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

Base de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
2.
Sci Adv ; 10(38): eadj4122, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39303039

RESUMEN

Sarcopenia is characterized by accelerated muscle mass and function loss, which burdens and challenges public health worldwide. Several studies indicated that selenium deficiency is associated with sarcopenia; however, the specific mechanism remains unclear. Here, we demonstrated that selenoprotein W (SELENOW) containing selenium in the form of selenocysteine functioned in sarcopenia. SELENOW expression is up-regulated in dexamethasone (DEX)-induced muscle atrophy and age-related sarcopenia mouse models. Knockout (KO) of SELENOW profoundly aggravated the process of muscle mass loss in the two mouse models. Mechanistically, SELENOW KO suppressed the RAC1-mTOR cascade by the interaction between SELENOW and RAC1 and induced the imbalance of protein synthesis and degradation. Consistently, overexpression of SELENOW in vivo and in vitro alleviated the muscle and myotube atrophy induced by DEX. SELENOW played a role in age-related sarcopenia and regulated the genes associated with aging. Together, our study uncovered the function of SELENOW in age-related sarcopenia and provides promising evidence for the prevention and treatment of sarcopenia.


Asunto(s)
Ratones Noqueados , Complejo de la Endopetidasa Proteasomal , Biosíntesis de Proteínas , Sarcopenia , Selenoproteína W , Ubiquitina , Animales , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratones , Sarcopenia/metabolismo , Sarcopenia/genética , Sarcopenia/patología , Ubiquitina/metabolismo , Selenoproteína W/genética , Selenoproteína W/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Dexametasona/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Modelos Animales de Enfermedad , Atrofia Muscular/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , Atrofia Muscular/inducido químicamente , Envejecimiento/metabolismo , Masculino , Transducción de Señal , Neuropéptidos
3.
Bone Res ; 12(1): 53, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242551

RESUMEN

Intervertebral disc degeneration (IDD) is a major cause of discogenic pain, and is attributed to the dysfunction of nucleus pulposus, annulus fibrosus, and cartilaginous endplate (CEP). Osteopontin (OPN), a glycoprotein, is highly expressed in the CEP. However, little is known on how OPN regulates CEP homeostasis and degeneration, contributing to the pathogenesis of IDD. Here, we investigate the roles of OPN in CEP degeneration in a mouse IDD model induced by lumbar spine instability and its impact on the degeneration of endplate chondrocytes (EPCs) under pathological conditions. OPN is mainly expressed in the CEP and decreases with degeneration in mice and human patients with severe IDD. Conditional Spp1 knockout in EPCs of adult mice enhances age-related CEP degeneration and accelerates CEP remodeling during IDD. Mechanistically, OPN deficiency increases CCL2 and CCL5 production in EPCs to recruit macrophages and enhances the activation of NLRP3 inflammasome and NF-κB signaling by facilitating assembly of IRAK1-TRAF6 complex, deteriorating CEP degeneration in a spatiotemporal pattern. More importantly, pharmacological inhibition of the NF-κB/NLRP3 axis attenuates CEP degeneration in OPN-deficient IDD mice. Overall, this study highlights the importance of OPN in maintaining CEP and disc homeostasis, and proposes a promising therapeutic strategy for IDD by targeting the NF-κB/NLRP3 axis.


Asunto(s)
Inflamasomas , Degeneración del Disco Intervertebral , Macrófagos , Ratones Noqueados , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , Osteopontina , Transducción de Señal , Adulto , Anciano , Anciano de 80 o más Años , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Adulto Joven , Cartílago/patología , Cartílago/metabolismo , Condrocitos/metabolismo , Condrocitos/patología , Inflamasomas/metabolismo , Degeneración del Disco Intervertebral/patología , Degeneración del Disco Intervertebral/metabolismo , Degeneración del Disco Intervertebral/genética , Macrófagos/metabolismo , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Osteopontina/metabolismo , Osteopontina/deficiencia , Osteopontina/genética
4.
Metabolism ; 160: 155980, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39053691

RESUMEN

BACKGROUND: The effect of coronavirus disease 2019 (COVID-19) on adrenal endocrine metabolism in critically ill patients remains unclear. This study aimed to investigate the alterations in adrenal steroidogenic activity, elucidate underlying mechanisms, provide in situ histopathological evidence, and examine the clinical implications. METHODS: The comparative analyses of the adrenal cortices from 24 patients with fatal COVID-19 and 20 matched controls were performed, excluding patients previously treated with glucocorticoids. SARS-CoV-2 and its receptors were identified and pathological alterations were examined. Furthermore, histological examinations, immunohistochemical staining and ultrastructural analyses were performed to assess corticosteroid biosynthesis. The zona glomerulosa (ZG) and zona fasciculata (ZF) were then dissected for proteomic analyses. The biological processes that affected steroidogenesis were analyzed by integrating histological, proteomic, and clinical data. Finally, the immunoreactivity and responsive genes of mineralocorticoid and glucocorticoid receptors in essential tissues were quantitatively measured to evaluate corticosteroid responsiveness. FINDINGS: The demographic characteristics of COVID-19 patients were comparable with those of controls. SARS-CoV-2-like particles were identified in the adrenocortical cells of three patients; however, these particles did not affect cellular morphology or steroid synthesis compared with SARS-CoV-2-negative specimens. Although the adrenals exhibited focal necrosis, vacuolization, microthrombi, and inflammation, widespread degeneration was not evident. Notably, corticosteroid biosynthesis was significantly enhanced in both the ZG and ZF of COVID-19 patients. The increase in the inflammatory response and cellular differentiation in the adrenal cortices of patients with critical COVID-19 was positively correlated with heightened steroidogenic activity. Additionally, the appearance of more dual-ZG/ZF identity cells in COVID-19 adrenals was in accordance with the increased steroidogenic function. However, activated mineralocorticoid and glucocorticoid receptors and their responsive genes in vital tissues were markedly reduced in patients with critical COVID-19. INTERPRETATION: Critical COVID-19 was characterized by potentiated adrenal steroidogenesis, associated with increased inflammation, enhanced differentiation and elevated dual-ZG/ZF identity cells, alongside suppressed corticosteroid responsiveness. These alterations implied the reduced effectiveness of conventional corticosteroid therapy and underscored the need for evaluation of the adrenal axis and corticosteroid sensitivity.


Asunto(s)
Corticoesteroides , COVID-19 , Enfermedad Crítica , Humanos , COVID-19/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Corticoesteroides/uso terapéutico , Corticoesteroides/biosíntesis , Anciano , SARS-CoV-2 , Zona Fascicular/metabolismo , Zona Fascicular/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Adulto , Corteza Suprarrenal/metabolismo , Corteza Suprarrenal/efectos de los fármacos , Corteza Suprarrenal/patología , Zona Glomerular/metabolismo , Zona Glomerular/efectos de los fármacos , Zona Glomerular/patología , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/efectos de los fármacos
5.
Cancer Cell ; 42(5): 815-832.e12, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38640932

RESUMEN

Monocyte-derived tumor-associated macrophages (Mo-TAMs) intensively infiltrate diffuse gliomas with remarkable heterogeneity. Using single-cell transcriptomics, we chart a spatially resolved transcriptional landscape of Mo-TAMs across 51 patients with isocitrate dehydrogenase (IDH)-wild-type glioblastomas or IDH-mutant gliomas. We characterize a Mo-TAM subset that is localized to the peri-necrotic niche and skewed by hypoxic niche cues to acquire a hypoxia response signature. Hypoxia-TAM destabilizes endothelial adherens junctions by activating adrenomedullin paracrine signaling, thereby stimulating a hyperpermeable neovasculature that hampers drug delivery in glioblastoma xenografts. Accordingly, genetic ablation or pharmacological blockade of adrenomedullin produced by Hypoxia-TAM restores vascular integrity, improves intratumoral concentration of the anti-tumor agent dabrafenib, and achieves combinatorial therapeutic benefits. Increased proportion of Hypoxia-TAM or adrenomedullin expression is predictive of tumor vessel hyperpermeability and a worse prognosis of glioblastoma. Our findings highlight Mo-TAM diversity and spatial niche-steered Mo-TAM reprogramming in diffuse gliomas and indicate potential therapeutics targeting Hypoxia-TAM to normalize tumor vasculature.


Asunto(s)
Adrenomedulina , Neoplasias Encefálicas , Glioblastoma , Macrófagos Asociados a Tumores , Humanos , Glioblastoma/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/irrigación sanguínea , Glioblastoma/genética , Glioblastoma/metabolismo , Animales , Adrenomedulina/genética , Adrenomedulina/metabolismo , Ratones , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Neovascularización Patológica/genética , Microambiente Tumoral , Isocitrato Deshidrogenasa/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Macrófagos/metabolismo , Hipoxia de la Célula
6.
Pathol Res Pract ; 252: 154920, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37948998

RESUMEN

Clinical data indicates that SARS-CoV-2 infection-induced respiratory failure is a fatal condition for severe COVID-19 patients. However, the pathological alterations of different types of respiratory failure remained unknown for severe COVID-19 patients. This study aims to evaluate whether there are differences in the performance of various types of respiratory failure in severe COVID-19 patients and investigate the pathological basis for these differences. The lung tissue sections of severe COVID-19 patients were assessed for the degree of injury and immune responses. Transcriptome data were used to analyze the molecular basis in severe COVID-19 patients. Severe COVID-19 patients with combined oxygenation and ventilatory failure presented more severe pulmonary fibrosis, airway obstruction, and prolonged disease course. The number of M2 macrophages increased with the degree of fibrosis in patients, suggesting that it may be closely related to the development of pulmonary fibrosis. The co-existence of pro-inflammatory and anti-inflammatory cytokines in the pulmonary environment could also participate in the progression of pulmonary fibrosis. Furthermore, the increased apoptosis in the lungs of COVID-19 patients with severe pulmonary fibrosis may represent a critical factor linking sustained inflammatory responses to fibrosis. Our findings indicate that during the extended phase of COVID-19, antifibrotic and antiapoptotic treatments should be considered in conjunction with the progression of the disease.


Asunto(s)
COVID-19 , Fibrosis Pulmonar , Insuficiencia Respiratoria , Humanos , COVID-19/complicaciones , COVID-19/patología , Fibrosis Pulmonar/patología , Autopsia , SARS-CoV-2 , Pulmón/patología , Macrófagos/patología , Insuficiencia Respiratoria/patología , Apoptosis
7.
Food Chem Toxicol ; 174: 113682, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36813151

RESUMEN

The objective of this study was to identify the key glutathione S-transferase (GST) isozymes involved in the detoxification of Aflatoxin B1 (AFB1) in ducks' primary hepatocytes. The full-length cDNA encoding the 10 GST isozymes (GST, GST3, GSTM3, MGST1, MGST2, MGST3, GSTK1, GSTT1, GSTO1 and GSTZ1) were isolated/synthesized from ducks' liver and cloned into the pcDNA3.1(+) vector. The results showed that pcDNA3.1(+)-GSTs plasmids were successfully transferred into the ducks' primary hepatocytes and the mRNA of the 10 GST isozymes were overexpressed by 1.9-3274.7 times. Compared to the control, 75 µg/L (IC30) or 150 µg/L (IC50) AFB1 treatment reduced the cell viability by 30.0-50.0% and increased the LDH activity by 19.8-58.2% in the ducks' primary hepatocytes. Notably, the AFB1-induced changes in cell viability and LDH activity were mitigated by overexpression of GST and GST3. Compared to the cells treated with AFB1, exo-AFB1-8,9-epoxide (AFBO)-GSH, as the major detoxified product of AFB1, was increased in the cells overexpression of GST and GST3. Moreover, the sequences, phylogenetic and domain analysis revealed that the GST and GST3 were orthologous to Meleagris gallopavo GSTA3 and GSTA4. In conclusion, this study found that the ducks' GST and GST3 were orthologous to Meleagris gallopavo GSTA3 and GSTA4, which were involved in the detoxification of AFB1 in ducks' primary hepatocytes.


Asunto(s)
Aflatoxina B1 , Patos , Animales , Isoenzimas/genética , Filogenia , Hígado , Glutatión Transferasa/genética , Glutatión/genética
8.
Free Radic Biol Med ; 197: 15-22, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36731804

RESUMEN

Selenium (Se) deficiency or excess impairs testicular development and spermatogenesis, while the underlying mechanisms in this regard remain unclear. This study was designed to explore the molecular biology of Se deficiency or excess in spermatogenesis in mice. Three-week-old male mice (n = 10 mice/diet) were fed with Se-deficient diet (SeD, 0.02 mg Se/kg), adequate-Se diet (SeA, 0.2 mg Se/kg), or excess-Se diet (SeE, 2.0 mg Se/kg) for 5 months. Compared with SeA, SeD reduced (P < 0.05) the body weight (10.4%) and sperm density (84.3%) but increased (P < 0.05) sperm deformity (32.8%); SeE decreased (P < 0.05) the sperm density (78.5%) and sperm motility (35.9%) of the mice. Meanwhile, both SeD and SeE increased (P < 0.05) serum FSH concentrations (10.4-25.6%) and induced testicular damage in mice in comparison with the SeA. Compared with SeA, SeD increased (P < 0.05) the 8-OHdG concentration by 25.5%; SeE increased (P < 0.05) both MDA and 8-OHdG concentrations by 118.8-180.3% in testis. Furthermore, transcriptome analysis showed that there 1325 and 858 transcripts were altered (P < 0.05) in the testis by SeD and SeE, respectively, compared with SeA. KEGG pathway analysis revealed that these differentially expressed genes were mainly enriched in the PI3K-AKT signaling pathway, which is regulated by oxidative stress. Moreover, western blotting analysis revealed that SeD and SeE dysregulated PI3K-AKT-mediated apoptosis and cell proliferation signaling, including upregulating (P < 0.05) caspase 3, cleaved-caspase 3, BCL-2 and (or) P53 and downregulating (P < 0.05) PI3K, p-AKT, p-mTOR, 4E-BP1, p-4E-BP1 and (or) p-p70S6K in the testis of mice compared with SeA. Additionally, compared with SeA, both SeD and SeE increased (P < 0.05) GPX3 and SELENOO; SeD decreased (P < 0.05) GPX1, TXRND3 and SELENOW, but SeE increased (P < 0.05) production of three selenoproteins in the testis. Conclusively, both Se deficiency and excess impairs male reproductive system in mice, potentially with the induction of oxidative stress and activation of PI3K/AKT-mediated apoptosis and cell proliferation signaling in the testis.


Asunto(s)
Selenio , Testículo , Masculino , Animales , Ratones , Selenio/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Caspasa 3/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Motilidad Espermática , Semen/metabolismo , Estrés Oxidativo , Apoptosis , Transducción de Señal , Proliferación Celular
9.
Int J Mol Sci ; 23(18)2022 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-36142341

RESUMEN

Oral squamous cell carcinoma (OSCC) is one of the top 15 most prevalent cancers worldwide. However, the current treatment models for OSCC (e.g., surgery, chemotherapy, radiotherapy, and combination therapy) present several limitations: damage to adjacent healthy tissue, possible recurrence, low efficiency, and severe side effects. In this context, nanomaterial-based photothermal therapy (PTT) has attracted extensive research attention. This paper reviews the latest progress in the application of biological nanomaterials for PTT in OSCC. We divide photothermal nanomaterials into four categories (noble metal nanomaterials, carbon-based nanomaterials, metal compounds, and organic nanomaterials) and introduce each category in detail. We also mention in detail the drug delivery systems for PTT of OSCC and briefly summarize the applications of hydrogels, liposomes, and micelles. Finally, we note the challenges faced by the clinical application of PTT nanomaterials and the possibility of further improvement, providing direction for the future research of PTT in OSCC treatment.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Nanoestructuras , Neoplasias , Carbono , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeza y Cuello/terapia , Humanos , Hidrogeles , Liposomas , Micelas , Neoplasias de la Boca/terapia , Nanoestructuras/uso terapéutico , Neoplasias/terapia , Terapia Fototérmica , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia
10.
Lab Invest ; 102(12): 1304-1313, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35882906

RESUMEN

Glioma progression is accompanied with increased tumor tissue stiffness, yet the underlying mechanisms are unclear. Herein, we employed atomic force microscopy analysis to show that tissue stiffness was higher in isocitrate dehydrogenase (IDH)-wild type gliomas than IDH-mutant gliomas. Bioinformatic analyses revealed that tissue inhibitor of metalloproteinase-1 (TIMP1) was one of the preferentially upregulated genes in IDH-wild type gliomas as compared to IDH-mutant gliomas, and its higher expression indicated worse prognosis of glioma patients. TIMP1 intensity determined by immunofluorescence staining on glioma tissues positively correlated with glioma tissue stiffness. Mechanistically, TIMP1 expression was positively correlated with the gene expression of two predominant extracellular matrix components, tenascin C and fibronectin, both of which were also highly expressed in IDH-wild type gliomas. By introducing IDH1-R132H-containing vectors into human IDH1-wild type glioma cells to obtain an IDH1-mutant cell line, we found that IDH1 mutation increased the TIMP1 promoter methylation through methylation-specific PCR. More importantly, IDH1-R132H mutation decreased both the expression of TIMP1, fibronectin, tenascin C, and the tumor tissue stiffness in IDH1-mutant glioma xenografts in contrast to IDH1-wild type counterparts. Moreover, TIMP1 knockdown in IDH-wild type glioma cells inhibited the expression of tenascin C and fibronectin, and decreased tissue stiffness in intracranial glioma xenografts. Conclusively, we revealed an IDH mutation status-mediated mechanism in regulating glioma tissue stiffness through modulating TIMP1 and downstream extracellular matrix components.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Fibronectinas/genética , Neoplasias Encefálicas/metabolismo , Tenascina/genética , Inhibidor Tisular de Metaloproteinasa-1/genética , Glioma/metabolismo , Mutación , Matriz Extracelular/metabolismo
11.
Oncogene ; 41(30): 3791-3803, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35764885

RESUMEN

Glioblastoma is a lethal primary brain tumor with abundant immune-suppressive glioblastoma-associated macrophage (GAM) infiltration. Skewing immune suppressive GAMs towards an immune-activating phenotype represents a promising immunotherapeutic strategy against glioblastoma. Herein, we reported that genetic deletion of miRNA-processing enzyme Dicer in macrophages inhibited the growth of GL261 murine glioblastoma xenografts and prolonged survival of tumor-bearing mice. Single cell RNA sequencing (scRNA-seq) of the tumor-infiltrating immune cells revealed that Dicer deletion in macrophages reduced the proportion of cell-cycling GAM cluster and reprogramed the remaining GAMs towards a proinflammatory activation state (enhanced phagocytotic and IFN-producing signature). Dicer-deficient GAMs showed reduced level of cyclin-dependent kinases (CDK1 and CDK2) and increased expression of CDK inhibitor p27 Kip1, thus manifesting impaired proliferation. Dicer knockout enhanced phagocytotic activity of GAMs to eliminate GL261 tumor cells. Increased proinflammatory GAM clusters in macrophage Dicer-deficient mice actively interacted with tumor-infiltrating T cells and NK cells through TNF paracrine signaling to create a pro-inflammatory immune microenvironment for tumor cell elimination. Our work identifies the role of Dicer deletion in macrophages in generating an immune-activating microenvironment, which could be further developed as a potential immunotherapeutic strategy against glioblastoma.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Neoplasias Encefálicas/patología , Proliferación Celular/genética , Glioblastoma/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Macrófagos/metabolismo , Ratones , Linfocitos T/metabolismo , Microambiente Tumoral/genética
12.
J Exp Clin Cancer Res ; 41(1): 74, 2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35193644

RESUMEN

BACKGROUND: Glioblastoma (GB) is the most common and highly malignant brain tumor characterized by aggressive growth and resistance to alkylating chemotherapy. Autophagy induction is one of the hallmark effects of anti-GB therapies with temozolomide (TMZ). However, the non-classical form of autophagy, autophagy-based unconventional secretion, also called secretory autophagy and its role in regulating the sensitivity of GB to TMZ remains unclear. There is an urgent need to illuminate the mechanism and to develop novel therapeutic targets for GB. METHODS: Cancer genome databases and paired-GB patient samples with or without TMZ treatment were used to assess the relationship between HMGB1 mRNA levels and overall patient survival. The relationship between HMGB1 protein level and TMZ sensitivity was measured by immunohistochemistry, ELISA, Western blot and qRT-PCR. GB cells were engineered to express a chimeric autophagic flux reporter protein consisting of mCherry, GFP and LC3B. The role of secretory autophagy in tumor microenvironment (TME) was analyzed by intracranial implantation of GL261 cells. Coimmunoprecipitation (Co-IP) and Western blotting were performed to test the RAGE-NFκB-NLRP3 inflammasome pathway. RESULTS: The exocytosis of HMGB1 induced by TMZ in GB is dependent on the secretory autophagy. HMGB1 contributed to M1-like polarization of tumor associated macrophages (TAMs) and enhanced the sensitivity of GB cells to TMZ. Mechanistically, RAGE acted as a receptor for HMGB1 in TAMs and through RAGE-NFκB-NLRP3 inflammasome pathway, HMGB1 enhanced M1-like polarization of TAMs. Clinically, the elevated level of HMGB1 in sera may serve as a beneficial therapeutic-predictor for GB patients under TMZ treatment. CONCLUSIONS: We demonstrated that enhanced secretory autophagy in GB facilitates M1-like polarization of TAMs to enhance TMZ sensitivity of GB cells. HMGB1 acts as a key regulator in the crosstalk between GB cells and tumor-suppressive M1-like TAMs in GB microenvironment and may be considered as an adjuvant for the chemotherapeutic agent TMZ.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Glioblastoma/tratamiento farmacológico , Macrófagos/metabolismo , Temozolomida/uso terapéutico , Animales , Antineoplásicos Alquilantes/farmacología , Apoptosis , Autofagia , Línea Celular Tumoral , Glioblastoma/patología , Humanos , Masculino , Ratones , Temozolomida/farmacología , Microambiente Tumoral
13.
Cancer Lett ; 533: 215605, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35219772

RESUMEN

Although the tumorigenic potential of glioma stem cells (GSCs) is associated with multiple molecular alterations, the gene amplification status of GSCs has not been elucidated. Overexpression of HomeoboxA5 (HOXA5) is associated with increased glioma malignancy. In this study, we identify the gene amplification and protein overexpression of HOXA5 in GSCs and its function in regulating GSC maintenance and the downstream transcriptional effector, to explore the significance of HOXA5 amplification/overexpression for GSC identification and prognostic determination. The HOXA5 gene is significantly amplified in glioblastoma (GBM) and is an independent prognostic factor for predicting worse patient outcomes. Specifically, HOXA5 gene amplification and the resultant protein overexpression are correlated with increased proportions of GSCs and enhanced self-renewal/invasiveness of these cells. Disruption of HOXA5 expression impairs GSC survival and GBM tumor propagation. Mechanistically, HOXA5 directly binds to the promoter region of protein tyrosine phosphatase receptor type Z1 (PTPRZ1), thereby upregulating this gene for GSC maintenance. Suppression of PTPRZ1 largely compromises the pro-tumoral effect of HOXA5 on GSCs. In summary, HOXA5 amplification serves as a genetic biomarker for predicting worse GBM outcome, by enhancing PTPRZ1-mediated GSC survival.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/patología , Carcinogénesis/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Glioblastoma/patología , Glioma/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Neoplásicas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo
14.
Oncoimmunology ; 11(1): 2030020, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35096487

RESUMEN

Glioblastoma (GBM) is an aggressive primary brain tumor with unique immunity predominated by myeloid cells. GBM cells have been implicated to evade immune attack through hijacking myeloid-affiliated transcriptional programs to establish an immunosuppressive microenvironment. However, molecular features of immune-evading GBM cells in heterogeneous GBMs and their interactions with immune cells remain unclear. Herein, we employed single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data to develop an in silico method for delineating GBM immune signature and identifying new molecular subsets for immunotherapy. We identified a new GBM cell subset, termed TC-6, that harbored immune-invading signature and actively interacted with tumor-associated macrophages (TAMs) to orchestrate an immune-suppressive niche. Proinflammatory transcriptional factors STAT1, STAT2, IRF1, IRF2, IRF3, and IRF7 were identified as the core regulons defining TC-6 subsets. Further immune transcriptome analyses revealed three immune subtypes (C1, C2, and C3). C3 subtype GBMs were enriched with TC-6 cells and immunosuppressive TAMs, and exhibited an immunomodulatory signature that associated with reduced efficacy of anti-PD-1 treatment. Interferon-related DNA damage resistance signaling was upregulated in C3 GBMs, predicting shortened survival of GBM patients who received chemo-radiation treatment. Treatment of OSI-930 as a molecular agent targeting c-kit and VEGFR2 tyrosine kinases may compromise the immunomodulatory signature of C3 GBMs and synergize with chemo-radiation therapy. We further developed a simplified 11-gene set for defining C3 GBMs. Our work identified TC-6 subset as an immune-evading hub that creates an immunomodulatory signature of C3 GBMs, gaining insights into the heterogeneity of GBM immune microenvironment and holding promise for optimized anti-GBM immunotherapy.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Perfilación de la Expresión Génica , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Inmunoterapia , Microambiente Tumoral/genética
15.
Lab Invest ; 102(7): 722-730, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34963686

RESUMEN

Glioma stem cells (GSCs) are self-renewing tumor cells with multi-lineage differentiation potential and the capacity of construct glioblastoma (GBM) heterogenicity. Mitochondrial morphology is associated with the metabolic plasticity of GBM cells. Previous studies have revealed distinct mitochondrial morphologies and metabolic phenotypes between GSCs and non-stem tumor cells (NSTCs), whereas the molecules regulating mitochondrial dynamics in GBM cells are largely unknown. Herein, we report that carnitine palmitoyltransferase 1A (CPT1A) is preferentially expressed in NSTCs, and governs mitochondrial dynamics and GSC differentiation. Expressions of CPT1A and GSC marker CD133 were mutually exclusive in human GBMs. Overexpression of CPT1A inhibited GSC self-renewal but promoted mitochondrial fusion. In contrast, disruption of CPT1A in NSTCs promoted mitochondrial fission and reprogrammed NSTCs toward GSC feature. Mechanistically, CPT1A overexpression increased the phosphorylation of dynamin-related protein 1 at Ser-637 to promote mitochondrial fusion. In vivo, CPT1A overexpression decreased the percentage of GSCs, impaired GSC-derived xenograft growth and prolonged tumor-bearing mice survival. Our work identified CPT1A as a critical regulator of mitochondrial dynamics and GSC differentiation, indicating that CPT1A could be developed as a molecular target for GBM cell-differentiation strategy.


Asunto(s)
Neoplasias Encefálicas , Carnitina O-Palmitoiltransferasa , Glioblastoma , Glioma , Dinámicas Mitocondriales , Animales , Neoplasias Encefálicas/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular Tumoral , Glioblastoma/metabolismo , Glioma/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/metabolismo
16.
Chemosphere ; 282: 130991, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34090006

RESUMEN

Phthalates are non-persistent chemicals used in products of daily necessities. The evidence on the relationship of prenatal phthalates exposure and preterm birth remain uncertain and dimed. We Searched Cochrane, EMBASE, PubMed and Qvid Medline and selected studies according to a priori defined inclusion criteria. A total of 20 relevant studies published before March 31, 2020, were included. The main methods to detect heterogeneity and publication bias of included studies were the Q-statistic and Begg's test. Overall summary estimates indicated positive association of prenatal exposure to di(2-ethylhexyl) phthalate [number of studies (n) = 11, odds ratio (OR) = 1.1; 95% confidence interval (CI): 0.89, 1.4], diethyl phthalate [n = 11, OR = 1.1; 95% CI: 0.92, 1.3], dibutyl phthalate [n = 10, OR = 1.1; 95% CI: 0.77, 1.4], butyl benzyl phthalate [n = 10, OR = 1.0; 95% CI: 0.91, 1.2], diisobutyl phthalate [n = 9, OR = 1.1; 95% CI: 0.92, 1.3], diisononyl phthalate [n = 4, OR = 1.1; 95% CI: 0.85, 1.3], and phthalates were negatively associated with gestational age except di(2-ethylhexyl) phthalate and diisononyl phthalate. Most of them were not statistically significant. Subgroup analysis indicated that the heterogeneity was mainly found in the studies conducted in China and matrix collected at first trimester of pregnancy. Substantial heterogeneity and inconsistency of research methods may lead to inconsistent results of maternal phthalates exposure with preterm birth. We recommend a multicenter cohort study with a consistent approach to unravel the complex associations of prenatal phthalates exposure with birth outcomes.


Asunto(s)
Ácidos Ftálicos , Nacimiento Prematuro , Efectos Tardíos de la Exposición Prenatal , Estudios de Cohortes , Femenino , Edad Gestacional , Humanos , Recién Nacido , Estudios Multicéntricos como Asunto , Ácidos Ftálicos/toxicidad , Embarazo , Nacimiento Prematuro/inducido químicamente , Nacimiento Prematuro/epidemiología , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/epidemiología
17.
Environ Int ; 153: 106538, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33839551

RESUMEN

BACKGROUND: Exposure to multiple metals is recognized as a common and real scenario in daily life. However, limited prospective studies have assessed associations between multiple metals exposure and hypertension. METHODS: In total, 2625 adults in a local area on the Yangtze River were investigated at baseline from 2014 to 2015 and followed up in 2019. We measured baseline urine levels of 22 metals and used multivariate logistic analysis and Bayesian kernel machine regression (BKMR) to explore associations between multiple metals exposure and the risk of hypertension. RESULTS: A total of 385 individuals (29.6%) were diagnosed with hypertension. Five metals (cadmium, copper, magnesium, molybdenum and zinc) were positively associated with hypertension in single-metal models. Cadmium and zinc remained significantly positive associations after adjusting for these five metals, with the odds ratio (OR) in the highest quartiles of 1.49 (95% CI: 1.01, 2.21; p-trend = 0.05) and 1.60 (95% CI: 1.08, 2.38; p-trend = 0.02), respectively. BKMR analysis showed a significant joint effect of multiple metals on hypertension when the concentrations of five metals were at or above their 55th percentile compared with their median values. A potential interaction between cadmium and zinc in increasing the risk of hypertension was observed with the ORint of 1.41 (95%CI: 1.05, 1.89). CONCLUSIONS: We identified the joint effect of multiple metals on hypertension and observed a significant interaction between cadmium and zinc. Further cohort studies are needed to clarify the health effects of multiple metals exposure in a larger population.


Asunto(s)
Hipertensión , Ríos , Adulto , Teorema de Bayes , China/epidemiología , Humanos , Hipertensión/inducido químicamente , Hipertensión/epidemiología , Estudios Prospectivos
18.
Gen Physiol Biophys ; 40(1): 17-29, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33655888

RESUMEN

Neuroblastoma (NB) is an extracranial solid malignancy in childhood. More and more studies have demonstrated that circRNAs are essential regulators of various tumors. This study conducted to explore the role and mechanism of circular RNA CUT-like homeobox 1 (circCUX1) in NB. The levels of circCUX1, miR-338-3p and plant homeodomain finger protein 20 (PHF20) were detected by qRT-PCR or Western blot. Cell proliferation and apoptosis were evaluated by colony formation assay, flow cytometry and Western blot analysis. Cell migration and invasion were examined via transwell assay. Glycolysis was expressed by measuring the extracellular acidification rate (ECAR). The interaction among circCUX1, miR-338-3p and PHF20 were validated by dual-luciferase reporter assay and RNA Immunoprecipitation assay. Besides, xenograft experiment was performed to assess tumor growth in vivo. circCUX1 and PHF20 were up-regulated, while miR-338-3p was down-regulated in NB tissues and cells. Knockdown of circCUX1 suppressed the progression and glycolysis of NB cells. circCUX1 triggered NB progression and glycolysis by regulating miR-338-3p. Additionally, down-regulation of miR-338-3p promoted NB progression and glycolysis via targeting PHF20. Moreover, circCUX1 sponged miR-338-3p to regulate PHF20 expression. Furthermore, circCUX1 silencing hindered tumor growth in vivo. circCUX1 depletion suppressed tumor progression and glycolysis in NB by regulating miR-338-3p/PHF20 axis, suggesting a potential biomarker for NB treatment.


Asunto(s)
MicroARNs , Neuroblastoma , Línea Celular Tumoral , Niño , Proteínas de Unión al ADN , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Neuroblastoma/genética , Factores de Transcripción
19.
J Cancer ; 12(3): 818-826, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33403039

RESUMEN

Glioma cells with stem cell-like properties are crucial for tumor initiation, progression and therapeutic resistance. Therefore, identifying specific factors in regulating stem-like traits is critical for the design of novel glioma therapeutics. Herein, we reported that ADP-Ribosylation Factor Like GTPase 4C (ARL4C) was highly expressed in glioma stem-like cells (GSLCs). GSLCs, determined by the efficiency of sphere formation in vitro and tumor growth in vivo, was increased by overexpression of ARL4C. ARL4C induced the tumorigenesis through ALDH1A3. Analyses of 325 patient specimens showed that ARL4C was highly expressed in glioblastoma (GBM) as compared with lower grade gliomas. In addition, higher level ARL4C expression in glioma was correlated with poorer progression-free survival and overall survival of patients. Therefore, ARL4C may act as a novel prognostic marker and a therapeutic target for GBM.

20.
Cancer Manag Res ; 12: 7635-7647, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32922073

RESUMEN

BACKGROUND: Hypoxia has been shown to induce the malignant progression of cancer, including non-small cell lung cancer (NSCLC). Circular RNA (circRNA) is considered to be an important regulator of cancer progression. However, the role of a newly discovered circRNA, circ_0000376, in the progression of NSCLC is unclear. METHODS: The relative expression levels of circ_0000376, miR-1182 and neuro-oncological ventral antigen 2 (NOVA2) were detected via quantitative real-time polymerase chain reaction (qRT-PCR). Glucose consumption and lactate production were determined using Glucose Assay Kit and Lactate Assay Kit, respectively. Moreover, the protein levels of glycolysis markers and NOVA2 were measured using Western blot (WB) analysis. Furthermore, 3-(4, 5-dimethyl-2 thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay was performed to assess cell viability, and transwell assay was employed to evaluate cell migration and invasion. The interaction between miR-1182 and circ_0000376 or NOVA2 was confirmed by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. In addition, animal experiments were conducted to assess the influence of circ_0000376 silencing on NSCLC tumor growth in vivo. RESULTS: Circ_0000376 was upregulated in NSCLC, and its high expression was related to the poor overall survival of NSCLC patients. Hypoxia could enhance circ_0000376 expression and promote the glycolysis, viability, migration, and invasion of NSCLC cells. However, silencing of circ_0000376 could inhibit the glycolysis, viability, migration, and invasion of hypoxia-induced NSCLC cells. Additionally, circ_0000376 could sponge miR-1182, and miR-1182 could target NOVA2. MiR-1182 silencing could reverse the inhibitory effect of circ_0000376 knockdown on NSCLC progression, and NOVA2 overexpression also could reverse the suppressive effect of miR-1182 overexpression on NSCLC progression. Meanwhile, miR-1182 inhibitor could invert the negative regulation effect of circ_0000376 silencing on NOVA2 expression. In addition, circ_0000376 knockdown inhibited the NSCLC tumor growth via regulating the miR-1182 and NOVA2 expression in vivo. CONCLUSION: Circ_0000376 promoted NSCLC progression by regulating the miR-1182/NOVA2 axis, suggesting that circ_0000376 might be a potential biomarker for NSCLC treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA