Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
BMC Med Genomics ; 17(1): 88, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38627714

RESUMEN

BACKGROUND: Liver cancer ranks sixth in incidence and third in mortality globally and hepatocellular carcinoma (HCC) accounts for 90% of it. Hypoxia, glycolysis, and lactate metabolism have been found to regulate the progression of HCC separately. However, there is a lack of studies linking the above three to predict the prognosis of HCC. The present study aimed to identify a hypoxia-glycolysis-lactate-related gene signature for assessing the prognosis of HCC. METHODS: This study collected 510 hypoxia-glycolysis-lactate genes from Molecular Signatures Database (MSigDB) and then classified HCC patients from TCGA-LIHC by analyzing their hypoxia-glycolysis-lactate genes expression. Differentially expressed genes (DEGs) were screened out to construct a gene signature by LASSO-Cox analysis. Univariate and multivariate regression analyses were used to evaluate the independent prognostic value of the gene signature. Analyses of immune infiltration, somatic cell mutations, and correlation heatmap were conducted by "GSVA" R package. Single-cell analysis conducted by "SingleR", "celldex", "Seurat", and "CellCha" R packages revealed how signature genes participated in hypoxia/glycolysis/lactate metabolism and PPI network identified hub genes. RESULTS: We classified HCC patients from TCGA-LIHC into two clusters and screened out DEGs. An 18-genes prognostic signature including CDCA8, CBX2, PDE6A, MED8, DYNC1LI1, PSMD1, EIF5B, GNL2, SEPHS1, CCNJL, SOCS2, LDHA, G6PD, YBX1, RTN3, ADAMTS5, CLEC3B, and UCK2 was built to stratify the risk of HCC. The risk score of the hypoxia-glycolysis-lactate gene signature was further identified as a valuable independent factor for estimating the prognosis of HCC. Then we found that the features of clinical characteristics, immune infiltration, somatic cell mutations, and correlation analysis differed between the high-risk and low-risk groups. Furthermore, single-cell analysis indicated that the signature genes could interact with the ligand-receptors of hepatocytes/fibroblasts/plasma cells to participate in hypoxia/glycolysis/lactate metabolism and PPI network identified potential hub genes in this process: CDCA8, LDHA, YBX1. CONCLUSION: The hypoxia-glycolysis-lactate-related gene signature we built could provide prognostic value for HCC and suggest several hub genes for future HCC studies.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Ácido Láctico , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Pronóstico , Hipoxia , Proteínas del Ojo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6 , Dineínas Citoplasmáticas
2.
Sci Adv ; 10(11): eadh9547, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38489372

RESUMEN

Solid tumors, especially those with aberrant MYCN activation, often harbor an immunosuppressive microenvironment to fuel malignant growth and trigger treatment resistance. Despite this knowledge, there are no effective strategies to tackle this problem. We found that chemokine-like factor (CKLF) is highly expressed by various solid tumor cells and transcriptionally up-regulated by MYCN. Using the MYCN-driven high-risk neuroblastoma as a model system, we demonstrated that as early as the premalignant stage, tumor cells secrete CKLF to attract CCR4-expressing CD4+ cells, inducing immunosuppression and tumor aggression. Genetic depletion of CD4+ T regulatory cells abolishes the immunorestrictive and protumorigenic effects of CKLF. Our work supports that disrupting CKLF-mediated cross-talk between tumor and CD4+ suppressor cells represents a promising immunotherapeutic approach to battling MYCN-driven tumors.


Asunto(s)
Quimiocinas , Proteínas con Dominio MARVEL , Proteína Proto-Oncogénica N-Myc , Neuroblastoma , Humanos , Línea Celular Tumoral , Quimiocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas con Dominio MARVEL/metabolismo , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuroblastoma/terapia , Microambiente Tumoral
3.
Anal Cell Pathol (Amst) ; 2023: 6007431, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38130905

RESUMEN

Currently, 80%-90% of liver cancers are hepatocellular carcinomas (HCC). HCC patients develop insidiously and have an inferior prognosis. The methyltransferase-like (METTL) family principal members are strongly associated with epigenetic and tumor progression. The present study mainly analyzed the value of METTLs (METTL1/13/18/21A/23/25/2A/2B/5/6/9) and associated mRNA risk signature for HCC. METTLs expression is upregulated in HCC and is a poor prognostic factor in HCC. METTLs were upregulated in patients older than 60 and associated with grade. Except for METTL25, the remaining 10 genes were associated with the HCC stage, invasion depth (T). In addition, METTLs showed an overall alteration rate of 50%. Except for METTL13/2A/25/9, the expression of the other seven genes was significantly associated with overall survival, disease-specific survival, and progression-free survival. Multivariate studies have shown that METTL21A/6 can be an independent prognostic marker in HCC. A total of 664 mRNAs were selected based on Pearson correlation coefficient (R > 0.5), unsupervised consensus clustering, weighted coexpression network analysis, and univariate Cox analysis. These mRNAs were significantly associated with METTLs and were poor prognostic factors in HCC patients. The least absolute shrinkage and selection operator (lasso) was used to construct the best METTLs associated with mRNA risk signature. The mRNA risk signature was significantly associated with age, stage, and t grade. The mRNA high-risk group had higher TP53 and RB1 mutations. This study constructed a nomogram with the mRNA risk profile and clinicopathological features, which could better predict the OS of individuals with HCC. We also analyzed associations between METTLs and mRNA risk signatures in epithelial-mesenchymal transition, immune checkpoints, immune cell infiltration, tumor mutational burden, microsatellite instability, cancer stem cells, tumor pathways, and drug sensitivity. In addition, this study constructed a protein interaction network network including METTLs and mRNA risk signature genes related to tumor microenvironment remodeling based on single-cell sequencing. In conclusion, this study provides a theoretical basis for the mechanism, biomarker screening, and treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Transición Epitelial-Mesenquimal , Mutación/genética , Células Madre Neoplásicas , Microambiente Tumoral , Metiltransferasas
4.
Cancer Sci ; 114(4): 1396-1409, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36562402

RESUMEN

Emerging evidence has suggested that circular RNAs (circRNAs) have vital functions during the initiation and progression of various diseases. However, circRNA potential mechanisms in colorectal cancer (CRC) are largely unknown. Here, we sought to investigate the role and underlying regulatory mechanism of circ0104103 in CRC. circ0104103 was validated by quantitative RT-PCR (qRT-PCR) and Sanger sequencing. Gain- and loss-of-function assays in cell lines and mouse xenograft models were utilized to investigate the effects of circ0104103 in CRC. RNA pull-down assays, RNA immunoprecipitation assays, bioinformatics analyses, RNA FISH, and luciferase reporter assays were used to elucidate the potential mechanism of circ0104103 in CRC. We identified circ0104103, which is strongly downregulated in CRC tissues and cell lines. Functional studies revealed that circ0104103 inhibited CRC cell growth, migration, and invasion both in vitro and in vivo. Mechanistically, circ0104103 binds to HuR, a functional RNA-binding protein commonly expressed in CRC. HuR binds to the 3'UTR of LACTB mRNA to facilitate stabilization and increase its expression. Moreover, circ0104103 was verified as a competing endogenous RNA (ceRNA) via negative regulation of miR-373-5p to increase LACTB expression, resulting in inhibiting the occurrence and progression of CRC. Taken together, our study revealed that circ0104103 acts as a tumor suppressor and may be a novel biomarker and therapeutic target in CRC.


Asunto(s)
Neoplasias Colorrectales , Proteína 1 Similar a ELAV , MicroARNs , ARN Circular , Animales , Humanos , Ratones , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Mitocondriales/metabolismo , Interferencia de ARN , ARN Circular/genética , Proteína 1 Similar a ELAV/genética , Proteína 1 Similar a ELAV/metabolismo
5.
J Immunol ; 208(8): 1912-1923, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35379745

RESUMEN

The mechanism regulating the life span of short-lived plasma cells (SLPCs) remains poorly understood. Here we demonstrated that the EP4-mediated activation of AKT by PGE2 was required for the proper control of inositol-requiring transmembrane kinase endoribonuclease-1α (IRE1α) hyperactivation and hence the endoplasmic reticulum (ER) homeostasis in IgM-producing SLPCs. Disruption of the PGE2-EP4-AKT signaling pathway resulted in IRE1α-induced activation of JNK, leading to accelerated death of SLPCs. Consequently, Ptger4-deficient mice (C57BL/6) exhibited a markedly impaired IgM response to T-independent Ags and increased susceptibility to Streptococcus pneumoniae infection. This study reveals a highly selective impact of the PGE2-EP4 signal on the humoral immunity and provides a link between ER stress response and the life span of SLPCs.


