Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncol Lett ; 27(5): 203, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38516684

RESUMEN

The 5-year survival rate of patients with advanced non-small cell lung cancer (NSCLC) remains low, despite recent advances in targeted therapy and immunotherapy. Therefore, there is a need to identify alternative strategies to improve treatment outcomes. Modern diagnostics can significantly facilitate the selection of treatment plans to improve patient outcomes. In the present study, multi-form diagnostic methodologies were adopted, including next-generation sequencing-based actionable gene sequencing, programmed death ligand 1 (PD-L1) immunohistochemistry, a circulating tumor cell (CTC) assay, flow cytometric analysis of lymphocyte subsets and computed tomography, to improve disease management in an 86-year-old female patient with relapsed metastatic NSCLC. High expression of PD-L1, elevated CTC tmutations, were observed. Based on these results, the patient was initially treated with the programmed death protein 1 blocking antibody sintilimab for two cycles, resulting in the stabilization of their condition, although the patient still exhibited severe pain and other symptoms, including fatigue, malaise, a loss of appetite and poor mental state. Informed by dynamic monitoring of the patient's response to treatment, the treatment plan was subsequently adjusted to a combination therapy with sintilimab and autologous cytokine-induced killer cell infusion, which eventually led to improved outcomes in both the management of the cancer and quality of life. In conclusion, multi-omics analysis may be used to establish patient-tailored therapies to improve clinical outcomes in hard-to-treat elderly patients with metastatic NSCLC.

2.
Front Oncol ; 14: 1341997, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38313801

RESUMEN

Background: According to GLOBOCAN 2020, lymphoma ranked as the 9th most common cancer and the 12th leading cause of cancer-related deaths worldwide. Traditional diagnostic methods rely on the invasive excisional lymph node biopsy, which is an invasive approach with some limitations. Most lymphoma patients are diagnosed at an advanced stage since they are asymptomatic at the beginning, which has significantly impacted treatment efficacy and prognosis of the disease. Method: This study assessed the performance and utility of a newly developed blood-based assay (SeekInCare) for lymphoma early detection. SeekInCare utilized protein tumor markers and a comprehensive set of cancer-associated genomic features, including copy number aberration (CNA), fragment size (FS), end motif, and lymphoma-related virus, which were profiled by shallow WGS of cfDNA. Results: Protein marker CA125 could be used for lymphoma detection independent of gender, and the sensitivity was 27.8% at specificity of 98.0%. After integrating these multi-dimensional features, 77.8% sensitivity was achieved at specificity of 98.0%, while its NPV and PPV were both more than 92% for lymphoma detection. The sensitivity of early-stage (I-II) lymphoma was up to 51.3% (47.4% and 55.0% for stage I and II respectively). After 2 cycles of treatment, the molecular response of SeekInCare was correlated with the clinical outcome. Conclusion: In summary, a blood-based assay can be an alternative to detect lymphoma with adequate performance. This approach becomes particularly valuable in cases where obtaining tissue biopsy is difficult to obtain or inconclusive.

3.
Sci Rep ; 10(1): 8056, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32415175

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous cells derived from immature myeloid cells (IMCs). MDSCs are known to play important roles in tumor immune evasion. While we know that there are a large number of circulating and tumor-infiltrating MDSCs existing in gastric cancer (GC) patients, the phenotypic characteristics and arginase 1 (ARG1) expression levels of these MDSCs remain very unclear. In our study, flow cytometric analysis of circulating MDSCs from 20 gastric adenocarcinoma (GAC) patients found that ≥80% ARG1-expressing MDSCs were mainly early-stage MDSCs (HLA-DR-CD33+CD14-CD15-MDSCs). In addition, our investigation showed that tumor-infiltrating MDSCs from 6 GAC patients consisted of >35% ARG1-expressing naïve MDSCs (HLA-DR-CD33-CD11b-CD14-CD15-MDSCs), >15% early-stage MDSCs and >40% monocytic MDSCs (HLA-DR-CD14+MDSCs). This preliminary study describes the phenotypic characteristics and ARG1 expression levels of MDSCs from GAC patients and shows that circulating and tumor-infiltrating ARG1-expressing cells were mainly immature and monocytic MDSCs, which provides information to better understand the mechanisms that allow gastric cancer cells to evade the immune system.


Asunto(s)
Adenocarcinoma/etiología , Adenocarcinoma/patología , Arginasa/genética , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias Gástricas/etiología , Neoplasias Gástricas/patología , Adenocarcinoma/metabolismo , Arginasa/metabolismo , Expresión Génica , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Monocitos/inmunología , Monocitos/metabolismo , Células Supresoras de Origen Mieloide/patología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neoplasias Gástricas/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
4.
Int J Oncol ; 56(1): 7-17, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31789408

RESUMEN

Although the majority of patients with follicular lymphoma (FL) harbor the t(14;18)(q32;q21) IGH/BCL2 gene rearrangement that leads to the overexpression of BCL2 protein, approximately 20% of FL cases lack t(14;18)(q32;q21). It is considered that BCL2 overexpression underscores the development of the majority of cases of FL and their transformation to more aggressive lymphoma [known as transformed FL (tFL)]. However, FL cases lacking the t(14;18)(q32;q21) translocation exhibit symptoms analogous to their t(14;18)­positive counterparts. An important goal of recent research on FL has been to clarify the distinctions between the two different forms of FL. Numerous studies have shed light onto the genetic and molecular features of t(14;18)­negative FL and the related clinical manifestations. In this review, we summarize the current knowledge of t(14;18)­negative FL occurring in the lymph nodes with an emphasis on the underlying molecular and clinical features. In addition, novel treatment directions are discussed.


Asunto(s)
Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 18/genética , Linfoma Folicular/genética , Linfoma Folicular/patología , Translocación Genética , Humanos , Pronóstico
5.
Cancer Manag Res ; 11: 4023-4040, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31190980

RESUMEN

  Background: Myeloid-derived suppressor cells (MDSCs) promote immunosuppression in the tumor microenvironment, support tumor growth and survival, and may contribute to immunotherapy resistance. Recent studies showed that tumor-derived exosomes (TDEs) can induce MDSCs accumulation and expansion, the mechanisms of which are largely unknown. Methods: The morphologies and sizes of the exosomes was observed by using a JEM-1400 transmission electron microscope. MicroRNA(miR)-107 and ARG1, DICER1, PTEN, PI3K, AKT, mTOR, and NF-kB mRNAs were quantified by quantitative reverse tanscription PCR. Dual-Luciferase Reports Assay were used to examine the expression of genes which was targeted by miR-107. The expression of proteins were analyzed by using western blot. Results: MiR-107 was not only overexpressed in gastric cancer cells but also enriched in their secreted TDEs. Also, these miR-107 enriched TDEs could be taken up by HLA-DR-CD33+MDSCs, where miR-107 was able to target and suppress expression of DICER1 and PTEN genes. Dampened DICER1 expression supported expansion of MDSCs , while decreased PTEN led to activation of the PI3K pathway, resulting in increased ARG1 expression. Furthemore, gastric cancer-derived miR-107 TDEs, when dosed intravenously into mice, were also capable of inducing expansion of CD11b+Gr1+/high MDSCs in mouse peripheral blood and altering expression of DICER1, PTEN, ARG1, and NOS2 in the MDSCs. Conclusions: Our findings demonstrate for the first time that gastric cancer-secreted exosomes are able to deliver miR-107 to the host MDSCs where they induce their expansion and activition by targeting DICER1 and PTEN genes, thereby may provide novel cancer therapeutics target for gastric cancer.

6.
Cancer Biol Ther ; 20(4): 402-407, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30359545

RESUMEN

Eosinophilia is a severe disease with increased eosinophil count. The transcript of FIP1L1-PDGFRA fusion gene is a genetic biomarker of clonal eosinophilia screened routinely by reverse transcript PCR (RT-PCR) during diagnosis. Another significant genetic biomarker is the PDGFRA gene alone as some of its mutations are targets of imatinib. In this study, we identified a patient who had typical symptoms of Eosinophilia but had no response to the first-line treatment of hormonotherapy. This patient also showed bone rupture and eosinophil bone infiltration, which are extremely rare among all known eosinophilia patients. We identified the FIP1L1-PDGFRA fusion gene via RT-PCR and Sanger sequencing. Using next generation sequencing (NGS), we detected point mutations in PDGFRA, MYOM2, and ASXL3. The patient then received imatinib therapy, leading to the complete disappearance of FIP1L1-PDGFRA fusion gene and mutated MYOM2. The level of PDGFRA point mutation was also decreased from pre-treatment: 57.86% down to 42.99% at 6 months and to 38.80% at one-year after treatment. The level of ASXL3 mutations did not change significantly. To the best of our knowledge, this is the first case in which the point mutation of PDGFRA has been identified at p.P6L in exon 2, likely making it sensitive to imatinib and thus should be further studied as a potential new molecular target of imatinib therapy.


Asunto(s)
Eosinofilia/tratamiento farmacológico , Eosinofilia/genética , Heterogeneidad Genética , Mesilato de Imatinib/uso terapéutico , Proteínas de Fusión Oncogénica/genética , Mutación Puntual , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Adulto , Antineoplásicos/uso terapéutico , Eosinofilia/patología , Humanos , Masculino , Pronóstico
7.
Biochim Biophys Acta Rev Cancer ; 1869(2): 85-96, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29337112

RESUMEN

Over half of patients with diffuse large B-cell lymphoma (DLBCL) can be cured by standard R-CHOP treatment (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone). However, the remaining patients are refractory and ultimately succumb to progressive or relapsed disease. During the past decade, there has been significant progress in the understanding of molecular mechanisms in DLBCL, largely owing to collaborative efforts in large-scale gene expression profiling and deep sequencing, which have identified genetic alterations critical in lymphomagenesis through activation of key signaling transduction pathways in DLBCL. These discoveries have not only led to the development of targeted therapies, including several currently in clinical trials, but also laid a solid foundation for the future identification of more effective therapies for patients not curable by R-CHOP. This review summarizes the recent advances in our understanding of the molecular characterization and pathogenesis of DLBCL and new treatment directions.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Heterogeneidad Genética , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Técnicas de Diagnóstico Molecular , Medicina de Precisión , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Toma de Decisiones Clínicas , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Linfoma de Células B Grandes Difuso/mortalidad , Linfoma de Células B Grandes Difuso/patología , Terapia Molecular Dirigida , Fenotipo , Valor Predictivo de las Pruebas , Factores de Riesgo , Transcriptoma
8.
Nat Commun ; 8(1): 1488, 2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29133852

RESUMEN

Transcription factor MEF2C regulates multiple genes linked to autism spectrum disorder (ASD), and human MEF2C haploinsufficiency results in ASD, intellectual disability, and epilepsy. However, molecular mechanisms underlying MEF2C haploinsufficiency syndrome remain poorly understood. Here we report that Mef2c +/-(Mef2c-het) mice exhibit behavioral deficits resembling those of human patients. Gene expression analyses on brains from these mice show changes in genes associated with neurogenesis, synapse formation, and neuronal cell death. Accordingly, Mef2c-het mice exhibit decreased neurogenesis, enhanced neuronal apoptosis, and an increased ratio of excitatory to inhibitory (E/I) neurotransmission. Importantly, neurobehavioral deficits, E/I imbalance, and histological damage are all ameliorated by treatment with NitroSynapsin, a new dual-action compound related to the FDA-approved drug memantine, representing an uncompetitive/fast off-rate antagonist of NMDA-type glutamate receptors. These results suggest that MEF2C haploinsufficiency leads to abnormal brain development, E/I imbalance, and neurobehavioral dysfunction, which may be mitigated by pharmacological intervention.


Asunto(s)
Trastorno Autístico/genética , Encéfalo/crecimiento & desarrollo , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Haploinsuficiencia , Memantina/análogos & derivados , Memantina/uso terapéutico , Animales , Trastorno Autístico/patología , Trastorno Autístico/fisiopatología , Conducta Animal , Biomarcadores/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Muerte Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Antagonistas de Aminoácidos Excitadores/farmacología , Perfilación de la Expresión Génica , Humanos , Potenciación a Largo Plazo/genética , Factores de Transcripción MEF2/genética , Memantina/farmacología , Ratones Endogámicos C57BL , Neurogénesis/genética , Neuronas/patología , Fenotipo , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Sinapsis/patología , Transmisión Sináptica/genética
9.
Oncotarget ; 8(47): 82244-82255, 2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-29137260

RESUMEN

Previous studies have shown that the prognosis of lung adenocarcinoma is associated with pathological characterization. In this study, we investigated whether pathology-based prognosis was further influenced by both tumor stage and oncogenic driver mutations. To this end, we recruited a cohort of 465 lung adenocarcinoma patients in China. These patients were classified into 6 pathology-defined subtypes i.e., lepidic-predominant adenocarcinoma (LPA), acinar-predominant adenocarcinoma (APA), papillary-predominant adenocarcinoma (PPA), micropapillary-predominant adenocarcinoma (MPA), solid-predominant adenocarcinoma (SPA), and invasive mucinous adenocarcinoma (IMA). Oncogenic mutations in EGFR, KRAS, ALK, RET, and BRAF genes were determined using fluorescent real-time RT-PCR. The associations of pathogenic subtype or oncogenic mutation with clinical characteristics were analyzed using Fisher's exact tests. The interactive effects on overall survival (OS) by pathologic subtype, oncogenic mutations, and tumor stage were also determined. We have found that pathogenic subtype of lung adenocarcinoma correlated with smoking habit and tumor cell differentiation. These pathology-defined subtypes can be regrouped into 3 pathology-based prognostic groups: PPG1 (LPA), PPG2 (IMA+APA+PPA), and PPG3 (MPA+SPA) with a favorable, intermediate, and poor OS, respectively. We further demonstrated that this pathology-determined OS can be affected by both tumor stage and status of oncogenic mutations in EGFR, KRAS, ALK, RET, and BRAF genes. Interestingly, the presence of genetic mutations related to ALK, RET and BRAF had an opposite effect on OS between PPG2 (worsen) and PPG3 (improved) patients, reversing the prognostic favorability for patients within these two groups. In conclusion, prognosis of lung adenocarcinoma was defined interactively by pathologic subtype, tumor stage and oncogenic mutation.

11.
Oncotarget ; 8(43): 74846-74855, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-29088828

RESUMEN

We previously showed that different pathologic subtypes were associated with different prognostic values in patients with stage IA lung adenocarcinoma (AC). We hypothesize that differential gene expression profiles of different subtypes may be valuable factors for prognosis in stage IA lung adenocarcinoma. We performed microarray gene expression profiling on tumor tissues micro-dissected from patients with acinar and solid predominant subtypes of stage IA lung adenocarcinoma. These patients had undergone a lobectomy and mediastinal lymph node dissection at the Shanghai Chest Hospital, Shanghai, China in 2012. No patient had preoperative treatment. We performed the Gene Set Enrichment Analysis (GSEA) analysis to look for gene expression signatures associated with tumor subtypes. The histologic subtypes of all patients were classified according to the 2015 WHO lung Adenocarcinoma classification. We found that patients with the solid predominant subtype are enriched for genes involved in RNA polymerase activity as well as inactivation of the p53 pathway. Further, we identified a list of genes that may serve as prognostic markers for stage IA lung adenocarcinoma. Validation in the TCGA database shows that these genes are correlated with survival, suggesting that they are novel prognostic factors for stage IA lung adenocarcinoma. In conclusion, we have uncovered novel prognostic factors for stage IA lung adenocarcinoma using gene expression profiling in combination with histopathology subtyping.

12.
Genet Test Mol Biomarkers ; 21(9): 547-554, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28799806

RESUMEN

AIMS: In this study, we determined whether serum tumor markers (STMs), including CA125, are associated with BRCA mutation status and if they can be used prognostically in sporadic ovarian cancer (SOC) and familial ovarian cancer (FOC). METHODS: BRCA gene mutations were screened using next-generation sequencing (NGS) in 31 FOC and 66 SOC patients enrolled between 2013 and 2014. The serum levels of STM CEA, CA125, CA199, and HE4 were also measured in these patients to determine the prognostic potential of these markers and their association with BRCA mutations. RESULTS: Elevated levels of CA125, but not the other three STMs, were associated with FOC and BRCA mutations. Median progression-free survival (PFS) was significantly longer in patients with FOC, higher CA125 expression (>2000 U/mL), and BRCA mutation. Strikingly, the median PFS was not reached in either BRCA+/higher CA125 or FOC/higher CA125 patient groups and these patients had significantly longer PFS than those in other groups. As reported previously, we also detected more BRCA mutations in FOC than in SOC. No significant differences were observed in onset age, menopausal status, tumor stage, and distant metastasis between FOC and SOC patients or between BRCA+ and BRCA- patients. CONCLUSIONS: Elevated levels of serum CA125 are associated with FOC and BRCA mutations, which can be further exploited as a prognostic marker in OC.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Antígeno Ca-125/sangre , Proteínas de la Membrana/sangre , Neoplasias Ováricas/genética , Adulto , Anciano , Proteína BRCA1/sangre , Proteína BRCA1/metabolismo , Proteína BRCA2/sangre , Proteína BRCA2/metabolismo , Biomarcadores de Tumor/sangre , Antígeno Ca-125/metabolismo , Supervivencia sin Enfermedad , Femenino , Humanos , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Mutación , Pronóstico
13.
Pathol Res Pract ; 213(7): 749-758, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28554755

RESUMEN

Lung cancer with EGFR mutation is often associated pathological characteristics and good responses to EGFR tyrosine kinase inhibitors (TKIs). However, certain types of rare EGFR mutations have be linked to cases with poor response to EGFR TKIs. Therefore, extensive molecular screening and pathological characterization are essential for accurate diagnosis and selection of effective treatment plans. Although a large body of studies have established the rate of EGFR mutations as a whole entity, the rates of each individual types of mutations, especially those rare ones, have not been precisely determined in large patient populations with uniform genetic background. To address this issue, we assembled a large cohort of 456 Chinese patients with lung cancers to determine the rate of both common and rare forms of EGFR mutations and associated clinicopathological features in this retrospective study. We have found single or double EGFR mutations in 200 (43.9%) patients, including exon 19 deletions (E19del) (20%), exon 21 L858R (17.1%) and L861Q (1.5%) point mutations, exon 20 T790M (1.3%) and other mutations (1,3%), exon 18 mutations (1.3%), and double mutations (1.3%). EGFR mutation as well as its subtypes E19del, L858R, or double mutations were associated with female patients or never-smokers. In contrast, rare mutations, especially EGFR TKI resistant exon 20 mutations, were not statistically associated with any clinicopathological features, implicating that tumorigenesis driven by different EGFR mutations were mechanistically different. In summary, we have determined occurring rate of EGFR subtype mutations and demonstrated that different mutations showed different clinicopathological manifestations in lung cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Mutación , Adulto , Pueblo Asiatico/genética , Biomarcadores de Tumor/análisis , China , Análisis Mutacional de ADN , Receptores ErbB/análisis , Femenino , Predisposición Genética a la Enfermedad , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/química , Neoplasias Pulmonares/etnología , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Tasa de Mutación , Fenotipo
14.
Cancer Biol Ther ; 17(10): 1003-1009, 2016 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-27611742

RESUMEN

Patients with BCR-ABL1 fusion genes are potential candidates for targeted therapy with tyrosine kinase inhibitor (TKI) imatinib. However, novel BCR-ABL1 fusion variants can be undetected by qRT-PCR-based routine molecular screening, affecting immediate patient management and proper treatment selection. In this study, we describe a case of chronic myeloid leukemia (CML) harboring a novel BCR-ABL1 variant gene. Although Fluorescent In situ Hybridization (FISH) analysis suggested Philadelphia (Ph) translocation, qRT-PCR screening failed to detect the presence of a functional fusion transcript, which is critical for selecting targeted therapy against BCR-ABL1 fusion with aberrant kinase activity. Meanwhile, G-band cytogenetic analysis was performed twice without a solid conclusion. To overcome the uncertainty whether TKIs should be used to treat this patient effectively, we performed whole genome sequencing (WGS) in a next-generation sequencing (NGS) platform and discovered an unusual e13a2-like BCR-ABL1 fusion with 9 ABL1 intron 1 nucleotides incorporated into the broken BCR exon 13 to form a novel chimeric exon, which has never been described previously based on the best of our knowledge. Based on FISH and NGS results, the patient was treated with imatinib, showing significant improvement. Moreover, we also detected novel genetic mutations in the known leukemic genes SETBP1, PAX5, and TP53, while their role in the leukemogenesis remains to be determined. In summary, we have identified BCR-ABL1 fusion and other genetic mutations in a diagnostically difficult case of CML, demonstrating that NGS is a powerful diagnostic tool when routine procedures are challenged.

15.
J Neurosci ; 36(30): 7996-8011, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27466343

RESUMEN

UNLABELLED: Proteolytic generation of amyloidogenic amyloid ß (Aß) fragments from the amyloid precursor protein (APP) significantly contributes to Alzheimer's disease (AD). Although amyloidogenic APP proteolysis can be affected by trafficking through genetically associated AD components such as SORLA, how SORLA functionally interacts with other trafficking components is yet unclear. Here, we report that SNX27, an endosomal trafficking/recycling factor and a negative regulator of the γ-secretase complex, binds to the SORLA cytosolic tail to form a ternary complex with APP. SNX27 enhances cell surface SORLA and APP levels in human cell lines and mouse primary neurons, and depletion of SNX27 or SORLA reduces APP endosome-to-cell surface recycling kinetics. SNX27 overexpression enhances the generation of cell surface APP cleavage products such as soluble alpha-APP C-terminal fragment (CTFα) in a SORLA-dependent manner. SORLA-mediated Aß reduction is attenuated by downregulation of SNX27. This indicates that an SNX27/SORLA complex functionally interacts to limit APP distribution to amyloidogenic compartments, forming a non-amyloidogenic shunt to promote APP recycling to the cell surface. SIGNIFICANCE STATEMENT: Many genes have been identified as risk factors for Alzheimer's disease (AD), and a large proportion of these genes function to limit production or toxicity of the AD-associated amyloid ß (Aß) peptide. Whether and how these genes precisely operate to limit AD onset remains an important question. We identify binding and trafficking interactions between two of these factors, SORLA and SNX27, and demonstrate that SNX27 can direct trafficking of SORLA and the Aß precursor APP to the cell surface to limit the production of Aß. Diversion APP to the cell surface through modulation of this molecular complex may represent a complimentary strategy for future development in AD treatment.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/biosíntesis , Proteínas de Transporte de Membrana/metabolismo , Neuronas/metabolismo , Receptores de LDL/metabolismo , Nexinas de Clasificación/metabolismo , Fracciones Subcelulares/metabolismo , Proteínas Amiloidogénicas/metabolismo , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Neuronas/citología , Unión Proteica , Transporte de Proteínas
16.
Cancer Biol Ther ; 17(8): 785-9, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27415155

RESUMEN

The 8p11 myeloproliferative syndrome (EMS), also known as 8p11 myeloproliferative neoplasm (8p11 MPN), is a collection of rare hematologic malignancies that are associated with fusion genes involving the tyrosine kinase receptor gene FGFR1 in chromosome 8p11. The entity is an aggressive disease with a high rate of transformation to acute myeloid leukemia (AML) and pathologically characterized by its associated eosinophilia. In this study, we reported a distinctive EMS case featuring an in-frame ZMYM2-FGFR1 fusion gene identified by next-generation sequencing technology (NGS). This patient exhibited not only typical EMS signs including elevated white blood cells in peripheral blood and hypercellular bone marrow with marked leukocytosis, but also exceptional characteristics including erythrocytosis in blood and bone marrow basophilia. Moreover, we detected 2 novel genomic mutations in 2 known leukemogenic genes, IKZF1 and ASXL1. Whether these 2 mutations play a part in EMS pathogenesis or contribute to its specific presentations clinically remain to be determined. In summary, we present a unique EMS case involving a ZMYM2-FGFR1 fusion with distinctive hematologic characteristics.


Asunto(s)
Proteínas de Unión al ADN/genética , Trastornos Mieloproliferativos/genética , Proteínas de Fusión Oncogénica/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Persona de Mediana Edad , Trastornos Mieloproliferativos/diagnóstico , Trastornos Mieloproliferativos/patología
17.
Sci Rep ; 6: 18668, 2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26733416

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia in the elderly. It is generally believed that ß-amyloidogenesis, tau-hyperphosphorylation, and synaptic loss underlie cognitive decline in AD. Rps23rg1, a functional retroposed mouse gene, has been shown to reduce Alzheimer's ß-amyloid (Aß) production and tau phosphorylation. In this study, we have identified its human homolog, and demonstrated that RPS23RG1 regulates synaptic plasticity, thus counteracting Aß oligomer (oAß)-induced cognitive deficits in mice. The level of RPS23RG1 mRNA is significantly lower in the brains of AD compared to non-AD patients, suggesting its potential role in the pathogenesis of the disease. Similar to its mouse counterpart, human RPS23RG1 interacts with adenylate cyclase, activating PKA/CREB, and inhibiting GSK-3. Furthermore, we show that human RPS23RG1 promotes synaptic plasticity and offsets oAß-induced synaptic loss in a PKA-dependent manner in cultured primary neurons. Overexpression of Rps23rg1 in transgenic mice consistently prevented oAß-induced PKA inactivation, synaptic deficits, suppression of long-term potentiation, and cognitive impairment as compared to wild type littermates. Our study demonstrates that RPS23RG1 may reduce the occurrence of key elements of AD pathology and enhance synaptic functions to counteract oAß-induced synaptic and cognitive deficits in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/metabolismo , Agregado de Proteínas , Agregación Patológica de Proteínas/metabolismo , Proteínas Ribosómicas/metabolismo , Sinapsis/metabolismo , Adenilil Ciclasas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Secuencia de Bases , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Clonación Molecular , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Disfunción Cognitiva/fisiopatología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Plasticidad Neuronal , Neuronas/metabolismo , Fosforilación , Unión Proteica , ARN Mensajero/genética , Proteínas Ribosómicas/química , Proteínas Ribosómicas/genética , Proteínas tau/genética , Proteínas tau/metabolismo
18.
J Mol Neurosci ; 58(1): 59-65, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26374431

RESUMEN

HIV-associated neurocognitive disorder (HAND) consists of motor and cognitive dysfunction in a relatively large percentage of patients with AIDS. Prior work has suggested that at least part of the neuronal and synaptic damage observed in HAND may occur due to excessive stimulation of NMDA-type glutamate receptors (NMDARs). Here, we compared pharmacological and genetic manipulation of NMDAR activity using an improved derivative of the NMDAR antagonist memantine, termed NitroMemantine, and the modulatory NMDAR subunit GluN3A in the HIV/gp120 transgenic (tg) mouse model of HAND. Interestingly, we found that while both NitroMemantine and GluN3A have been shown to inhibit NMDAR activity, NitroMemantine protected synapses in gp120-tg mice, but overexpression of GluN3A augmented the damage. Given recent findings in the field, one explanation for this apparently paradoxical result is the location of the NMDARs primarily affected, with NitroMemantine inhibiting predominantly extrasynaptic pathologically activated NMDARs, but GluN3A disrupting normal NMDAR-mediated neuroprotective activity via inhibition of synaptic NMDARs.


Asunto(s)
Complejo SIDA Demencia/terapia , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Memantina/uso terapéutico , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Complejo SIDA Demencia/tratamiento farmacológico , Complejo SIDA Demencia/etiología , Animales , Antagonistas de Aminoácidos Excitadores/farmacología , Terapia Genética , Proteína gp120 de Envoltorio del VIH/toxicidad , Memantina/farmacología , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Receptores de N-Metil-D-Aspartato/metabolismo
19.
Mol Neurodegener ; 9: 48, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25394486

RESUMEN

Alzheimer's disease (AD) is a devastating disease characterized by synaptic and neuronal loss in the elderly. Compelling evidence suggests that soluble amyloid-ß peptide (Aß) oligomers induce synaptic loss in AD. Aß-induced synaptic dysfunction is dependent on overstimulation of N-methyl-D-aspartate receptors (NMDARs) resulting in aberrant activation of redox-mediated events as well as elevation of cytoplasmic Ca2+, which in turn triggers downstream pathways involving phospho-tau (p-tau), caspases, Cdk5/dynamin-related protein 1 (Drp1), calcineurin/PP2B, PP2A, Gsk-3ß, Fyn, cofilin, and CaMKII and causes endocytosis of AMPA receptors (AMPARs) as well as NMDARs. Dysfunction in these pathways leads to mitochondrial dysfunction, bioenergetic compromise and consequent synaptic dysfunction and loss, impaired long-term potentiation (LTP), and cognitive decline. Evidence also suggests that Aß may, at least in part, mediate these events by causing an aberrant rise in extrasynaptic glutamate levels by inhibiting glutamate uptake or triggering glutamate release from glial cells. Consequent extrasynaptic NMDAR (eNMDAR) overstimulation then results in synaptic dysfunction via the aforementioned pathways. Consistent with this model of Aß-induced synaptic loss, Aß synaptic toxicity can be partially ameliorated by the NMDAR antagonists (such as memantine and NitroMemantine). PSD-95, an important scaffolding protein that regulates synaptic distribution and activity of both NMDA and AMPA receptors, is also functionally disrupted by Aß. PSD-95 dysregulation is likely an important intermediate step in the pathological cascade of events caused by Aß. In summary, Aß-induced synaptic dysfunction is a complicated process involving multiple pathways, components and biological events, and their underlying mechanisms, albeit as yet incompletely understood, may offer hope for new therapeutic avenues.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Enfermedad de Alzheimer/patología , Animales , Humanos
20.
J Neurosci ; 34(14): 5023-8, 2014 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-24695719

RESUMEN

Oligomerized amyloid-ß (Aß) peptide is thought to contribute to synaptic damage, resulting in dysfunctional neuronal networks in patients with Alzheimer's disease. It has been previously suggested that Aß may be detrimental to neuronal health, at least in part, by triggering oxidative/nitrosative stress. However, the mechanisms underlying this process remain to be elucidated. Here, using rat primary cerebrocortical cultures, we demonstrate that Aß1-42 oligomers trigger a dramatic increase in intracellular nitric oxide (NO) concentration via a process mediated by activation of NMDA-type glutamate receptors (NMDARs). Considering that synaptic NMDARs and extrasynaptic NMDARs (eNMDARs) can have opposite effects on neuronal viability, we explored their respective roles in Aß-induced increases in NO levels. Surprisingly, after pharmacological isolation of eNMDARs, we discovered that eNMDARs are primarily responsible for the increase in neuronal NO triggered by Aß oligomers. Moreover, we found that the eNMDAR-mediated increase in NO can produce S-nitrosylation of Drp1 (dynamin-related protein 1) and Cdk5 (cyclin-dependent kinase 5), targets known to contribute to Aß-induced synaptic damage. These results suggest that pharmacological intervention specifically aimed at eNMDARs may decrease Aß-induced nitrosative stress and thus ameliorate neurotoxic damage to synapses.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Corteza Cerebelosa/citología , Neuronas/efectos de los fármacos , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Fluoresceínas/metabolismo , Humanos , NG-Nitroarginina Metil Éster/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA