanism to keep cell death in stimulated PBMCs in patients with sepsis.


Asunto(s)
Apoptosis/fisiología , Leucocitos Mononucleares/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Sepsis/inmunología , Receptor Toll-Like 4/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología , Anciano , Antígenos de Neoplasias/fisiología , Caspasas/metabolismo , Células Cultivadas , Femenino , Proteína HMGB1/farmacología , Humanos , Lipopolisacáridos/farmacología , Masculino , Proteínas Quinasas Activadas por Mitógenos/fisiología , Piroptosis , Sepsis/patología , Receptor Toll-Like 4/fisiología
3.
Int Immunopharmacol ; 101(Pt A): 108183, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34649092

RESUMEN

Glioma is the most commonly diagnosed primary tumor of central nervous system. Previous studies found that the six-transmembrane epithelial antigen of prostate (STEAP) family can regulate the biological behaviors of several cancers. However, the role of STEAP family in glioma remains unclear. Here, we systematically evaluated the relationship between STEAP family and prognosis of glioma patients in multiple cohorts. The analysis showed that dysregulation of STEAP family may affect cancer-immunity cycle, immune infiltration and phenotypes resulting in an immunosuppressive microenvironment in glioma. To accurately predict the prognosis of glioma patients, gene-based risk models were established based on the expression of STEAP1, 2 and 3. Multivariate and univariate Cox analyses demonstrated that the risk models could independently predict the prognosis of glioma. Finally, chemotherapy and immune therapy responses for high- and low-risk patients were predicted. In conclusion, this study systematically analyzed the role of STEAP family in glioma and established a model for predicting therapy response in patients with glioma.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Glioma/metabolismo , Proteínas de la Membrana/metabolismo , Oxidorreductasas/metabolismo , Microambiente Tumoral/inmunología , Antígenos de Neoplasias/fisiología , Antineoplásicos/uso terapéutico , Proteínas de Ciclo Celular/fisiología , Línea Celular Tumoral , Expresión Génica , Glioma/tratamiento farmacológico , Glioma/inmunología , Glioma/terapia , Humanos , Inmunoterapia , Proteínas de la Membrana/fisiología , Oxidorreductasas/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
Drug Dev Res ; 82(8): 1096-1110, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34462935

RESUMEN

Trophoblast cell-surface antigen 2 (Trop 2) is a transmembrane glycoprotein that is highly expressed in various cancer types with relatively low or no baseline expression in most normal tissues. Its overexpression is associated with tumor growth and poor prognosis; Trop 2 is, therefore, an ideal therapeutic target for epithelial cancers. Several Trop 2 targeted therapeutics have recently been developed for the treatment of cancers, such as anti-Trop 2 antibodies and antibody-drug conjugates (ADCs), as well as Trop 2-specific cell therapy. In particular, the safety and clinical benefit of Trop 2-based ADCs have been demonstrated in clinical trials across multiple tumor types, including those with limited treatment options, such as triple-negative breast cancer, platinum-resistant urothelial cancer, and heavily pretreated non-small cell lung cancer. In this review, we elaborate on recent advances in Trop 2 targeted modalities and provide an overview of novel insights for future developments in this field.


Asunto(s)
Moléculas de Adhesión Celular/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/fisiología , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Humanos , Inmunoconjugados/uso terapéutico , Inmunoterapia Adoptiva
5.
Nucleic Acids Res ; 49(12): 6849-6862, 2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34157123

RESUMEN

Circular RNAs (circRNAs) are highly expressed in the brain and their expression increases during neuronal differentiation. The factors regulating circRNAs in the developing mouse brain are unknown. NOVA1 and NOVA2 are neural-enriched RNA-binding proteins with well-characterized roles in alternative splicing. Profiling of circRNAs from RNA-seq data revealed that global circRNA levels were reduced in embryonic cortex of Nova2 but not Nova1 knockout mice. Analysis of isolated inhibitory and excitatory cortical neurons lacking NOVA2 revealed an even more dramatic reduction of circRNAs and establishes a widespread role for NOVA2 in enhancing circRNA biogenesis. To investigate the cis-elements controlling NOVA2-regulation of circRNA biogenesis, we generated a backsplicing reporter based on the Efnb2 gene. We found that NOVA2-mediated backsplicing of circEfnb2 was impaired when YCAY clusters located in flanking introns were mutagenized. CLIP (cross-linking and immunoprecipitation) and additional reporter analyses demonstrated the importance of NOVA2 binding sites located in both flanking introns of circRNA loci. NOVA2 is the first RNA-binding protein identified to globally promote circRNA biogenesis in the developing brain.


Asunto(s)
Antígenos de Neoplasias/fisiología , Encéfalo/metabolismo , Neuronas/metabolismo , ARN Circular/metabolismo , Proteínas de Unión al ARN/fisiología , Empalme Alternativo , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Sitios de Unión , Encéfalo/embriología , Efrina-B2/genética , Exones , Regulación de la Expresión Génica , Células HEK293 , Humanos , Intrones , Ratones Noqueados , Antígeno Ventral Neuro-Oncológico , Motivos de Nucleótidos , ARN Circular/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946884

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive breast tumor subtype characterized by poor clinical outcome. In recent years, numerous advancements have been made to better understand the biological landscape of TNBC, though appropriate targets still remain to be determined. In the present study, we have determined that the expression levels of FGF2 and S100A4 are higher in TNBC with respect to non-TNBC patients when analyzing "The Invasive Breast Cancer Cohort of The Cancer Genome Atlas" (TCGA) dataset. In addition, we have found that the gene expression of FGF2 is positively correlated with S100A4 in TNBC samples. Performing quantitative PCR, Western blot, CRISPR/Cas9 genome editing, promoter studies, immunofluorescence analysis, subcellular fractionation studies, and ChIP assays, we have also demonstrated that FGF2 induces in TNBC cells the upregulation and secretion of S100A4 via FGFR1, along with the ERK1/2-AKT-c-Rel transduction signaling. Using conditioned medium from TNBC cells stimulated with FGF2, we have also ascertained that the paracrine activation of the S100A4/RAGE pathway triggers angiogenic effects in vascular endothelial cells (HUVECs) and promotes the migration of cancer-associated fibroblasts (CAFs). Collectively, our data provide novel insights into the action of the FGF2/FGFR1 axis through S100A4 toward stimulatory effects elicited in TNBC cells.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Proteínas de Neoplasias/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Proteína de Unión al Calcio S100A4/fisiología , Transducción de Señal/fisiología , Neoplasias de la Mama Triple Negativas/fisiopatología , Antígenos de Neoplasias/fisiología , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Neovascularización Patológica/fisiopatología , Comunicación Paracrina , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-rel/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Células Tumorales Cultivadas
7.
Toxicol Lett ; 348: 50-58, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34052307

RESUMEN

Antimony is a common environmental contaminant that causes biological toxicity in exposed populations worldwide. Previous studies have revealed that antimony promotes prostate cancer growth by stabilizing the c-Myc protein and mimicking androgen activity. However, the role of lncRNAs in the regulation of antimony-induced carcinogenesis remains unknown, and the precise mechanisms need to be explored. In the present study, we found that chronic exposure to antimony promoted cell growth and lipid metabolic disequilibrium in prostate cancer. Mechanistically, we identified a long noncoding RNA molecule, PCA3, that was substantially upregulated in LNCaP cells in response to long-term antimony exposure. Functional studies indicated that abnormal PCA3 expression modulated antimony-induced proliferation and cellular triglyceride and cholesterol levels. In addition, PCA3 levels were found to be inversely correlated with MIR-132-3 P levels by acting as a decoy for MIR-132-3P. Besides, SREBP1 directly interacted with MIR-132-3 P to increase cell growth and disrupt lipid metabolism by targeting its 3'UTR regions. Taken together, our results revealed that lncRNA PCA3 promotes antimony-induced lipid metabolic disorder in prostate cancer by targeting MIR-132-3 P/SREBP1 signaling.


Asunto(s)
Antígenos de Neoplasias/fisiología , Antimonio/toxicidad , Metabolismo de los Lípidos/efectos de los fármacos , MicroARNs/fisiología , Neoplasias de la Próstata/metabolismo , ARN Largo no Codificante/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/fisiología
8.
Clin Cancer Res ; 27(15): 4410-4421, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34031055

RESUMEN

PURPOSE: Gemcitabine-based chemotherapy regimens are first-line for several advanced cancers. Because of better tolerability, gemcitabine + cisplatin is a preferred neoadjuvant, adjuvant, and/or palliative chemotherapy regimen for advanced bladder cancer. Nevertheless, predicting treatment failure and overcoming resistance remain unmet clinical needs. We discovered that splice variant (V1) of HYAL-4 is a first-in-class eukaryotic chondroitinase (Chase), and CD44 is its major substrate. V1 is upregulated in bladder cancer and drives a malignant phenotype. In this study, we investigated whether V1 drives chemotherapy resistance. EXPERIMENTAL DESIGN: V1 expression was measured in muscle-invasive bladder cancer (MIBC) specimens by qRT-PCR and IHC. HYAL-4 wild-type (Wt) and V1 were stably expressed or silenced in normal urothelial and three bladder cancer cell lines. Transfectants were analyzed for chemoresistance and associated mechanism in preclinical models. RESULTS: V1 levels in MIBC specimens of patients who developed metastasis, predicted response to gemcitabine + cisplatin adjuvant/salvage treatment and disease-specific mortality. V1-expressing bladder cells were resistant to gemcitabine but not to cisplatin. V1 expression neither affected gemcitabine influx nor the drug-efflux transporters. Instead, V1 increased gemcitabine metabolism and subsequent efflux of difluorodeoxyuridine, by upregulating cytidine deaminase (CDA) expression through increased CD44-JAK2/STAT3 signaling. CDA inhibitor tetrahydrouridine resensitized V1-expressing cells to gemcitabine. While gemcitabine (25-50 mg/kg) inhibited bladder cancer xenograft growth, V1-expressing tumors were resistant. Low-dose combination of gemcitabine and tetrahydrouridine abrogated the growth of V1 tumors with minimal toxicity. CONCLUSIONS: V1/Chase drives gemcitabine resistance and potentially predicts gemcitabine + cisplatin failure. CDA inhibition resensitizes V1-expressing tumors to gemcitabine. Because several chemotherapy regimens include gemcitabine, our study could have broad significance.


Asunto(s)
Antígenos de Neoplasias/fisiología , Antimetabolitos Antineoplásicos/uso terapéutico , Condroitinasas y Condroitín Liasas/fisiología , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/fisiología , Histona Acetiltransferasas/fisiología , Hialuronoglucosaminidasa/fisiología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Desoxicitidina/uso terapéutico , Humanos , Ratones , Pronóstico , Insuficiencia del Tratamiento , Gemcitabina
9.
Anticancer Res ; 41(4): 2053-2058, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33813413

RESUMEN

AIM: To investigate potential associations between selected oncomarkers [carcinoembryonic antigen (CEA), C-terminus of cytokeratin 19 (CYFRA 21-1, CYFRA), and squamous cell carcinoma antigen (SCC)] and outcomes in patients with NSCLC treated with bevacizumab plus chemotherapy. PATIENTS AND METHODS: We retrospectively analysed 105 patients with NSCLC from the Czech TULUNG registry treated at University Hospital in Pilsen with bevacizumab plus chemotherapy. Response to therapy was tested by Fisher's exact test. Survival statistics were evaluated using the Kaplan-Meier method and Cox analysis. RESULTS: Only normal values of CYFRA (not CEA or SCC) were associated with significantly better overall and progression-free survival in univariate analysis. We also observed a trend for a better disease control rate in patients with normal levels of CYFRA. In a multivariate Cox model, only CYFRA was associated with significantly better overall but not progression-free survival. CONCLUSION: In our retrospective study, we point out the possibility of using CYFRA as a prognostic marker in patients with NSCLC treated with chemotherapy plus bevacizumab.


Asunto(s)
Antígenos de Neoplasias/fisiología , Antineoplásicos/uso terapéutico , Bevacizumab/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Queratina-19/fisiología , Neoplasias Pulmonares/tratamiento farmacológico , Anciano , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/sangre , Bevacizumab/efectos adversos , Biomarcadores Farmacológicos/análisis , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/fisiología , Antígeno Carcinoembrionario/análisis , Antígeno Carcinoembrionario/sangre , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Progresión de la Enfermedad , Femenino , Humanos , Queratina-19/análisis , Queratina-19/sangre , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Pronóstico , Estudios Retrospectivos , Serpinas/análisis , Serpinas/sangre , Resultado del Tratamiento
10.
Biochim Biophys Acta Mol Cell Res ; 1868(7): 119015, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33741433

RESUMEN

An essential requirement for cells to sustain a high proliferating rate is to be paired with enhanced protein synthesis through the production of ribosomes. For this reason, part of the growth-factor signaling pathways, are devoted to activate ribosome biogenesis. Enhanced production of ribosomes is a hallmark in cancer cells, which is boosted by different mechanisms. Here we report that the nucleolar tumor-protein MageB2, whose expression is associated with cell proliferation, also participates in ribosome biogenesis. Studies carried out in both siRNA-mediated MageB2 silenced cells and CRISPR/CAS9-mediated MageB2 knockout (KO) cells showed that its expression is linked to rRNA transcription increase independently of the cell proliferation status. Mechanistically, MageB2 interacts with phospho-UBF, a protein which causes the recruitment of RNA Pol I pre-initiation complex required for rRNA transcription. In addition, cells expressing MageB2 displays enhanced phospho-UBF occupancy at the rDNA gene promoter. Proteomic studies performed in MageB2 KO cells revealed impairment in ribosomal protein (RPs) content. Functionally, enhancement in rRNA production in MageB2 expressing cells, was directly associated with an increased dynamic in protein synthesis. Altogether our results unveil a novel function for a tumor-expressed protein from the MAGE-I family. Findings reported here suggest that nucleolar MageB2 might play a role in enhancing ribosome biogenesis as part of its repertoire to support cancer cell proliferation.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , Ribosomas/metabolismo , Antígenos de Neoplasias/fisiología , Línea Celular Tumoral , Nucléolo Celular/metabolismo , Proliferación Celular/genética , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Células HCT116 , Células HEK293 , Humanos , Proteínas de Neoplasias/fisiología , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Regiones Promotoras Genéticas/genética , Biosíntesis de Proteínas , Procesamiento Proteico-Postraduccional , Proteómica , ARN Polimerasa I/metabolismo , ARN Ribosómico/biosíntesis , Ribosomas/genética , Transcripción Genética/genética
11.
Mol Biol Cell ; 32(8): 675-689, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33625872

RESUMEN

A range of severe human diseases called ciliopathies is caused by the dysfunction of primary cilia. Primary cilia are cytoplasmic protrusions consisting of the basal body (BB), the axoneme, and the transition zone (TZ). The BB is a modified mother centriole from which the axoneme, the microtubule-based ciliary scaffold, is formed. At the proximal end of the axoneme, the TZ functions as the ciliary gate governing ciliary protein entry and exit. Since ciliopathies often develop due to mutations in genes encoding proteins that localize to the TZ, the understanding of the mechanisms underlying TZ function is of eminent importance. Here, we show that the ciliopathy protein Rpgrip1l governs ciliary gating by ensuring the proper amount of Cep290 at the vertebrate TZ. Further, we identified the flavonoid eupatilin as a potential agent to tackle ciliopathies caused by mutations in RPGRIP1L as it rescues ciliary gating in the absence of Rpgrip1l.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cilios/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Antígenos de Neoplasias/fisiología , Axonema/metabolismo , Cuerpos Basales/metabolismo , Proteínas de Ciclo Celular/fisiología , Centriolos/metabolismo , Cilios/fisiología , Ciliopatías/metabolismo , Ciliopatías/fisiopatología , Proteínas del Citoesqueleto/fisiología , Células HEK293 , Humanos , Ratones , Mutación , Células 3T3 NIH , Transducción de Señal
12.
Sci Rep ; 11(1): 425, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33432108

RESUMEN

Hypoxia is prevalent in atherosclerotic plaques, promoting plaque aggravation and subsequent cardiovascular disease (CVD). Transmembrane protein carbonic anhydrase IX (CAIX) is hypoxia-induced and can be shed into the circulation as soluble CAIX (sCAIX). As plaque macrophages are hypoxic, we hypothesized a role for CAIX in macrophage function, and as biomarker of hypoxic plaque burden and CVD. As tumor patients with probable CVD are treated with CAIX inhibitors, this study will shed light on their safety profile. CAIX co-localized with macrophages (CD68) and hypoxia (pimonidazole), and correlated with lipid core size and pro-inflammatory iNOS+ macrophages in unstable human carotid artery plaques. Although elevated pH and reduced lactate levels in culture medium of CAIX knock-out (CAIXko) macrophages confirmed its role as pH-regulator, only spare respiratory capacity of CAIXko macrophages was reduced. Proliferation, apoptosis, lipid uptake and expression of pro- and anti-inflammatory genes were not altered. Plasma sCAIX levels and plaque-resident CAIX were below the detection threshold in 50 and 90% of asymptomatic and symptomatic cases, respectively, while detectable levels did not associate with primary or secondary events, or intraplaque hemorrhage. Initial findings show that CAIX deficiency interferes with macrophage metabolism. Despite a correlation with inflammatory macrophages, plaque-resident and sCAIX expression levels are too low to serve as biomarkers of future CVD.


Asunto(s)
Antígenos de Neoplasias/fisiología , Anhidrasa Carbónica IX/fisiología , Enfermedades Cardiovasculares , Macrófagos/metabolismo , Anciano , Animales , Antígenos de Neoplasias/genética , Aterosclerosis/diagnóstico , Aterosclerosis/genética , Aterosclerosis/metabolismo , Biomarcadores/metabolismo , Anhidrasa Carbónica IX/genética , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Células Cultivadas , Estudios de Cohortes , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
13.
Mol Cell Biochem ; 476(1): 1-10, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32935296

RESUMEN

Prostate cancer (PC) is the most common reproductive cancer in men and the third leading cause of cancer death among men worldwide. Recently targeted therapy showed a significant therapeutic effect on PC, whereas finding more PC therapeutic target is still urgently needed. Melanoma-associated antigen-encoding C2 (MAGE-C2/CT10), which have significant homology with the MAGE-C1/CT-7 gene, was known to be involved in the development of a variety of tumors. However, the role and mechanism of MAGE-C2/CT10 in prostate cancer remains unclear. Herein, we found the high levels of MAGE-C2/CT10 in highly metastatic prostate cancer. Our findings confirmed that the depletion of MAGE-C2/CT10 suppressed the growth of PC cells, and restrained PC cell migration and invasion in vitro. We noticed MAGE-C2/CT10 could stimulate c-Myc expression via FBP1, and further contributed to PC cell proliferation and motility. Performing in vivo assays, we demonstrated MAGE-C2/CT10 promoted tumor growth and metastasis of PC cells in mice. Collectively, we found the abnormal expression of MAGE-C2/CT10 in PC, and revealed the regulatory mechanism underlying MAGE-C2/CT10 promoting PC progression and metastasis.


Asunto(s)
Antígenos de Neoplasias/fisiología , Regulación Neoplásica de la Expresión Génica , Metástasis de la Neoplasia , Proteínas de Neoplasias/fisiología , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Fructosa-Bifosfatasa/metabolismo , Humanos , Masculino , Melanoma/patología , Ratones , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Activación Transcripcional , Regulación hacia Arriba
14.
Respirology ; 26(5): 461-468, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33336433

RESUMEN

BACKGROUND AND OBJECTIVE: The course of systemic sclerosis-associated interstitial lung disease (SSc-ILD) is highly variable, and accurate prognostic markers are needed. KL-6 is a mucin-like glycoprotein (MUC1) expressed by type II pneumocytes, while CYFRA 21-1 is expressed by alveolar and bronchiolar epithelial cells. Both are released into the blood from cell injury. METHODS: Serum KL-6 and CYFRA 21-1 levels were measured in a retrospective (n = 189) and a prospective (n = 118) cohort of SSc patients. Genotyping of MUC1 rs4072037 was performed. Linear mixed-effect models were used to evaluate the relationship with change in lung function parameters over time, while association with survival was evaluated with Cox proportional hazard analysis. RESULTS: In both cohorts, KL-6 and CYFRA 21-1 were highest in patients with lung involvement, and in patients with extensive rather than limited ILD. KL-6 was higher in patients carrying the MUC1 rs4072037 G allele in both cohorts. In patients with SSc-ILD, serum KL-6, but not CYFRA 21-1, was significantly associated with DLCO decline in both cohorts (P = 0.001 and P = 0.004, respectively), and with FVC decline in the retrospective cohort (P = 0.005), but not the prospective cohort. When combining the cohorts and subgrouping by severity (median CPI = 45.97), KL-6 remained predictive of decline in DLCO in both milder (P = 0.007) and more severe disease (P = 0.02) on multivariable analysis correcting for age, gender, ethnicity, smoking history and MUC1 allele carriage. CONCLUSION: Our results suggest serum KL-6 predicts decline in lung function in SSc, suggesting its clinical utility in risk stratification for progressive SSc-ILD.


Asunto(s)
Antígenos de Neoplasias/inmunología , Queratina-19/inmunología , Enfermedades Pulmonares Intersticiales , Pulmón/fisiología , Esclerodermia Sistémica , Antígenos de Neoplasias/fisiología , Biomarcadores , Progresión de la Enfermedad , Humanos , Queratina-19/fisiología , Enfermedades Pulmonares Intersticiales/etiología , Estudios Prospectivos , Estudios Retrospectivos , Esclerodermia Sistémica/complicaciones
15.
Mol Biol Cell ; 31(24): 2687-2702, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32966160

RESUMEN

SLC45A2 encodes a putative transporter expressed primarily in pigment cells. SLC45A2 mutations cause oculocutaneous albinism type 4 (OCA4) and polymorphisms are associated with pigmentation variation, but the localization, function, and regulation of SLC45A2 and its variants remain unknown. We show that SLC45A2 localizes to a cohort of mature melanosomes that only partially overlaps with the cohort expressing the chloride channel OCA2. SLC45A2 expressed ectopically in HeLa cells localizes to lysosomes and raises lysosomal pH, suggesting that in melanocytes SLC45A2 expression, like OCA2 expression, results in the deacidification of maturing melanosomes to support melanin synthesis. Interestingly, OCA2 overexpression compensates for loss of SLC45A2 expression in pigmentation. Analyses of SLC45A2- and OCA2-deficient mouse melanocytes show that SLC45A2 likely functions later during melanosome maturation than OCA2. Moreover, the light skin-associated SLC45A2 allelic F374 variant restores only moderate pigmentation to SLC45A2-deficient melanocytes due to rapid proteasome-dependent degradation resulting in lower protein expression levels in melanosomes than the dark skin-associated allelic L374 variant. Our data suggest that SLC45A2 maintains melanosome neutralization that is initially orchestrated by transient OCA2 activity to support melanization at late stages of melanosome maturation, and that a common allelic variant imparts reduced activity due to protein instability.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Melanocitos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Pigmentación de la Piel/fisiología , Animales , Antígenos de Neoplasias/fisiología , Proteínas Portadoras/metabolismo , Línea Celular , Canales de Cloruro/metabolismo , Células HeLa , Humanos , Lisosomas/metabolismo , Masculino , Melanosomas/metabolismo , Proteínas de Transporte de Membrana/fisiología , Ratones , Pigmentación/fisiología , Estabilidad Proteica , Piel/metabolismo
16.
Biol Cell ; 112(12): 383-397, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32894882

RESUMEN

The main function of carbonic anhydrases (CAs) in cancer cells is the pH regulation through a conversion of H2 O and CO2 to H+ and HCO3 - . However, the data of in vitro and in vivo studies have demonstrated that transmembrane isoforms of CA IX and CA XII are involved in various steps of cancer cell migration, invasion and metastasis. According to literature, inhibition of these CAs can affect the expression of multiple proteins. Some scientific groups have reported the possible interactions between CA IX and E-cadherin-catenin system, CA IX and integrins, CA IX, CA XII and ion transporters, which all are highly involved in cell-to-cell adhesion, the formation of membrane protrusions and focal adhesions. Nevertheless, CA IX and CA XII have a high impact on tumour growth and metastases formation. The data discussed in this review are quite recent. It highly support the role of CA IX and CA XII in various cancer metastasis processes through their interactions to other invasion proteins. Nevertheless, all findings show the great potential of these CAs in the context of research and application in clinical use.


Asunto(s)
Antígenos de Neoplasias/fisiología , Anhidrasa Carbónica IX/fisiología , Anhidrasas Carbónicas/fisiología , Metástasis de la Neoplasia , Neoplasias , Antígenos CD/metabolismo , Cadherinas/metabolismo , Adhesión Celular , Movimiento Celular , Humanos , Neoplasias/metabolismo , Neoplasias/patología
17.
JCI Insight ; 5(17)2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32879135

RESUMEN

Prader-Willi syndrome (PWS) is a developmental disorder caused by loss of maternally imprinted genes on 15q11-q13, including melanoma antigen gene family member L2 (MAGEL2). The clinical phenotypes of PWS suggest impaired hypothalamic neuroendocrine function; however, the exact cellular defects are unknown. Here, we report deficits in secretory granule (SG) abundance and bioactive neuropeptide production upon loss of MAGEL2 in humans and mice. Unbiased proteomic analysis of Magel2pΔ/m+ mice revealed a reduction in components of SG in the hypothalamus that was confirmed in 2 PWS patient-derived neuronal cell models. Mechanistically, we show that proper endosomal trafficking by the MAGEL2-regulated WASH complex is required to prevent aberrant lysosomal degradation of SG proteins and reduction of mature SG abundance. Importantly, loss of MAGEL2 in mice, NGN2-induced neurons, and human patients led to reduced neuropeptide production. Thus, MAGEL2 plays an important role in hypothalamic neuroendocrine function, and cellular defects in this pathway may contribute to PWS disease etiology. Moreover, these findings suggest unanticipated approaches for therapeutic intervention.


Asunto(s)
Antígenos de Neoplasias/fisiología , Hipotálamo/patología , Neuronas/patología , Neuropéptidos/metabolismo , Síndrome de Prader-Willi/fisiopatología , Proteínas/metabolismo , Proteínas/fisiología , Vesículas Secretoras/patología , Animales , Femenino , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Fenotipo , Transporte de Proteínas , Proteínas/genética , Proteoma/análisis , Proteoma/metabolismo , Vesículas Secretoras/metabolismo
18.
Cells ; 9(8)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32781650

RESUMEN

TROP1 (EpCAM) and TROP2 are homologous cell surface proteins that are widely expressed, and often co-expressed, in developing and adult epithelia. Various functions have been ascribed to EpCAM and TROP2, but responsible mechanisms are incompletely characterized and functional equivalence has not been examined. Adult intestinal epithelial cells (IEC) express high levels of EpCAM, while TROP2 is not expressed. EpCAM deficiency causes congenital tufting enteropathy (CTE) in humans and a corresponding lethal condition in mice. We expressed TROP2 and EpCAM in the IEC of EpCAM-deficient mice utilizing a villin promoter to assess EpCAM and TROP2 function. Expression of EpCAM or TROP2 in the IEC of EpCAM knockout mice prevented CTE. TROP2 rescue (T2R) mice were smaller than controls, while EpCAM rescue (EpR) mice were not. Abnormalities were observed in the diameters and histology of T2R small intestine, and Paneth and stem cell markers were decreased. T2R mice also exhibited enlarged mesenteric lymph nodes, enhanced permeability to 4 kDa FITC-dextran and increased sensitivity to detergent-induced colitis, consistent with compromised barrier function. Studies of IEC organoids and spheroids revealed that stem cell function was also compromised in T2R mice. We conclude that EpCAM and TROP2 exhibit functional redundancy, but they are not equivalent.


Asunto(s)
Antígenos de Neoplasias/fisiología , Moléculas de Adhesión Celular/fisiología , Diarrea Infantil/genética , Molécula de Adhesión Celular Epitelial/fisiología , Células Epiteliales/metabolismo , Síndromes de Malabsorción/genética , Animales , Células HEK293 , Humanos , Mucosa Intestinal/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas
19.
Br J Cancer ; 123(7): 1145-1153, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32624579

RESUMEN

BACKGROUND: Wisteria floribunda agglutinin (WFA)+ Mac-2-binding protein (M2BPGi) is a novel serum marker for liver fibrosis. Although an elevated serum level of M2BPGi can predict development of hepatocellular carcinoma (HCC), the effect of M2BPGi on HCC remains unclear. There are no reports about the association of M2BPGi with HCC aggressiveness. We aimed to clarify the significance of M2BPGi in HCC. METHODS: The protein expression of M2BPGi and galectin-3, a ligand of M2BP, and the mRNA expression of M2BP were evaluated in surgically resected human HCC samples. M2BPGi-regulating signals in HCC cells were investigated using transcriptome analysis. The effects of M2BPGi on HCC properties and galectin-3/mTOR signaling were evaluated. RESULTS: M2BPGi and galectin-3 proteins co-localised in HCC cells, while M2BP mRNA was detected in cirrhotic liver stromal cells. mTOR signaling was upregulated in M2BPGi-treated HCC cells. Moreover, M2BPGi treatment induced tumour-promoting effects on HCC in vitro by activated mTOR signaling. In addition, M2BPGi bound to galectin-3 to induce membranous galectin-3 expression in HCC cells. In vivo, M2BPGi enhanced the growth of xenografted HCC. CONCLUSIONS: M2BPGi is produced in stromal cells of the cirrhotic liver. Furthermore, M2BPGi enhances the progression of HCC through the galectin-3/mTOR pathway.


Asunto(s)
Antígenos de Neoplasias/fisiología , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Glicoproteínas de Membrana/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Antígenos de Neoplasias/análisis , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Galectina 3/análisis , Galectina 3/fisiología , Humanos , Glicoproteínas de Membrana/análisis , Ratones , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...