Asunto(s)
Supervivencia Celular , Dinoprostona , Estrés del Retículo Endoplásmico , Endorribonucleasas , Células Plasmáticas , Proteínas Serina-Treonina Quinasas , Animales , Supervivencia Celular/inmunología , Dinoprostona/inmunología , Estrés del Retículo Endoplásmico/inmunología , Endorribonucleasas/inmunología , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos C57BL , Células Plasmáticas/inmunología , Prostaglandinas/inmunología , Prostaglandinas E/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología
6.
Cancer Res ; 81(17): 4417-4430, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34233924

RESUMEN

High-risk neuroblastoma remains therapeutically challenging to treat, and the mechanisms promoting disease aggression are poorly understood. Here, we show that elevated expression of dihydrolipoamide S-succinyltransferase (DLST) predicts poor treatment outcome and aggressive disease in patients with neuroblastoma. DLST is an E2 component of the α-ketoglutarate (αKG) dehydrogenase complex, which governs the entry of glutamine into the tricarboxylic acid cycle (TCA) for oxidative decarboxylation. During this irreversible step, αKG is converted into succinyl-CoA, producing NADH for oxidative phosphorylation (OXPHOS). Utilizing a zebrafish model of MYCN-driven neuroblastoma, we demonstrate that even modest increases in DLST expression promote tumor aggression, while monoallelic dlst loss impedes disease initiation and progression. DLST depletion in human MYCN-amplified neuroblastoma cells minimally affected glutamine anaplerosis and did not alter TCA cycle metabolites other than αKG. However, DLST loss significantly suppressed NADH production and impaired OXPHOS, leading to growth arrest and apoptosis of neuroblastoma cells. In addition, multiple inhibitors targeting the electron transport chain, including the potent IACS-010759 that is currently in clinical testing for other cancers, efficiently reduced neuroblastoma proliferation in vitro. IACS-010759 also suppressed tumor growth in zebrafish and mouse xenograft models of high-risk neuroblastoma. Together, these results demonstrate that DLST promotes neuroblastoma aggression and unveils OXPHOS as an essential contributor to high-risk neuroblastoma. SIGNIFICANCE: These findings demonstrate a novel role for DLST in neuroblastoma aggression and identify the OXPHOS inhibitor IACS-010759 as a potential therapeutic strategy for this deadly disease.


Asunto(s)
Aciltransferasas/metabolismo , Neoplasias Encefálicas/metabolismo , Neuroblastoma/metabolismo , Fosforilación Oxidativa , Animales , Apoptosis , Línea Celular Tumoral , Colágeno/química , Modelos Animales de Enfermedad , Combinación de Medicamentos , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Concentración 50 Inhibidora , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Laminina/química , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Trasplante de Neoplasias , Oxígeno/metabolismo , Proteoglicanos/química , Interferencia de ARN , Riesgo , Smegmamorpha , Resultado del Tratamiento , Ácidos Tricarboxílicos/metabolismo , Pez Cebra
7.
Front Cell Dev Biol ; 9: 660969, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34095125

RESUMEN

The future of improved immunotherapy against cancer depends on an in-depth understanding of the dynamic interactions between the immune system and tumors. Over the past two decades, the zebrafish has served as a valuable model system to provide fresh insights into both the development of the immune system and the etiologies of many different cancers. This well-established foundation of knowledge combined with the imaging and genetic capacities of the zebrafish provides a new frontier in cancer immunology research. In this review, we provide an overview of the development of the zebrafish immune system along with a side-by-side comparison of its human counterpart. We then introduce components of the adaptive immune system with a focus on their roles in the tumor microenvironment (TME) of teleosts. In addition, we summarize zebrafish models developed for the study of cancer and adaptive immunity along with other available tools and technology afforded by this experimental system. Finally, we discuss some recent research conducted using the zebrafish to investigate adaptive immune cell-tumor interactions. Without a doubt, the zebrafish will arise as one of the driving forces to help expand the knowledge of tumor immunity and facilitate the development of improved anti-cancer immunotherapy in the foreseeable future.

8.
Nat Commun ; 12(1): 1736, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741957

RESUMEN

Metastasis is the leading cause of cancer-related death. Despite the recent advancements in cancer treatment, there is currently no approved therapy for metastasis. The present study reveals a potent and selective activity of PRAK in the regulation of tumor metastasis. While showing no apparent effect on the growth of primary breast cancers or subcutaneously inoculated tumor lines, Prak deficiency abrogates lung metastases in PyMT mice or mice receiving intravenous injection of tumor cells. Consistently, PRAK expression is closely associated with metastatic risk in human cancers. Further analysis indicates that loss of function of PRAK leads to a pronounced inhibition of HIF-1α protein synthesis, possibly due to reduced mTORC1 activities. Notably, pharmacological inactivation of PRAK with a clinically relevant inhibitor recapitulates the anti-metastatic effect of Prak depletion, highlighting the therapeutic potential of targeting PRAK in the control of metastasis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Neoplasias de la Mama , Línea Celular Tumoral , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Neoplasias/terapia , Proteínas Serina-Treonina Quinasas/genética
9.
Quant Imaging Med Surg ; 11(3): 983-997, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33654671

RESUMEN

BACKGROUND: Conventional light sheet fluorescence microscopy (LSFM), or selective plane illumination microscopy (SPIM), enables high-resolution 3D imaging over a large volume by using two orthogonally aligned objective lenses to decouple excitation and emission. The recent development of oblique plane microscopy (OPM) simplifies LSFM design with only one single objective lens, by using off-axis excitation and remote focusing. However, most reports on OPM have a limited microscopic field of view (FOV), typically within 1×1 mm2. Our goal is to overcome the limitation with a new variant of OPM to achieve a mesoscopic FOV. METHODS: We implemented an optical design of mesoscopic scanning OPM to allow the use of low numerical aperture (NA) objective lenses. The angle of the intermediate image before the remote focusing system was increased by a demagnification under Scheimpflug condition such that the light collecting efficiency in the remote focusing system was significantly improved. A telescope composed of cylindrical lenses was used to correct the distorted image caused by the demagnification design. We characterized the 3D resolutions and imaging volume by imaging fluorescent microspheres, and demonstrated the volumetric imaging on intact whole zebrafish larvae, mouse cortex, and multiple Caenorhabditis elegans (C. elegans). RESULTS: We demonstrate a mesoscopic FOV up to ~6×5×0.6 mm3 volumetric imaging, the largest reported FOV by OPM so far. The angle of the intermediate image plane is independent of the magnification as long as the size of the pupil aperture of the objectives is the same. As a result, the system is highly versatile, allowing simple switching between different objective lenses with low (10×, NA 0.3) and median NA (20×, NA 0.5). Detailed microvasculature in zebrafish larvae, mouse cortex, and neurons in C. elegans are clearly visualized in 3D. CONCLUSIONS: The proposed mesoscopic scanning OPM allows using low NA objectives such that centimeter-level FOV volumetric imaging can be achieved. With the extended FOV, simple sample mounting protocol, and the versatility of changeable FOVs/resolutions, our system will be ready for the varieties of applications requiring in vivo volumetric imaging over large length scales.

10.
Crit Rev Biochem Mol Biol ; 55(4): 322-353, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32633575

RESUMEN

During malignant transformation and cancer progression, tumor cells face both intrinsic and extrinsic stress, endoplasmic reticulum (ER) stress in particular. To survive and proliferate, tumor cells use multiple stress response pathways to mitigate ER stress, promoting disease aggression and treatment resistance. Among the stress response pathways is ER-associated degradation (ERAD), which consists of multiple components and steps working together to ensure protein quality and quantity. In addition to its established role in stress responses and tumor cell survival, ERAD has recently been shown to regulate tumor immunity. Here we summarize current knowledge on how ERAD promotes protein degradation, regulates immune cell development and function, participates in antigen presentation, exerts paradoxical roles on tumorigenesis and immunity, and thus impacts current cancer therapy. Collectively, ERAD is a critical protein homeostasis pathway intertwined with cancer development and tumor immunity. Of particular importance is the need to further unveil ERAD's enigmatic roles in tumor immunity to develop effective targeted and combination therapy for successful treatment of cancer.


Asunto(s)
Carcinogénesis/inmunología , Estrés del Retículo Endoplásmico/inmunología , Degradación Asociada con el Retículo Endoplásmico/inmunología , Neoplasias/inmunología , Proteolisis , Animales , Carcinogénesis/patología , Humanos , Neoplasias/patología , Neoplasias/terapia
11.
J Vis Exp ; (159)2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32449736

RESUMEN

Developing nanoparticles capable of detecting, targeting, and destroying cancer cells is of great interest in the field of nanomedicine. In vivo animal models are required for bridging the nanotechnology to its biomedical application. The mouse represents the traditional animal model for preclinical testing; however, mice are relatively expensive to keep and have long experimental cycles due to the limited progeny from each mother. The zebrafish has emerged as a powerful model system for developmental and biomedical research, including cancer research. In particular, due to its optical transparency and rapid development, zebrafish embryos are well suited for real-time in vivo monitoring of the behavior of cancer cells and their interactions with their microenvironment. This method was developed to sequentially introduce human cancer cells and functionalized nanoparticles in transparent Casper zebrafish embryos and monitor in vivo recognition and targeting of the cancer cells by nanoparticles in real time. This optimized protocol shows that fluorescently labeled nanoparticles, which are functionalized with folate groups, can specifically recognize and target metastatic human cervical epithelial cancer cells labeled with a different fluorochrome. The recognition and targeting process can occur as early as 30 min postinjection of the nanoparticles tested. The whole experiment only requires the breeding of a few pairs of adult fish and takes less than 4 days to complete. Moreover, zebrafish embryos lack a functional adaptive immune system, allowing the engraftment of a wide range of human cancer cells. Hence, the utility of the protocol described here enables the testing of nanoparticles on various types of human cancer cells, facilitating the selection of optimal nanoparticles in each specific cancer context for future testing in mammals and the clinic.


Asunto(s)
Nanopartículas/química , Neoplasias/genética , Dióxido de Silicio/química , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Microambiente Tumoral , Pez Cebra
12.
Nanoscale ; 11(46): 22316-22327, 2019 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-31724677

RESUMEN

New ultrabright fluorescent silica nanoparticles capable of the fast targeting of epithelial tumors in vivo are presented. The as-synthesized folate-functionalized ultrabright particles of 30-40 nm are 230 times brighter than quantum dots (QD450) and 50% brighter than the polymer dots with similar spectra (excitation 365 nm and emission 486 nm). To decrease non-specific targeting, particles are coated with polyethylene glycol (PEG). We demonstrate the in vivo targeting of xenographic human cervical epithelial tumors (HeLa cells) using zebrafish as a model system. The particles target tumors (and probably even individual HeLa cells) as small as 10-20 microns within 20-30 minutes after blood injection. To demonstrate the advantages of ultrabrightness, we repeated the experiments with similar but 200× less bright particles. Compared to those, ultrabright particles showed ∼3× faster tumor detection and ∼2× higher relative fluorescent contrast of tumors/cancer cells.


Asunto(s)
Nanopartículas/química , Neoplasias/diagnóstico por imagen , Dióxido de Silicio/química , Animales , Femenino , Ácido Fólico/química , Células HeLa , Humanos , Imagen Óptica , Tamaño de la Partícula , Polietilenglicoles/química , Porosidad , Trasplante Heterólogo , Pez Cebra
13.
Nanoscale ; 11(14): 6784-6793, 2019 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-30907913

RESUMEN

Dual-modal fluorescence-magnetic resonance imaging (MRI) techniques have gained great interest in biomedical research and clinical practice, since they integrate the advantages of both imaging techniques and provide a useful approach to simultaneously investigate both molecular and anatomical information at the same biological structures. Herein, we report the construction of a dual-modal time-gated luminescence (TGL)/MRI nanoprobe, BHHBB-Eu3+@MnO2, for glutathione (GSH) by anchoring luminescent ß-diketone-Eu3+ complexes on layered MnO2 nanosheets. The fabricated nanoprobe exhibited very week luminescence and MR signals since the luminescence of the Eu3+ complex was quenched by MnO2 nanosheets and Mn atoms were isolated from water. Upon exposure to GSH, the MnO2 nanosheets were rapidly and selectively reduced to Mn2+ ions, resulting in remarkable enhancements of TGL and MR signals simultaneously. The combination of TGL and MR detection modes enables the nanoprobe to be used for detecting GSH in a wide concentration range (1-1000 µM) and imaging GSH at different resolutions and depths ranging from the subcellular level to the whole body. Furthermore, the as-prepared nanoprobe exhibited a low cytotoxicity and good biocompatibility, rapid response rate, long-lived luminescence, and high sensitivity and selectivity for responding to GSH. These features allowed it to be successfully used for the TGL detection of GSH in human sera, TGL imaging of GSH in living cells and zebrafish, as well as dual-modal TGL/MR imaging of GSH in tumor-bearing mice. All of these results highlighted the applicability and advantages of the nanoprobe for detecting GSH in vitro and in vivo.


Asunto(s)
Complejos de Coordinación/química , Europio/química , Glutatión/química , Imagen por Resonancia Magnética , Compuestos de Manganeso/química , Nanocompuestos/química , Óxidos/química , Animales , Femenino , Glutatión/sangre , Glutatión/metabolismo , Células HeLa , Humanos , Mediciones Luminiscentes , Ratones , Ratones Endogámicos BALB C , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Trasplante Heterólogo , Pez Cebra
14.
Leukemia ; 33(6): 1475-1486, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30573782

RESUMEN

B7-H3 (CD276) is broadly overexpressed by multiple human cancers. It plays a vital role in tumor progression and has been accepted as one of the inhibitory B7 family checkpoint molecules. To identify the functions and underlying mechanisms of B7-H3 in multiple myeloma, we analyzed B7-H3 expression in myeloma patients and used siRNAs and overexpression plasmid of B7-H3 to investigate its roles and downstream signaling molecules in myeloma cell lines. The results showed that surface expression of B7-H3 was upregulated in myeloma samples and cell lines. Lower expression of B7-H3 in myeloma cells was associated with better progression-free survival. Myeloma cell survival, drug resistance, and tumor growth could be promoted by B7-H3. The molecular basis for these functional roles of B7-H3 involved the activation of JAK2/STAT3 via redox-mediated oxidation and activation of Src. We further identified a STAT3-promoting signaling pathway by which oxidant-mediated Src phosphorylation led to secondary activation of the E3 ubiquitin ligase c-Cbl. Activated c-Cbl subsequently caused specific proteasomal degradation of SOCS3, a negative regulator of JAK2/STAT3. These data indicate B7-H3's important role in the activation of ROS/Src/c-Cbl pathway in multiple myeloma which integrates redox regulation and sustained STAT3 activation at the level of degradation of STAT3 suppressor.


Asunto(s)
Apoptosis , Antígenos B7/metabolismo , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple/patología , Especies Reactivas de Oxígeno/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos , Antígenos B7/genética , Biomarcadores de Tumor/genética , Estudios de Casos y Controles , Resistencia a Antineoplásicos , Femenino , Estudios de Seguimiento , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
15.
ACS Appl Mater Interfaces ; 10(33): 27681-27691, 2018 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-30058801

RESUMEN

Bimodal fluorescence-magnetic resonance imaging (MRI) technique has shown great utilities in bioassays because it combines the advantages of both optical imaging and MRI to provide more sufficient information over any modality alone. In this work, on the basis of a MnO2 nanosheet-Ru(II) complex nanoarchitecture, a bimodal phosphorescence-MRI nanoprobe for glutathione (GSH) has been constructed. The nanoprobe, Ru(BPY)3@MnO2, was constructed by integrating MnO2 nanosheets with a phosphorescent Ru(II) complex [Ru(BPY)3](PF6)2 (BPY = 2,2'-bipyridine), which resulted in complete phosphorescence quenching of the Ru(II) complex, accompanied by very low longitudinal and transverse relaxivity. Upon exposure to GSH, the reduction of MnO2 nanosheets by GSH triggers a recovery of phosphorescence and simultaneously produces a number of Mn2+ ions, a perfect MRI contrast agent. The as-prepared nanoprobe showed good water dispersion and biocompatibility and a rapid, selective, and sensitive response toward GSH in the phosphorescence and MR detection modes. The practicability of the nanoprobe was proved by time-gated luminescence assay of GSH in human serum, phosphorescent imaging of endogenous GSH in living cells, zebrafish, and tumor-bearing mice, as well as the MRI of GSH in tumor-bearing mice. The research outcomes suggested the potential of Ru(BPY)3@MnO2 for the bimodal phosphorescence-MRI sensing of GSH in vitro and in vivo.


Asunto(s)
Nanoestructuras , Animales , Medios de Contraste , Glutatión , Humanos , Iones , Imagen por Resonancia Magnética , Compuestos de Manganeso , Ratones , Imagen Óptica , Óxidos , Compuestos de Rutenio
16.
Mol Immunol ; 99: 87-94, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29730547

RESUMEN

Degeneration of the thymic epithelium is believed to be the primary cause of age-associated thymic involution. In order to investigate the molecular events during the early phase of thymic involution, RNA-seq was performed to gain the transcriptional profiles of medullary thymic epithelial cells (mTEC) from mice of 2, 6 and 10 weeks of age. We confirmed and extended the previous observation of declined expression of cell cycle-related genes and diminished E2F3 activity during thymic involution, showing that it occurred as early as 2-6 weeks after birth. Moreover, we demonstrated that mTEC aging was coupled with augmented expression of inflammatory chemokines and cytokines, reminiscent of the senescence-associated secretory phenotype. Impaired cell cycling and proinflammatoty response therefore represent two predominant transcriptional signatures during the very early phase of thymic involution. Taken together, the present study provides not only complimentary information about, but also new insight into the molecular mechanisms underlying age-related degeneration of thymic epithelial cells.


Asunto(s)
Células Epiteliales/fisiología , Timo/fisiología , Transcriptoma/genética , Envejecimiento/genética , Animales , Diferenciación Celular/genética , Citocinas/genética , Femenino , Perfilación de la Expresión Génica/métodos , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Transcripción Genética/genética
17.
Sci Rep ; 7: 45305, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28345605

RESUMEN

Reelin is an extracellular matrix protein that is crucial for neuron migration, adhesion, and positioning. We examined the expression of Reelin in a large cohort of multiple myeloma patients recorded in Gene Expression Omnibus (GEO) database and used over-expression and siRNA knockdown of Reelin to investigate the role of Reelin in myeloma cell growth. We find that Reelin expression is negatively associated with myeloma prognosis. Reelin promotes myeloma cell proliferation in vitro as well as in vivo. The Warburg effect, evidenced by increased glucose uptake and lactate production, is also enhanced in Reelin-expressing cells. The activation of FAK/Syk/Akt/mTOR and STAT3 pathways contributes to Reelin-induced cancer cell growth and metabolic reprogramming. Our findings further reveal that activated Akt and STAT3 pathways induce the upregulation of HIF1α and its downstream targets (LDHA and PDK1), leading to increased glycolysis in myeloma cells. Together, our results demonstrate the critical contributions of Reelin to myeloma growth and metabolism. It presents an opportunity for myeloma therapeutic intervention by inhibiting Reelin and its signaling pathways.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Proliferación Celular/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Glucólisis/fisiología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Glucosa/metabolismo , Humanos , Ácido Láctico/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pronóstico , ARN Interferente Pequeño/metabolismo , Proteína Reelina , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
18.
Oncotarget ; 7(9): 9844-58, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26848618

RESUMEN

Reelin is an extracellular matrix (ECM) protein that is essential for neuron migration and positioning. The expression of reelin in multiple myeloma (MM) cells and its association with cell adhesion and survival were investigated. Overexpression, siRNA knockdown, and the addition of recombinant protein of reelin were used to examine the function of reelin in MM cells. Clinically, high expression of reelin was negatively associated with progression-free survival and overall survival. Functionally, reelin promoted the adhesion of MM cells to fibronectin via activation of α5ß1 integrin. The resulting phosphorylation of Focal Adhesion Kinase (FAK) led to the activation of Src/Syk/STAT3 and Akt, crucial signaling molecules involved in enhancing cell adhesion and protecting cells from drug-induced cell apoptosis. These findings indicate reelin's important role in the activation of integrin-ß1 and STAT3/Akt pathways in multiple myeloma and highlight the therapeutic potential of targeting reelin/integrin/FAK axis.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Adhesión Celular/fisiología , Resistencia a Antineoplásicos/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Integrina alfa5beta1/metabolismo , Integrina beta1/metabolismo , Mieloma Múltiple/patología , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción STAT3/metabolismo , Serina Endopeptidasas/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Supervivencia sin Enfermedad , Activación Enzimática , Proteínas de la Matriz Extracelular/genética , Fibronectinas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Proteínas del Tejido Nervioso/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteína Reelina , Serina Endopeptidasas/genética , Quinasa Syk/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA