Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Pathol ; 259(2): 205-219, 2023 02.
Article in English | MEDLINE | ID: mdl-36373776

ABSTRACT

Colorectal cancer (CRC) is one of the most common cancers worldwide. The tumor microenvironment exerts crucial effects in driving CRC progression. Cancer-associated fibroblasts (CAFs) serve as one of the most important tumor microenvironment components promoting CRC progression. This study aimed to elucidate the novel molecular mechanisms of CAF-secreted insulin-like growth factor (IGF) 2 in colorectal carcinogenesis. Our results indicated that IGF2 was a prominent factor upregulated in CAFs compared with normal fibroblasts. CAF-derived conditioned media (CM) promoted tumor growth, migration, and invasion of HCT 116 and DLD-1 cells. IGF1R expression is significantly increased in CRC, serving as a potent receptor in response to IGF2 stimulation and predicting unfavorable outcomes for CRC patients. Apart from the PI3K-AKT pathway, RNA-seq analysis revealed that the YAP1-target signature serves as a prominent downstream effector to mediate the oncogenic signaling of IGF2-IGF1R. By single-cell RNA sequencing (scRNA-seq) and immunohistochemical validation, IGF2 was found to be predominantly secreted by CAFs, whereas IGF1R was expressed mainly by cancer cells. IGF2 triggers the nuclear accumulation of YAP1 and upregulates YAP1 target signatures; however, these effects were abolished by either IGF1R knockdown or inhibition with picropodophyllin (PPP), an IGF1R inhibitor. Using CRC organoid and in vivo studies, we found that cotargeting IGF1R and YAP1 with PPP and verteporfin (VP), a YAP1 inhibitor, enhanced antitumor effects compared with PPP treatment alone. In conclusion, this study revealed a novel molecular mechanism by which CAFs promote CRC progression. The findings highlight the translational potential of the IGF2-IGF1R-YAP1 axis as a prognostic biomarker and therapeutic target for CRC. © 2022 The Pathological Society of Great Britain and Ireland.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Humans , Cancer-Associated Fibroblasts/pathology , Phosphatidylinositol 3-Kinases/metabolism , Cell Line, Tumor , Signal Transduction , Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Cell Proliferation , Tumor Microenvironment , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor II/pharmacology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology
2.
J Pathol ; 260(4): 402-416, 2023 08.
Article in English | MEDLINE | ID: mdl-37272544

ABSTRACT

Homeobox genes include HOX and non-HOX genes. HOX proteins play fundamental roles during ontogenesis by interacting with other non-HOX gene-encoded partners and performing transcriptional functions, whereas aberrant activation of HOX family members drives tumorigenesis. In this study, gastric cancer (GC) expression microarray data indicated that HOXB9 is a prominent upregulated HOX member in GC samples significantly associated with clinical outcomes and advanced TNM stages. However, the functional role of HOXB9 in GC remains contradictory in previous reports, and the regulatory mechanisms are elusive. By in silico and experimental analyses, we found that HOXB9 was upregulated by a vital cell cycle-related transcription factor, E2F1. Depleting HOXB9 causes G1-phase cell cycle arrest by downregulating CDK6 and a subset of cell cycle-related genes. Meanwhile, HOXB9 contributes to cell division and maintains the cytoskeleton in GC cells. We verified that HOXB9 interacts with PBX2 to form a heterodimer, which transcriptionally upregulates CDK6. Knocking down CDK6 can phenocopy the tumor-suppressive effects caused by HOXB9 depletion. Blocking HOXB9 can enhance the anti-tumor effect of CDK6 inhibitors. In conclusion, we elucidate the oncogenic role of HOXB9 in GC and reveal CDK6 as its potent downstream effector. The E2F1-HOXB9/PBX2-CDK6 axis represents a novel mechanism driving gastric carcinogenesis and conveys prognostic and therapeutic implications. © 2023 The Pathological Society of Great Britain and Ireland.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Genes, Homeobox , Cell Line, Tumor , Carcinogenesis/pathology , Transcription Factors/genetics , Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Gene Expression Regulation, Neoplastic , Cell Proliferation/physiology , Proto-Oncogene Proteins/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism
3.
Int J Cancer ; 152(8): 1510-1525, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36093588

ABSTRACT

The use of cell cycle inhibitors has necessitated a better understanding of the cell cycle in tumor biology to optimize the therapeutic approach. Cell cycle aberrations are common in cancers, and it is increasingly acknowledged that these aberrations exert oncogenic effects beyond the cell cycle. Multiple facets such as cancer metabolism, immunity and metastasis are also affected, all of which are beyond the effect of cell proliferation alone. This review comprehensively summarized the important recent findings and advances in these interrelated processes. In cancer metabolism, cell cycle regulators can modulate various pathways in aerobic glycolysis, glucose uptake and gluconeogenesis, mainly through transcriptional regulation and kinase activities. Amino acid metabolism is also regulated through cell cycle progression. On cancer metastasis, metabolic plasticity, immune evasion, tumor microenvironment adaptation and metastatic site colonization are intricately related to the cell cycle, with distinct regulatory mechanisms at each step of invasion and dissemination. Throughout the synthesis of current understanding, knowledge gaps and limitations in the literature are also highlighted, as are new therapeutic approaches such as combinational therapy and challenges in tackling emerging targeted therapy resistance. A greater understanding of how the cell cycle modulates diverse aspects of cancer biology can hopefully shed light on identifying new molecular targets by harnessing the vast potential of the cell cycle.


Subject(s)
Neoplasms , Humans , Neoplasms/pathology , Citric Acid Cycle , Carbohydrate Metabolism , Cell Division , Cell Cycle , Glycolysis , Tumor Microenvironment
4.
Int J Cancer ; 152(4): 558-571, 2023 02 15.
Article in English | MEDLINE | ID: mdl-35983734

ABSTRACT

Accumulating evidence has underscored the importance of the Hippo-YAP1 signaling in lung tissue homeostasis, whereas its deregulation induces tumorigenesis. YAP1 and its paralog TAZ are the key downstream effectors tightly controlled by the Hippo pathway. YAP1/TAZ exerts oncogenic activities by transcriptional regulation via physical interaction with TEAD transcription factors. In solid tumors, Hippo-YAP1 crosstalks with other signaling pathways such as Wnt/ß-catenin, receptor tyrosine kinase cascade, Notch and TGF-ß to synergistically drive tumorigenesis. As YAP1/TAZ expression is significantly correlated with unfavorable outcomes for the patients, small molecules have been developed for targeting YAP1/TAZ to get a therapeutic effect. In this review, we summarize the recent findings on the deregulation of Hippo-YAP1 pathway in nonsmall cell lung carcinoma, discuss the molecular mechanisms of its dysregulation in leading to tumorigenesis, explore the therapeutic strategies for targeting YAP1/TAZ, and provide the research directions for deep investigation. We believe that detailed delineation of Hippo-YAP1 regulation in tumorigenesis provides novel insight for accurate therapeutic intervention.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma , Lung Neoplasms , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Trans-Activators/metabolism , YAP-Signaling Proteins , Precision Medicine , Carcinoma, Non-Small-Cell Lung/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Lung Neoplasms/genetics , Lung/metabolism
5.
Cell Biol Toxicol ; 39(4): 1297-1317, 2023 08.
Article in English | MEDLINE | ID: mdl-36070022

ABSTRACT

Autophagy is typically activated in cancer cells as a rescue strategy in response to cellular stress (e.g., chemotherapy). Herein, we found that Berbamine Hydrochloride (Ber) can act as an effective inhibitor of the late stage of autophagic flux, thereby potentiating the killing effect of chemotherapy agents. Lung carcinoma cells exposed to Ber exhibited increased autophagosomes, marked by LC3-II upregulation. The increased level of p62 after Ber treatment indicated that the autophagic flux was blocked at the late stage. The lysosome staining assay and cathepsin maturation detection indicated impaired lysosomal acidification. We found that Nox2 exhibited intensified co-localization with lysosomes in Ber-treated cells. Nox2 is a key enzyme for superoxide anion production capable of transferring electrons into the lysosomal lumen, thereby neutralizing the inner protons; this might explain the aberrant acidification. This hypothesis is further supported by the observed reversal of lysosomal cathepsin maturation by Nox2 inhibitors. Finally, Ber combined with cisplatin exhibited a synergistic killing effect on lung carcinoma cells. Further data suggested that lung carcinoma cells co-treated with Ber and cisplatin accumulated excessive reactive oxygen species (ROS), which typically activated MAPK-mediated mitochondria-dependent apoptosis. The enhanced anti-cancer effect of Ber combined with cisplatin was also confirmed in an in vivo xenograft mouse model. These findings indicate that Ber might be a promising adjuvant for enhancing the cancer cell killing effect of chemotherapy via the inhibition of autophagy. In this process, Nox2 might be a significant mediator of Ber-induced aberrant lysosomal acidification.


Subject(s)
Antineoplastic Agents , Carcinoma , Lung Neoplasms , Humans , Animals , Mice , Reactive Oxygen Species/metabolism , Cisplatin/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy , Apoptosis , Lysosomes/metabolism , Lung/metabolism , Hydrogen-Ion Concentration , Cathepsins/metabolism , Cathepsins/pharmacology , Cathepsins/therapeutic use , Carcinoma/drug therapy , Carcinoma/metabolism
6.
Phytother Res ; 37(9): 3820-3838, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37421347

ABSTRACT

We conducted a meta-analysis to evaluate the clinical efficacy of berberine (BBR) in treating acute ischemic stroke (AIS), explore its anti-inflammatory effects, and assess its potential applications for AIS patients. We comprehensively searched nine databases from inception until July 1, 2022, to identify clinical trials investigating the use of BBR in treating AIS. We performed statistical analyses using RevMan5.4 software and focused on primary outcomes such as inflammatory markers as well as secondary outcomes including immune system indicators, relevant biomarkers, carotid artery atherosclerosis, and adverse reactions. Our analysis included data from 17 clinical trials involving 1670 patients with AIS. Our results revealed that BBR in combination with conventional treatment significantly reduced levels of high-sensitivity C-reactive protein (hs-CRP), macrophage migration inhibitory factor (MIF), interleukin-6 (IL-6), complement C3, hypoxic inducible factor-1 α (HIF-1α), cysteine protease-3 (Caspase-3), the national institutes of health stroke scale (NIHSS), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), carotid intima-media thickness (IMT), the number of unstable plaques, and carotid crouse score on ultrasound when compared with conventional treatment alone. Furthermore, combining BBR with conventional treatment may improve the overall effective rate. Therefore, our findings suggest that BBR can be used as an adjuvant therapy for AIS due to its ability to reduce inflammatory cytokine levels, providing a novel therapeutic option for AIS. However, larger randomized controlled trials are necessary to confirm these results.


Subject(s)
Atherosclerosis , Berberine , Ischemic Stroke , Stroke , Humans , Ischemic Stroke/drug therapy , Berberine/therapeutic use , Carotid Intima-Media Thickness , Triglycerides , Atherosclerosis/drug therapy , Stroke/drug therapy
7.
Int J Cancer ; 151(8): 1195-1215, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35603909

ABSTRACT

Lung cancer is the common and leading cause of cancer death worldwide. The tumor microenvironment has been recognized to be instrumental in tumorigenesis. To have a deep understanding of the molecular mechanism of nonsmall cell lung carcinoma (NSCLC), cancer-associated fibroblasts (CAFs) have gained increasing research interests. CAFs belong to the crucial and dominant cell population in the tumor microenvironment to support the cancer cells. The interplay and partnership between cancer cells and CAFs contribute to each stage of tumorigenesis. CAFs exhibit prominent heterogeneity and secrete different kinds of cytokines and chemokines, growth factors and extracellular matrix proteins involved in cancer cell proliferation, invasion, metastasis and chemoresistance. Many studies focused on the protumorigenic functions of CAFs, yet many challenges about the heterogeneity of CAFS remain unresolved. This review comprehensively summarized the tumor-promoting role and molecular mechanisms of CAFs in NSCLC, including their origin, phenotypic changes and heterogeneity and their functional roles in carcinogenesis. Meanwhile, we also highlighted the updated molecular classifications based on the molecular features and functional roles of CAFs. With the development of cutting-edge platforms and further investigations of CAFs, novel therapeutic strategies for accurately targeting CAFs in NSCLC may be developed based on the increased understanding of the relevant molecular mechanisms.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Cancer-Associated Fibroblasts/metabolism , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Transformation, Neoplastic/metabolism , Fibroblasts/pathology , Humans , Lung Neoplasms/pathology , Tumor Microenvironment
8.
Mol Carcinog ; 58(6): 875-886, 2019 06.
Article in English | MEDLINE | ID: mdl-30644610

ABSTRACT

The protein EPHB4 plays a vital role in various tumor types. However, few studies into the function of circ-EPHB4 (hsa_circ_0001730) in tumors have been conducted. This study aimed to investigate the functions of circ-EPHB4 and the underlying mechanism of circ-EPHB4 in regulating hepatocellular carcinoma (HCC). The expression of circ-EPHB4 was found to be downregulated in HCC tumor tissues, whereas circ-EPHB4 overexpression suppressed cell viability, induced apoptosis, and inhibited cell migration and invasion in Huh7 and HepG2 cells. circ-EPHB4 levels were negatively correlated with tumor weight, size, and metastasis foci in nude mouse models, suggesting circ-EPHB4 inhibits tumorigenesis, tumor development, and metastasis. In addition, HIF-1α and PI3K-AKT pathways were markedly affected by circ-EPHB4 overexpression. HIF-1α could potentially be the target of circ-EPHB4. By overexpressing both HIF-1α and circ-EPHB4, the antitumor effect of circ-EPHB4 should be most probably correlated with HIF-1α. In conclusion, circ-EPHB4 is a tumor inhibitor in HCC and functions by inhibiting HIF-1α expression.


Subject(s)
Carcinoma, Hepatocellular/pathology , Gene Expression Profiling/methods , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Neoplasms/pathology , RNA/genetics , Animals , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Down-Regulation , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Circular
9.
Int J Mol Sci ; 20(14)2019 Jul 11.
Article in English | MEDLINE | ID: mdl-31336784

ABSTRACT

The main mechanistic function of most chemotherapeutic drugs is mediated by inducing mitochondria-dependent apoptosis. Tumor cells usually respond to upregulate autophagy to eliminate impaired mitochondria for survival. Hypothetically, inhibiting autophagy might promote mitochondria-dependent apoptosis, thus enhancing the efficacy of chemotherapeutic therapies. We previously identified N-methylparoxetine (NMP) as an inducer of mitochondrial fragmentation with subsequent apoptosis in non-small cell lung cancer (NSCLC) cells. We discovered that ROS was accumulated in NMP-treated NSCLC cells, followed by c-Jun N-terminal kinase (JNK) and p38 MAP kinase (p38) activation. This was reversed by the application of a reactive oxygen species (ROS) scavenger, N-acetylcysteine (NAC), leading to a reduction in apoptosis. Our data suggested that NMP induced apoptosis in NSCLC cells by activating mitogen-activated protein kinase (MAPK) pathway. We further speculated that the remarkable increase of ROS in NMP-treated NSCLC cells might result from an inhibition of autophagy. Our current data confirmed that NMP blocked autophagy flux at late stage wherein lysosomal acidification was inhibited. Taken together, this study demonstrated that NMP could exert dual apoptotic functions-mitochondria impairment and, concomitantly, autophagy inhibition. NMP-related excessive ROS accumulation induced apoptosis by activating the MAPK pathway in NSCLC cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , Paroxetine/pharmacology , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lysosomes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Paroxetine/analogs & derivatives , Paroxetine/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Molecules ; 24(12)2019 Jun 14.
Article in English | MEDLINE | ID: mdl-31197076

ABSTRACT

Dioscin is a natural steroidal saponin that can be isolated from Chinese medicine, such as Dioscoreae rhizoma. It has wild range of pharmacological activities such as hepatoprotection, a lipid-lowering effect, and anti-inflammation. Recently, mounting studies reported the anticancer effect of dioscin on a variety of tumor cells. However, the potential effect of dioscin on the epithelial-mesenchymal transition (EMT) of HepG2 cells is unclear. In the present study, dioscin was identified to inhibit transforming growth factor-ß1 (TGF-ß1) and induced invasive and migratory behavior of HepG2 cells. Consistently, the expression of the epithelial marker E-cadherin and gap junction proteins increased following dioscin treatment, while mesenchymal markers decreased, including N-cadherin, Vimentin, Snail, and Slug. Furthermore, we discovered that TGF-ß1 induces phosphorylation of JNK, p38, and Erk, whereas the activation of these kinases was reversed by dioscin treatment in a dose-dependent manner. With the addition of Asiatic acid, a p38 activator, the inhibitory effect of dioscin on EMT was reversed. Taken together, these data indicated that dioscin inhibits EMT in HepG2 cells, which is mediated in large part by inhibition of the p38-MAPK signaling.


Subject(s)
Cell Proliferation/drug effects , Diosgenin/analogs & derivatives , Liver Neoplasms/drug therapy , Transforming Growth Factor beta1/genetics , Cell Movement/drug effects , Diosgenin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplasm Invasiveness/genetics , Pentacyclic Triterpenes/pharmacology , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
11.
Int J Mol Sci ; 19(10)2018 Oct 18.
Article in English | MEDLINE | ID: mdl-30340379

ABSTRACT

Chemoresistance is a major limiting factor that impairs the outcome of non-small cell lung cancer (NSCLC) chemotherapy. Paclitaxel (Tax) induces protective autophagy in NSCLC cells, leading to the development of drug resistance. We recently identified a new autophagy inhibitor (alpha-hederin) and hypothesized that it may promote the killing effect of Tax on NSCLC cells. We found that alpha-hederin (α-Hed) could block late autophagic flux in NSCLC cells by altering lysosomal pH and inhibiting lysosomal cathepsin D maturation. Combination treatment of α-Hed and Tax synergistically reduced NSCLC cell proliferation and increased NSCLC cell apoptosis compared with treatment with α-Hed or Tax alone. Furthermore, α-Hed plus Tax enhanced the accumulation of intracellular reactive oxygen species (ROS) in NSCLC cells, while the ROS inhibitor N-acetylcysteine reversed the inhibitory effect of the combination treatment. Our findings suggest that α-Hed can increase the killing effect of Tax on NSCLC cells by promoting ROS accumulation, and that combining α-Hed with classical Tax represents a novel strategy for treating NSCLC.


Subject(s)
Autophagy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Oleanolic Acid/analogs & derivatives , Paclitaxel/pharmacology , Reactive Oxygen Species/metabolism , Saponins/metabolism , Apoptosis/drug effects , Autophagosomes/metabolism , Cathepsins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Hydrogen-Ion Concentration , Lysosomes/metabolism , Oleanolic Acid/metabolism , Paclitaxel/chemistry , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
13.
Biomed Pharmacother ; 177: 116952, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38917754

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH) and liver fibrosis are progressive conditions associated with non-alcoholic fatty liver disease (NAFLD), characterized by hepatocyte pyroptosis and hepatic stellate cell (HSC) activation. Gentiopicroside (GPS) has emerged as a potential treatment for NASH, yet its underlying mechanism remains unclear. AIM: To confirm that GPS can improve NASH and liver fibrosis by blocking the NLRP3 signaling pathway STUDY DESIGN: Initially, different animal models were used to study the effects and mechanisms of GPS on NASH and fibrosis. Subsequent in vitro experiments utilized co-cultures and other techniques to delve deeper into its mechanism, followed by validation of the findings in mouse liver tissues. METHODS: C57BL/6 mice were fed high-fat, high-cholesterol (HFHC), or methionine-choline-deficient (MCD) diets to induce NASH and fibrosis. RAW264.7 cells and born marrow bone marrow-derived macrophages (BMDMs) were stimulated with LPS and ATP to induce inflammation, then co-cultured with primary hepatocytes and HSCs, treated with GPS, and its efficacy and mechanism were analyzed. RESULTS: In vivo, GPS alleviated NASH and liver fibrosis by inhibiting the NLRP3 pathway. In vitro, GPS attenuated inflammation induced by BMDMs by inhibiting TLR4 and NLRP3 signaling pathways, and Co-culture studies suggested that GPS reduced hepatocyte pyroptosis and HSC activation, which was also confirmed in liver tissues CONCLUSION: GPS improves NASH and liver fibrosis by inhibiting the TLR4 and NLRP3 signaling pathways. The specific mechanism may be related to the suppression of macrophage-mediated inflammatory responses, thereby reducing hepatocyte pyroptosis and HSC activation.


Subject(s)
Iridoid Glucosides , Liver Cirrhosis , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Non-alcoholic Fatty Liver Disease , Signal Transduction , Toll-Like Receptor 4 , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Toll-Like Receptor 4/metabolism , Mice , Signal Transduction/drug effects , Iridoid Glucosides/pharmacology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Male , RAW 264.7 Cells , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Disease Models, Animal , Diet, High-Fat
14.
Pharmacol Ther ; 253: 108576, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38065232

ABSTRACT

RNA ac4C modification is a novel and rare chemical modification observed in mRNA. Traditional biochemical studies had primarily associated ac4C modification with tRNA and rRNA until in 2018, Arango D et al. first reported the presence of ac4C modification on mRNA and demonstrated its critical role in mRNA stability and translation regulation. Furthermore, they established that the ac4C modification on mRNA is mediated by the classical N-acetyltransferase NAT10. Subsequent studies have underscored the essential implications of NAT10 and mRNA ac4C modification across both physiological and pathological regulatory processes. In this review, we aimed to explore the discovery history of RNA ac4C modification, its detection methods, and its regulatory mechanisms in disease and physiological development. We offer a forward-looking examination and discourse concerning the employment of RNA ac4C modification as a prospective therapeutic strategy across diverse diseases. Furthermore, we comprehensively summarize the functions and mechanisms of NAT10 in gene expression regulation and pathogenesis independent of RNA ac4C modification.


Subject(s)
Mammals , N-Terminal Acetyltransferases , Animals , Humans , RNA, Messenger , Mammals/genetics
15.
Oncogene ; 43(2): 77-91, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38081962

ABSTRACT

Gastrointestinal (GI) cancer is a formidable malignancy with significant morbidity and mortality rates. Recent studies have shed light on the complex interplay between the nervous system and the GI system, influencing various aspects of GI tumorigenesis, such as the malignance of cancer cells, the conformation of tumor microenvironment (TME), and the resistance to chemotherapies. The discussion in this review first focused on exploring the intricate details of the biological function of the nervous system in the development of the GI tract and the progression of tumors within it. Meanwhile, the cancer cell-originated feedback regulation on the nervous system is revealed to play a crucial role in the growth and development of nerve cells within tumor tissues. This interaction is vital for understanding the complex relationship between the nervous system and GI oncogenesis. Additionally, the study identified various components within the TME that possess a significant influence on the occurrence and progression of GI cancer, including microbiota, immune cells, and fibroblasts. Moreover, we highlighted the transformation relationship between non-neuronal cells and neuronal cells during GI cancer progression, inspiring the development of strategies for nervous system-guided anti-tumor drugs. By further elucidating the deep mechanism of various neuroregulatory signals and neuronal intervention, we underlined the potential of these targeted drugs translating into effective therapies for GI cancer treatment. In summary, this review provides an overview of the mechanisms of neuromodulation and explores potential therapeutic opportunities, providing insights into the understanding and management of GI cancers.


Subject(s)
Antineoplastic Agents , Gastrointestinal Neoplasms , Humans , Gastrointestinal Neoplasms/pathology , Carcinogenesis , Cell Transformation, Neoplastic , Antineoplastic Agents/therapeutic use , Neurons , Tumor Microenvironment
16.
Signal Transduct Target Ther ; 9(1): 6, 2024 01 03.
Article in English | MEDLINE | ID: mdl-38169461

ABSTRACT

Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.


Subject(s)
Biological Phenomena , Zinc , Humans , Zinc/metabolism , Homeostasis , Membrane Transport Proteins , Disease Progression
17.
Adv Sci (Weinh) ; 11(18): e2307899, 2024 May.
Article in English | MEDLINE | ID: mdl-38460164

ABSTRACT

Gastric cancer (GC) presents a formidable global health challenge, and conventional therapies face efficacy limitations. Ubiquitin-specific protease 7 (USP7) plays pivotal roles in GC development, immune response, and chemo-resistance, making it a promising target. Various USP7 inhibitors have shown selectivity and efficacy in preclinical studies. However, the mechanistic role of USP7 has not been fully elucidated, and currently, no USP7 inhibitors have been approved for clinical use. In this study, DHPO is identified as a potent USP7 inhibitor for GC treatment through in silico screening. DHPO demonstrates significant anti-tumor activity in vitro, inhibiting cell viability and clonogenic ability, and preventing tumor migration and invasion. In vivo studies using orthotopic gastric tumor mouse models validate DHPO's efficacy in suppressing tumor growth and metastasis without significant toxicity. Mechanistically, DHPO inhibition triggers ferroptosis, evidenced by mitochondrial alterations, lipid Reactive Oxygen Species (ROS), Malondialdehyde (MDA) accumulation, and iron overload. Further investigations unveil USP7's regulation of Stearoyl-CoA Desaturase (SCD) through deubiquitination, linking USP7 inhibition to SCD degradation and ferroptosis induction. Overall, this study identifies USP7 as a key player in ferroptosis of GC, elucidates DHPO's inhibitory mechanisms, and highlights its potential for GC treatment by inducing ferroptosis through SCD regulation.


Subject(s)
Ferroptosis , Stearoyl-CoA Desaturase , Stomach Neoplasms , Ubiquitin-Specific Peptidase 7 , Animals , Humans , Mice , Cell Line, Tumor , Disease Models, Animal , Ferroptosis/drug effects , Ferroptosis/genetics , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/genetics
18.
Cancer Res ; 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39047223

ABSTRACT

The Hippo-YAP1 pathway is an evolutionally conserved signaling cascade that controls organ size and tissue regeneration. Dysregulation of Hippo-YAP1 signaling promotes initiation and progression of several types of cancer, including gastric cancer (GC). As the Hippo-YAP1 pathway regulates expression of thousands of genes, it is important to establish which target genes contribute to the oncogenic program driven by YAP1 to identify strategies to circumvent it. Here, we identified a vital role of FOXP4 in YAP1-driven gastric carcinogenesis by maintaining stemness and promoting peritoneal metastasis. Loss of FOXP4 impaired GC spheroid formation and reduced stemness marker expression, while FOXP4 upregulation potentiated cancer cell stemness. RNA-seq analysis revealed SOX12 as downstream target of FOXP4, and functional studies established that SOX12 supports stemness in YAP1-induced carcinogenesis. A small molecule screen identified 42-(2-Tetrazolyl)rapamycin as a FOXP4 inhibitor, and targeting FOXP4 suppressed GC tumor growth and enhanced the efficacy of 5-FU chemotherapy in vivo. Collectively, these findings revealed that FOXP4 upregulation by YAP1 in GC regulates stemness and tumorigenesis by upregulating SOX12. Targeting the YAP1-FOXP4-SOX12 axis represents a potential therapeutic strategy for GC.

19.
Cancers (Basel) ; 15(9)2023 May 06.
Article in English | MEDLINE | ID: mdl-37174105

ABSTRACT

Despite the decline in incidence and mortality rates, gastric cancer (GC) is the fifth leading cause of cancer deaths worldwide. The incidence and mortality of GC are exceptionally high in Asia due to high H. pylori infection, dietary habits, smoking behaviors, and heavy alcohol consumption. In Asia, males are more susceptible to developing GC than females. Variations in H. pylori strains and prevalence rates may contribute to the differences in incidence and mortality rates across Asian countries. Large-scale H. pylori eradication was one of the effective ways to reduce GC incidences. Treatment methods and clinical trials have evolved, but the 5-year survival rate of advanced GC is still low. Efforts should be put towards large-scale screening and early diagnosis, precision medicine, and deep mechanism studies on the interplay of GC cells and microenvironments for dealing with peritoneal metastasis and prolonging patients' survival.

20.
J Ethnopharmacol ; 307: 116216, 2023 May 10.
Article in English | MEDLINE | ID: mdl-36736714

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Traditional Chinese medicine (TCM) has extensive healing effects on inflammatory diseases with few side effects. Reduning injection (RDNI), a TCM prescription composed of Lonicera japonica Thunb., Gardenia jasminoides Ellis. and Artemisia annua L., is wildly used for treating inflammatory diseases. However, the mechanism of action of RDNI, like most TCM prescriptions, is unclear due to the complexity of relationships between components and their curative effects. AIM OF THE STUDY: To develop a universal systems pharmacology protocol for mechanism modeling of TCM and apply it to reveal the real-time anti-inflammatory effect of Reduning Injection. MATERIALS AND METHODS: Combined with database mining and references, a regulatory mechanism network of inflammation was constructed. A quantitative model was established afterwards by integrating pharmacokinetic data and two network parameters, namely Network Efficiency and Network Flux. The time-dependent and dose-response relationship of RDNI on the regulation of inflammation was then quantitatively evaluated. ELISA tests were performed to verify the reliability of the model. RESULTS: Three cytokines, namely IL-1ß, IL-6 and TNF-α were screened out to be markers for evaluation of the anti-inflammatory effect of RDNI. An HPLC method for the simultaneous determination of 10 RDNI compounds in SD rat plasma was established and then applied to pharmacokinetic studies. Based on compound activity and pharmacokinetic data, the time-dependent effect of RDNI were quantitatively predicted by the pathway network-based modeling procedure. CONCLUSIONS: The quantitative model established in this work was successfully applied to predict a TCM prescription's real-time dynamic healing effect after administration. It is qualified to provide novel insights into the time-dependent and dose-effect relationship of drugs in an intricate biological system and new strategies for investigating the detailed molecular mechanisms of TCM.


Subject(s)
Drugs, Chinese Herbal , Rats , Animals , Reproducibility of Results , Rats, Sprague-Dawley , Drugs, Chinese Herbal/pharmacology , Medicine, Chinese Traditional/methods , Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL