Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
BMC Gastroenterol ; 23(1): 420, 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38030976

RESUMEN

BACKGROUND: Single-incision plus one-port laparoscopic surgery (SILS + 1) has been demonstrated to be minimally invasive while possessing better cosmesis and less pain compared with conventional laparoscopic surgery (CLS). However, SILS + 1 as an alternative to CLS for colorectal cancer is still controversial. METHODS: A total of 1071 patients who underwent curative laparoscopic surgery for colon cancer between 2015 and 2018 were included. Of these patients, 258 SILS + 1 cases and 516 CLS cases were analyzed using propensity score matching. The baseline characteristics, surgical outcomes, pathologic findings and recovery course, morbidity and mortality within postoperative 30 days and 3-year disease-free and overall survival were compared. RESULTS: Baseline characteristics were balanced between the groups. The mean operating time was significantly shorter in SILS + 1 group, with less estimated blood loss. Tumor size, tumor differentiation, number of harvested lymph nodes, resection margin and pathologic T, N, TNM stage was similar between the groups. There was no significant difference in overall perioperative complications. Uni- and multivariate analyses revealed that SILS + 1 was not a risk factor for complications. Postoperatively, SILS + 1 group showed faster recovery than CLS group in terms of ambulation, bowel function, oral intake and discharge. The 3-year disease-free survival rates of SILS + 1 and CLS groups were 90.1% and 87.3%(p = 0.59), respectively and the 3-year overall survival rates were 93.3% vs. 89.8%(p = 0.172). DISCUSSION: Our study revealed that SILS + 1 is safe, feasible, oncologically efficient, and may be considered as a surgical option for selected patients with colorectal cancer.


Asunto(s)
Neoplasias del Colon , Laparoscopía , Humanos , Resultado del Tratamiento , Estudios de Cohortes , Neoplasias del Colon/cirugía , Laparoscopía/efectos adversos , Colectomía/efectos adversos , Tiempo de Internación , Tempo Operativo
2.
Br J Cancer ; 127(5): 811-823, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35637411

RESUMEN

BACKGROUND: Multiple mechanisms have been proposed that lead to reduced effectiveness of trastuzumab in HER2-positive gastric cancer (GC), yet resistance to trastuzumab remains a challenge in clinics. METHODS: We established trastuzumab-resistant cells and patient-derived xenografts models to measure metabolic levels and vascular density and shape. The HER2-positive GC patient samples were used to determine clinical significance. We also measured protein expression and phosphorylation modifications to determine those alterations related to resistance. In vivo studies combining inhibitor of PFKFB3 with trastuzumab corroborated the in vitro findings. RESULTS: The 6-phosphofructo-2-kinase (PFKFB3)-mediated trastuzumab resistance pathways in HER2-positive GC by activating the glycolytic pathway. We also found vessels are chaotic and destabilised in the tumour during the trastuzumab resistance process. Inhibition of PFKFB3 significantly diminished tumour proliferation and promoted vessel normalisation in the patient-derived xenograft model. Mechanistically, PFKFB3 promoted the secretion of CXCL8 into the tumour microenvironment, and phosphorylated Ser1151 of ERBB2, enhancing the transcription of CXCL8 by activating the PI3K/AKT/NFκB p65 pathway. CONCLUSIONS: Our current findings discover that PFKFB3 inhibitors might be effective tools to overcome adjuvant therapy resistance in HER2-positive GC and reshaping the microenvironment by normalising tumour vessels is a novel strategy to overcome trastuzumab resistance.


Asunto(s)
Fosfofructoquinasa-2 , Neoplasias Gástricas , Trastuzumab , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfofructoquinasa-2/antagonistas & inhibidores , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/farmacología , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Trastuzumab/farmacología , Trastuzumab/uso terapéutico , Microambiente Tumoral
3.
Arch Biochem Biophys ; 722: 109216, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35421356

RESUMEN

BACKGROUND: FAM98A is a microtubule-associated protein involved in cell proliferation and migration, and is frequently dysregulated in epithelial cancers. But its role in the development of colorectal cancer (CRC) cancer remains unknown. METHODS: Immunohistochemical analysis was performed to examine the expression of FAM98A in CRC samples. We also investigated the effects of abnormal expression on the biological behavior of colorectal cancer cells both in vitro and in vivo. Immunoblotting and immunoprecipitation were used to screen FAM98A-related signalling pathways and downstream factors. RESULTS: FAM98A was upregulated in CRC tissues and CRC cell lines. Overexpression of FAM98A promoted cell proliferation and recovered 5-FU suppressed CRC cell proliferation both in vitro and in vivo. In addition, the Enhanced expression of FAM98A inhibited ferroptosis in CRC cells by activating the translation of xCT in stress granules (SGs). Furthermore, we identified that metformin could reverse FAM98A-mediated 5-FU resistance in CRC cells. CONCLUSIONS: Our results for the first time indicate that FAM98A plays a critical role in promoting CRC progression, which provides a novel target for clinical drug resistance of colorectal cancer. And metformin may sensitize 5-FU in the treatment of colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Ferroptosis , Metformina , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas/metabolismo
4.
Bioorg Med Chem ; 71: 116962, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35987104

RESUMEN

A series of novel quinazoline analogs were designed and synthesized based on ARS-1620 and LLK-10 (a KRAS inhibitor reported by us recently) as KRAS G12C inhibitors with a 5-nitrofuran-2-carboxylic acid warhead. Most of the newly synthesized compounds exhibited antiproliferative activities similar to or better than ARS-1620 and LLK-10. Among them, compound KS-19 showed the highest activity (IC50 = 460 âˆ¼ 870 nM) and reasonable selectivity (3 to 27-fold) for inhibiting the proliferation of KRAS G12C-mutated cells (NCI-H358 and NCI-H23) over other KRAS mutant (e.g. G13D, G12D, G12S, G12V, WT) cancer cells. ITC, KRAS-GTP pull-down assay and western blot analysis demonstrated that KS-19 could bind to KRAS G12C protein with high affinity (KD = 97 nM), thus decreasing the active form of KRAS G12C (KRAS G12C-GTP) and phosphorylated Erk, and leading to NCI-H358 tumor cell apoptosis. In addition, KS-19 was able to suppress the formation of NCI-H358 and NCI-H23 tumor colonies in a dose-dependent manner. Moreover, in vivo efficacy studies indicated that KS-19 (40 mg/kg) was effective in suppressing tumor growth in nude mice bearing NCI-H358 tumor xenografts with a TGI (tumor growth inhibition) of 47 %, comparable to that of ARS-1620 (50 %). Lastly, KS-19 possessed a benign toxicity profile without causing bone marrow suppression and any obvious morphological abnormalities in major organs of mice. Collectively, these results suggest that KS-19 represents a novel inhibitor of KRAS G12C worthy of further investigation as a potential anticancer agent.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Antineoplásicos/farmacología , Guanosina Trifosfato , Humanos , Ratones , Ratones Desnudos , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Quinazolinas/farmacología
5.
Arch Biochem Biophys ; 702: 108838, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33727040

RESUMEN

The antimetabolite 5-fluorouracil (5-FU) is a widely used chemotherapy regimen for the treatment of gastric cancer (GC). However, resistance to 5-FU remains a major drawback in the clinical use. The treatments of anti-tumor chemo-agents recruit tumor associated macrophages (TAMs) which are highly implicated in the chemoresistance development, but the underlying molecular mechanism is unclear. Here, we demonstrate that YAP1 is overexpressed in resistant GC tissues compared to sensitive GC tissues. Further, IL-3 secreted by YAP1-overexpressed GC could skew macrophage polarization to M2-like phenotype and inducing GLUT3-depended glycolysis program. Meanwhile, polarized M2 macrophages enhance 5-FU resistance in tumor cells by secreting CCL8 and activating phosphorylation of JAK1/STAT3 signaling pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Fluorouracilo/farmacología , Transportador de Glucosa de Tipo 3/metabolismo , Neoplasias Gástricas/patología , Factores de Transcripción/metabolismo , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/metabolismo , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/metabolismo , Proteínas Señalizadoras YAP
6.
Cancer Cell Int ; 20: 503, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33061855

RESUMEN

BACKGROUND: Tumors display a high rate of glucose metabolism and the SLC2A (also known as GLUT) gene family may be central regulators of cellular glucose uptake. However, roles of SLC2A family in mechanism of metabolite communication with immunity in gastric cancer remains unknown. METHODS: Bioinformatics analysis and IHC staining were used to reveal the expression of SLC2A3 in gastric cancer and the correlation with survival prognosis. Real-time PCR, western blots, OCR, ECAR, lactate production and glucose uptake assays were applied to determine the effect of SLC2A3 on glycolysis reprogramming. We then investigated the consequences of SLC2A3 upregulation or inhibition on aerobic glycolysis, also explored the underlying mechanism. Bioinformatics analysis and in vitro and in vivo research were used to reveal the role of SLC2A3 in macrophage infiltration and transition. RESULTS: Here, we show that SLC2A3 acts as a tumor promoter and accelerates aerobic glycolysis in GC cells. Mechanistically, the SLC2A3-STAT3-SLC2A3 feedback loop could promote phosphorylation of the STAT3 signaling pathway and downstream glycolytic targeting genes. Moreover, SLC2A3 potentially contributes to M2 subtype transition of macrophage infiltration in the GC microenvironment. CONCLUSIONS: SLC2A3 could be used as a prognostic biomarker to determine prognosis and immune infiltration in GC and may provide an intervention strategy for GC therapy.

8.
Front Microbiol ; 15: 1367630, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38952444

RESUMEN

Purpose: To investigate the therapeutic efficiency of a novel drink termed "Ferment" in cases of ulcerative colitis (UC) and its influence on the gut microbiota. Method: In this study, we developed a complex of mixed fruit juice and lactic acid bacteria referred to as Ferment. Ferment was fed to mice for 35 days, before inducing UC with Dextran Sulfate Sodium Salt. We subsequently investigated the gut microbiome composition using 16S rRNA sequencing. Result: After Ferment treatment, mouse body weight increased, and animals displayed less diarrhea, reduced frequency of bloody stools, and reduced inflammation in the colon. Beneficial bacteria belonging to Ileibacterium, Akkermansia, and Prevotellacea were enriched in the gut after Ferment treatment, while detrimental organisms including Erysipelatoclostridium, Dubosiella, and Alistipes were reduced. Conclusion: These data place Ferment as a promising dietary candidate for enhancing immunity and protecting against UC.

9.
Int Immunopharmacol ; : 112990, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39223062

RESUMEN

Inflammatory bowel diseases (IBDs) are chronic, relapsing, and inflammatory disorders of the gastrointestinal tract characterized by abnormal immune responses. Recently, STING has emerged as a promising therapeutic target for various autoinflammatory diseases. However, few STING-selective small molecules have been investigated as novel strategies for IBD. In this study, we sought to examine the effects of PROTAC-based STING degrader SP23 on acute colitis and explore its underlying mechanism. SP23 treatment notably alleviates dextran sulfate sodium (DSS)-induced colitis. Pharmacological degradation of STING significantly reduced the production of inflammatory cytokines, such as TNF-α, IL-1ß, and IL-6, and inhibited macrophage polarization towards the M1 type. Furthermore, SP23 administration decreased the loss of tight junction proteins, including ZO-1, occludin, and claudin-1, and downregulated STING and NLRP3 signaling pathways in intestinal inflammation. In vitro, STING activated NLRP3 inflammasome-mediated pyroptosis in intestinal epithelial cells, which could be abrogated by SP23 and STING siRNA intervention. In conclusion, these findings provide new evidence for STING as a novel therapeutic target for IBD, and reveal that hyperactivation of STING could exaggerate colitis by inducing NLRP3/Caspase-1/GSDMD axis mediated intestinal epithelial cells pyroptosis.

10.
Plast Reconstr Surg ; 2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37285020

RESUMEN

BACKGROUND: Fat grafting has an unsatisfactory retention rate for breast reconstruction due to poor recipient conditions. The contribution of the recipient site to fat grafts is unknown. In this study, we hypothesize that tissue expansion could improve fat graft retention by preconditioning the recipient fat fad. METHODS: Over-expansion was achieved using 10 ml cylindrical soft-tissue expanders implanted beneath the left inguinal fat flaps of 16 Sprague-Dawley rats (weighing 250-300 g), whose contralateral parts were implanted with a silicone sheet as control. After 7-days expansion, the implants were removed and both inguinal fat flaps received 1 ml of fat grafts from 8 donor rats. Fluorescent dye-labeled mesenchymal stromal cells (MSCs) were injected into rats and tracked in vivo by fluorescence imaging. Transplanted adipose tissue was harvested at 4 and 10 weeks (n = 8 per time point). RESULTS: After 7-days expansion, OCT4+ (p = 0.0002) and Ki67+ (p = 0.0004) positive area were increased with up-regulated expression of CXCL12 in recipient adipose flaps. An increasing number of DiI-positive MSCs were observed in the expanded fat pad. At 10 weeks after fat grafting, retention rate, measured using the Archimedes principle, was much higher in the expanded group than that in the non-expanded group (0.3019 ± 0.0680 vs. 0.1066 ± 0.0402, p = 0.0005). Histologic and transcriptional analyses revealed that angiogenesis was enhanced, and macrophage infiltration was decreased in the expanded group. CONCLUSIONS: Internal expansion preconditioning increased circulating stem cells into recipient fat pad and contributed to improved fat graft retention.

11.
Plast Reconstr Surg ; 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37734113

RESUMEN

BACKGROUND: The unstable recipient conditions after fat grafting remains an obstacle for tissue volumization. The interaction between fat grafts and recipient sites is not fully understood. We hypothesize that recipient-derived adipocytes undergo dedifferentiation and migrate into fat grafts in tissue regeneration. METHODS: To observe the participation from recipient fat pad, we established a recipient adipocyte-tracing model where 0.2 ml inguinal fat from ten 8-week-old C57BL/6 mice was grafted to ten tamoxifen-treated AdipoqCre;mT/mG mice. Next, to evaluate the impact of physical force on recipient fat and fat graft, murine internal expansion model was established by implanting a 1 ml internal expander upon the inguinal fat pad of the lineage tracing mice that received fat graft from C57BL/6 mice. Transplanted adipose tissue was collected and analyzed by immunostaining of GFP, tdTomato, perilipin, CD31. RESULTS: In the observing model, immunostaining revealed that both GFP+ and tdTomato+ cells from recipient fat pad presented in fat grafts. Among the GFP+ cells, most of them were perilipin+ adipocytes and other perilipin- cells co-expressed OCT4, indicating dedifferentiated adipocytes. In the internal expansion model, internal expansion increased GFP+ cells in fat graft. Both OCT4+/GFP+ (0.23 ± 0.01 vs. 0.12 ± 0.04) and perilipin+/GFP+ (0.17 ± 0.02 vs. 0.06 ± 0.01) cells were increased in the expanded group, compared with control. CONCLUSIONS: Host-derived adipocytes participate in fat graft regeneration through migration and dedifferentiation, which could be enhanced by internal expansion to increase fat graft retention rate. Further study using larger animal model is needed, since this is a murine study.

12.
Artículo en Inglés | MEDLINE | ID: mdl-32982961

RESUMEN

In our previous study, we have shown that CRLF1 can promote proliferation and metastasis of papillary thyroid carcinoma (PTC); however, the mechanism is unclear. Herein, we investigated whether the interaction of CRLF1 and MYH9 regulates proliferation and metastasis of PTC cells via the ERK/ETV4 axis. Immunohistochemistry (IHC), qPCR, and Western blotting assays were performed on PTC cells and normal thyroid cells to profile specific target genes. In vitro assays and in vivo assays were also conducted to examine the molecular mechanism. Results showed that CRLF1 directly bound MYH9 to enhance the stability of CRLF1 protein. Inhibition of MYH9 in PTC cells overexpressing CRLF1 significantly reversed malignant phenotypes, and CRLF1 overexpression activated ERK pathway, in vitro, and in vivo. RNA-sequencing revealed that ETV4 is a downstream target gene of CRLF1, which was up-regulated following ERK activation. Moreover, it was revealed that ETV4 is highly expressed in PTC tissues and is associated with poor prognosis. Finally, the ChIP assays showed that ETV4 induces the expression of matrix metalloproteinase 1 (MMP1) by binding to its promoter on PTC cells. Altogether, our study demonstrates that CRLF1 interacts with MYH9, promoting cell proliferation and metastasis via the ERK/ETV4 axis in PTC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proliferación Celular , Sistema de Señalización de MAP Quinasas , Cadenas Pesadas de Miosina/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Receptores de Citocinas/metabolismo , Cáncer Papilar Tiroideo/secundario , Adolescente , Adulto , Anciano , Animales , Apoptosis , Biomarcadores de Tumor/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Cadenas Pesadas de Miosina/genética , Pronóstico , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-ets/genética , Receptores de Citocinas/genética , Tasa de Supervivencia , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
13.
Cell Death Dis ; 11(3): 202, 2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-32205841

RESUMEN

Tumor microenvironment plays vital roles in shaping cancer diversity, and CD73 (ecto-5'-nucleotidase; NT5E) is an emerging immune checkpoint in modulating cancer progression via conversion of immunostimulatory ATP into immunosuppressive adenosine. However, how the CD73 is regulated and how it functions in the progression of cancer are largely unknown. Here, we showed that CD73 was overexpressed and correlated with poor prognosis of gastric cancer. CD73 links adenosinergic signaling in microenvironment switching to induction of epithelial-to-mesenchymal transition phenotype in gastric cancer during metastasis. Further pathway and gene set enrichment analysis of transcriptome data revealed the modulation role of CD73 in RICS/RhoA signaling by its extracellular function in adenosinergic pathway, which subsequently inhibited phosphorylation of LIMK/cofilin and promoted ß-catenin activation. Pharmacological inhibition of CD73 adenosinergic signaling was found to induce RICS dysfunction. Dissemination and hematogenous metastasis model showed that targeting CD73 in gastric cancer could suppress experimental metastasis. To conclude, it substantiates CD73 as a target for treatment of gastric cancer metastasis and verifies RICS as an intracellular functional molecule linking CD73/adenosinergic signaling switching to RhoA/LIMK/cofilin pathway.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Neoplasias Gástricas/metabolismo , Tetraspaninas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Movimiento Celular/fisiología , Citoesqueleto/metabolismo , Citoesqueleto/patología , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Transducción de Señal , Neoplasias Gástricas/patología , Transfección , Microambiente Tumoral
14.
J Cancer ; 9(21): 3839-3849, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30410586

RESUMEN

Driver genes conducing to peritoneal metastasis in advanced gastric cancer remain to be clarified. S100A4 is suggested to evolve in metastasis of gastrointestinal cancer, we aim to explore the role of S100A4 plays in metastasis of advanced gastric cancer and the potential mechanism. Transfection of siRNA or cDNA was applied to alter the expression of protein S100A4 and MYH9, investigation of the expression of epithelial and mesenchymal transition (EMT) associated markers was followed. Cell migration assay was used to screen the alteration of migration ability regulated by S100A4 and MYH9. IHC analysis for tissue sample microarray was performed to reveal their relationship with clinical pathological parameters and potential capacity of predicting survival. Consistent overexpression of S100A4 and MYH9 were found in peritoneal metastasis and primary site compared with adjacent normal tissue. Low expression of S100A4 led to increased epithelial markers as wells as decline of mesenchymal makers, while overexpression of S100A4 led to inverse impact. S100A4 expression was closely correlated with increased migration ability and EMT process induced by TGF-ß stimulation. Interference of S100A4 led to downregulation of MYH9 and inactivation of Smad pathway through participating in EMT process, which could be reversed by overexpression of MYH9. Moreover, co-expression of S100A4 and MYH9 was identified in tissue microarray and confirmed by immunofluorescence assay. In conclusion, overexpression of S100A4 and downstream molecular MYH9 in advanced gastric cancer predicted poor prognosis; oncogene S100A4 facilitate EMT process induced by TGF-ß stimulation, suggesting a potential target in management of peritoneal metastasis of gastric cancer.

15.
Cell Death Dis ; 9(7): 749, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29970879

RESUMEN

Epithelial-mesenchymal transition (EMT)-induced metastasis contributes to human colorectal cancer (CRC) progression, especially in advanced CRC. However, the underlying mechanism of ß-catenin in this process is elusive. We identified that LIM domain kinase (LIMK)2 was progressively downregulated with tumor progression from precancerous lesions to advanced cancer. Gain- and loss-of-function assays revealed that LIMK2 inhibits cell proliferation via cell cycle arrest at the G1-S transition and suppresses the ability of cell metastasis by restricting the EMT process. Reduced LIMK2 expression enhanced the nuclear accumulation of ß-catenin and activated the Wnt signaling pathway, thus contributing to tumor progression. A homolog of the LIMK family, LIMK1, which was overexpressed throughout tumor progression, served as a competitive inhibitor of LIMK2 via ß-catenin nuclear translocation. The imbalanced expression of LIMK1 and LIMK2 is important in CRC progression, and the combined effects provide a new insight into the mechanism of CRC progression. These findings provide a new understanding for LIMK-based anticancer therapy.


Asunto(s)
Núcleo Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Quinasas Lim/metabolismo , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/fisiología , Animales , Proliferación Celular/genética , Proliferación Celular/fisiología , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Quinasas Lim/genética , Masculino , Ratones , Ratones Desnudos , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , beta Catenina/genética
16.
Oncogene ; 37(22): 3022-3038, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29535422

RESUMEN

Trastuzumab is the only target to be approved as the first-line treatment of HER2 positive metastatic gastric cancer, but ubiquitous resistance decreases its therapeutic benefit. In this study, we found HER4, phosphorylation HER4 (p-HER4) and the mesenchymal marker Vimentin increased in trastuzumab-resistant cells (MKN45TR and NCI-N87TR), while epithelial markers expressions in trastuzumab-resistant cell lines and animal models decreased. Additionally, silencing HER4 prevented the epithelial-mesenchymal transition and led to decreased proliferation and migration in vitro and in vivo. The expression of YAP1, a vital downstream interacted target of HER4, decreased when HER4 was knocked down. Interestingly, stimulation of NRG1 could compromise the inhibitory impact and rescue cell survival; whereas, transfection of siYAP1 sensitized trastuzumab-treated cells. Expression analysis of the proteins in patient-derived xenograft model (PDX) mice showed that HER4, p-HER4, YAP1, and Vimentin were clearly upregulated in the trastuzumab-resistant mice compared to mice without trastuzumab resistance. However, HER2 and E-cadherin were downregulated in response to continuous treatment with trastuzumab. These findings elucidated that the central role of the HER4-YAP1 axis in trastuzumab resistance of HER2-positive gastric cancer cells through induction of EMT. Hence, regulating the HER4-YAP1 axis might be a promising strategy for clinical interventions in patients with HER2-positive gastric cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Resistencia a Antineoplásicos , Fosfoproteínas/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-4/genética , Receptor ErbB-4/metabolismo , Neoplasias Gástricas/patología , Regulación hacia Arriba , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Trasplante de Neoplasias , Fosforilación , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Factores de Transcripción , Trastuzumab , Vimentina/metabolismo , Proteínas Señalizadoras YAP
17.
Cancer Lett ; 373(2): 203-13, 2016 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-26820227

RESUMEN

Constitutive overactivation of TGFß signaling is a common event in human cancer progression and acts as a major inducer of epithelial-mesenchymal transition (EMT). In pre-metastatic colorectal cancer (CRC) cells, however, this cascade is tightly controlled and the underlying mechanism in TGFß stimulated hyperactivation of downstream Smad pathway remains elusive. In this study, expression of miR-187 was downregulated in colorectal cancer (CRC) compared with adjacent normal tissues. miR-187 could suppress the formation of aggressive phenotype in CRC and inactivate Smad pathway, thus preventing EMT. TGFß stimulation significantly suppressed the expression of miR-187, and overexpressed miR-187 counteracted the influence of TGFß on cell phenotype and downstream pathway. Furthermore, we found that miR-187 directly suppressed the expression of SOX4, NT5E and PTK6, which were identified as essential upstream effectors of Smad pathway. Together with the fact that high SOX4 or NT5E levels were associated with poor prognosis, we also demonstrated that downregulation of miR-187 was closely related to tumor metastasis and poor prognosis in CRC. These findings revealed a plausible mechanism for sustained TGFß activation in cancer progression and might have suggested a novel miR-187-based clinical intervention target for patients with advanced CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal , MicroARNs/fisiología , Transducción de Señal/fisiología , Proteínas Smad/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/fisiología , 5'-Nucleotidasa/fisiología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Proteínas Ligadas a GPI/fisiología , Perfilación de la Expresión Génica , Humanos , Proteínas de Neoplasias/fisiología , Proteínas Tirosina Quinasas/fisiología , Factores de Transcripción SOXC/fisiología
18.
Sci Rep ; 6: 25631, 2016 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-27156963

RESUMEN

LIM and SH3 protein 1 (LASP1) can promote colorectal cancer (CRC) progression and metastasis, but the direct evidence that elucidates the molecular mechanism remains unclear. Here, our proteomic data showed that LASP1 interacted with 14-3-3σ and decreased the expression of 14-3-3σ in CRC. Deletion of 14-3-3σ was required for LASP1-mediated CRC cell aggressiveness. In vitro gain- and loss-of-function assays showed that 14-3-3σ suppressed the ability of cell migration and decreased the phosphorylation of AKT in CRC cells. We further observed clearly co-localization between AKT and 14-3-3σ in CRC cells. Treatment of PI3K inhibitor LY294002 markedly prevented phosphorylation of AKT and subsequently counteract aggressive phenotype mediated by siRNA of 14-3-3σ. Clinically, 14-3-3σ is frequently down-regulated in CRC tissues. Down-regulation of 14-3-3σ is associated with tumor progression and poor prognosis of patients with CRC. Multivariate analysis confirmed low expression of 14-3-3σ as an independent prognostic factor for CRC. A combination of low 14-3-3σ and high LASP1 expression shows a worse trend with overall survival of CRC patients. Our research paves the path to future investigation of the LASP1-14-3-3σ axis as a target for novel anticancer therapies of advanced CRC.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas del Citoesqueleto/metabolismo , Progresión de la Enfermedad , Proteínas con Dominio LIM/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Línea Celular Tumoral , Movimiento Celular , Regulación hacia Abajo , Electroforesis en Gel Bidimensional , Humanos , Modelos Biológicos , Invasividad Neoplásica , Fosforilación , Pronóstico , Unión Proteica
19.
Clin Cancer Res ; 20(22): 5835-47, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25252758

RESUMEN

PURPOSE: The expression of LIM and SH3 protein 1 (LASP1) was upregulated in colorectal cancer cases, thereby contributing to the aggressive phenotypes of colorectal cancer cells. However, we still cannot decipher the underlying molecular mechanism associated with colorectal cancer metastasis. EXPERIMENTAL DESIGN: In this study, IHC was performed to investigate the expression of proteins in human colorectal cancer tissues. Western blot analysis was used to assess the LASP1-induced signal pathway. Two-dimensional difference gel electrophoresis was performed to screen LASP1-modulated proteins and uncover the molecular mechanism of LASP1. TGFß was used to induce an epithelial-mesenchymal transition (EMT). RESULTS: LASP1 expression was correlated with the mesenchymal marker vimentin and was inversely correlated with epithelial markers, namely, E-cadherin and ß-catenin, in clinical colorectal cancer samples. The gain- and loss-of-function assay showed that LASP1 induces EMT-like phenotypes in vitro and in vivo. S100A4, identified as a LASP1-modulated protein, was upregulated by LASP1. Moreover, it is frequently coexpressed with LASP1 in colorectal cancer. S100A4 was required for EMT, and an increased cell invasiveness of colorectal cancer cell is induced by LASP1. Furthermore, the stimulation of TGFß resulted in an activated Smad pathway that increased the expression of LASP1 and S100A4. The depletion of LASP1 or S100A4 expression inhibited the TGFß signaling pathway. Moreover, it significantly weakened the proinvasive effects of TGFß on colorectal cancer cells. CONCLUSION: These findings elucidate the central role of LASP1 in the TGFß-mediated EMT process and suggest a potential target for the clinical intervention in patients with advanced colorectal cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Proteínas con Dominio LIM/metabolismo , Proteínas S100/genética , Factor de Crecimiento Transformador beta/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Humanos , Invasividad Neoplásica , Proteína de Unión al Calcio S100A4 , Transducción de Señal , Regulación hacia Arriba , Vimentina/genética , Vimentina/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA