Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Biomater Sci ; 12(15): 3826-3840, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38758027

RESUMEN

Due to the immunosuppressive tumor microenvironment (TME) and potential systemic toxicity, chemotherapy often fails to elicit satisfactory anti-tumor responses, so how to activate anti-tumor immunity to improve the therapeutic efficacy remains a challenging problem. Photothermal therapy (PTT) serves as a promising approach to activate anti-tumor immunity by inducing the release of tumor neoantigens in situ. In this study, we designed tetrasulfide bonded mesoporous silicon nanoparticles (MSNs) loaded with the traditional drug doxorubicin (DOX) inside and modified their outer layer with polydopamine (DOX/MSN-4S@PDA) for comprehensive anti-tumor studies in vivo and in vitro. The MSN core contains GSH-sensitive tetrasulfide bonds that enhance DOX release while generating hydrogen sulfide (H2S) to improve the therapeutic efficacy of DOX. The polydopamine (PDA) coating confers acid sensitivity and mild photothermal properties upon exposure to near-infrared (NIR) light, while the addition of hyaluronic acid (HA) to the outermost layer enables targeted delivery to CD44-expressing tumor cells, thereby enhancing drug accumulation at the tumor site and reducing toxic side effects. Our studies demonstrate that DOX/MSN@PDA-HA can reverse the immunosuppressive tumor microenvironment in vivo, inducing potent immunogenic cell death (ICD) of tumor cells and improving anti-tumor efficacy. In addition, DOX/MSN@PDA-HA significantly suppresses tumor metastasis to the lung and liver. In summary, DOX/MSN@PDA-HA exhibits controlled drug release, excellent biocompatibility, and remarkable tumor inhibition capabilities through synergistic chemical/photothermal combined therapy.


Asunto(s)
Doxorrubicina , Indoles , Nanopartículas , Terapia Fototérmica , Polímeros , Silicio , Silicio/química , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/administración & dosificación , Animales , Ratones , Indoles/química , Indoles/farmacología , Indoles/administración & dosificación , Porosidad , Nanopartículas/química , Nanopartículas/administración & dosificación , Polímeros/química , Línea Celular Tumoral , Microambiente Tumoral/efectos de los fármacos , Humanos , Liberación de Fármacos , Portadores de Fármacos/química , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Femenino , Terapia Combinada , Ratones Endogámicos BALB C
2.
Neoplasia ; 49: 100973, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38277817

RESUMEN

F-box only protein 38 (FBXO38) is a member of the F-box family that mediates the ubiquitination and proteasome degradation of programmed death 1 (PD-1), and thus has important effects on T cell-related immunity. While its powerful role in adaptive immunity has attracted much attention, its regulatory roles in innate immune pathways remain unknown. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is an important innate immune pathway that regulates type I interferons. STING protein is the core component of this pathway. In this study, we identified that FBXO38 deficiency enhanced tumor proliferation and reduced tumor CD8+ T cells infiltration. Loss of FBXO38 resulted in reduced STING protein levels in vitro and in vivo, further leading to preventing cGAS-STING pathway activation, and decreased downstream product IFNA1 and CCL5. The mechanism of reduced STING protein was associated with lysosome-mediated degradation rather than proteasomal function. Our results demonstrate a critical role for FBXO38 in the cGAS-STING pathway.


Asunto(s)
Neoplasias , Transducción de Señal , Humanos , Linfocitos T CD8-positivos/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Lisosomas/metabolismo , Inmunidad Innata
3.
Eur Radiol ; 34(7): 4516-4526, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38112763

RESUMEN

OBJECTIVES: To investigate the pathological interplay between immunity and the visual processing system (VPS) in thyroid eye disease (TED). METHODS: A total of 24 active patients (AP), 26 inactive patients (IP) of TED, and 27 healthy controls (HCs) were enrolled. Orbital magnetic resonance imaging (MRI) and resting-state functional MRI (rs-fMRI) were conducted for each participant. Multiple MRI parameters of the intraorbital optic nerve (ON) were assessed. The amplitude of low-frequency fluctuations (ALFF) and regional homogeneity (ReHo) were calculated. Correlation analyses were carried out on the above parameters and clinical characteristics. RESULTS: Visual functioning scores differentiated between the AP and IP groups. The ON subarachnoid space and ON sheath diameter were significantly higher in AP than in IP. Six vision-related brain regions were identified in TED patients compared with HCs, including right calcarine (CAL.R), right cuneus (CUN.R), left postcentral gyrus (PoCG.L), right middle temporal gyrus (MTG.R), left superior frontal gyrus (SFG.L), and left caudate (CAU.L). The brain activity of MTG.R, SFG.L, and CAU.L differentiated between the AP and IP groups. The correlation analysis revealed a close association among the vision-related brain regions, MRI parameters of ON, and clinical characteristics in AP and IP, respectively. CONCLUSIONS: Combined orbital and brain neuroimaging revealed abnormalities of the VPS in TED, which had a close correlation with immune statuses. Vision-related brain regions in TED might be possibly altered by peripheral immunity via a direct or indirect approach. CLINICAL RELEVANCE STATEMENT: The discovery of this study explained the disparity of visual dysfunction in TED patients with different immune statuses. With the uncovered neuroimaging markers, early detection and intervention of visual dysfunction could be achieved and potentially benefit TED patients. KEY POINTS: • Patients with different immune statuses of thyroid eye disease varied in the presentation of visual dysfunction. • The combined orbital and brain neuroimaging study identified six altered vision-related brain regions, which had a significant correlation with the MRI parameters of the intraorbital optic nerve and immunological characteristics. • Peripheral immunity might possibly give rise to alterations in the central nervous system part of the visual processing system via a direct or indirect approach.


Asunto(s)
Oftalmopatía de Graves , Imagen por Resonancia Magnética , Neuroimagen , Humanos , Masculino , Femenino , Imagen por Resonancia Magnética/métodos , Oftalmopatía de Graves/diagnóstico por imagen , Oftalmopatía de Graves/complicaciones , Oftalmopatía de Graves/inmunología , Persona de Mediana Edad , Adulto , Neuroimagen/métodos , Encéfalo/diagnóstico por imagen , Trastornos de la Visión/diagnóstico por imagen , Trastornos de la Visión/etiología , Trastornos de la Visión/fisiopatología , Estudios de Casos y Controles , Nervio Óptico/diagnóstico por imagen , Nervio Óptico/patología , Órbita/diagnóstico por imagen
4.
Exp Eye Res ; 230: 109436, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36914000

RESUMEN

Thyroid-associated ophthalmopathy (TAO), also known as Graves' ophthalmopathy, is an autoimmune disease that is usually accompanied by hyperthyroidism. Its pathogenesis involves the activation of autoimmune T lymphocytes by a cross-antigen reaction of thyroid and orbital tissues. The thyroid-stimulating hormone receptor (TSHR) is known to play an important role in the development of TAO. Because of the difficulty of orbital tissue biopsy, the establishment of an ideal animal model is important for developing novel clinical therapies of TAO. To date, TAO animal modeling methods are mainly based on inducing experimental animals to produce anti-thyroid-stimulating hormone receptor antibodies (TRAbs) and then recruit autoimmune T lymphocytes. Currently, the most common methods are hTSHR-A subunit plasmid electroporation and hTSHR-A subunit adenovirus transfection. These animal models provide a powerful tool for exploring the internal relationship between local and systemic immune microenvironment disorders of the TAO orbit, facilitating the development of new drugs. However, existing TAO modeling methods still have some defects, such as low modeling rate, long modeling cycles, low repetition rate, and considerable differences from human histology. Hence, the modeling methods require further innovation, improvement, and in-depth exploration.


Asunto(s)
Enfermedades Autoinmunes , Oftalmopatía de Graves , Animales , Humanos , Oftalmopatía de Graves/patología , Órbita/patología , Receptores de Tirotropina , Modelos Animales de Enfermedad , Hormonas
5.
Mol Med Rep ; 27(5)2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36960863

RESUMEN

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that, for the Transwell invasion assay experiments with the SK­MES­1 cell line shown in Fig. 4A on p. 1748, the 'mimic'NC' and 'inhibitor­NC' data panels showed overlapping sections, such that these data may have been derived from the same original source even though they were intending to show the results of different experiments. The authors have consulted their original data, and realize that the 'inhibitor­NC' data panel was inadvertently selected incorrectly for Fig. 4A. The revised version of Fig. 4, showing the correct data for the 'inhibitor­NC' experiment, is shown on the next page. Note that the error made during the assembly of Fig. 4 did not significantly affect either the results or the conclusions reported in this paper, and all the authors agree to this Corrigendum. The authors are grateful to the Editor of Molecular Medicine Reports for allowing them the opportunity to publish this corrigendum, and apologize to the readership for any inconvenience caused. [Molecular Medicine Reports 17: 1742­1752, 2018; DOI: 10.3892/mmr.2017.8050].

6.
J Nanobiotechnology ; 21(1): 20, 2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36658649

RESUMEN

The slightest change in the extra/intracellular concentration of metal ions results in amplified effects by signaling cascades that regulate both cell fate within the tumor microenvironment and immune status, which influences the network of antitumor immunity through various pathways. Based on the fact that metal ions influence the fate of cancer cells and participate in both innate and adaptive immunity, they are widely applied in antitumor therapy as immune modulators. Moreover, nanomedicine possesses the advantage of precise delivery and responsive release, which can perfectly remedy the drawbacks of metal ions, such as low target selectivity and systematic toxicity, thus providing an ideal platform for metal ion application in cancer treatment. Emerging evidence has shown that immunotherapy applied with nanometallic materials may significantly enhance therapeutic efficacy. Here, we focus on the physiopathology of metal ions in tumorigenesis and discuss several breakthroughs regarding the use of nanometallic materials in antitumor immunotherapeutics. These findings demonstrate the prominence of metal ion-based nanomedicine in cancer therapy and prophylaxis, providing many new ideas for basic immunity research and clinical application. Consequently, we provide innovative insights into the comprehensive understanding of the application of metal ions combined with nanomedicine in cancer immunotherapy in the past few years.


Asunto(s)
Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Metales/uso terapéutico , Inmunoterapia/métodos , Transducción de Señal , Iones , Nanomedicina/métodos , Microambiente Tumoral
7.
Front Oncol ; 11: 620924, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34249677

RESUMEN

BACKGROUND: Brain metastasis is extremely rare but predicts dismal prognosis in papillary thyroid cancer (PTC). Dynamic evaluation of stepwise metastatic lesions was barely conducted to identify the longitudinal genomic evolution of brain metastasis in PTC. METHOD: Chronologically resected specimen was analyzed by whole exome sequencing, including four metastatic lymph nodes (lyn 1-4) and brain metastasis lesion (BM). Phylogenetic tree was reconstructed to infer the metastatic pattern and the potential functional mutations. RESULTS: Contrasting with lyn1, ipsilateral metastatic lesions (lyn2-4 and BM) with shared biallelic mutations of TSC2 indicated different genetic originations from multifocal tumors. Lyn 3/4, particularly lyn4 exhibited high genetic similarity with BM. Besides the similar mutational compositions and signatures, shared functional mutations (CDK4 R24C , TP53R342*) were observed in lyn3/4 and BM. Frequencies of these mutations gradually increase along with the metastasis progression. Consistently, TP53 knockout and CDK4 R24C introduction in PTC cells significantly decreased radioiodine uptake and increased metastatic ability. CONCLUSION: Genomic mutations in CDK4 and TP53 during the tumor evolution may contribute to the lymph node and brain metastasis of PTC.

8.
Mol Med Rep ; 17(1): 1742-1752, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29138830

RESUMEN

SRY-box 9 (SOX9) is an important transcription factor required for development, which has additionally been reported to be an independent prognostic indicator for the survival of patients with non­small cell lung cancer (NSCLC). Accumulating evidence has indicated that dysregulation of microRNAs (miRNAs/miRs) may contribute to the initiation and progression of cancer. SOX9 may be regulated by a number of miRNAs in different types of cancer, including in NSCLC. The present study sought to identify novel candidate miRNAs associated with SOX9 in NSCLC using online tools, and investigated the detailed functions of miR­185, which suppressed SOX9 mRNA expression most strongly out of the candidate miRNAs. It was observed that ectopic miR­185 expression significantly suppressed NSCLC cell proliferation, invasion and migration. Using luciferase reporter gene and RNA immunoprecipitation assays, SOX9 was confirmed to be a direct target of miR­185. In addition, the downstream Wnt signaling­associated factors ß­catenin and c­Myc proto­oncogene protein (Myc) were demonstrated to be inhibited by miR­185 overexpression. SOX9, ß­catenin and c­Myc mRNA expression was significantly upregulated in NSCLC tissues, and was inversely correlated with miR­185 expression. The results of the present study demonstrated that rescuing miR­185 expression in NSCLC, thereby inhibiting SOX9 expression and the downstream Wnt signaling, and leading to the suppression of NSCLC cell proliferation, invasion and migration, may be a promising strategy for the treatment of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Factor de Transcripción SOX9/genética , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Interferencia de ARN , Factor de Transcripción SOX9/metabolismo , Vía de Señalización Wnt
9.
Haematologica ; 103(3): 406-416, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29269522

RESUMEN

The microRNAs miR-144 and -451 are encoded by a bicistronic gene that is strongly induced during red blood cell formation (erythropoiesis). Ablation of the miR-144/451 gene in mice causes mild anemia under baseline conditions. Here we show that miR-144/451-/- erythroblasts exhibit increased apoptosis during recovery from acute anemia. Mechanistically, miR-144/451 depletion increases the expression of the miR-451 target mRNA Cab39, which encodes a co-factor for the serine-threonine kinase LKB1. During erythropoietic stress, miR-144/451-/- erythroblasts exhibit abnormally increased Cab39 protein, which activates LKB1 and its downstream AMPK/mTOR effector pathway. Suppression of this pathway via drugs or shRNAs enhances survival of the mutant erythroblasts. Thus, miR-144/451 facilitates recovery from acute anemia by repressing Cab39/AMPK/mTOR. Our findings suggest that miR-144/451 is a key protector of erythroblasts during pathological states associated with dramatically increased erythropoietic demand, including acute blood loss and hemolytic anemia.


Asunto(s)
Anemia/sangre , Células Eritroides/citología , MicroARNs/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Enfermedad Aguda , Animales , Proteínas de Unión al Calcio/genética , Supervivencia Celular , Eritropoyesis , Redes y Vías Metabólicas , Ratones , ARN Mensajero , Serina-Treonina Quinasas TOR/metabolismo
10.
Oncol Lett ; 13(6): 4526-4532, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28599453

RESUMEN

Ultraconserved regions (UCRs) are non-protein-coding gene sequences that are strictly conserved across numerous distinct species. It has been demonstrated previously that UCRs encoding non-coding RNAs serve as regulators of gene expression. In recent decades, there has been increasing evidence for the involvement of UCRs in carcinogenesis. In previous studies, the non-coding RNA transcribed ultraconserved element 338 (TUC338) was identified to serve an oncogenic role in hepatocellular cancer; however, thus far, the role of TUC338 in cervical cancer (CC) remains undefined. The results of the present study revealed that TUC338 is significantly upregulated in CC tissues and cell lines, and that the upregulation of TUC338 is associated with lymph node metastasis. Transfection with small interfering RNA (siRNA) against TUC338 could markedly inhibit cell migration and invasion in HeLa and C33A CC cell lines. Using a dual-luciferase reporter assay, tissue inhibitor of metalloproteinase 1 (TIMP1) was demonstrated to be negatively regulated by TUC338 at the post-transcriptional level, via a specific target site within the 3' untranslated region. The expression of TIMP1 was also observed to be inversely associated with TUC338 expression in CC tissues. Overexpression of TIMP1 with MigRI-TIMP1-green fluorescent protein inhibited CC cell migration and invasion and downregulated matrix metalloproteinase 9, resembling the effects of TUC338 siRNA. Therefore, the results of the present study suggest that TUC338 acts as a novel oncogene by targeting the TIMP1 gene, and inhibiting CC cell migration and invasion.

11.
Oncotarget ; 8(63): 107109-107124, 2017 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-29291015

RESUMEN

One fundamental issue in public health is the safety of food products derived from plants and animals. A recent study raised a concern that microRNAs, which widely exist in everyday foods, may alter consumers' functions. However, some studies have strongly questioned the likelihood of dietary uptake of functional microRNAs in mammals. Here we use a microRNA gene knockout animal model to show that miR-144/451 null mice can orally uptake miR-451 from a daily chow diet, and ingestion of wild type blood, that contains abundant miR-451, also enhances the level of miR-451 in the circulating blood of knockout mice. Moreover, reducing miR-451 level in miR-144/451 knockout blood by consuming food lacking miR-451 reduces the anti-oxidant capacity of miR-144/451 null red blood cells by targeting the 14-3-3ζ/Foxo3 pathway, while increasing miR-451 level via gavage-feeding of wild type blood increases the anti-oxidant capacity of miR-144/451 null red blood cells. We conclude that 1) some miRNAs in food can pass through the gastrointestinal tract into the blood to affect consumers' function and 2) microRNA knockout animals such as miR-144/451 null mice can acquire the deleted genetic information from daily foods, which might alter the results and conclusions from the studies using such animals.

12.
Brain Res ; 1650: 232-242, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27637156

RESUMEN

Post-stroke cognitive impairment (PSCI), commonly seen in the clinical practice, is a major factor impeding patient rehabilitation. Enriched environment (EE) intervention is a simple and effective way to improve cognitive impairment, partially due to the rebalancing of the basal forebrain-hippocampus cholinergic signaling pathway. Epigenetic changes have been identified in many cognitive disorders. However, studies on the effects of EE on epigenetic regulation of cholinergic circuits in PSCI animal models have not yet been reported. In this study, we established a photothrombotic mouse PSCI model and showed that after EE intervention, mice with PSCI had significantly improved water maze performance, better induction of hippocampal long-term potentiation (LTP), enhanced function of the basal forebrain-hippocampus cholinergic circuits of contralateral side of stroke and relatively balanced acetylation homeostasis compared to those of PSCI mice in standard environments (SE). In addition, PSCI mice in EE expressed much higher levels of p-CREB and CBP than in SE, and the chromatins bound to M-type promoter of ChAT gene were more acetylated. These results demonstrate that EE plays an important role in the improvement of PSCI and the underlying mechanism may involve in the acetylation of histones bound to the ChAT gene promoter in cholinergic circuits.


Asunto(s)
Neuronas Colinérgicas/fisiología , Disfunción Cognitiva/terapia , Accidente Cerebrovascular/terapia , Acetilación , Acetilcolina/metabolismo , Animales , Colinérgicos/uso terapéutico , Cromatina , Trastornos del Conocimiento/metabolismo , Modelos Animales de Enfermedad , Ambiente , Epigénesis Genética , Hipocampo/metabolismo , Histonas/metabolismo , Homeostasis/fisiología , Potenciación a Largo Plazo , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Accidente Cerebrovascular/metabolismo
13.
Dev Growth Differ ; 57(6): 466-473, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26098172

RESUMEN

Long noncoding RNAs (LncRNAs) are longer than 200 nucleotide noncoding RNAs without apparent functional coding capacity that function as regulators of cell growth and development. In recent years, increasing evidence implicates the involvement of LncRNAs in erythropoiesis. shlnc-EC6 is a LncRNA associated with erythroid differentiation but the mechanism remains undefined. In this study, we found that knockdown of shlnc-EC6 in purified mouse fetal liver erythroid progenitor and hematopoietic stem cells (FLEPHSCs) significantly blocked erythroid enucleation. We also showed that Rac1 was negatively regulated by shlnc-EC6 at the posttranscriptional level via specific binding to sites within the 3'UTR of Rac1 mRNA. Moreover, we found that knockdown of shlnc-EC6 led to upregulation of Rac1, followed by the activation of the downstream protein PIP5K, and subsequently resulted in the inhibition of enucleation in cultured mouse fetal erythroblasts. Thus, our findings suggest that shlnc-EC6 acts as a novel modulator to regulate mouse erythropoiesis via Rac1/PIP5K signaling pathway.


Asunto(s)
Núcleo Celular/genética , Células Eritroides/metabolismo , Células Precursoras Eritroides/fisiología , Eritropoyesis/genética , ARN Largo no Codificante/fisiología , ARN Interferente Pequeño/fisiología , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Embrión de Mamíferos , Eritrocitos/fisiología , Células Eritroides/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , ARN Largo no Codificante/farmacología , ARN Interferente Pequeño/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA