Due to its difficulty in early diagnosis and lack of sensitivity to chemotherapy and radiotherapy,renal cell carcinoma(RCC)remains to be a frequent cause of cancer-related death.Here,we probed into new targets for its ...Due to its difficulty in early diagnosis and lack of sensitivity to chemotherapy and radiotherapy,renal cell carcinoma(RCC)remains to be a frequent cause of cancer-related death.Here,we probed into new targets for its early diagnosis and treatment for RCC.microRNA(miRNA)data of M2-EVs and RCC were searched on the Gene Expression Omnibus database,followed by the prediction of the potential downstream target.Expression of target genes was measured via RT-qPCR and Western blot,respectively.M2 macrophage was obtained viaflow cytometry with M2-EVs extracted.The binding ability of miR-342-3p to NEDD4L and to CEP55 ubiquitination was studied with their roles in the physical abilities of RCC cells assayed.Subcutaneous tumor-bearing mouse models and lung metastasis models were prepared to observe in vivo role of target genes.M2-EVs induced RCC growth and metastasis.miR-342-3p showed high expression in both M2-EVs and RCC cells.M2-EVs carrying miR-342-3p promoted RCC cell abilities to proliferate,invade and migrate.In RCC cells,M2-EV-derived miR-342-3p could specifically bind to NEDD4L and consequently elevate CEP55 protein expression via suppressing NEDD4L,thereby exerting tumor-promoting effects.CEP55 could be degraded by ubiquitination under the function of NEDD4L,and miR-342-3p delivered by M2-EVs facilitated the RCC occurrence and development by activating the PI3K/AKT/mTOR signaling pathway.In conclusion,M2-EVs promote RCC growth and metastasis by delivering miR-342-3p to suppress NEDD4L and subsequently inhibit CEP55 ubiquitination and degradation via activation of the PI3K/AKT/mTOR signaling pathway,strongly driving the proliferative,migratory and invasive of RCC cells.展开更多
Objective:Lung cancer is the most common cause of cancer-related deaths worldwide.Platinum-based chemotherapy is one of the main treatment options for patients with non-small cell lung cancer(NSCLC)but the effectivene...Objective:Lung cancer is the most common cause of cancer-related deaths worldwide.Platinum-based chemotherapy is one of the main treatment options for patients with non-small cell lung cancer(NSCLC)but the effectiveness of chemotherapy is encumbered by drug resistance.Therefore,understanding the molecular mechanisms underlying chemotherapy resistance is crucial in improving treatment outcomes and prognosis.Methods:The cell viability assay and apoptosis were used to analyze chemoresistance.Western blot analysis and wound healing testing were used to evaluate the epithelial-to-mesenchymal transition(EMT).Immunoprecipitation was used for analysis of protein modification.Promoter activity was determined using the luciferase reporter assay.Immunofluorescence staining was used to determine reactive oxygen species levels.The expression patterns of EMT markers and carnitine palmitoyltransferase 1C(CPT1C)were determined by Western blot analysis.Results:CPT1C,which was shown to be highly expressed in lung cancer,is associated with cisplatin resistance in NSCLC cells.CPT1C depletion increased NSCLC cell sensitivity to cisplatin,while overexpression of CPT1C increased NSCLC cell resistance to cisplatin.Induction of EMT mediated CPT1C-induced cisplatin resistance.Ectopic expression of Snail reversed the increase in cisplatin sensitivity triggered by CPT1C knockdown.Moreover,CPT1C was shown to be regulated at the post-translational level and an E3-ubiquitin ligase,NEDD4L,was shown to be a major regulator of CPT1C stability and activity.Conclusions:These data provide evidence for the first time that the lipid metabolism enzyme,CPT1C,mediates resistance to chemotherapy.Therefore,the use of combination therapy with a CPT1C inhibitor may be a promising new avenue in lung cancer treatment.展开更多
Background:Nuclear Yes1-associated transcriptional regulator(YAP1)promotes tumor progression.However,the function of cytoplasmic YAP1 in breast cancer cells and its impact on the survival of breast cancer patients rem...Background:Nuclear Yes1-associated transcriptional regulator(YAP1)promotes tumor progression.However,the function of cytoplasmic YAP1 in breast cancer cells and its impact on the survival of breast cancer patients remain unclear.Our research aimed to explore the biological function of cytoplasmic YAP1 in breast cancer cells and the possibility of cytoplasmic YAP1 as a predictive marker of breast cancer survival.Methods:We constructed cell mutant models,including NLS-YAP15SA(nuclear localized),YAP1S94A(incapable of binding to the TEA domain transcription factor family)and YAP1S127D(cytoplasmic localized),and used Cell Counting Kit-8(CCK-8)assays,5-ethynyl-2’-deoxyuridine(EdU)incorporation assays,and Western blotting(WB)analysis to detect cell proliferation and apoptosis.The specific mechanism of cytoplasmic YAP1-mediated endosomal sorting complexes required for transport III(ESCRT-III)assembly was studied by co-immunoprecipitation,immunofluorescence staining,and WB analysis.Epigallocatechin gallate(EGCG)was used to simulate YAP1 retention in the cytoplasm in in vitro and in vivo experiments to study the function of cytoplasmic YAP1.YAP1 binding to NEDD4-like E3 ubiquitin protein ligase(NEDD4L)was identified using mass spectrometry and was verified in vitro.Breast tissue microarrays were used to analyze the relationship between cytoplasmic YAP1 expression and the survival of breast cancer patients.Results:YAP1 was mainly expressed in the cytoplasm in breast cancer cells.Cytoplasmic YAP1 promoted autophagic death of breast cancer cells.Cytoplasmic YAP1 bound to the ESCRT-III complex subunits charged multivesicular body protein 2B(CHMP2B)and vacuolar protein sorting 4 homolog B(VPS4B),promoting assembly of CHMP2B-VPS4B and activating autophagosome formation.EGCG retained YAP1 in the cytoplasm,promoting the assembly of CHMP2B-VPS4B to promote autophagic death of breast cancer cells.YAP1 bound to NEDD4L,and NEDD4L mediated ubiquitination and degradation of YAP1.Breast tissue microarrays revealed that high levels of cytoplasmic YAP1 were beneficial to the survival of breast cancer patients.Conclusions:Cytoplasmic YAP1 mediated autophagic death of breast cancer cells by promoting assembly of the ESCRT-III complex;furthermore,we established a new breast cancer survival prediction model based on cytoplasmic YAP1 expression.展开更多
文摘目的探讨神经前体细胞表达发育调控样蛋白(Nedd4L)通过泛素化降解癌蛋白神经生长因子酪氨酸激酶受体(Trk A)抑制老年前列腺癌的分子作用机制。方法收集手术切除的43例老年前列腺癌组织及对应癌旁组织标本。免疫组化染色检测Nedd4L及Trk A的表达,在人前列腺癌LNCa P细胞中转染Nedd4L特异性si RNA后,采用q RT-PCR及Western印迹检测LNCa P细胞内Trk A m RNA及蛋白的表达变化。结果Nedd4L蛋白在老年前列腺癌组织中表达明显降低,而Trk A表达显著升高(P<0.05);二者呈显著负相关(P<0.05),Nedd4L的表达降低与Trk A表达升高均与肿瘤淋巴结转移、高TNM分期显著相关(P<0.05);在体外,下调Nedd4L表达后可显著升高Trk A蛋白的表达水平(P<0.05),而对Trk A m RNA表达无明显影响(P>0.05)。结论 Nedd4L及Trk A在老年前列腺癌组织中异常表达,Nedd4L可能通过泛素化降解癌蛋白Trk A实现其抗肿瘤作用。
基金supported by the Science and Technology Department of Sichuan Province(2015SZ0117,2019YJ0701,and 2021YJ0239).
文摘Due to its difficulty in early diagnosis and lack of sensitivity to chemotherapy and radiotherapy,renal cell carcinoma(RCC)remains to be a frequent cause of cancer-related death.Here,we probed into new targets for its early diagnosis and treatment for RCC.microRNA(miRNA)data of M2-EVs and RCC were searched on the Gene Expression Omnibus database,followed by the prediction of the potential downstream target.Expression of target genes was measured via RT-qPCR and Western blot,respectively.M2 macrophage was obtained viaflow cytometry with M2-EVs extracted.The binding ability of miR-342-3p to NEDD4L and to CEP55 ubiquitination was studied with their roles in the physical abilities of RCC cells assayed.Subcutaneous tumor-bearing mouse models and lung metastasis models were prepared to observe in vivo role of target genes.M2-EVs induced RCC growth and metastasis.miR-342-3p showed high expression in both M2-EVs and RCC cells.M2-EVs carrying miR-342-3p promoted RCC cell abilities to proliferate,invade and migrate.In RCC cells,M2-EV-derived miR-342-3p could specifically bind to NEDD4L and consequently elevate CEP55 protein expression via suppressing NEDD4L,thereby exerting tumor-promoting effects.CEP55 could be degraded by ubiquitination under the function of NEDD4L,and miR-342-3p delivered by M2-EVs facilitated the RCC occurrence and development by activating the PI3K/AKT/mTOR signaling pathway.In conclusion,M2-EVs promote RCC growth and metastasis by delivering miR-342-3p to suppress NEDD4L and subsequently inhibit CEP55 ubiquitination and degradation via activation of the PI3K/AKT/mTOR signaling pathway,strongly driving the proliferative,migratory and invasive of RCC cells.
基金supported by the National Natural Science Foundation of China(Grant No.81872371)the Open Project Program of State Key Laboratory of Molecular Oncology(Grant No.SKL-KF-2019-11)+3 种基金the Program for Excellent Sci-tech Innovation Teams of Universities in Anhui Province(Grant No.2023AH010073)the Provincial-level Quality Project in Higher Education Institutions of Anhui Province(Grant No.2022jyxm1710)the College Students Innovation and Entrepreneurship Training Program(Grant No.S202310368028 and S202410368035)the Health Science Research Project of Anhui Province(Grant No.AHWJ2022a028)。
文摘Objective:Lung cancer is the most common cause of cancer-related deaths worldwide.Platinum-based chemotherapy is one of the main treatment options for patients with non-small cell lung cancer(NSCLC)but the effectiveness of chemotherapy is encumbered by drug resistance.Therefore,understanding the molecular mechanisms underlying chemotherapy resistance is crucial in improving treatment outcomes and prognosis.Methods:The cell viability assay and apoptosis were used to analyze chemoresistance.Western blot analysis and wound healing testing were used to evaluate the epithelial-to-mesenchymal transition(EMT).Immunoprecipitation was used for analysis of protein modification.Promoter activity was determined using the luciferase reporter assay.Immunofluorescence staining was used to determine reactive oxygen species levels.The expression patterns of EMT markers and carnitine palmitoyltransferase 1C(CPT1C)were determined by Western blot analysis.Results:CPT1C,which was shown to be highly expressed in lung cancer,is associated with cisplatin resistance in NSCLC cells.CPT1C depletion increased NSCLC cell sensitivity to cisplatin,while overexpression of CPT1C increased NSCLC cell resistance to cisplatin.Induction of EMT mediated CPT1C-induced cisplatin resistance.Ectopic expression of Snail reversed the increase in cisplatin sensitivity triggered by CPT1C knockdown.Moreover,CPT1C was shown to be regulated at the post-translational level and an E3-ubiquitin ligase,NEDD4L,was shown to be a major regulator of CPT1C stability and activity.Conclusions:These data provide evidence for the first time that the lipid metabolism enzyme,CPT1C,mediates resistance to chemotherapy.Therefore,the use of combination therapy with a CPT1C inhibitor may be a promising new avenue in lung cancer treatment.
基金National Natural Science Foundation of China,Grant/Award Numbers:81573001,81773295Haiyan Research Fund Project of Harbin Medical University Cancer Hospital,Grant/Award Number:JJZD2023-04Beijing Kechuang Medical Development Foundation,Grant/Award Number:KC2021-JF-0055-06。
文摘Background:Nuclear Yes1-associated transcriptional regulator(YAP1)promotes tumor progression.However,the function of cytoplasmic YAP1 in breast cancer cells and its impact on the survival of breast cancer patients remain unclear.Our research aimed to explore the biological function of cytoplasmic YAP1 in breast cancer cells and the possibility of cytoplasmic YAP1 as a predictive marker of breast cancer survival.Methods:We constructed cell mutant models,including NLS-YAP15SA(nuclear localized),YAP1S94A(incapable of binding to the TEA domain transcription factor family)and YAP1S127D(cytoplasmic localized),and used Cell Counting Kit-8(CCK-8)assays,5-ethynyl-2’-deoxyuridine(EdU)incorporation assays,and Western blotting(WB)analysis to detect cell proliferation and apoptosis.The specific mechanism of cytoplasmic YAP1-mediated endosomal sorting complexes required for transport III(ESCRT-III)assembly was studied by co-immunoprecipitation,immunofluorescence staining,and WB analysis.Epigallocatechin gallate(EGCG)was used to simulate YAP1 retention in the cytoplasm in in vitro and in vivo experiments to study the function of cytoplasmic YAP1.YAP1 binding to NEDD4-like E3 ubiquitin protein ligase(NEDD4L)was identified using mass spectrometry and was verified in vitro.Breast tissue microarrays were used to analyze the relationship between cytoplasmic YAP1 expression and the survival of breast cancer patients.Results:YAP1 was mainly expressed in the cytoplasm in breast cancer cells.Cytoplasmic YAP1 promoted autophagic death of breast cancer cells.Cytoplasmic YAP1 bound to the ESCRT-III complex subunits charged multivesicular body protein 2B(CHMP2B)and vacuolar protein sorting 4 homolog B(VPS4B),promoting assembly of CHMP2B-VPS4B and activating autophagosome formation.EGCG retained YAP1 in the cytoplasm,promoting the assembly of CHMP2B-VPS4B to promote autophagic death of breast cancer cells.YAP1 bound to NEDD4L,and NEDD4L mediated ubiquitination and degradation of YAP1.Breast tissue microarrays revealed that high levels of cytoplasmic YAP1 were beneficial to the survival of breast cancer patients.Conclusions:Cytoplasmic YAP1 mediated autophagic death of breast cancer cells by promoting assembly of the ESCRT-III complex;furthermore,we established a new breast cancer survival prediction model based on cytoplasmic YAP1 expression.