Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
PLoS Genet ; 19(12): e1011084, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38157491

RESUMEN

mDia formin proteins regulate the dynamics and organization of the cytoskeleton through their linear actin nucleation and polymerization activities. We previously showed that mDia1 deficiency leads to aberrant innate immune activation and induces myelodysplasia in a mouse model, and mDia2 regulates enucleation and cytokinesis of erythroblasts and the engraftment of hematopoietic stem and progenitor cells (HSPCs). However, whether and how mDia formins interplay and regulate hematopoiesis under physiological and stress conditions remains unknown. Here, we found that both mDia1 and mDia2 are required for HSPC regeneration under stress, such as serial plating, aging, and reconstitution after myeloid ablation. We showed that mDia1 and mDia2 form hetero-oligomers through the interactions between mDia1 GBD-DID and mDia2 DAD domains. Double knockout of mDia1 and mDia2 in hematopoietic cells synergistically impaired the filamentous actin network and serum response factor-involved transcriptional signaling, which led to declined HSPCs, severe anemia, and significant mortality in neonates and newborn mice. Our data demonstrate the potential roles of mDia hetero-oligomerization and their non-rodent functions in the regulation of HSPCs activity and orchestration of hematopoiesis.


Asunto(s)
Actinas , Proteínas Portadoras , Ratones , Animales , Forminas/genética , Forminas/metabolismo , Actinas/genética , Actinas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Citoesqueleto de Actina/metabolismo , Microtúbulos/metabolismo
2.
Angew Chem Int Ed Engl ; 63(24): e202402611, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38607929

RESUMEN

METTL3, a primary methyltransferase catalyzing the RNA N6-methyladenosine (m6A) modification, has been identified as an oncogene in several cancer types and thus nominated as a potentially effective target for therapeutic inhibition. However, current options using this strategy are limited. In this study, we targeted protein-protein interactions at the METTL3-METTL14 binding interface to inhibit complex formation and subsequent catalysis of the RNA m6A modification. Among candidate peptides, RM3 exhibited the highest anti-cancer potency, inhibiting METTL3 activity while also facilitating its proteasomal degradation. We then designed a stapled peptide inhibitor (RSM3) with enhanced peptide stability and formation of the α-helical secondary structure required for METTL3 interaction. Functional and transcriptomic analysis in vivo indicated that RSM3 induced upregulation of programmed cell death-related genes while inhibiting cancer-promoting signals. Furthermore, tumor growth was significantly suppressed while apoptosis was enhanced upon RSM3 treatment, accompanied by increased METTL3 degradation, and reduced global RNA methylation levels in two in vivo tumor models. This peptide inhibitor thus exploits a mechanism distinct from other small-molecule competitive inhibitors to inhibit oncogenic METTL3 activity. Our findings collectively highlight the potential of targeting METTL3 in cancer therapies through peptide-based inhibition of complex formation and proteolytic degradation.


Asunto(s)
Antineoplásicos , Metiltransferasas , Péptidos , Metiltransferasas/metabolismo , Metiltransferasas/antagonistas & inhibidores , Humanos , Péptidos/química , Péptidos/farmacología , Péptidos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Adenosina/análogos & derivados , Adenosina/química , Adenosina/metabolismo , Adenosina/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Línea Celular Tumoral , Apoptosis/efectos de los fármacos
3.
Biol Pharm Bull ; 46(8): 1145-1151, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37286513

RESUMEN

Inositol hexaphosphate (IP6), a widely found natural bioactive substance in grains, effectively inhibits the progression of colorectal cancer (CRC) when used in combination with inositol (INS). We previously showed that supplementation of IP6 and INS upregulated the claudin 7 gene in orthotropic CRC xenografts in mice. The aim of this study was to elucidate the role of claudin 7 in the inhibition of CRC metastasis by IP6 and INS, and explore the underlying mechanisms. We found that IP6, INS and their combination inhibited the epithelial-mesenchymal transition (EMT) of colon cancer cell lines (SW480 and SW620), as indicated by upregulation of claudin 7 and E-cadherin, and downregulation of N-cadherin. The effect of IP6 and INS was stronger compared to either agent alone (combination index < 1). Furthermore, the silencing of the claudin 7 gene diminished the anti-metastatic effects of IP6 and INS on SW480 and SW620 cells. Consistent with in vitro findings, the combination of IP6 and INS suppressed CRC xenograft growth in a mouse model, which was neutralized by claudin 7. Taken together, the combination of IP6 and INS can inhibit CRC metastasis by blocking EMT of tumor cells through upregulation of claudin 7.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Ratones , Animales , Neoplasias Colorrectales/metabolismo , Ácido Fítico/farmacología , Ácido Fítico/uso terapéutico , Inositol/farmacología , Inositol/uso terapéutico , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Claudinas/genética
4.
Invest New Drugs ; 39(5): 1411-1418, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33765213

RESUMEN

Primary acinar soft part sarcoma of the lung (ASPS) is a rare malignancy with unique cellular structure and clinical and genetic characteristics. Most patients do not exhibit clear clinical symptoms, with only a few developing respiratory symptoms. The typical histological characteristics are acinoid or organ-like structures. Immunofluorescence in situ hybridization suggests a rearrangement of the transcription factor E3 gene. Patients respond poorly to chemotherapy and are, thus, primarily treated with surgery and targeted therapy. We report herein a unique case of primary alveolar soft part sarcoma of the lung. The patient was a 24-year-old man with metastases to multiple organs, such as the brain, lungs, pancreas, and liver. The craniocerebral lesions attained partial remission after whole-brain radiotherapy and targeted combined immunotherapy, and other distant metastases completely disappeared after targeted combined immunotherapy (anlotinib and camrelizumab), indicating significant treatment efficacy. Anlotinib is an oral multi-target tyrosine kinase inhibitor (TKI) that exerts its anti-tumor effects by acting on various kinases. Camrelizumab is a humanized immunoglobulin G4 monoclonal antibody that can target PD-1 to block the interaction between PD-L1 and programmed death ligand 2, ultimately causing an anti-tumor effect. This is the first report of successful use of anlotinib combined with camrelizumab in the treatment of advanced primary ASPS. The treatment benefit provides preliminary evidence that targeted therapy, combined with immunotherapy, may be a safe and effective approach to treat primary pulmonary ASPS patients, thus warranting further investigation.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Indoles/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Quinolinas/uso terapéutico , Sarcoma de Parte Blanda Alveolar/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica , Humanos , Indoles/administración & dosificación , Indoles/efectos adversos , Neoplasias Pulmonares/patología , Masculino , Metástasis de la Neoplasia , Quinolinas/administración & dosificación , Quinolinas/efectos adversos , Adulto Joven
5.
BMC Womens Health ; 20(1): 194, 2020 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-32891135

RESUMEN

BACKGROUND: To investigate the impact of the elevation of tumor-infiltrating lymphocytes (TILs) in different molecular subtypes of primary breast cancer, i.e. each 10% increment of TILs and high-level TILs (TILs≥50%) in tumor, on overall survival (OS) and pathological complete response (pCR) and to compare the presentation of high-level TILs across these molecular subtypes. METHODS: Citation retrieval was performed in the PubMed, Cochrane Library, Embase and Web of Science databases. All statistical calculations were performed by the software of StataSE version 12.0. RESULTS: Twenty-two eligible clinical trials including 15,676 unique patients were included for meta-analysis. Each 10% increment of TILs significantly improved OS in human epidermal growth factor receptor 2 (HER2)-overexpression (pooled Hazard ratio (HR), 0.92; 95% CI, 0.89-0.95) and triple-negative (TN) (pooled HR, 0.90; 95% CI, 0.89-0.92) breast tumors but not in luminal tumor subtype (pooled HR, 1.06; 95% CI, 0.99-1.13). It was also associated with an increased pCR rate in breast cancers (pooled Odds ratio (OR), 1.27; 95% CI, 1.19-13.5). High-level TILs were significantly related with a higher pCR rate (pooled OR, 2.73; 95% CI, 2.40-3.01) than low-level TILs. The HER2-amplified (pooled OR, 3.14; 95% CI, 1.95-5.06) and TN (pooled OR, 4.09; 95% CI, 2.71-6.19) phenotypes of breast cancers expressed significantly more high-level TILs than the luminal tumor subtype, although the presentation of those between the former two subsets was not significantly different (pooled OR, 1.30; 95%CI, 0.83-2.04). CONCLUSIONS: The elevation of TILs in breast tumors predicts favorable prognostic outcomes, particularly in the HER2-overexpression and TN subtypes.


Asunto(s)
Neoplasias de la Mama/patología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Neoplasias de la Mama Triple Negativas/diagnóstico , Antígeno B7-H1 , Biomarcadores de Tumor , Neoplasias de la Mama/sangre , Neoplasias de la Mama/mortalidad , Femenino , Factores de Transcripción Forkhead , Humanos , Pronóstico , Receptor ErbB-2 , Análisis de Supervivencia , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología
6.
BMC Womens Health ; 20(1): 17, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32005117

RESUMEN

BACKGROUND: To investigate the efficacy of neoadjuvant chemotherapy (NCT), neoadjuvant endocrine therapy (NET) and neoadjuvant chemoendocrine therapy (NCET) on the tumour response, including pathological complete response (pCR) rate and overall response rate (ORR), in postmenopausal women with hormone receptor (HR)-positive breast cancer. METHODS: Based on a PRISMA-IPD statement, the PubMed, Embase and Cochrane Library databases were used to identify eligible trials published from inception to 7 May 2019. Pooled odds ratio (OR) with 95% confidential interval (CI) was calculated to assess the pCR rate and ORR of tumours among those three treatments via fixed- or random-effect Mantel-Haenszel models in terms of a Heterogeneity Chi2 test with a significant level of p < 0.1. All statistical tests were performed by the software of StataSE, version 12.0. RESULTS: The analysed data consisted of 10 eligible clinical trials with 971 unique HR-positive breast cancer patients. The pooled results indicated that the pCR rate of those patients undergoing NET was significantly lower than those undergoing NCT (pooled OR, 0.48; 95% CI, 0.26-0.90), whereas the difference of ORR between both therapies was not statistically significant (pooled OR, 1.05; 95% CI, 0.73-1.52). The combined paradigm of NCET compared with the monotherapy of NET or NCT did not present a significantly improved pCR rate or ORR (pooled OR, 2.61; 95% CI, 0.94-7.25; and 2.25; 95% CI, 0.39-13.05; respectively). CONCLUSION: Postmenopausal HR-positive breast cancer patients after NCT may have better tumour response than those after NET, while those undergoing NCET may not manifest the apparently improved clinical efficacies compared to those receiving monotherapy.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Anciano , Neoplasias de la Mama/metabolismo , Quimioterapia Adyuvante , Femenino , Humanos , Persona de Mediana Edad , Terapia Neoadyuvante , Posmenopausia , Receptores de Estrógenos/metabolismo , Resultado del Tratamiento
7.
Tumour Biol ; 39(3): 1010428317695961, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28349820

RESUMEN

Recurrence of breast cancer after radiotherapy may be partly explained by the presence of radioresistant cells. Thus, it would be desirable to develop an effective therapy against radioresistant cells. In this study, we demonstrated the intense antitumor activity of cytokine-induced killer cells against MCF-7 and radioresistant MCF-7 cells, as revealed by cytokine-induced killer-mediated cytotoxicity, tumor cell proliferation, and tumor invasion. Radioresistant MCF-7 cells were more susceptible to cytokine-induced killer cell killing. The stronger cytotoxicity of cytokine-induced killer cells against radioresistant MCF-7 cells was dependent on the expression of major histocompatibility complex class I polypeptide-related sequence A/B on radioresistant MCF-7 cells after exposure of cytokine-induced killer cells to sensitized targets. In addition, we demonstrated that cytokine-induced killer cell treatment sensitized breast cancer cells to chemotherapy via the downregulation of TK1, TYMS, and MDR1. These results indicate that cytokine-induced killer cell treatment in combination with radiotherapy and/or chemotherapy may induce synergistic antitumor activities and represent a novel strategy for breast cancer.


Asunto(s)
Neoplasias de la Mama/radioterapia , Tratamiento Basado en Trasplante de Células y Tejidos , Células Asesinas Inducidas por Citocinas/metabolismo , Recurrencia Local de Neoplasia/radioterapia , Subfamilia B de Transportador de Casetes de Unión a ATP/biosíntesis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Células Asesinas Inducidas por Citocinas/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de la radiación , Femenino , Humanos , Células MCF-7 , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Tolerancia a Radiación , Timidina Quinasa/biosíntesis , Timidilato Sintasa/biosíntesis
8.
BMC Cancer ; 17(1): 899, 2017 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-29282026

RESUMEN

BACKGROUND: To examine the effects of recombinant human endostatin combined with radiotherapy on colorectal cancer HCT-116 cell xenografts in nude mice. METHODS: Forty male BALB/c nude mice were injected with human colorectal cancer HCT-116 cells to form xenografts and then randomized into the following 4 groups (each group comprised ten mice): a control group, an endostatin group (20 mg/kg endostatin once a day for 10 days), a radiotherapy group (a 6-Gy dose was administered via a 6-MV X-ray on day 5 post-inoculation), and a combination therapy group (radiotherapy with endostatin treatment). The tumor growth inhibition rate were detected. CD31, vascular endothelial growth factor (VEGF), and hypoxia inducible factor-1α (HIF-1α) expression and microvascular density (MVD) were evaluated by immunohistochemistry. The expression of VEGF protein was also detected by western blotting. RESULTS: The tumor growth inhibition rate in the radiotherapy with endostatin treatment group was significantly higher than those in endostatin group or radiotherapy group (77.67% vs 12.31% and 38.59%; n = 8 per group, P < 0.05). The results of immunohistochemistry showed that treatment with radiotherapy induced significant increases in CD31, VEGF, and HIF-1α expression and MVD compared with treatment with saline, while treatment with endostatin or radiotherapy with endostatin induced reductions in CD31, VEGF, and HIF-1α expression and MVD compared with treatment with saline (n = 8 per group, P < 0.05). The results of western blotting showed that VEGF protein expression in radiotherapy group was significantly increased compared with that in the control group. However, VEGF protein expression in the endostatin or radiotherapy with endostatin groups was significantly decreased compared with that in the control group (n = 8 per group, P < 0.05). CONCLUSIONS: Endostatin combined with radiotherapy can significantly inhibit HCT-116 cell xenograft growth, possibly by inhibiting angiogenesis and attenuating tumor cell hypoxia.


Asunto(s)
Adenocarcinoma/terapia , Inhibidores de la Angiogénesis/administración & dosificación , Quimioradioterapia , Neoplasias Colorrectales/terapia , Endostatinas/administración & dosificación , Neovascularización Patológica/prevención & control , Proteínas Recombinantes/administración & dosificación , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Biochemistry (Mosc) ; 82(4): 438-445, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28371600

RESUMEN

Long noncoding RNAs (lncRNAs) have been recently regarded as systemic regulators in multiple biological processes including tumorigenesis. In this study, we report an ultra-highly expressed lncRNA, lnc-Sox5, in tongue tumor tissues. The results imply that lnc-Sox5 may play vital role in tongue carcinoma progression. We observed that the growth of Tca8113 cells was suppressed by lnc-Sox5 downregulation. Additionally, lnc-Sox5 knockdown simultaneously increased Tca8113 cell apoptosis, but the cell cycle was arrested. RNA immunoprecipitation suggested that HuR directly bound to and stabilized lnc-Sox5 RNA. Consistently, HuR knockdown reduced the level of lnc-Sox5 in Tca8113 cells. However, overexpression of HuR induced more lnc-Sox5 in Tca8113 cells. Both lnc-Sox5 knockdown and HuR knockdown suppressed Tca8113 cell tumorigenesis in xenograft models. These results suggest that lnc-Sox5, which was stabilized by HuR, could regulate carcinogenesis of tongue cancer and may serve as a predicted target for tongue carcinoma therapies.


Asunto(s)
Carcinoma de Células Escamosas/patología , Proteína 1 Similar a ELAV , ARN Largo no Codificante/genética , ARN Mensajero/genética , Factores de Transcripción SOXD/genética , Neoplasias de la Lengua/patología , Animales , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Lengua/genética
10.
Tumour Biol ; 36(11): 8925-30, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26076812

RESUMEN

Gastric carcinoma (GC) is a leading cause of cancer-related death in China. Dysregulation of microRNAs (miRNAs) has been shown to contribute to the development of GC, whereas the role of miR-133 in GC is unknown. Here, we analyzed the levels of miR-133 in GC tissues by reverse and quantitative transcription polymerase chain reaction (RT-qPCR). We overexpressed or inhibited miR-133 in GC cells. Cell growth was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and cell apoptosis was evaluated by fluorescence-activated cell sorting (FACS) analysis. Targeted genes were predicted by a bioinformatics algorithm and confirmed by a dual-luciferase reporter assay. We detected lower miR-133 levels in GC tissues compared with normal gastric tissue. Moreover, the low miR-133 levels were correlated with low survival rate. Overexpression of miR-133 inhibited cell growth and promoted apoptosis, while depletion of miR-133 increased cell growth and suppressed apoptosis. Moreover, the 3'-untranslated region (3'UTR) of Her-2, the epidermal growth factor receptor (EGFR) that transduces cell growth signals, appeared to be targeted by miR-133. Together, these data suggest that reduced miR-133 levels in GC tissues promote GC growth, which possibly contributes to a low survival rate of GC patients. MiR-133 may target Her-2 to suppress GC cell growth.


Asunto(s)
Carcinoma/genética , MicroARNs/biosíntesis , Receptor ErbB-2/genética , Neoplasias Gástricas/genética , Apoptosis/genética , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular/genética , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , MicroARNs/genética , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal/genética , Neoplasias Gástricas/patología
11.
Biotechnol Lett ; 37(12): 2387-94, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26318721

RESUMEN

OBJECTIVES: To investigate the role of microRNAs in doxorubicin resistance of breast cancer cells. RESULTS: MicroRNA-134 was down-regulated in doxorubicin-resistant and doxorubicin-sensitive breast cancer samples and MCF-7/ADR cell lines. After transfection of miR-134 mimics, an MTT assay confirmed that the MCF-7/ADR cell proliferation was inhibited. In the presence of doxorubicin, there was inhibition of cell proliferation. Transfection of miR-134 mimics induced MCF-7/ADR cell apoptosis. The expression of ABCC1 was significantly upregulated in doxorubicin-resistant or -sensitive breast cancer tissues and MCF-7/ADR cell lines. Over-expression of miR-134 decreased the expression of ABCC1 at the protein level. CONCLUSION: MicroRNA-134 modulates resistance to doxorubicin in breast cancer cells by downregulating the expression of ABCC1 which is known to encode the multidrug resistance-associated protein 1.


Asunto(s)
Antibióticos Antineoplásicos/metabolismo , Neoplasias de la Mama/fisiopatología , Regulación hacia Abajo , Doxorrubicina/metabolismo , Resistencia a Antineoplásicos , MicroARNs/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Formazáns/análisis , Humanos , Modelos Biológicos , Sales de Tetrazolio/análisis
12.
Front Endocrinol (Lausanne) ; 15: 1379607, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38686204

RESUMEN

Background: Hepatobiliary cancer (HBC), including hepatocellular carcinoma (HCC) and biliary tract cancer (BTC), is currently one of the malignant tumors that mainly cause human death. Many HBCs are diagnosed in the late stage, which increases the disease burden, indicating that effective prevention strategies and identification of risk factors are urgent. Many studies have reported the role of thyroid hormones on HBC. Our research aims to assess the causal effects and investigate the mediation effects between thyroid function and HBC. Methods: Utilizing the Mendelian randomization (MR) approach, the study employs single nucleotide polymorphisms (SNPs) as instrumental variables (IVs) to explore causal links between thyroid function [free thyroxine (FT4), thyroid stimulating hormone (TSH), hyperthyroidism and hypothyroidism] and HBC. Data were sourced from the ThyroidOmic consortium and FinnGen consortium. The analysis included univariable and multivariable MR analysis, followed by mediation analysis. Results: The study found a significant causal association between high FT4 levels and the reduced risk of BTC, but not HCC. However, TSH, hyperthyroidism and hypothyroidism had no causal associations with the risk of HBC. Notably, we also demonstrated that only higher FT4 levels with the reference range (FT4-RR) could reduce the risk of BTC because this protective effect no longer existed under the conditions of hyperthyroidism or hypothyroidism. Finally, we found that the protective effect of FT4-RR on BTC was mediated partially by decreasing the risk of metabolic syndrome (MetS) and reducing the waist circumference (WC). Conclusion: The findings suggest that higher FT4-RR may have a protective effect against BTC, which is partially mediated by decreased risk of MetS and a reduction in WC. This study highlights the potential role of FT4 in the pathogenesis of BTC and underscores that MetS and WC may play mediation effects as two mediators in this process.


Asunto(s)
Neoplasias del Sistema Biliar , Análisis de la Aleatorización Mendeliana , Polimorfismo de Nucleótido Simple , Tiroxina , Humanos , Neoplasias del Sistema Biliar/genética , Neoplasias del Sistema Biliar/sangre , Neoplasias del Sistema Biliar/epidemiología , Neoplasias del Sistema Biliar/prevención & control , Tiroxina/sangre , Análisis de Mediación , Factores de Riesgo , Hipotiroidismo/genética , Hipotiroidismo/sangre , Femenino , Masculino , Hipertiroidismo/genética , Hipertiroidismo/sangre , Carcinoma Hepatocelular/sangre , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/prevención & control , Carcinoma Hepatocelular/etiología
13.
J Mater Chem B ; 12(9): 2294-2303, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38344907

RESUMEN

Diketopyrrolopyrrole (DPP) is an excellent photosensitizer and photothermal agent with the advantages of good planarity, strong electron affinity, high electron mobility, easy purification, easy structural modification and high molar absorption coefficient. It is regarded as one of the ideal choices for the design and synthesis of efficient organic photovoltaic materials. Therefore, two kinds of donor-acceptor (D-A) conjugated polymers were designed and synthesized with DPP as the acceptor, and their optical properties and applications in the near-infrared region were studied. The quantum yield (QY) of PBDT-DPP is 0.46%, and the highest temperature reached within 10 minutes after irradiation with a 660 nm laser is 60 °C. Another polymer, EDOT-DPP, has a QY of 0.48%, and its semiconductor polymer nanoparticle aqueous solution can reach 60 °C within 12 minutes under laser irradiation, achieving photothermal treatment of nude mice tumors. Both polymer NPs have good biocompatibility and promising applications in bioimaging and photothermal therapy.


Asunto(s)
Cetonas , Fototerapia , Polímeros , Pirroles , Animales , Ratones , Fototerapia/métodos , Polímeros/química , Ratones Desnudos , Imagen Óptica/métodos
14.
Eur J Med Res ; 29(1): 217, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38570887

RESUMEN

BACKGROUND: Malignant esophageal fistula (MEF), which occurs in 5% to 15% of esophageal cancer (EC) patients, has a poor prognosis. Accurate identification of esophageal cancer patients at high risk of MEF is challenging. The goal of this study was to build and validate a model to predict the occurrence of esophageal fistula in EC patients. METHODS: This study retrospectively enrolled 122 esophageal cancer patients treated by chemotherapy or chemoradiotherapy (53 with fistula, 69 without), and all patients were randomly assigned to a training (n = 86) and a validation (n = 36) cohort. Radiomic features were extracted from pre-treatment CTs, clinically predictors were identified by logistic regression analysis. Lasso regression model was used for feature selection, and radiomics signature building. Multivariable logistic regression analysis was used to develop the clinical nomogram, radiomics-clinical nomogram and radiomics prediction model. The models were validated and compared by discrimination, calibration, reclassification, and clinical benefit. RESULTS: The radiomic signature consisting of ten selected features, was significantly associated with esophageal fistula (P = 0.001). Radiomics-clinical nomogram was created by two predictors including radiomics signature and stenosis, which was identified by logistic regression analysis. The model showed good discrimination with an AUC = 0.782 (95% CI 0.684-0.8796) in the training set and 0.867 (95% CI 0.7461-0.987) in the validation set, with an AIC = 101.1, and good calibration. When compared to the clinical prediction model, the radiomics-clinical nomogram improved NRI by 0.236 (95% CI 0.153, 0.614) and IDI by 0.125 (95% CI 0.040, 0.210), P = 0.004. CONCLUSION: We developed and validated the first radiomics-clinical nomogram for malignant esophageal fistula, which could assist clinicians in identifying patients at high risk of MEF.


Asunto(s)
Fístula Esofágica , Neoplasias Esofágicas , Humanos , Fístula Esofágica/diagnóstico por imagen , Fístula Esofágica/etiología , Neoplasias Esofágicas/complicaciones , Neoplasias Esofágicas/diagnóstico por imagen , Modelos Estadísticos , Nomogramas , Pronóstico , Radiómica , Estudios Retrospectivos
15.
Aging (Albany NY) ; 16(3): 2161-2180, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-38277212

RESUMEN

Immune checkpoint blockade (ICB) therapy has resulted in improved overall survival in kidney renal clear cell carcinoma (KIRC), but most treated patients fail to show durable clinical responses. Lymphocyte activation gene-3 (LAG3) is a novel inhibitory immune checkpoint, but its expression pattern, prognostic value, and immunological role in KIRC remain unknown. In this study, we utilized TCGA_KIRC RNA-sequencing data to analyze the relationship between LAG3 expression and clinical features. The single-cell sequencing data and tissue immunofluorescence are employed to investigate the subcellular localization of LAG3 in KIRC. Kaplan-Meier plotter, TIMER, and TISIDB were used to assess the association between LAG3 expression and prognosis, as well as its correlation with immune-related components. We constructed the LAG3 interaction network by using STRING, GeneMANIA, BioGRID, and HitPredict databases. We found that LAG3 is upregulated and correlates with poor prognostic phenotype in KIRC. LAG3 is predominantly expressed on exhausted CD8+ T cells and shows strong co-expression with PDCD1 in KIRC. Moreover, our findings indicated that LAG3 not only inhibits T cell activation but also potentially regulates cell adhesion in KIRC. In conclusion, our study implies that LAG3 can serve as a potential prognostic biomarker for KIRC. Furthermore, blocking both LAG3 and PDCD1 may alleviate resistance to anti-PDCD1 therapy, providing novel insights for immunotherapy decision-making in KIRC patients.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Pronóstico , Linfocitos T CD8-positivos , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Riñón
16.
Food Funct ; 15(7): 3463-3478, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38456333

RESUMEN

Currently, the clinical efficacy of anti-PD-1/PD-L1 monotherapy strategies against breast cancer is limited, and low response rates need to be improved. Gut microbiota plays a crucial role in the sensitization process of immunotherapy. As a natural dietary supplement, fucoidan has been reported to have immunomodulatory effects, while some studies have found that oral fucoidan may act as a potential prebiotic to modulate the gut microbiota. Therefore, this study investigated whether fucoidan could enhance the effects of anti-PD-1 monoclonal antibody antitumor immunotherapy by modulating gut microbiota and its metabolites. We found that the anti-tumor effect of the combination treatment was significantly enhanced, while fucoidan significantly improved the composition of the gut microbiota by increasing the number of potentially beneficial bacteria, such as Bifidobacterium, Faecalibaculum and Lactobacillus. Interference with the gut microbiota by antibiotics revealed impaired antitumor efficacy, confirming the necessity of gut microbiota in the antitumor effects of fucoidan in vivo. Metabolomics further revealed that fucoidan may have reversed the metabolic disturbances induced by the breast cancer model through tryptophan metabolism and glycerophospholipid metabolism pathways, with the most significant increase in the content of short-chain fatty acids, especially acetic and butyric acids. These modulations improved the function of effector T cells and suppressed Treg cell production. Thus, our findings suggest that fucoidan combined with the anti-PD-1 monoclonal antibody may be a novel strategy to sensitize breast cancer patients to anti-PD-1 monoclonal antibody immunotherapy. Meanwhile, the gut microbiota might serve as a new biomarker to predict the anti-PD-1 monoclonal antibody response to breast cancer immunotherapy.


Asunto(s)
Neoplasias de la Mama , Microbioma Gastrointestinal , Polisacáridos , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Inmunoterapia , Anticuerpos Monoclonales/farmacología
17.
Cell Death Dis ; 15(4): 295, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664392

RESUMEN

Abnormal Transmembrane protein 9 (TMEM9) expression has been identified in various human tumors. However, the prognostic potential and mechanistic role of TMEM9 in lung adenocarcinoma (LUAD) remain unclear. Here, we first found a significant upregulation of TMEM9 in LUAD tissues, and TMEM9 expression was positively correlated with microvessel density (MVD), T stage, and clinical stage. Survival analysis demonstrated TMEM9 was an independent indicator of poor prognosis in LUAD patients. In addition, downregulation of TMEM9 suppressed tumor growth and metastasis in vitro and in vivo models, and reduced HUVEC proliferation, migration, and tube formation in a cancer cell/HUVEC coculture model. Furthermore, TMEM9 upregulated VEGF expression, and VEGF-neutralizing antibodies reversed HUVEC angiogenesis and cancer cell migration ability caused by overexpression of TMEM9. In contrast, recombinant VEGF (rVEGF) abolished the inhibitory effect of TMEM9-knockdown LUAD cells on HUVEC angiogenesis and tumor cell migration. Moreover, we showed that TMEM9 upregulated VEGF expression by activating the mitogen-activated protein kinase/extracellular signal-regulated kinase/STAT3 (MEK/ERK/STAT3) pathway. Together, our study provides mechanistic insights into the role of TMEM9 in LUAD and highlights the potential of targeting the TMEM9/MEK/ERK/STAT3/VEGF pathway as a novel therapy for preventing LUAD progression.


Asunto(s)
Adenocarcinoma del Pulmón , Progresión de la Enfermedad , Neoplasias Pulmonares , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana , Factor de Transcripción STAT3 , Factor A de Crecimiento Endotelial Vascular , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Células A549 , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/genética , Factor de Transcripción STAT3/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Clin Exp Med ; 23(6): 1973-1979, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36913035

RESUMEN

Forkhead box M1 (FoxM1), a proliferation specific transcriptional modulator, plays a principal role in many physiological and pathological processes. FoxM1-mediated oncogenic processes have been well addressed. However, functions of FoxM1 in immune cells are less summarized. The literatures about the expression of FoxM1 and its regulation on immune cells were searched on PubMed and Google Scholar. In this review, we provide an overview on the roles of FoxM1 in regulating functions of immune cells, including T cells, B cells, monocytes, macrophages, and dendritic cells, and discuss their contributions to diseases.


Asunto(s)
Proteína Forkhead Box M1 , Monocitos , Humanos , Proteína Forkhead Box M1/genética , Macrófagos/metabolismo , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Proliferación Celular
19.
Front Microbiol ; 14: 1321386, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38298540

RESUMEN

Cancer is the most common cause of human death worldwide, posing a serious threat to human health and having a negative impact on the economy. In the past few decades, significant progress has been made in anticancer therapies, but traditional anticancer therapies, including radiation therapy, surgery, chemotherapy, molecular targeted therapy, immunotherapy and antibody-drug conjugates (ADCs), have serious side effects, low specificity, and the emergence of drug resistance. Therefore, there is an urgent need to develop new treatment methods to improve efficacy and reduce side effects. Antimicrobial peptides (AMPs) exist in the innate immune system of various organisms. As the most promising alternatives to traditional drugs for treating cancers, some AMPs also have been proven to possess anticancer activities, which are defined as anticancer peptides (ACPs). These peptides have the advantages of being able to specifically target cancer cells and have less toxicity to normal tissues. More and more studies have found that marine and terrestrial animals contain a large amount of ACPs. In this article, we introduced the animal derived AMPs with anti-cancer activity, and summarized the types of tumor cells inhibited by ACPs, the mechanisms by which they exert anti-tumor effects and clinical applications of ACPs.

20.
Transl Res ; 255: 159-170, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36347490

RESUMEN

Clonal hematopoiesis (CH) occurs in hematopoietic stem cells with increased risks of progressing to hematologic malignancies. CH mutations are predominantly found in aged populations and correlate with an increased incidence of cardiovascular and other diseases. Increased lines of evidence demonstrate that CH mutations are closely related to the inflammatory bone marrow microenvironment. In this review, we summarize the recent advances in this topic starting from the discovery of CH and its mutations. We focus on the most commonly mutated and well-studied genes in CH and their contributions to the innate immune responses and inflammatory signaling, especially in the hematopoietic cells of bone marrow. We also aimed to discuss the interrelationship between inflammatory bone marrow microenvironment and CH mutations. Finally, we provide our perspectives on the challenges in the field and possible future directions to help understand the pathophysiology of CH.


Asunto(s)
Médula Ósea , Hematopoyesis Clonal , Humanos , Anciano , Hematopoyesis Clonal/genética , Hematopoyesis/genética , Evolución Clonal , Inflamación/genética , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA