19 results on '"Lin, Nengming"'
Search Results
2. A Pluripotential Neutrophil-Mimic Nanovehicle Modulates Immune Microenvironment with Targeted Drug Delivery for Augmented Antitumor Chemotherapy.
- Author
-
Wu, Jiahe, Ma, Teng, Zhu, Manning, Mu, Jiafu, Huang, Tianchen, Xu, Donghang, Lin, Nengming, and Gao, Jianqing
- Published
- 2024
- Full Text
- View/download PDF
3. SLC15A3 plays a crucial role in pulmonary fibrosis by regulating macrophage oxidative stress
- Author
-
Luo, Jun, Li, Ping, Dong, Minlei, Zhang, Yingqiong, Lu, Shuanghui, Chen, Mingyang, Zhou, Hui, Lin, Nengming, Jiang, Huidi, and Wang, Yuqing
- Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal and irreversible disease with few effective treatments. Alveolar macrophages (AMs) are involved in the development of IPF from the initial stages due to direct exposure to air and respond to external oxidative damage (a major inducement of pulmonary fibrosis). Oxidative stress in AMs plays an indispensable role in promoting fibrosis development. The oligopeptide histidine transporter SLC15A3, mainly expressed on the lysosomal membrane of macrophages and highly expressed in the lung, has proved to be involved in innate immune and antiviral signaling pathways. In this study, we demonstrated that during bleomycin (BLM)- or radiation-induced pulmonary fibrosis, the recruitment of macrophages induced an increase of SLC15A3 in the lung, and the deficiency of SLC15A3 protected mice from pulmonary fibrosis and maintained the homeostasis of the pulmonary microenvironment. Mechanistically, deficiency of SLC15A3 resisted oxidative stress in macrophages, and SLC15A3 interacted with the scaffold protein p62 to regulate its expression and phosphorylation activation, thereby regulating p62-nuclear factor erythroid 2-related factor 2 (NRF2) antioxidant stress pathway protein, which is related to the production of reactive oxygen species (ROS). Overall, our data provided a novel mechanism for targeting SLC15A3 to regulate oxidative stress in macrophages, supporting the therapeutic potential of inhibiting or silencing SLC15A3 for the precautions and treatment of pulmonary fibrosis.
- Published
- 2024
- Full Text
- View/download PDF
4. MFN2 deficiency affects calcium homeostasis in lung adenocarcinoma cells via downregulation of UCP4.
- Author
-
Zhang, Jingjing, Pan, Lifang, Zhang, Qiang, Zhao, Yanyan, Wang, Wenwen, Lin, Nengming, Zhang, Shirong, and Wu, Qiong
- Subjects
HOMEOSTASIS ,CALCIUM ,LUNGS ,ADENOCARCINOMA ,REACTIVE oxygen species ,CALCIUM channels ,INTRACELLULAR calcium - Abstract
Mitofusin‐2 (MFN2) is a transmembrane GTPase that regulates mitochondrial fusion and thereby modulates mitochondrial function. However, the role of MFN2 in lung adenocarcinoma remains controversial. Here, we investigated the effect of MFN2 regulation on mitochondria in lung adenocarcinoma. We found that MFN2 deficiency resulted in decreased UCP4 expression and mitochondrial dysfunction in A549 and H1975 cells. UCP4 overexpression restored ATP and intracellular calcium concentration, but not mtDNA copy number, mitochondrial membrane potential or reactive oxygen species level. Furthermore, mass spectrometry analysis identified 460 overlapping proteins after independent overexpression of MFN2 and UCP4; these proteins were significantly enriched in the cytoskeleton, energy production, and calponin homology (CH) domains. Moreover, the calcium signaling pathway was confirmed to be enriched in KEGG pathway analysis. We also found by protein–protein interaction network analysis that PINK1 may be a key regulator of MFN2‐ and UCP4‐mediated calcium homeostasis. Furthermore, PINK1 increased MFN2/UCP4‐mediated intracellular Ca2+ concentration in A549 and H1975 cells. Finally, we demonstrated that low expression levels of MFN2 and UCP4 in lung adenocarcinoma are associated with poor clinical prognosis. In conclusion, our data suggest not only a potential role of MFN2 and UCP4 in co‐regulating calcium homeostasis in lung adenocarcinoma but also their potential use as therapeutic targets in lung cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Engineering cells or cellular outer membranes as multifaceted vehicles for reinforcing nanomedicine-based pulmonary disease treatment.
- Author
-
Wang, Nan, Wu, Jiahe, Gao, Jianqing, and Lin, Nengming
- Abstract
Pulmonary diseases are seriously threatening the health of human body, placing a significant financial burden on society. The development of nanotechnology has attracted increasing attention in recent pulmonary disease treatment owing to its distinctive advantages in conquering the limitations of conventional pharmacotherapeutic treatments. However, the inherent "foreign" and difficulties in adding biointerfacing capabilities to nanoparticles (NPs) in the complicated biological environment via synthetic techniques may hinder its further clinical applications. Mammalian cells that possess unique functions in complex and dynamic bioenvironments are encouraging the rational design of cell-nano integrated systems by leveraging the innate biointerfacing properties of cells and the distinct characterizations of NPs. Here, a comprehensive review is presented to introduce a promising bio-nano-cooperated platform for targeted pulmonary disease treatment. The multifaceted vehicles from different source cells and the application-specific design of corresponding bio-nano-cooperated platforms against various pulmonary diseases are introduced. By directly hitchhiking the entire cells or their outer membranes, the NPs can be endowed with enhanced biointerfacing and biointeracting abilities, befriending physiological barriers and achieving targeted drug delivery to specifically execute therapeutic performance with minimized side effects in vivo. Moreover, these vehicles also exhibit multifaceted therapeutic potential, which synergistically improve the therapeutic efficiency in pulmonary disease treatment. Despite its attractive applications, various challenges in the scope of future perspectives need to be addressed. [Display omitted] • Cells and cellular membranes-cooperated nanoparticles (NPs) for high-performance pulmonary disease treatment are overviewed. • The bio-nano-integrated systems can leverage the biointerfacing properties of cells and the distinct characterizations of NPs. • The bio-nano-integrated systems can befriend physiological barriers and achieve targeted drug delivery. • The "therapeutic bonus" of vehicular cells and cell membranes for synergistic pulmonary disease treatment is presented. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Recent nanotechnology-based strategies for interfering with the life cycle of bacterial biofilms
- Author
-
Wu, Jiahe, Zhang, Bo, Lin, Nengming, and Gao, Jianqing
- Abstract
Biofilm formation plays an important role in the resistance development in bacteria to conventional antibiotics. Different properties of the bacterial strains within biofilms compared with their planktonic states and the protective effect of extracellular polymeric substances contribute to the insusceptibility of bacterial cells to conventional antimicrobials. Although great effort has been devoted to developing novel antibiotics or synthetic antibacterial compounds, their efficiency is overshadowed by the growth of drug resistance. Developments in nanotechnology have brought various feasible strategies to combat biofilms by interfering with the biofilm life cycle. In this review, recent nanotechnology-based strategies for interfering with the biofilm life cycle according to the requirements of different stages are summarized. Additionally, the importance of strategies that modulate the bacterial biofilm microenvironment is also illustrated with specific examples. Lastly, we discussed the remaining challenges and future perspectives on nanotechnology-based strategies for the treatment of bacterial infection.
- Published
- 2023
- Full Text
- View/download PDF
7. Deubiquitinase JOSD2 stabilizes YAP/TAZ to promote cholangiocarcinoma progression.
- Author
-
Qian, Meijia, Yan, Fangjie, Wang, Weihua, Du, Jiamin, Yuan, Tao, Wu, Ruilin, Zhao, Chenxi, Wang, Jiao, Lu, Jiabin, Zhang, Bo, Lin, Nengming, Dong, Xin, Dai, Xiaoyang, Dong, Xiaowu, Yang, Bo, Zhu, Hong, and He, Qiaojun
- Subjects
CHOLANGIOCARCINOMA ,DEUBIQUITINATING enzymes ,PROTEIN stability - Abstract
Cholangiocarcinoma (CCA) has emerged as an intractable cancer with scanty therapeutic regimens. The aberrant activation of Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are reported to be common in CCA patients. However, the underpinning mechanism remains poorly understood. Deubiquitinase (DUB) is regarded as a main orchestrator in maintaining protein homeostasis. Here, we identified Josephin domain-containing protein 2 (JOSD2) as an essential DUB of YAP/TAZ that sustained the protein level through cleavage of polyubiquitin chains in a deubiquitinase activity-dependent manner. The depletion of JOSD2 promoted YAP/TAZ proteasomal degradation and significantly impeded CCA proliferation in vitro and in vivo. Further analysis has highlighted the positive correlation between JOSD2 and YAP abundance in CCA patient samples. Collectively, this study uncovers the regulatory effects of JOSD2 on YAP/TAZ protein stabilities and profiles its contribution in CCA malignant progression, which may provide a potential intervention target for YAP/TAZ-related CCA patients. JOSD2, a deubiquitinating enzyme, increases YAP/TAZ abundance and reinforces their signaling through the cleavage of polyubiquitin chains, observably contributing to cholangiocarcinoma progression. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
8. Recent progress of ferroptosis in cancers and drug discovery
- Author
-
Wang, Xiang, Ren, Xinxin, Lin, Xu, Li, Qi, Zhang, Yingqiong, Deng, Jun, Chen, Binxin, Ru, Guoqing, Luo, Ying, and Lin, Nengming
- Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron dependence and lipid peroxidation. Ferroptosis is involved in a range of pathological processes, such as cancer. Many studies have confirmed that ferroptosis plays an essential role in inhibiting cancer cell proliferation. In addition, a series of small-molecule compounds have been developed, including erastin, RSL3, and FIN56, which can be used as ferroptosis inducers. The combination of ferroptosis inducers with anticancer drugs can produce a significant synergistic effect in cancer treatment, and patients treated with these combinations exhibit a better prognosis than patients receiving traditional therapy. Therefore, a thorough understanding of the roles of ferroptosis in cancer is of great significance for the treatment of cancer. This review mainly elaborates the molecular biological characteristics and mechanism of ferroptosis, summarizes the function of ferroptosis in cancer development and treatment,illustrates the application of ferroptosis in patient's prognosis prediction and drug discovery, and discusses the prospects of targeting ferroptosis.
- Published
- 2024
- Full Text
- View/download PDF
9. Autophagic degradation of CCN2 (cellular communication network factor 2) causes cardiotoxicity of sunitinib
- Author
-
Xu, Zhifei, Jin, Ying, Gao, Zizheng, Zeng, Yan, Du, Jiangxia, Yan, Hao, Chen, Xueqin, Ping, Li, Lin, Nengming, Yang, Bo, He, Qiaojun, and Luo, Peihua
- Abstract
ABSTRACTExcessive macroautophagy/autophagy is one of the causes of cardiomyocyte death induced by cardiovascular diseases or cancer therapy, yet the underlying mechanism remains unknown. We and other groups previously reported that autophagy might contribute to cardiomyocyte death caused by sunitinib, a tumor angiogenesis inhibitor that is widely used in clinic, which may help to understand the mechanism of autophagy-induced cardiomyocyte death. Here, we found that sunitinib-induced autophagy leads to apoptosis of cardiomyocyte and cardiac dysfunction as the cardiomyocyte-specific Atg7−/+heterozygous mice are resistant to sunitinib. Sunitinib-induced maladaptive autophagy selectively degrades the cardiomyocyte survival mediator CCN2 (cellular communication network factor 2) through the TOLLIP (toll interacting protein)-mediated endosome-related pathway and cardiomyocyte-specific knockdown of Ccn2through adeno-associated virus serotype 9 (AAV9) mimics sunitinib-induced cardiac dysfunction in vivo, suggesting that the autophagic degradation of CCN2 is one of the causes of sunitinib-induced cardiotoxicity and death of cardiomyocytes. Remarkably, deletion of Hmgb1(high mobility group box 1) inhibited sunitinib-induced cardiomyocyte autophagy and apoptosis, and the HMGB1-specific inhibitor glycyrrhizic acid (GA) significantly mitigated sunitinib-induced autophagy, cardiomyocyte death and cardiotoxicity. Our study reveals a novel target protein of autophagic degradation in the regulation of cardiomyocyte death and highlights the pharmacological inhibitor of HMGB1 as an attractive approach for improving the safety of sunitinib-based cancer therapy.
- Published
- 2022
- Full Text
- View/download PDF
10. Bilirubin Reduces the Uptake of Estrogen Precursors and the Followed Synthesis of Estradiol in Human Placental Syncytiotrophoblasts via Inhibition and Downregulation of Organic Anion Transporter 4
- Author
-
Zhang, Yingqiong, Chen, Yujia, Dai, Binxin, Bai, Mengru, Lu, Shuanghui, Lin, Nengming, Zhou, Hui, and Jiang, Huidi
- Abstract
Estrogen biosynthesis in human placental trophoblasts requires the human organic anion transporter 4 (hOAT4)-mediated uptake of fetal derived precursors such as dehydroepiandrosterone-3-sulfate (DHEAS) and 16α-hydroxy-DHEA-S (16α-OH-DHEAS). Scant information is available concerning the contribution of fetal metabolites on the impact of placental estrogen precursor transport and the followed estrogen synthesis. This study substantiated the roles of bilirubin as well as bile acids (taurochenodeoxycholic acid, taurocholic acid, glycochenodeoxycholic acid, chenodeoxycholic acid) on the inhibition of hOAT4-mediated uptake of probe substrate 6-carboxylfluorescein and DHEAS in stably transfected hOAT4-Chinese hamster ovary cells, with the IC50of 1.53 and 0.98 μM on 6-carboxylfluorescein and DHEAS, respectively, for bilirubin, and 90.2, 129, 16.4, and 12.3 μM on 6-CF for taurochenodeoxycholic acid, glycochenodeoxycholic acid, taurocholic acid, and chenodeoxycholic acid. Bilirubin (2.5–10 μM) concentration-dependently inhibited the accumulation of estradiol precursor DHEAS in human choriocarcinoma JEG-3 cells (reduced by 60% at 10 μM) and primary human trophoblast cells (reduced by 80% at 10 μM). Further study confirmed that bilirubin (0.625–2.5 μM) concentration-dependently reduced the synthesis and secretion of estradiol in primary human trophoblast cells, among which 2.5 μM of bilirubin reduced the synthesis of estradiol by 30% and secretion by 35%. In addition, immunostaining and Western blot results revealed a distinct downregulation of hOAT4 protein expression in primary human trophoblast cells pretreated with 2.5 μM of bilirubin. In conclusion, this study demonstrated that bilirubin reduced the uptake of estrogen precursors and the followed synthesis of estradiol in human placenta viainhibition and downregulation of organic anion transporter 4.SIGNIFICANCE STATEMENTFetal metabolites, especially bilirubin, were first identified with significant inhibitory effects on the hOAT4-mediated uptake of estrogen precursor DHEAS in hOAT4-CHO, JEG-3 and PHTCs. Bilirubin concentration-dependently suppressed the estradiol synthesis and secretion in PHTCs treated with DHEAS, which was synchronized with the decline of hOAT4 protein expression. Additionally, those identified bile acids exhibited a weaker inhibitory effect on the secretion of estradiol.
- Published
- 2022
- Full Text
- View/download PDF
11. PLK1 (polo like kinase 1)-dependent autophagy facilitates gefitinib-induced hepatotoxicity by degrading COX6A1 (cytochrome c oxidase subunit 6A1)
- Author
-
Luo, Peihua, Yan, Hao, Du, Jiangxia, Chen, Xueqin, Shao, Jinjin, Zhang, Ying, Xu, Zhifei, Jin, Ying, Lin, Nengming, Yang, Bo, and He, Qiaojun
- Abstract
ABSTRACTLiver dysfunction is an outstanding dose-limiting toxicity of gefitinib, an EGFR (epidermal growth factor receptor)-tyrosine kinase inhibitor (TKI), in the treatment of EGFR mutation-positive non-small cell lung cancer (NSCLC). We aimed to elucidate the mechanisms underlying gefitinib-induced hepatotoxicity, and provide potentially effective intervention strategy. We discovered that gefitinib could sequentially activate macroautophagy/autophagy and apoptosis in hepatocytes. The inhibition of autophagy alleviated gefitinib-induced apoptosis, whereas the suppression of apoptosis failed to lessen gefitinib-induced autophagy. Moreover, liver-specific Atg7+/−heterozygous mice showed less severe liver injury than vehicle, suggesting that autophagy is involved in the gefitinib-promoted hepatotoxicity. Mechanistically, gefitinib selectively degrades the important anti-apoptosis factor COX6A1 (cytochrome c oxidase subunit 6A1) in the autophagy-lysosome pathway. The gefitinib-induced COX6A1 reduction impairs mitochondrial respiratory chain complex IV (RCC IV) function, which in turn activates apoptosis, hence causing liver injury. Notably, this autophagy-promoted apoptosis is dependent on PLK1 (polo like kinase 1). Both AAV8-mediated Plk1knockdown and PLK1 inhibitor BI-2536 could mitigate the gefitinib-induced hepatotoxicity in vivoby abrogating the autophagic degradation of the COX6A1 protein. In addition, PLK1 inhibition could not compromise the anti-cancer activity of gefitinib. In conclusion, our findings reveal the gefitinib-hepatotoxicity pathway, wherein autophagy promotes apoptosis through COX6A1 degradation, and highlight pharmacological inhibition of PLK1 as an attractive therapeutic approach toward improving the safety of gefitinib-based cancer therapy.Abbreviations:3-MA: 3-methyladenine; AAV8: adeno-associated virus serotype 8; ATG5: autophagy related 5; ATG7: autophagy related 7; B2M: beta-2-microglobulin; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CHX: cycloheximide; COX6A1: cytochrome c oxidase subunit 6A1; c-PARP: cleaved poly(ADP-ribose) polymerase; CQ: chloroquine; GOT1/AST: glutamic-oxaloacetic transaminase 1, soluble; GPT/ALT: glutamic pyruvic transaminase, soluble; HBSS: Hanks´ balanced salt solution; H&E: hematoxylin and eosin; MAP1LC3/LC3: microtubule associated proteins 1 light chain 3; PLK1: polo like kinase 1; RCC IV: respiratory chain complex IV; ROS: reactive oxygen species; TUBB8: tubulin beta 8 class VIII
- Published
- 2021
- Full Text
- View/download PDF
12. Efficacy and safety of daptomycin versus linezolid treatment in patients with vancomycin-resistant enterococcal bacteraemia: An updated systematic review and meta-analysis
- Author
-
Shi, Changcheng, Jin, Weizhong, Xie, Yaping, Zhou, Dongmei, Xu, Shuang, Li, Qingyu, and Lin, Nengming
- Abstract
•Compared with linezolid treatment, daptomycin treatment showed comparable clinical and microbiological outcomes but a lower incidence of thrombocytopenia.•High-dose daptomycin treatment should be considered for patients with VRE bacteraemia.•More studies comparing diverse high-dose daptomycin and linezolid regimens are needed to determine if a proper high-dose regimen exists.
- Published
- 2020
- Full Text
- View/download PDF
13. The Ibr‐7 derivative of ibrutinib exhibits enhanced cytotoxicity against non‐small cell lung cancer cells via targeting of mTORC1/S6 signaling.
- Author
-
Zhang, Bo, Wang, Linling, Zhang, Qi, Yan, Youyou, Jiang, Hong, Hu, Runlei, Zhou, Xinglu, Liu, Xingguo, Feng, Jianguo, and Lin, Nengming
- Abstract
Ibrutinib is a small molecule drug that targets Bruton's tyrosine kinase in B‐cell malignancies and is highly efficient at killing mantle cell lymphoma and chronic lymphocytic leukemia. However, the anti‐cancer activity of ibrutinib against solid tumors, such as non‐small cell lung cancer (NSCLC), remains low. To improve the cytotoxicity of ibrutinib towards lung cancer, we synthesized a series of ibrutinib derivatives, of which Ibr‐7 exhibited superior anti‐cancer activity to ibrutinib, especially against epithelial growth factor receptor (EGFR) wild‐type NSCLC cell lines. Ibr‐7 was observed to dramatically suppress the mammalian target of Rapamycin complex 1 (mTORC1)/S6 signaling pathway, which is only slightly affected by ibrutinib, thus accounting for the superior anti‐cancer activity of Ibr‐7 towards NSCLC. Ibr‐7 was shown to overcome the elevation of Mcl‐1 caused by ABT‐199 mono‐treatment, and thus exhibited a significant synergistic effect when combined with ABT‐199. In conclusion, we used a molecular substitution method to generate a novel ibrutinib derivative, termed Ibr‐7, which exhibits enhanced anti‐cancer activity against NSCLC cells as compared with the parental compound. Ibr‐7 exhibits antitumor activity against EGFR wild‐type and mutant NSCLC cells via inhibition of the mTOR‐S6 pathway; the parent compound of Ibr‐7, ibrutinib, does not affect this pathway. The anti‐proliferation effect of Ibr‐7 against freshly cultured primary lung cancer cells was greater than that of ibrutinib and AZD‐9291, irrespective of pathological patterns or EGFR mutation status. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
14. Discovery of a Highly Potent and Selective BRD9 PROTAC Degrader Based on E3 Binder Investigation for the Treatment of Hematological Tumors
- Author
-
Duan, Haiting, Zhang, Jingyu, Gui, Renzhao, Lu, Yang, Pang, Ao, Chen, Beijing, Shen, Liteng, Yu, Hengyuan, Li, Jia, Xu, Tengfei, Wang, Yuwei, Yao, Xiaojun, Zhang, Bo, Lin, Nengming, Dong, Xiaowu, Zhou, Yubo, and Che, Jinxin
- Abstract
BRD9 is a pivotal epigenetic factor involved in cancers and inflammatory diseases. Still, the limited selectivity and poor phenotypic activity of targeted agents make it an atypically undruggable target. PROTAC offers an alternative strategy for overcoming the issue. In this study, we explored diverse E3 ligase ligands for the contribution of BRD9 PROTAC degradation. Through molecular docking, binding affinity analysis, and structure–activity relationship study, we identified a highly potent PROTAC E5, with excellent BRD9 degradation (DC50= 16 pM) and antiproliferation in MV4–11 cells (IC50= 0.27 nM) and OCI-LY10 cells (IC50= 1.04 nM). E5can selectively degrade BRD9 and induce cell cycle arrest and apoptosis. Moreover, the therapeutic efficacy of E5was confirmed in xenograft tumor models, accompanied by further RNA-seq analysis. Therefore, these results may pave the way and provide the reference for the discovery and investigation of highly effective PROTAC degraders.
- Published
- 2024
- Full Text
- View/download PDF
15. A Pluripotential Neutrophil-Mimic Nanovehicle Modulates Immune Microenvironment with Targeted Drug Delivery for Augmented Antitumor Chemotherapy
- Author
-
Wu, Jiahe, Ma, Teng, Zhu, Manning, Mu, Jiafu, Huang, Tianchen, Xu, Donghang, Lin, Nengming, and Gao, Jianqing
- Abstract
The limited therapeutic outcomes and severe systemic toxicity of chemotherapy remain major challenges to the current clinical antitumor therapeutic regimen. Tumor-targeted drug delivery that diminishes the undifferentiated systemic distribution is a practical solution to ameliorating systemic toxicity. However, the tumor adaptive immune microenvironment still poses a great threat that compromises the therapeutic efficacy of chemotherapy by promoting the tolerance of the tumor cells. Herein, a pluripotential neutrophil-mimic nanovehicle (Neutrosome(L)) composed of an activated neutrophil membrane-incorporated liposome is proposed to modulate the immune microenvironment and synergize antitumor chemotherapy. The prominent tumor targeting capability inherited from activated neutrophils and the improved tumor penetration ability of Neutrosome(L) enable considerable drug accumulation in tumor tissues (more than sixfold that of free drug). Importantly, Neutrosome(L) can modulate the immune microenvironment by restricting neutrophil infiltration in tumor tissue, which may be attributed to the neutralization of inflammatory cytokines, thus potentiating antitumor chemotherapy. As a consequence, the treatment of cisplatin-loaded Neutrosome(L) performs prominent tumor suppression effects, reduces systemic drug toxicity, and prolongs the survival period of tumor-bearing mice. The pluripotential neutrophil-mimic nanovehicle proposed in this study can not only enhance the tumor accumulation of chemotherapeutics but also modulate the immune microenvironment, providing a compendious strategy for augmented antitumor chemotherapy.
- Published
- 2024
- Full Text
- View/download PDF
16. Targeting the cell signaling pathway Keap1-Nrf2 as a therapeutic strategy for adenocarcinomas of the lung
- Author
-
Zhang, Bo, Ma, Zhiyuan, Tan, Biqin, and Lin, Nengming
- Abstract
ABSTRACTIntroduction: Kelch-like ECH associated protein 1/Nuclear factor erythroid 2-like factor 2 (Keap1-Nrf2) signaling plays a pivotal role in response to oxidative stress in lung cancer. Mutations in KEAP1/NFE2L2 genes always cause persistent Nrf2 activation in lung cancer cells that confer therapeutic resistance and aggressive tumorigenic activity, dictating either poor prognosis or short duration of response to chemotherapy in clinical observations.Areas covered: We provide a review of the mechanisms underlying the regulation of Keap1-Nrf2 at different stages, including genetic mutations, epigenetic modifications, translational/post-translational alterations, and protein–protein interactions. Based on the current knowledge, we discuss the possibilities of intervening Keap1-Nrf2 in lung adenocarcinoma as a therapeutic target.Expert opinion: It is prevalently conceived that Keap1-Nrf2 signaling plays different roles at diverse stages of cancer. Although various Nrf2 or Keap1 inhibitors have been reported during the last decades, none of these inhibitors are currently under clinical studies or in clinical applications, suggesting that sole inhibition of Nrf2 might not be sufficient to suppress tumor growth. On the basis of current studies, we suggest that the rational combination of Nrf2 suppression with chemical agents which cause enhanced oxidative imbalance or abnormal metabolism would be promising in the treatment of lung adenocarcinoma.
- Published
- 2019
- Full Text
- View/download PDF
17. Histone H3 Lysine 4 Trimethylation, Lysine 27 Trimethylation, and Lysine 27 Acetylation Contribute to the Transcriptional Repression of Solute Carrier Family 47 Member 2 in Renal Cell Carcinoma
- Author
-
Yu, Qinqin, Liu, Yanqing, Zheng, Xiaoli, Zhu, Qianying, Shen, Zhuowei, Wang, Hua, He, Huadong, Lin, Nengming, Jiang, Huidi, Yu, Lushan, and Zeng, Su
- Abstract
In recent years, finding effective biomarkers for identifying early stage cancer and predicating prognosis is crucial for renal cell carcinoma (RCC) diagnosis and treatment. In this study, a dramatic decrease of the solute carrier family 47 member 2 (SLC47A2) mRNA in RCC comparing with the paired adjacent nontumor tissues from patients at low Tumor Node Metastasis stage was observed. Thus, patients with SLC47A2transcriptional repression are susceptible to RCC. Little is known about the regulation mechanism of SLC47A2. We found that it was a bivalent gene that was enriched with both histone H3 lysine 4 trimethylation (H3K4me3) and lysine 27 trimethylation (H3K27me3). Loss of mixed lineage leukemia 1 binding at the gene promoter caused decreased H3K4me3 enrichment and H3K4me3/H3K27me3 ratio, and subsequently repressed the expression of SLC47A2. These two epigenetic markers modulated the expression of SLC47A2simultaneously, suggesting the regulation pattern for bivalent genes. Histone H3 lysine 27 acetylation also contributed to the expression of SLC47A2. An E2F1-histone deacetylase 10 complex catalyzed deacetylation of H3K27, then prevented the enrichment of H3K4me3, and finally reduced SLC47A2expression. Consequently, the combined effect of all these factors determined SLC47A2transcriptional repression in RCC tissues.
- Published
- 2017
- Full Text
- View/download PDF
18. Nanotechnology reinforced neutrophil-based therapeutic strategies for inflammatory diseases therapy.
- Author
-
Wu, Jiahe, Ma, Teng, Zhu, Manning, Huang, Tianchen, Zhang, Bo, Gao, Jianqing, and Lin, Nengming
- Subjects
NANOTECHNOLOGY ,NEUTROPHILS ,BIOAVAILABILITY ,THERAPEUTICS ,SMALL molecules ,IMMUNE system ,IMMUNOREGULATION - Abstract
Misguided attack of immune systems on self-tissue or multifactorial uncontrolled inflammation is capable of causing inflammatory diseases. Neutrophils play a critical role in the initiation of inflammation and pathophysiological progression, and the pathological context can influence the biology of neutrophils. Moreover, they usually respond faster than other immune cells to pathological variation. Recently, due to their immunomodulation capability and intrinsic tropism to inflammatory sites, neutrophil-based therapeutic strategies, including neutrophil manipulation and development of neutrophil-based delivery systems, are emerging as a promising solution for the treatment of inflammation-related diseases. However, obstacles in neutrophil manipulation using small molecules or antibodies (e.g., lack of targeting ability to the inflammatory site, low bioavailability, and fast clearance) and the concerns about developing neutrophil-based delivery systems (e.g., stability of biopharmaceuticals, activity maintenance, and cargo loading efficiency) hindered the therapeutic outcomes of neutrophil-based therapeutic strategies. Nanotechnology, which has offered multiple resolutions for disease treatment, can be utilized to reinforce neutrophil-based therapeutic strategies. In order to afford a comprehensive overview of the state-of-the-art progress of this emerging field, the biology of neutrophils and their function in a certain inflammatory context is briefly introduced. Then the current research progresses about nanomaterials augmented efficient neutrophils manipulation and developing neutrophil-based carriers utilizing nanotechnology for inflammation-targeted treatment are illustrated with specific research examples. Finally, perspectives in light of the current challenges and future opportunities in this field are discussed. [Display omitted] • Progress of nanotechnology-reinforced neutrophil-based therapeutic strategies is summarized. • Nanotechnology assists neutrophil manipulation for inflammatory diseases therapy. • Nanotechnology augments neutrophil-based carriers for inflammation-targeted therapy. • The challenges and perspectives of neutrophil-based therapeutic strategies are discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
19. Vernodalin Suppresses Tumor Proliferation and Increases Apoptosis of Gastric Cancer Cells Through Attenuation of FAK/PI3K/AKT/mTOR and MAPKs Signaling Pathways
- Author
-
Luo, Ying, Zhang, Dan, Hou, Leping, and Lin, Nengming
- Abstract
Background: Gastric cancer (GC) is the most aggressive malignant tumor with limited treatment alternatives post metastasis. Vernodalin (VN) induced apoptosis has been reported in various types of human cancer cells. However, the precise molecular mechanisms underlying the anti-metastasis action of VN on GC cells are yet to be elucidated. Objective: In this study, we investigated the anti-metastatic and apoptotic effects of VN on SGC- 7901 and AGS cells, with a purpose of gaining a deeper understanding of the anti-metastatic mechanisms of VN on gastric carcinoma. To attenuate the activation of PI3K/AKT/mTOR and mitogen-activated protein kinase (MAPK) signaling pathways by VN in GC cells. Methods: We employed VN and gastric cancer cells in experiments such as MTT assay, apoptosis, MMP, DAPI, Rh-123, cell adhesion assay, and western blot analysis on GC SGC-7901 and AGS cells. Results: Our results revealed that VN inhibits cell proliferation, adhesion, and metastasis and induces apoptosis of both GC cells. VN potentially reduced the protein expressions of MMP-2, MMP-9, and uPA, whereas intensified expressions of TIMP-1 and TIMP-2. Also, VN attenuates the expression of FAK, p-PI3K, p-AKT, p-mTOR, p-JNK, p-p38MAPK, and p-ERK. Thus, it is inferred that VN treatment reduced the activities of MMP-2 and MMP-9 via the FAK/PI3K/AKT/ mTOR, and MAPKs signaling pathways. Our results confirm that VN prevented GC growth, invasion and metastasis and induce apoptosis in GC cells. Conclusion: Our findings suggest that VN is a potential natural therapeutic compound as a new remedy for GC chemotherapy treatment.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.