Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Base de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PeerJ ; 12: e16911, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38371373

RESUMEN

Objective: E2F transcription factors are associated with tumor development, but their underlying mechanisms in gastric cancer (GC) remain unclear. This study explored whether E2Fs determine the prognosis or immune and therapy responses of GC patients. Methods: E2F regulation patterns from The Cancer Genome Atlas (TCGA) were systematically investigated and E2F patterns were correlated with the characteristics of cellular infiltration in the tumor microenvironment (TME). A principal component analysis was used to construct an E2F scoring model based on prognosis-related differential genes to quantify the E2F regulation of a single tumor. This scoring model was then tested in patient cohorts to predict effects of immunotherapy. Results: Based on the expression profiles of E2F transcription factors in GC, two different regulatory patterns of E2F were identified. TME and survival differences emerged between the two clusters. Lower survival rates in the Cluster2 group were attributed to limited immune function due to stromal activation. The E2F scoring model was then constructed based on the E2F-related prognostic genes. Evidence supported the E2F score as an independent and effective prognostic factor and predictor of immunotherapy response. A gene-set analysis correlated E2F score with the characteristics of immune cell infiltration within the TME. The immunotherapy cohort database showed that patients with a higher E2F score demonstrated better survival and immune responses. Conclusions: This study found that differences in GC prognosis might be related to the E2F patterns in the TME. The E2F scoring system developed in this study has practical value as a predictor of survival and treatment response in GC patients.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Microambiente Tumoral/genética , Inmunoterapia , Bases de Datos Factuales , Factores de Transcripción E2F
2.
Antioxidants (Basel) ; 13(2)2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38397811

RESUMEN

Ulcerative colitis is an inflammatory bowel disease with multiple pathogeneses. Here, we aimed to study the therapeutic role of ulinastatin (UTI), an anti-inflammatory bioagent, and its associated mechanisms in treating colitis. Dextran sulfate sodium was administrated to induce colitis in mice, and a subgroup of colitis mice was treated with UTI. The gut barrier defect and inflammatory manifestations of colitis were determined via histological and molecular experiments. In addition, transcriptomics, metagenomics, and metabolomics were employed to explore the possible mechanisms underlying the effects of UTI. We found that UTI significantly alleviated the inflammatory manifestations and intestinal barrier damage in the mice with colitis. Transcriptome sequencing revealed a correlation between the UTI treatment and JAK-STAT signaling pathway. UTI up-regulated the expression of SOCS1, which subsequently inhibited the phosphorylation of JAK2 and STAT3, thus limiting the action of inflammatory mediators. In addition, 16S rRNA sequencing illustrated that UTI maintained a more stable intestinal flora, protecting the gut from dysbiosis in colitis. Moreover, metabolomics analysis demonstrated that UTI indeed facilitated the production of some bile acids and short-chain fatty acids, which supported intestinal homeostasis. Our data provide evidence that UTI is effective in treating colitis and support the potential use of UTI treatment for patients with ulcerative colitis.

3.
J Cancer ; 15(3): 809-824, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38213725

RESUMEN

Background: Colorectal cancer (CRC) is one of the most common malignant tumors and has high morbidity and mortality rates. Previous studies have shown that TSPEAR mutations are involved in the development and progression of gastric cancer and liver cancer. However, the role of TSPEAR in CRC is still unclear. Methods: In The Cancer Genome Atlas (TCGA) database, 590 CRC patients with complete survival information were analyzed. We assessed TSPEAR expression in a pan-cancer dataset from the TCGA database. Cox regression analysis was performed to evaluate factors associated with prognosis. Enrichment analysis via the R package "clusterProfiler" was used to explore the potential function of TSPEAR. The single-sample GSEA (ssGSEA) method from the R package "GSVA" and the TIMER database were used to investigate the association between the immune infiltration level and TSPEAR expression in CRC. The R package "maftools" was used to explore the association between tumour mutation burden (TMB) and TSPEAR expression in CRC. CCK-8 assays and cell invasion assays were used to detect the effect of TSPEAR and TGIF2 on the biological behavior of CRC cells. Results: Pan-cancer analysis revealed that TSPEAR was upregulated in CRC tissues compared to normal tissues and that high TSPEAR expression was associated with poorer overall survival (OS) (p=0.0053). The expression of TSPEAR increased with increasing TNM stage, T stage, N stage, and M stage. The nomogram constructed with TSPEAR, age, and TNM stage showed better predictive value than TSPEAR, age, or TNM stage alone. Immune cell infiltration analysis revealed that high expression of TSPEAR was associated with lower immune cell infiltration. Tumor mutation burden (TMB) analysis indicated that high expression of TSPEAR was associated with lower TMB (p=0.005), and high TMB was associated with shorter OS (p=0.02). CCK-8 assays and cell invasion assays indicated that in vitro knockdown of TSPEAR inhibited the proliferation, migration, and invasion of CRC cells. In addition, TSPEAR expression may be regulated by the upstream transcription factor TGIF2. Conclusion: TSPEAR expression was higher in CRC tissues than in normal tissues. Its upregulation was significantly associated with a poor prognosis. Additionally, TSPEAR plays a significant role in tumor immunity and the biological behavior of CRC cells. Thus, TSPEAR may become a promising prognostic biomarker and therapeutic target for CRC patients.

4.
Clin Transl Oncol ; 26(3): 630-643, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37480430

RESUMEN

PURPOSE: Metabolic reprogramming is a novel hallmark and therapeutic target of cancer. Our study aimed to establish fatty acid metabolism-associated scores based on gene signature and investigated its effects on immunotherapy in colon cancer. METHODS: Gene expression and clinical information were collected from Gene Expression Omnibus (GEO) database to identify a gene signature by non-negative matrix factorization (NMF) clustering and Cox regression analysis. Subsequently, we constructed the fatty acid metabolism score (FA-score) model by principal component analysis (PCA) and explored its relativity of prognosis and the response to immunotherapy in colon cancer. Finally, the Cancer Genome Atlas (TCGA) database was introduced and in vitro study was performed for verification. RESULTS: The FA-score-high group had a higher level of fatty acid metabolism and was associated with worse patient overall survival. Significantly, FA-score correlated closely with the biomarkers of immunotherapy, and the FA-score-high group had a poorer therapeutic efficacy of immune checkpoint blockade. In vitro experiments demonstrated that ACSL5 may be a critical metabolic regulatory target. CONCLUSIONS: Our study provided a comprehensive analysis of the heterogeneity of fatty acid metabolism in colon cancer. We highlighted the potential clinical utility of fatty acid metabolism-related genes to be biomarkers of colon cancer prognosis and targets to improve the effect of immunotherapy.


Asunto(s)
Neoplasias del Colon , Humanos , Pronóstico , Neoplasias del Colon/genética , Neoplasias del Colon/terapia , Inmunoterapia , Biomarcadores , Ácidos Grasos
5.
Front Oncol ; 13: 1234045, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37564935

RESUMEN

Background: As the most common gastrointestinal malignancy worldwide, liver metastases occur in half colorectal cancer (CRC) patients. Early detection can help treat them early and reduce mortality in patients with colorectal cancer liver metastases (CRLM). Finding useful biomarkers for CRLM is thus essential. Methods: The TCGA and GEO databases were used to download the expression profiles and clinical data of the patients. Differential analysis screened for genes associated with CRLM, and univariate Cox regression analysis identified genes associated with prognosis. The least absolute shrinkage and selection operator (LASSO) method further preferred genes to construct a prognostic signature. Kaplan-Meier survival curves were used to show patients' overall survival (OS). Receiver operating characteristic (ROC) curves showed the accuracy of the model. Risk scores and clinical characteristics of patients were included in multivariate Cox regression analysis to identify independent risk factors, and a nomogram was constructed. The proportion of immune cells and infiltration were assessed using the 'CIBERSORT' package and the 'ESTIMATE' package. Results: We constructed a signature consisting of seven CRLM-associated genes, and signature-based risk scores have great potential in estimating the prognosis of CRC patients. Moreover, the poor response to immunotherapy in high-risk patients might contribute to the poor prognosis of individuals. Furthermore, we found that overexpression of Hepcidin antimicrobial peptide (HAMP), the only gene highly expressed in CRC and liver metastatic tissues, promoted CRC development and that it was associated with tumor mutation burden (TMB), DNA mismatch repair (MMR) genes, and microsatellite instability (MSI) in various tumors. Finally, we found that in CRC patients, low expression of HAMP also represented a better immunotherapeutic outcome, reflecting the critical role of HAMP in guiding immunotherapy. Conclusion: We identified a prognostic signature containing 7 CRLM-associated genes, and the signature was specified as an independent predictor and a nomogram containing the risk score was built accordingly. In addition, the derived gene HAMP could help guide the exploration of profitable immunotherapeutic strategies.

6.
Front Oncol ; 13: 1190229, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37223685

RESUMEN

Background: Given the key role of integrins in maintaining intestinal homeostasis, anti-integrin biologics in inflammatory bowel disease (IBD) are being investigated in full swing. However, the unsatisfactory efficacy and safety of current anti-integrin biologics in clinical trials limit their widespread use in clinic. Therefore, it is particularly important to find a target that is highly and specifically expressed in the intestinal epithelium of patients with IBD. Methods: The function of integrin αvß6 in IBD and colitis-associated carcinoma (CAC) with the underlying mechanisms has been less studied. In the present study, we detected the level of integrin ß6 within inflammation including colitis tissues in human and mouse. To investigate the role of integrin ß6 in IBD and CAC, integrin ß6 deficient mice were hence generated based on the construction of colitis and CAC model. Results: We noted that integrin ß6 was significantly upregulated in inflammatory epithelium of patients with IBD. Integrin ß6 deletion not only reduced infiltration of pro-inflammatory cytokines, but also attenuated disruption of tight junctions between colonic epithelial cells. Meanwhile, lack of integrin ß6 affected macrophage infiltration in mice with colitis. This study further revealed that lack of integrin ß6 could inhibit tumorigenesis and tumor progression in CAC model by influencing macrophage polarization, which was also involved in attenuating the degree of intestinal symptoms and inflammatory responses in mice suffering from colitis. Conclusions: The present research provides a potentially new perspective and option for the treatment of IBD and CAC.

7.
Cells ; 11(13)2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35805163

RESUMEN

Integrin ß3 plays a key role in the resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKI), but the development of integrin ß3 inhibitors has been stalled due to the failure of phase III clinical trials for cancer treatment. Therefore, it is imperative to find a potentially effective solution to the problem of acquired resistance to EGFR-TKI for patients with integrin-ß3 positive non-small-cell lung cancer (NSCLC) by exploring novel downstream targets and action mechanisms of integrin ß3. In the present study, we observed that the expression of integrin ß3 and AXL was significantly upregulated in erlotinib-resistant NSCLC cell lines, which was further confirmed clinically in tumor specimens from patients with NSCLC who developed acquired resistance to erlotinib. Through ectopic expression or knockdown, we found that AXL expression was positively regulated by integrin ß3. In addition, integrin ß3 promoted erlotinib resistance in NSCLC cells by upregulating AXL expression. Furthermore, the YAP pathway, rather than pathways associated with ERK or AKT, was involved in the regulation of AXL by integrin ß3. To investigate the clinical significance of this finding, the current well-known AXL inhibitor R428 was tested, demonstrating that R428 significantly inhibited resistance to erlotinib, colony formation, epithelial-mesenchymal transformation and cell migration induced by integrin ß3. In conclusion, integrin ß3 could promote resistance to EGFR-TKI in NSCLC by upregulating the expression of AXL through the YAP pathway. Patients with advanced NSCLC, who are positive for integrin ß3, might benefit from a combination of AXL inhibitors and EGFR-TKI therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/farmacología , Clorhidrato de Erlotinib/uso terapéutico , Humanos , Integrina beta3/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
8.
Int J Biol Sci ; 18(10): 3944-3960, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35844799

RESUMEN

Our understanding of coding gene functions in lung cancer leads to the development of multiple generations of targeted drugs. Noncoding RNAs, including circular RNAs (circRNAs), have been demonstrated to play a vital role in tumorigenesis. Uncovering the functions of circRNAs in tumorigenesis and their underlying regulatory mechanisms may shed new light on the development of novel diagnostic and therapeutic strategies for human cancer. Here we report the important role of circFAT1 in lung adenocarcinoma (LUAD) progression and the potential impact of circFAT1 on LUAD treatment. We found that circFAT1 was one of the top expressed circRNAs in A549 cells by circRNA-seq and was significantly upregulated in human LUAD tissues. Multiple cellular assays with A549 and PC9 LAUD cell lines under both gain-of-function and loss-of-function conditions demonstrated that circFAT1 promoted proliferation of LUAD cells in vitro and in vivo. At molecular level, circFAT1 sequestered miR-7 to upregulate IRS2, which in turn regulated downstream ERK1/2 phosphorylation and CCND1 expression, ultimately promoting tumor progression. In addition, we showed that DDP treatment was much more effective in circFAT1 knockdown tumor cells in vitro and in a xenograft tumor model. Our results indicate that circFAT1 promote tumorigenesis in LUAD through sequestering miR-7, consequently upregulating IRS2-ERK1/2-mediated CCND1 expression, and can be a valuable therapeutic target and an important parameter for precision treatment in LUAD patients.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , MicroARNs , Adenocarcinoma del Pulmón/metabolismo , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica , Ciclina D1/genética , Ciclina D1/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Neoplasias Pulmonares/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética
9.
Dalton Trans ; 51(24): 9218-9222, 2022 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-35670316

RESUMEN

A C,S bonded quasi-two-coordinate Cr(II) complex, Cr(SAr*)2 (HSAr* = HSC6H3-2,6(C6H2-2,4,6-Pri3)2), has been synthesized according to literature precedent. Magnetic measurements, high-frequency/field electron paramagnetic resonance (HF-EPR) experiments and ab initio calculation studies show that the field-induced slow magnetic relaxation behaviour is caused by relatively weak axial magnetic anisotropy.

11.
Cardiovasc Toxicol ; 22(4): 311-325, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35211833

RESUMEN

Hypertension is one of the most prevalent cardiovascular disorders worldwide, affecting 1.13 billion people, or 14% of the global population. Hypertension is the single biggest risk factor for cerebrovascular dysfunction. According to the American Heart Association, high blood pressure (BP), especially in middle-aged individuals (~ 40 to 60 years old), is associated with an increased risk of dementia, later in life. Alzheimer's disease and cerebrovascular disease are the two leading causes of dementia, accounting for around 80% of the total cases and usually combining mixed pathologies from both. Little is known regarding how hypertension affects cognitive function, so the impact of its treatment on cognitive impairment has been difficult to assess. The brain renin-angiotensin system (RAS) is essential for BP regulation and overactivity of this system has been established to precede the development and maintenance of hypertension. Angiotensin II (Ang-II), the main peptide within this system, induces vasoconstriction and impairs neuro-vascular coupling by acting on brain Ang-II type 1 receptors (AT1R). In this review, we systemically analyzed the association between RAS and biological mechanisms of cognitive impairment, from the perspective of AT1R located in the central nervous system. Additionally, the possible contribution of brain AT1R to global cognition decline in COVID-19 cases will be discussed as well.


Asunto(s)
COVID-19 , Disfunción Cognitiva , Hipertensión , Adulto , Angiotensina II/metabolismo , Presión Sanguínea/fisiología , COVID-19/complicaciones , Disfunción Cognitiva/diagnóstico , Humanos , Hipertensión/diagnóstico , Persona de Mediana Edad , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina
12.
Small ; 17(20): e2100394, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33870652

RESUMEN

In nature, cells rely on a structural framework called the "cytoskeleton" to maintain their shape and polarity. Based on this, herein a new class of cell-mimicking nanomedicine using bionic skeletons constituted by the oligomeric Au(I)-peptide complex is developed. The peptide function of degrading pathological MDM2 and MDMX is used to synthesize an oligomeric Au(I)-PMIV precursor capable of self-assembling into a clustered spherical bionic skeleton. Through coating by erythrocyte membrane, an erythrocyte-mimicking nano-cell (Nery-PMIV) is developed with depressed macrophage uptakes, increased colloidal stability, and prolonged blood circulation. Nery-PMIV potently restores p53 and p73 in vitro and in vivo by degrading MDM2/MDMX. More importantly, Nery-PMIV effectively augments antitumor immunity elicited by anti-PD1 therapy in a murine orthotopic allograft model for LUAD and a humanized patient-derived xenograft (PDX) mouse model for LUAD, while maintaining a favorable safety profile. Taken together, this work not only presents evidence showing that MDM2/MDMX degradation is a potentially viable therapeutic paradigm to synergize anti-PD1 immunotherapy toward LUAD carrying wild-type p53; it also suggests that cell-mimicking nanoparticles with applicable bionic skeletons hold tremendous promise in offering new therapies to revolutionize nanomedicine in the treatment of a myriad of human diseases.


Asunto(s)
Adenocarcinoma , Proteínas Proto-Oncogénicas c-mdm2 , Animales , Biomimética , Proteínas de Ciclo Celular , Eritrocitos/metabolismo , Inmunoterapia , Ratones , Péptidos/metabolismo , Conducta Predatoria , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Esqueleto/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
13.
Transl Cancer Res ; 10(6): 2790-2800, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35116589

RESUMEN

BACKGROUND: A growing number of evidence has revealed the vital role of autophagy in pathological processes of cancer, including gastric cancer (GC). However, many previous studies only focused on exploring single pathway or limited genes of interest in GC, which only reflected partial functions of autophagy. The present study aimed to construct an autophagy-related risk signature for GC. METHODS: Differentially expressed autophagy-related genes (ARGs) in GC and non-tumor samples were screened through The Cancer Genome Atlas (TCGA) database, followed by bioinformatics analysis using the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) platforms. Prognosis-related ARGs were generated by univariate and multivariate Cox regression test. RESULTS: A total of seven prognosis-related ARGs (HSPB8, NRG2, GABARAPL1, TMEM74, DLC1, MAP1LC3C and NRG3) were determined to establish a prognostic index (PI) model, which was demonstrated to be an independent prognostic indicator for patients with GC. More importantly, it was successfully validated in an external cohort of patients from the GSE15460 dataset, indicating the useful reproducibility of this signature. In addition, the PI model was associated with immune cell infiltration estimates in GC. CONCLUSIONS: Taken together, the present study suggested that the seven ARGs-related signature could serve as an independent prognostic indicator for patients with GC.

14.
Cancer Manag Res ; 12: 9599-9608, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33061645

RESUMEN

PURPOSE: The development of esophageal squamous cell carcinoma (ESCC) is a complicated process in which cell adhesion and motility, mediated by integrins, are involved through connecting the cytoskeleton to extracellular matrix. Different mechanisms via which integrin ß6 participates in cancer invasion and metastasis have been described by numerous studies; however, the expression and clinical significance of integrin ß6 in ESCC remain unknown. METHODS: To investigate the differential expression of integrin ß6 in ESCC, qPCR and immunohistochemistry assays were performed in 10 paired human samples. A total of 137 ESCC samples were further enrolled to evaluate the expression levels of integrin ß6 and its endocytic trafficking regulator HS1-associated protein X-1 (HAX-1), followed by the evaluation of their correlation with clinicopathological parameters. The overall survival was analyzed using the Kaplan-Meier method, with significant variables further evaluated by multivariate Cox regression analyses. RESULTS: The expression of integrin ß6 was markedly increased in ESCC compared with matched adjacent normal tissues. Among the ESCC samples, positive expression of integrin ß6 was observed in 41.6% tumors, which was associated with histological differentiation, lymph node metastasis and TNM stage. High expression of HAX-1 was detected in 47.4% tumors, and there was a positive relationship between the expression levels of integrin ß6 and HAX-1. Furthermore, the expression of integrin ß6 and HAX-1 were independent unfavorable indicators for prognosis. Patients with positive integrin ß6 and high HAX-1 expression demonstrated worst outcomes. CONCLUSION: The present findings suggested the predictive value of integrin ß6 and HAX-1 as independent indicators of poor prognosis for patients with ESCC, both of which may contribute to the tumor proliferation and metastasis, leading to ESCC progression. Therefore, combined targeting of integrin ß6 and HAX-1 may provide a potential novel approach for the treatment of ESCC.

15.
Oxid Med Cell Longev ; 2020: 8032187, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32855767

RESUMEN

The metastatic potential of colorectal cancer (CRC) is intensively promoted by the tumor microenvironment (TME) in a paracrine manner. As a pleiotropic inflammatory cytokine, Interleukin-6 (IL-6) is produced and involved in CRC, the same scenario where integrin αvß6 also becomes upregulated. However, the relationship between IL-6 and integrin αvß6 as well as their involvement in the crosstalk between CRC and TME remains largely unclear. In the present study, we demonstrated a positive correlation between the expression of IL-6 and integrin ß6 in CRC samples. The mutually promotive interaction between CRC and TME was further determined by an indirect coculture system. CRC cells could augment the secretion of IL-6 from fibroblasts, which in return induced invasion and integrin ß6 expression of CRC cells. Through the classic IL-6 receptor/STAT-3 signaling pathway, IL-6 mediated the upregulation of integrin ß6, which was involved in the invasion and epithelial-mesenchymal transition of CRC cells induced by IL-6. Taken together, our results reveal a paracrine crosstalk between IL-6 signals originating from the TME and increased the integrin ß6 level of CRC. IL-6 induces CRC invasion via upregulation of integrin ß6 through the IL-6 receptor/STAT-3 signaling pathway. Combined inhibition of IL-6 along with integrin ß6-targeted strategy may indicate new directions for antitumor strategies for CRC.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Cadenas beta de Integrinas/genética , Interleucina-6/metabolismo , Regulación hacia Arriba , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Receptores de Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Regulación hacia Arriba/genética
16.
Theranostics ; 10(19): 8513-8527, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32754260

RESUMEN

Peptide-derived nanocomposites have been exhibiting fascinating biological advantages, including but not limited to excellent biocompatibility, biological degradation, high targetability and subsequent potent therapeutic efficacy. While some successes have been achieved in the nanoengineering of peptide-based architectures with defined dimensions and medical functions, enormous challenges remain about clinical nano-pharmaceutics of peptides, especially those modulating intracellular protein-protein interactions (PPIs). Methods: We developed a general method to translate intracellular-PPI-targeted peptides into a bioavailable peptide-auric spheroidal nanohybrid (SNH), for which polymeric peptide-Auric precursors [Au1+-S-peptide]n are in-situ reduced on the surface of gold nanoseeds via a simple and mild reaction. As proofs of concept, three cytomembrane-impenetrable peptides with different physicochemical properties were successfully engineered into stable and tumor-specific SNH respectively. Results: To highlight the advantage of SNH, PMI, a hydrophobic and enzyme-intolerant peptide capable of p53 restoration, was selected to challenge the power of SNH in a colon tumor xenografts model. PMI-Au SNH in vivo suppressed tumor growth potently after three administrations: intravenous injection, intraperitoneal injection and gastric perfusion, and maintained a favorable therapeutic safety. Conclusion: This therapeutically feasible strategy of peptide nanoengineering will allow us to fabricate a series of nanomedicines to modulate carcinogenic PPIs that hide and multiply inside cells, and in all likelihood reinvigorate the development of peptide drug against wide varieties of human diseases.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Oro/química , Péptidos/administración & dosificación , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Células HCT116 , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Nanopartículas del Metal , Ratones , Nanocompuestos , Péptidos/química , Péptidos/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Pathol Res Pract ; 216(7): 153022, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32534716

RESUMEN

OBJECTIVE: Integrin αvß6 is associated with an extremely aggressive cancer phenotype. However, little is known about the clinicopathological significance and prognostic value of integrin αvß6 in human hilar cholangiocarcinoma. METHODS: In the present study, bioinformatics analysis demonstrated a significant increase of integrin ß6 gene expression in cholangiocarcinoma tissues compared to non-tumorous tissues, which was further validated in clinical samples through RT-qPCR and western blotting analyses. Integrin αvß6 was observed to be expressed in 48.6% of tumors, and its expression was related to a poor tumor differentiation (p = 0.002), lymph node metastasis (p<0.001) and advanced TNM stage (p=0.001). Furthermore, patients who were αvß6-positive showed a significantly shorter overall survival period than those who were αvß6-negative (p=0.004). Multivariate analysis confirmed that integrin αvß6 was an independent prognostic factor (p=0.002). In addition, loss- and gain-of-function assays showed integrin αvß6 not only played an important role in colony formation, but also protected cholangiocarcinoma cells from cisplatin-induced growth inhibition and apoptosis. ERK/MAPK signaling pathway was involved in integrin αvß6-mediated resistance of cholangiocarcinoma cells to cisplatin. CONCLUSIONS: Taken together, the present findings revealed that integrin αvß6 could serve as a potential prognostic predictor and contribute to cisplatin resistance, which might prove to be a promising target candidate for the clinical intervention of human hilar cholangiocarcinoma.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Neoplasias de los Conductos Biliares/patología , Biomarcadores de Tumor/análisis , Integrinas/metabolismo , Tumor de Klatskin/patología , Anciano , Antígenos de Neoplasias/genética , Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Integrinas/genética , Masculino , Persona de Mediana Edad , Pronóstico
18.
Cell Mol Immunol ; 17(3): 261-271, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-30911117

RESUMEN

OBJECTIVES: Anti-citrullinated protein antibodies (ACPAs) are a group of autoantibodies targeted against citrullinated proteins/peptides and are informative rheumatoid arthritis (RA) biomarkers. ACPAs also play a crucial role in RA pathogenesis, and their underlying mechanism merits investigation. METHODS: Immunohistochemical (IHC) assays were carried out to determine IL-1ß levels in ACPA+ and ACPA- RA patients. PBMC-derived monocytes were differentiated into macrophages before stimulation with ACPAs purified from RA patients. The localization and interaction of molecules were analyzed by confocal microscopy, co-IP, and surface plasmon resonance. RESULTS: In our study, we found that IL-1ß levels were elevated in ACPA+ RA patients and that ACPAs promoted IL-1ß production by PBMC-derived macrophages. ACPAs interacted with CD147 to enhance the interaction between CD147 and integrin ß1 and, in turn, activate the Akt/NF-κB signaling pathway. The nuclear localization of p65 promoted the expression of NLRP3 and pro-IL-1ß, resulting in priming. Moreover, ACPA stimulation activated pannexin channels, leading to ATP release. The accumulated ATP bound to the P2X7 receptor, leading to NLRP3 inflammasome activation. CONCLUSIONS: Our study suggests a new hypothesis regarding IL-1ß production in RA involving ACPAs, which may be a potential therapeutic target in RA treatment.


Asunto(s)
Anticuerpos Antiproteína Citrulinada/inmunología , Artritis Reumatoide/inmunología , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Artritis Reumatoide/patología , Femenino , Humanos , Macrófagos/patología , Masculino , Células THP-1
19.
J Exp Clin Cancer Res ; 38(1): 449, 2019 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-31684995

RESUMEN

BACKGROUND: Interleukin-8 (IL-8) plays a vital role in the invasion and metastasis of hepatocellular carcinoma (HCC), and is closely associated with poor prognosis of HCC patients. Integrin αvß3, a member of the integrin family, has been reported to be overexpressed in cancer tissues and mediate the invasion and metastasis of HCC cells. However, the relationship between IL-8 and integrin αvß3 in HCC and the underlying mechanism of IL-8 and integrin αvß3 in the invasion of HCC remains unclear. METHODS: The expression of IL-8, integrin αv and integrin ß3 in HCC cells and tissues was detected by quantitative real-time PCR, Western blot and immunohistochemistry. Transwell assay and Western blot was used to detect the invasiveness, the expression of integrin ß3 and the activation of PI3K/Akt pathway of HCC cells pretreated with IL-8 knockdown or exogenous IL-8. RESULTS: IL-8, integrin αv and integrin ß3 were overexpressed in highly metastatic HCC cell lines compared with low metastatic cell lines. There was a positive correlation between integrin ß3 and IL-8 expression in HCC tissues. IL-8 siRNA transfection reduced HCC cell invasion and the levels of integrin ß3, p-PI3K and p-Akt. IL-8 induced HCC cell invasion and integrin ß3 expression was significantly inhibited by transfection with CXCR1 siRNA or CXCR2 siRNA. When we stimulated HCC cells with exogenous IL-8, cell invasion and the levels of integrin ß3, p-PI3K, and p-Akt increased, which could be effectively reversed by adding PI3K inhibitor LY294002. CONCLUSIONS: Our results suggest that IL-8 promotes integrin ß3 upregulation and the invasion of HCC cells through activation of the PI3K/Akt pathway. The IL-8/CXCR1/CXCR2/PI3K/Akt/integrin ß3 axis may serve as a potential treatment target for patients with HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Integrina beta3/genética , Integrina beta3/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Neoplasias Hepáticas/metabolismo , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Integrinas/genética , Integrinas/metabolismo , Neoplasias Hepáticas/genética , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Regulación hacia Arriba
20.
J Biochem Mol Toxicol ; 33(11): e22400, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31593355

RESUMEN

Inflammatory bowel disease (IBD) is a continual ailment condition which engrosses the entire alimentary canal. The IBD can be primarily distinguished into two forms, ulcerative colitis, and Crohn's disease. The major symptoms of IBD include pustules or abscesses, severe abdominal pain, diarrhea, fistula, and stenosis, which may directly affect the patient's quality of life. A variety of mediators can stimulate the circumstances of IBD, some examples include infections by microbes such as bacteria, perturbation of the immune system and the surrounding environment of the intestines. Severe colitis was stimulated in the experimental animals through administering 4% dextran sulfate sodium (DSS) which is mixed in water ad libitum for 6 days. Eriocitrin (30 mg/kg) was then administered to the experimental animals followed by the induction of severe colitis to evaluate the therapeutic prospective of eriocitrin against the colon inflammation stimulated by DSS. In this study, eriocitrin (30 mg/kg) demonstrated significant (P < .05) attenuation activity against the DSS-stimulated severe colitis in experimental animals. Eriocitrin counteracted all of the clinical deleterious effects induced by DSS, such as body-weight loss, colon shortening, histopathological injury, accretion of infiltrated inflammatory cells at the inflamed region and the secretion of inflammatory cytokines. The results clearly showed that eriocitrin effectively attenuated DSS-induced acute colitis in experimental animals.


Asunto(s)
Antiinflamatorios/uso terapéutico , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Sulfato de Dextran/farmacología , Flavanonas/uso terapéutico , Extractos Vegetales/uso terapéutico , Animales , Antiinflamatorios/administración & dosificación , Citrus/química , Colon/efectos de los fármacos , Colon/patología , Ciclooxigenasa 2/análisis , Citocinas/metabolismo , Modelos Animales de Enfermedad , Flavanonas/administración & dosificación , Inflamación/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/análisis , Peroxidasa/metabolismo , Extractos Vegetales/administración & dosificación , Índice de Severidad de la Enfermedad , Pérdida de Peso/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA