Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Mol Cell ; 82(1): 159-176.e12, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34847357

RESUMEN

The MYCN oncoprotein drives the development of numerous neuroendocrine and pediatric tumors. Here we show that MYCN interacts with the nuclear RNA exosome, a 3'-5' exoribonuclease complex, and recruits the exosome to its target genes. In the absence of the exosome, MYCN-directed elongation by RNA polymerase II (RNAPII) is slow and non-productive on a large group of cell-cycle-regulated genes. During the S phase of MYCN-driven tumor cells, the exosome is required to prevent the accumulation of stalled replication forks and of double-strand breaks close to the transcription start sites. Upon depletion of the exosome, activation of ATM causes recruitment of BRCA1, which stabilizes nuclear mRNA decapping complexes, leading to MYCN-dependent transcription termination. Disruption of mRNA decapping in turn activates ATR, indicating transcription-replication conflicts. We propose that exosome recruitment by MYCN maintains productive transcription elongation during S phase and prevents transcription-replication conflicts to maintain the rapid proliferation of neuroendocrine tumor cells.


Asunto(s)
Núcleo Celular/enzimología , Proliferación Celular , Replicación del ADN , Exosomas/enzimología , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/enzimología , ARN Polimerasa II/metabolismo , Transcripción Genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Núcleo Celular/genética , Roturas del ADN de Doble Cadena , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Exosomas/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Proteína Proto-Oncogénica N-Myc/genética , Células 3T3 NIH , Neuroblastoma/genética , Neuroblastoma/patología , Regiones Promotoras Genéticas , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , ARN Polimerasa II/genética , Terminación de la Transcripción Genética
2.
Inflammation ; 44(6): 2531-2542, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34637033

RESUMEN

Salivary exosomes contain various components and may play important roles in oral diseases. The purpose of this study was to verify the possible function of miR-223-3p from salivary exosomes in periodontitis. We isolated the salivary exosomes and found that the miR-223-3p content of salivary exosomes from periodontitis was less than the healthy control. Furthermore, we performed dual-luciferase reporter assay and real-time PCR to verify that (NOD)-like receptor (NLR) pyrin domain-containing 3 (NLRP3) was the target of miR-223-3p. When we knocked down the miR-223-3p expression in THP-1-derived macrophages, the expression of NLRP3 and the downstream inflammatory mediators interleukin-1ß (IL-1ß) and IL-6 were upregulated. By using integrated bioinformatics analysis, we found that pyroptosis and cytokine secretion participated in inflammatory gingival tissues. In addition, NLRP3, and the pyroptosis executioner, gasdermin D (GSDMD) was highly active in inflammatory gingival tissues compared with healthy controls by western blotting and immunohistochemistry. In summary, we speculated that miR-223-3p in salivary exosomes might regulate GSDMD-mediated pyroptosis by targeting NLRP3 in periodontitis. Detection of miR-223-3p expression in salivary exosomes could be used as an important non-invasive method to diagnose and evaluate the severity of periodontitis.


Asunto(s)
Caspasa 1/metabolismo , Exosomas/enzimología , Inflamasomas/metabolismo , Macrófagos/enzimología , MicroARNs/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Periodontitis/enzimología , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Piroptosis , Saliva/metabolismo , Estudios de Casos y Controles , Caspasa 1/genética , Exosomas/genética , Exosomas/patología , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/patología , MicroARNs/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Periodontitis/genética , Periodontitis/patología , Transducción de Señal , Células THP-1
3.
Elife ; 102021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34590578

RESUMEN

Astrocytes are essential cells of the central nervous system, characterized by dynamic relationships with neurons that range from functional metabolic interactions and regulation of neuronal firing activities, to the release of neurotrophic and neuroprotective factors. In Parkinson's disease (PD), dopaminergic neurons are progressively lost during the course of the disease, but the effects of PD on astrocytes and astrocyte-to-neuron communication remain largely unknown. This study focuses on the effects of the PD-related mutation LRRK2 G2019S in astrocytes generated from patient-derived induced pluripotent stem cells. We report the alteration of extracellular vesicle (EV) biogenesis in astrocytes and identify the abnormal accumulation of key PD-related proteins within multivesicular bodies (MVBs). We found that dopaminergic neurons internalize astrocyte-secreted EVs and that LRRK2 G2019S EVs are abnormally enriched in neurites and fail to provide full neurotrophic support to dopaminergic neurons. Thus, dysfunctional astrocyte-to-neuron communication via altered EV biological properties may participate in the progression of PD.


Asunto(s)
Astrocitos/enzimología , Comunicación Celular , Neuronas Dopaminérgicas/enzimología , Exosomas/enzimología , Células Madre Pluripotentes Inducidas/enzimología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Células-Madre Neurales/enzimología , Enfermedad de Parkinson/enzimología , Animales , Astrocitos/ultraestructura , Atrofia , Estudios de Casos y Controles , Línea Celular , Neuronas Dopaminérgicas/patología , Endocitosis , Exosomas/genética , Exosomas/ultraestructura , Humanos , Células Madre Pluripotentes Inducidas/ultraestructura , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Células-Madre Neurales/ultraestructura , Biogénesis de Organelos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología
4.
Appl Immunohistochem Mol Morphol ; 29(10): 720-727, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34433181

RESUMEN

The human aspartyl ß-hydroxylase (ASPH) is overexpressed in tumor tissues. Bronchoalveolar lavage (BAL) is a diagnostic procedure for infections and malignancies. The aim of this study was to investigate whether tumor exosomes carrying ASPH gene marker were present in bronchoalveolar fluid of patients with non-small cell lung cancer (NSCLC). A tissue microarray analysis was applied to explore the expression of ASPH in different histologic NSCLC. The human NSCLC cell lines and normal bronchial cell lines were used to study exosomal ASPH exprerssion. A total of 27 NSCLC, 21 benign tumor, and 15 healthy controls underwent BAL. Immunohistochemistry was performed to study the ASPH expression in malignant and normal lung tissues. The expression characteristics of ASPH in different NSCLC and normal bronchial cells and pneumocytes were confirmed by cell blocks. A reverse transcription-quantitative polymerase chain reaction was carried out to study the levels of exosomal ASPH expression. Immunohistochemical staining of tissue microarray demonstrated that overexpression of ASPH was found in NSCLC tissues including adenocarcinoma, large cell carcinoma, and squamous cell carcinoma, but absent in adjacent normal tissues. All NSCLC specimens exhibited high levels of ASPH immunoreactivity, while nonmalignant and normal lung tissues exhibited a very low level of expression. Overexpression of ASPH was found in exosomes from NSCLC cell lines but absent from the normal bronchial cell line NL-20. ASPH level from BAL exosomes was significantly increased in NSCLC patients compared with that from nonmalignant or health group. Our method of isolation of BAL exosomes was easily performed in the clinical laboratory. BAL exosomal ASPH can be a potential biomarker for NSCLC diagnosis.


Asunto(s)
Lavado Broncoalveolar , Proteínas de Unión al Calcio/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas , Exosomas/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , Proteínas de la Membrana/biosíntesis , Oxigenasas de Función Mixta/biosíntesis , Proteínas Musculares/biosíntesis , Proteínas de Neoplasias/biosíntesis , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología
5.
J Immunol Res ; 2021: 8243378, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34327245

RESUMEN

The extracellular matrix (ECM) is a highly dynamic and complex network structure, which exists in almost all tissues and is the microenvironment that cells rely on for survival. ECM interacts with cells to regulate diverse functions, including differentiation, proliferation, and migration. Neutrophils are the most abundant immune cells in circulation and play key roles in orchestrating a complex series of events during inflammation. Neutrophils can also mediate ECM remodeling by providing specific matrix-remodeling enzymes (such as neutrophil elastase and metalloproteinases), generating neutrophil extracellular traps, and releasing exosomes. In turn, ECM can remodel the inflammatory microenvironment by regulating the function of neutrophils, which drives disease progression. Both the presence of ECM and the interplay between neutrophils and their extracellular matrices are considered an important and outstanding mechanistic aspect of inflammation. In this review, the importance of ECM will be considered, together with the discussion of recent advances in understanding the underlying mechanisms of the intricate interplay between ECM and neutrophils. A better comprehension of immune cell-matrix reciprocal dependence has exciting implications for the development of new therapeutic options for neutrophil-associated infectious and inflammatory diseases.


Asunto(s)
Matriz Extracelular/inmunología , Infecciones/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Exosomas/enzimología , Exosomas/inmunología , Matriz Extracelular/enzimología , Trampas Extracelulares/enzimología , Trampas Extracelulares/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Infecciones/microbiología , Elastasa de Leucocito/metabolismo , Metaloproteasas/metabolismo , Neutrófilos/enzimología
6.
Asian Pac J Cancer Prev ; 22(3): 861-869, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33773551

RESUMEN

OBJECTIVE: The aim of the study was to develop a model for predicting cancer risk in colorectal polyps' patients (CPPs), as well as to reveal additional prognosis factors for Stage III colorectal cancer based on differences in subpopulations of tetraspanins, tetraspanin-associated and tetraspanin-non-associated proteases in blood plasma exosomes of CPPs and colorectal cancer patients (CRCPs). METHODS: The subpopulations of CD151- and Tspan8-positive exosomes, the subpopulations of metalloproteinase at the surface of СD9-positive exosomes and the level of 20S proteasomes in plasma exosomes in 15 CPPs (tubulovillous adenomas) and 60 CRCPs were evaluated using flow cytometry and Western blotting. Logistic regression analysis was performed to predict cancer risk of CPPs. RESULTS: The levels of 20S proteasomes in exosomes, MMP9+, MMP9+/MMP2+/EMMPRIN+ in CD9-positive blood plasma exosomes are associated with the risk of malignant transformation of colorectal tubulovillous adenomas.  In patients with Stage III CRC, the levels of 20S proteasomes (less than 2 units) and MMP9+ subpopulations (more than 61%) in plasma exosomes are unfavorable prognostic factors for overall survival. The levels of 20S proteasomes and ADAM10+/ADAM17- subpopulations in CD9-positive blood plasma exosomes are the most significant values for predicting relapse-free survival. CONCLUSION: Protease cargo in CD9-positive blood plasma exosomes is prognostic biomarker for colorectal polyps and colorectal cancer.


Asunto(s)
Adenoma/enzimología , Carcinoma/enzimología , Pólipos del Colon/enzimología , Neoplasias Colorrectales/enzimología , Exosomas/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Adenoma/metabolismo , Adenoma/patología , Adenoma Velloso/enzimología , Adenoma Velloso/metabolismo , Adenoma Velloso/patología , Basigina/metabolismo , Carcinoma/metabolismo , Carcinoma/patología , Pólipos del Colon/metabolismo , Pólipos del Colon/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Exosomas/metabolismo , Femenino , Humanos , Pólipos Intestinales/enzimología , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Péptido Hidrolasas/metabolismo , Pronóstico , Tetraspanina 24/metabolismo , Tetraspaninas/metabolismo
7.
J Biol Chem ; 296: 100569, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33753167

RESUMEN

The apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1), the main AP-endonuclease of the DNA base excision repair pathway, is a key molecule of interest to researchers due to its unsuspected roles in different nonrepair activities, such as: i) adaptive cell response to genotoxic stress, ii) regulation of gene expression, and iii) processing of microRNAs, which make it an excellent drug target for cancer treatment. We and others recently demonstrated that APE1 can be secreted in the extracellular environment and that serum APE1 may represent a novel prognostic biomarker in hepatocellular and non-small-cell lung cancers. However, the mechanism by which APE1 is released extracellularly was not described before. Here, using three different approaches for exosomes isolation: commercial kit, nickel-based isolation, and ultracentrifugation methods and various mammalian cell lines, we elucidated the mechanisms responsible for APE1 secretion. We demonstrated that APE1 p37 and p33 forms are actively secreted through extracellular vesicles (EVs), including exosomes from different mammalian cell lines. We then observed that APE1 p33 form is generated by proteasomal-mediated degradation and is enzymatically active in EVs. Finally, we revealed that the p33 form of APE1 accumulates in EVs upon genotoxic treatment by cisplatin and doxorubicin, compounds commonly found in chemotherapy pharmacological treatments. Taken together, these findings provide for the first time evidence that a functional Base Excision Repair protein is delivered through exosomes in response to genotoxic stresses, shedding new light into the complex noncanonical biological functions of APE1 and opening new intriguing perspectives on its role in cancer biology.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Exosomas/enzimología , Animales , Línea Celular , Reparación del ADN , Humanos
8.
Cancer Lett ; 500: 98-106, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33307156

RESUMEN

Esophageal carcinoma stem cells (ECSCs) are responsible for the initiation and therapy-resistance of esophageal cancer. Nutrient sensor O-GlcNAc transferase (OGT) promoted the growth and metastasis of cancer cells. However, the contributions of OGT to the tumorigenesis of ECSCs remain largely uncover. In the present study, as compared to matched non-stem cancer cells, the expression of OGT was higher in ALDH+ ECSCs. Knock down of OGT by lentivirus system reduced the self-renewal capacities and tumorigenicity of ALDH+ ECSCs. In addition, OGT in exosome derived from ALDH+ ECSCs was taken up by neighboring CD8+ T cells and increased the expression of PD-1 in CD8+ T cells. Down-regulation of OGT increased the apoptosis of ALDH+ ECSCs induced by CD8+ T cells, which could be blocked by overexpression of PD-1 in CD8+ T cells. Together, OGT in exosome from ECSCs protects ECSCs from CD8+ T cells through up-regulation of PD-1.


Asunto(s)
Aldehído Deshidrogenasa/genética , Carcinoma/genética , Neoplasias Esofágicas/genética , N-Acetilglucosaminiltransferasas/genética , Receptor de Muerte Celular Programada 1/genética , Apoptosis/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Carcinogénesis/genética , Carcinoma/patología , Línea Celular Tumoral , Neoplasias Esofágicas/patología , Exosomas/enzimología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Terapia de Inmunosupresión , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
9.
Curr Mol Med ; 21(6): 476-483, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33222670

RESUMEN

Tumor-derived exosomes contain biological contents such as proteins, lipids, RNA (miRNAs, mRNAs, lncRNA), and DNA for intracellular communication. Meanwhile, studies have shown the role of exosomes in cancer progression, metastasis, and therapeutic resistance. Furthermore, tumor exosomes have received growing attention due to their potential as novel therapeutic protocols for the treatment of cancers. Adenosine nucleoside, which is a derivative of ATP, is highly elevated in the tumor microenvironment by CD39 and CD73 enzymatic activity. Recently, it is distinguished that cancer cell-derived exosomes carry CD39 and CD73 on their surface and may contribute to rising adenosine levels in the tumor microenvironment. In this review, we summarize the evidence of CD39/CD73-bearing exosomes and their role in cancer development, progression, invasion, angiogenesis, metastasis and their application in the selection of the appropriate strategy to treat different types of cancer.


Asunto(s)
5'-Nucleotidasa , Reducción Gradual de Medicamentos , Exosomas/enzimología , Proteínas de Neoplasias , Neoplasias , Neovascularización Patológica , 5'-Nucleotidasa/antagonistas & inhibidores , 5'-Nucleotidasa/metabolismo , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/enzimología , Transducción de Señal , Microambiente Tumoral
10.
Oncology ; 98(10): 734-742, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32726790

RESUMEN

BACKGROUND: There has been no clinically useful diagnostic or prognostic biomarker for renal cell carcinoma (RCC). Serum γ-glutamyltransferase (GGT) activity has been reported to be a prognostic marker for several types of cancer including RCC. Exosomes or small extracellular vesicles present in body fluids have potential as a biomarker. We have recently demonstrated that GGT activity on exosomes isolated from serum is useful for the differential diagnosis of prostate cancer and benign prostate hyperplasia. In this study, we aimed to examine if serum exosomal GGT activity could be a marker for RCC. METHODS: We examined GGT1 expression and GGT activity in cell lysates and exosomes from culture medium of HK-2 proximal tubule epithelial and RCC cell lines. GGT activity was measured using a fluorescent probe for GGT, γ-glutamyl hydroxymethyl rhodamine green. Serum and serum exosomal GGT activities were measured in patients with RCC. GGT1 expression in RCC tissues was evaluated by immunohistochemical staining. RESULTS: GGT1 levels in exosomes from KMRC-1, OS-RC-2 and 786-O cells were elevated compared with those from HK-2 cells. In exosomes, GGT1 expression correlated with GGT activity determined using a fluorescent probe for GGT. In RCC patients, serum exosomal GGT activity was elevated in those with advanced stages (III/IV vs. I/II, p = 0.037) and those with microvascular invasion (with vs. without, p = 0.034). Immunohistochemical analysis showed that membranous GGT1 expression was increased in RCC with microvascular invasion. Notably, preoperative serum exosomal GGT activity could predict the likelihood of having microvascular invasion diagnosed by pathological examination of surgically resected specimens. CONCLUSIONS: Our results suggest that serum exosomal GGT activity could be a clinically useful marker for advanced clinicopathological features of RCC patients, and its combined use with conventional diagnostic modalities may improve the diagnosis and treatment of patients.


Asunto(s)
Carcinoma de Células Renales/enzimología , Exosomas/enzimología , Neoplasias Renales/enzimología , gamma-Glutamiltransferasa/sangre , Anciano , Carcinoma de Células Renales/sangre , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Neoplasias Renales/sangre , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , gamma-Glutamiltransferasa/biosíntesis
11.
J Nanobiotechnology ; 18(1): 68, 2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375794

RESUMEN

BACKGROUND: Enhancing angiogenesis is critical for accelerating wound healing. Application of different types of exosomes (Exos) to promote angiogenesis represents a novel strategy for enhanced wound repair. Saliva is known to accelerate wound healing, but the underlying mechanisms remain unclear. RESULTS: Our results have demonstrated that saliva-derived exosomes (saliva-Exos) induce human umbilical vein endothelial cells (HUVEC) proliferation, migration, and angiogenesis in vitro, and promote cutaneous wound healing in vivo. Further experiments documented that Ubiquitin-conjugating enzyme E2O (UBE2O) is one of the main mRNAs of saliva-Exos, and activation of UBE2O has effects similar to those of saliva-Exos, both in vitro and in vivo. Mechanistically, UBE2O decreases the level of SMAD family member 6 (SMAD6), thereby activating bone morphogenetic protein 2 (BMP2), which, in turn, induces angiogenesis. CONCLUSIONS: The present work suggests that administration of saliva-Exos and UBE2O represents a promising strategy for enhancing wound healing through promotion of angiogenesis.


Asunto(s)
Exosomas/enzimología , Neovascularización Fisiológica/efectos de los fármacos , Saliva/enzimología , Proteína smad6/metabolismo , Enzimas Ubiquitina-Conjugadoras , Animales , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero/farmacología , Saliva/citología , Piel/lesiones , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enzimas Ubiquitina-Conjugadoras/farmacología , Cicatrización de Heridas/efectos de los fármacos
12.
Mol Cell Biochem ; 469(1-2): 77-87, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32297178

RESUMEN

Lung cancer has been recognized as the leading cause of cancer-related death worldwide. Despite the improvements of treatment, the distant metastasis and recurrence of lung cancer caused by therapy resistance is the biggest challenge in clinical management. Extracellular vesicles named exosomes play crucial roles in intercellular communication as signaling mediators and are involved in tumor development. In this study, we isolated exosomes from irradiated lung cancer cells and co-cultured the exosomes with other lung cancer cells. It was found that cellular growth and motility of recipient cells were facilitated. High-throughput LC-MS/MS assay of exosomal proteins and Gene Ontology enrichment analyses indicated that the metabolic enzymes ALDOA and ALDH3A1 had potential contribution in exosome-enhanced motility of recipient cells, and clinical survival analysis demonstrated the close correlations between ALDOA or ALDH3A1 expression and poor prognosis of lung cancer patients. After co-culturing with exosomes derived from irradiated cancer cells, the expressions of these metabolic enzymes were elevated and the glycolytic activity was promoted in recipient cancer cells. In conclusion, our data suggested that exosomes from irradiated lung cancer cells regulated the motility of recipient cells by accelerating glycolytic process, where exosomal ALDOA and ALDH3A1 proteins were important signaling factors.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Movimiento Celular/genética , Proliferación Celular/genética , Exosomas/metabolismo , Fructosa-Bifosfato Aldolasa/metabolismo , Glucólisis , Neoplasias Pulmonares/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Aldehído Deshidrogenasa/genética , Comunicación Celular/genética , Línea Celular Tumoral , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Cromatografía Liquida , Técnicas de Cocultivo , Exosomas/enzimología , Exosomas/efectos de la radiación , Exosomas/ultraestructura , Fructosa-Bifosfato Aldolasa/genética , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Microscopía Electrónica de Transmisión , Recurrencia Local de Neoplasia/enzimología , Recurrencia Local de Neoplasia/genética , Pronóstico , Proteómica , Radiación , Espectrometría de Masas en Tándem
13.
Stem Cells Transl Med ; 9(4): 499-517, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31943851

RESUMEN

Mesenchymal stem cells (MSCs) have been used in clinical studies to treat neurological diseases and damage. However, implanted MSCs do not achieve their regenerative effects by differentiating into and replacing neural cells. Instead, MSC secretome components mediate the regenerative effects of MSCs. MSC-derived extracellular vesicles (EVs)/exosomes carry cargo responsible for rescuing brain damage. We previously showed that EP4 antagonist-induced MSC EVs/exosomes have enhanced regenerative potential to rescue hippocampal damage, compared with EVs/exosomes from untreated MSCs. Here we show that EP4 antagonist-induced MSC EVs/exosomes promote neurosphere formation in vitro and increase neurogenesis and neuritogenesis in damaged hippocampi; basal MSC EVs/exosomes do not contribute to these regenerative effects. 2',3'-Cyclic nucleotide 3'-phosphodiesterase (CNP) levels in EP4 antagonist-induced MSC EVs/exosomes are 20-fold higher than CNP levels in basal MSC EVs/exosomes. Decreasing elevated exosomal CNP levels in EP4 antagonist-induced MSC EVs/exosomes reduced the efficacy of these EVs/exosomes in promoting ß3-tubulin polymerization and in converting toxic 2',3'-cAMP into neuroprotective adenosine. CNP-depleted EP4 antagonist-induced MSC EVs/exosomes lost the ability to promote neurogenesis and neuritogenesis in damaged hippocampi. Systemic administration of EV/exosomes from EP4 -antagonist derived MSC EVs/exosomes repaired cognition, learning, and memory deficiencies in mice caused by hippocampal damage. In contrast, CNP-depleted EP4 antagonist-induced MSC EVs/exosomes failed to repair this damage. Exosomal CNP contributes to the ability of EP4 antagonist-elicited MSC EVs/exosomes to promote neurogenesis and neuritogenesis in damaged hippocampi and recovery of cognition, memory, and learning. This experimental approach should be generally applicable to identifying the role of EV/exosomal components in eliciting a variety of biological responses.


Asunto(s)
2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Lesiones Encefálicas/terapia , Región CA1 Hipocampal/metabolismo , Cognición , Exosomas/enzimología , Aprendizaje , Células Madre Mesenquimatosas/enzimología , Neuritas/metabolismo , Neurogénesis , Animales , Lesiones Encefálicas/patología , Cognición/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas de Dominio Doblecortina , Exosomas/efectos de los fármacos , Humanos , Isoindoles/farmacología , Aprendizaje/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuritas/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuropéptidos/metabolismo , Polimerizacion , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Sulfonamidas/farmacología , Tubulina (Proteína)/metabolismo
14.
Clin Chim Acta ; 503: 203-209, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31794764

RESUMEN

BACKGROUND: Lactate dehydrogenase C4 (LDH-C4) as a cancer/testis antigen (CTA) is abnormally expressed in some malignant tumors. However, the expression and clinical significance of LDH-C4 in breast cancer (BC) has not been characterized. METHODS: We determined LDHC mRNA expression in serum and serum-derived exosomes of BC patients by quantitative RT-PCR. We also evaluated the protein expression of LDH-C4 in BC tissues using high-throughput tissue microarray analysis and immunohistochemistry. RESULTS: Our results showed high mRNA expression level of LDHC in serum and serum-derived exosomes of BC patients. The LDHC level in serum and exosomes could distinguish BC cases from healthy individuals based on their AUCs of 0.9587 and 0.9464, respectively. Besides, the LDHC level in exosomes of BC patients associated with tumor size, and positively correlated with HER2 and Ki-67 expressions (all with P < 0.05). Serum and exosomal level of LDHC negatively correlated with medical treatment and positively with the recurrence of BC. Survival analysis showed that LDH-C4 expression negatively correlated with BC prognosis. CONCLUSION: Serum and exosomal LDHC may be an effective indicator for the diagnosis, efficacy evaluation, and monitoring the recurrence of BC. LDH-C4 may act as a biomarker that predicts BC prognosis.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias de la Mama/enzimología , L-Lactato Deshidrogenasa/sangre , Adulto , Área Bajo la Curva , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/mortalidad , Estudios de Casos y Controles , Exosomas/enzimología , Femenino , Humanos , Isoenzimas/análisis , Isoenzimas/sangre , Isoenzimas/genética , L-Lactato Deshidrogenasa/análisis , L-Lactato Deshidrogenasa/genética , Masculino , Persona de Mediana Edad , Pronóstico , ARN Mensajero/sangre , Recurrencia , Análisis de Supervivencia , Resultado del Tratamiento
15.
Matrix Biol ; 88: 53-68, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31812535

RESUMEN

Heparanase is known to enhance the progression of many cancer types and is associated with poor patient prognosis. We recently reported that after patients with multiple myeloma were treated with high dose chemotherapy, the tumor cells that emerged upon relapse expressed a much higher level of heparanase than was present prior to therapy. Because tumor cells having stemness properties are thought to seed tumor relapse, we investigated whether heparanase had a role in promoting myeloma stemness. When plated at low density and grown in serum-free conditions that support survival and expansion of stem-like cells, myeloma cells expressing a low level of heparanase formed tumor spheroids poorly. In contrast, cells expressing a high level of heparanase formed significantly more and larger spheroids than did the heparanase low cells. Importantly, heparanase-low expressing cells exhibited plasticity and were induced to exhibit stemness properties when exposed to recombinant heparanase or to exosomes that contained a high level of heparanase cargo. The spheroid-forming heparanase-high cells had elevated expression of GLI1, SOX2 and ALDH1A1, three genes known to be associated with myeloma stemness. Inhibitors that block the heparan sulfate degrading activity of heparanase significantly diminished spheroid formation and expression of stemness genes implying a direct role of the enzyme in regulating stemness. Blocking the NF-κB pathway inhibited spheroid formation and expression of stemness genes demonstrating a role for NF-κB in heparanase-mediated stemness. Myeloma cells made deficient in heparanase exhibited decreased stemness properties in vitro and when injected into mice they formed tumors poorly compared to the robust tumorigenic capacity of cells expressing higher levels of heparanase. These studies reveal for the first time a role for heparanase in promoting cancer stemness and provide new insight into its function in driving tumor progression and its association with poor prognosis in cancer patients.


Asunto(s)
Regulación hacia Abajo , Glucuronidasa/genética , Mieloma Múltiple/patología , Células Madre Neoplásicas/patología , Familia de Aldehído Deshidrogenasa 1/genética , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Exosomas/enzimología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Mieloma Múltiple/genética , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Retinal-Deshidrogenasa/genética , Factores de Transcripción SOXB1/genética , Esferoides Celulares/citología , Proteína con Dedos de Zinc GLI1/genética
16.
Cancer Metastasis Rev ; 38(3): 455-468, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31773432

RESUMEN

Rapidly increasing scientific reports of exosomes and their biological effects have improved our understanding of their cellular sources and their cell-to-cell communication. These nano-sized vesicles act as potent carriers of regulatory bio-macromolecules and can induce regulatory functions by delivering them from its source to recipient cells. The details of their communication network are less understood. Recent studies have shown that apart from delivering its cargo to the cells, it can directly act on extracellular matrix (ECM) proteins and growth factors and can induce various remodeling events. More importantly, exosomes carry many surface-bound proteases, which can cleave different ECM proteins and carbohydrates and can shed cell surface receptors. These local extracellular events can modulate signaling cascades, which consequently influences the whole tissue and organ. This review aims to highlight the critical roles of exosomal proteases and their mechanistic insights within the cellular and extracellular environment.


Asunto(s)
Exosomas/enzimología , Neoplasias/enzimología , Neoplasias/patología , Péptido Hidrolasas/metabolismo , Animales , Comunicación Celular/fisiología , Progresión de la Enfermedad , Matriz Extracelular/enzimología , Humanos
17.
Int J Biol Sci ; 15(11): 2320-2329, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31595150

RESUMEN

Solid tumors consist of various types of stromal cells in addition to cancer cells. Cancer-associated fibroblasts (CAFs) are a major component of the tumor stroma and play an essential role in tumor progression and metastasis in a variety of malignancies, including gastric cancer. However, the effects of CAFs on gastric cancer cells' progression and metastasis are not well studied. Here we show that matrix metalloproteinase 11 (MMP11) in exosomes secreted from CAFs can be delivered into gastric cancer cells. Gastric CAFs promote gastric cancer cell migration partially through exosomal MMP11. Moreover, MMP11 is overexpressed in exosomes purified from plasma of gastric cancer patients and tumor tissues and associated with overall survival of gastric patients. We also find that MMP11 is negatively regulated by exosomal miR-139 in the CAFs of gastric cancer. Exosomal miR-139 inhibits tumor growth and metastasis of gastric cancer cells by decreasing the expression of MMP11 in vitro and in vivo. Thus, we propose that exosomal miR-139 derived from gastric CAFs could inhibit the progression and metastasis of gastric cancer by decreasing MMP11 in tumor microenvironment.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Exosomas/genética , Metaloproteinasa 11 de la Matriz/genética , MicroARNs/metabolismo , Neoplasias Gástricas/genética , Animales , Fibroblastos Asociados al Cáncer/enzimología , Movimiento Celular/genética , Progresión de la Enfermedad , Exosomas/enzimología , Exosomas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metaloproteinasa 11 de la Matriz/metabolismo , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
18.
Cancer Lett ; 457: 47-59, 2019 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-31059752

RESUMEN

We previously reported that Vps4A acted as a tumor suppressor by influencing the microRNA profiles of exosomes and their parental cells in hepatocellular carcinoma (HCC). However, the underlying mechanism and if Vps4A contributes to sorting proteins into exosomes are not well known. Here, we performed mass spectrometry analysis of the immunoprecipitated Vps4A complex and confirmed that Vps4A was associated with ß-catenin and CHMP4B. Through this interaction, Vps4A promoted the plasma membrane (PM) localization and exosome release of ß-catenin. Silencing Vps4A or CHMP4B decreased the PM localization and exosome sorting of ß-catenin. Vps4A overexpression decreased ß-catenin signaling pathway and inhibited epithelial-mesenchymal transition (EMT) and motility of HCC cells. And, silencing Vps4A or CHMP4B promoted EMT in HCC. Furthermore, the expression of Vps4A was significantly related to that of several EMT markers in HCC tissues and the level of exosomal ß-catenin in patients with metastatic HCC was significantly lower compared to that of control patients. In conclusion, through the interaction with CHMP4B and ß-catenin, Vps4A regulates the PM localization and exosome sorting of ß-catenin, consequently decreases ß-catenin signaling, and thereby inhibits EMT and metastasis in HCC.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Carcinoma Hepatocelular/enzimología , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Transición Epitelial-Mesenquimal , Exosomas/enzimología , Neoplasias Hepáticas/enzimología , ATPasas de Translocación de Protón Vacuolares/metabolismo , beta Catenina/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/secundario , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Exosomas/genética , Exosomas/patología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Transporte de Proteínas , Transducción de Señal , ATPasas de Translocación de Protón Vacuolares/genética , beta Catenina/genética
19.
Clin Exp Nephrol ; 23(8): 1013-1021, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30955187

RESUMEN

BACKGROUND: Previously we found that kidney tissue and urinary exosomes from patients of diabetic kidney disease showed high levels of ceruloplasmin (CP). Because CP is an acute-phase protein of kidney origin, it could be an early marker of many other kidney diseases. To investigate this hypothesis, we first measured urine exosomal and kidney expression of CP in non-diabetic chronic kidney disease (CKD) patients (membranous nephropathy, focal segmental glomerulosclerosis, lupus nephritis and IgA nephropathy) followed by a longitudinal study in rat passive Heymann nephritis (PHN), a model of human membranous nephropathy. METHODS: Urinary exosomes were isolated from urine of patients (and rats) by differential centrifugation. The exosomal extracts were used for measuring CP using ELISA. Kidney expression of CP was evaluated by immune-staining biopsy tissues. Similar techniques were applied in rat PHN model (produced by injection of anti-gp600 antiserum) to analyze urine exosomal and kidney CP. RESULTS: Urine exosomal CP levels were 10-20 times higher in CKD patients than in controls; consistent with this we found high immune-reactive CP localized in tubules and collecting ducts of biopsies of CKD patients. In the PHN model urinary exosomal CP level was significantly higher prior to the onset of proteinuria. Early rise of urine exosomal CP, which preceded proteinuria, correlated with high immunoreactive CP found in rat kidneys at this time. CONCLUSION: We propose that urine exosomal CP, observed to increase prior to proteinuria, makes it a potential urinary biomarker to diagnose early kidney disease.


Asunto(s)
Ceruloplasmina/orina , Exosomas/enzimología , Glomerulonefritis Membranosa/orina , Riñón/enzimología , Proteinuria/orina , Insuficiencia Renal Crónica/orina , Adulto , Animales , Biomarcadores/orina , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Diagnóstico Precoz , Exosomas/patología , Femenino , Glomerulonefritis Membranosa/diagnóstico , Glomerulonefritis Membranosa/enzimología , Humanos , Riñón/patología , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Proteinuria/diagnóstico , Proteinuria/enzimología , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/enzimología , Regulación hacia Arriba
20.
Sci Rep ; 9(1): 3265, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30824759

RESUMEN

c-Src is a membrane-associated tyrosine kinase that has key roles in the signaling transduction that controls cell growth, adhesion, and migration. In the early stage of carcinogenesis, c-Src is activated under the plasma membrane and transduces oncogenic signals. Here we show that c-Src localized to the endosomal membrane has unique functions in c-Src-transformed cells. Our results indicate that activated c-Src in the endosomal membrane promoted the secretion of exosomes, in which c-Src was encapsulated. In addition, the ESCRT-interacting molecule, Alix was identified as a c-Src-interacting protein in exosomes. We revealed that the interaction between the SH3 domain of c-Src and the proline-rich region of Alix activates ESCRT-mediated intra-luminal vesicle (ILV) formation, resulting in the upregulation of exosome secretion in c-Src-transformed cells. We observed also a correlation between malignant phenotypes and Alix-dependent aberrant exosome secretion in Src-upregulated cancer cells. Collectively, our findings provide a unique mechanism for the upregulation of exosomes in cancer cells, as well as new insights into the significance of exosome secretion in cancer progression.


Asunto(s)
Proteína Tirosina Quinasa CSK/metabolismo , Exosomas/enzimología , Membranas Intracelulares/enzimología , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Proteína Tirosina Quinasa CSK/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Exosomas/genética , Exosomas/patología , Células HCT116 , Células HT29 , Humanos , Membranas Intracelulares/patología , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA