Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46.435
Filter
2.
Vet Res ; 55(1): 84, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965634

ABSTRACT

Pseudorabies virus (PRV) has evolved multiple strategies to evade host antiviral responses to benefit virus replication and establish persistent infection. Recently, tripartite motif 26 (TRIM26), a TRIM family protein, has been shown to be involved in a broad range of biological processes involved in innate immunity, especially in regulating viral infection. Herein, we found that the expression of TRIM26 was significantly induced after PRV infection. Surprisingly, the overexpression of TRIM26 promoted PRV production, while the depletion of this protein inhibited virus replication, suggesting that TRIM26 could positively regulate PRV infection. Further analysis revealed that TRIM26 negatively regulates the innate immune response by targeting the RIG-I-triggered type I interferon signalling pathway. TRIM26 was physically associated with MAVS independent of viral infection and reduced MAVS expression. Mechanistically, we found that NDP52 interacted with both TRIM26 and MAVS and that TRIM26-induced MAVS degradation was almost entirely blocked in NDP52-knockdown cells, demonstrating that TRIM26 degrades MAVS through NDP52-mediated selective autophagy. Our results reveal a novel mechanism by which PRV escapes host antiviral innate immunity and provide insights into the crosstalk among virus infection, autophagy, and the innate immune response.


Subject(s)
Adaptor Proteins, Signal Transducing , Autophagy , Immunity, Innate , Animals , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Swine , Virus Replication , Humans , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
3.
Elife ; 132024 Jul 12.
Article in English | MEDLINE | ID: mdl-38995680

ABSTRACT

Truncation of the protein-protein interaction SH3 domain of the membrane remodeling Bridging Integrator 1 (BIN1, Amphiphysin 2) protein leads to centronuclear myopathy. Here, we assessed the impact of a set of naturally observed, previously uncharacterized BIN1 SH3 domain variants using conventional in vitro and cell-based assays monitoring the BIN1 interaction with dynamin 2 (DNM2) and identified potentially harmful ones that can be also tentatively connected to neuromuscular disorders. However, SH3 domains are typically promiscuous and it is expected that other, so far unknown partners of BIN1 exist besides DNM2, that also participate in the development of centronuclear myopathy. In order to shed light on these other relevant interaction partners and to get a holistic picture of the pathomechanism behind BIN1 SH3 domain variants, we used affinity interactomics. We identified hundreds of new BIN1 interaction partners proteome-wide, among which many appear to participate in cell division, suggesting a critical role of BIN1 in the regulation of mitosis. Finally, we show that the identified BIN1 mutations indeed cause proteome-wide affinity perturbation, signifying the importance of employing unbiased affinity interactomic approaches.


Subject(s)
Adaptor Proteins, Signal Transducing , Myopathies, Structural, Congenital , Nuclear Proteins , Tumor Suppressor Proteins , src Homology Domains , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/genetics , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Protein Binding , Dynamin II/metabolism , Dynamin II/genetics , Mutation
4.
Oncol Rep ; 52(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38963044

ABSTRACT

Lysine methyltransferase 5A (KMT5A) is the sole mammalian enzyme known to catalyse the mono­methylation of histone H4 lysine 20 and non­histone proteins such as p53, which are involved in the occurrence and progression of numerous cancers. The present study aimed to determine the function of KMT5A in inducing docetaxel (DTX) resistance in patients with breast carcinoma by evaluating glucose metabolism and the underlying mechanism involved. The upregulation or downregulation of KMT5A­related proteins was examined after KMT5A knockdown in breast cancer (BRCA) cells by Tandem Mass Tag proteomics. Through differential protein expression and pathway enrichment analysis, the upregulated key gluconeogenic enzyme fructose­1,6­bisphosphatase 1 (FBP1) was discovered. Loss of FBP1 expression is closely related to the development and prognosis of cancers. A dual­luciferase reporter gene assay confirmed that KMT5A inhibited the expression of FBP1 and that overexpression of FBP1 could enhance the chemotherapeutic sensitivity to DTX through the suppression of KMT5A expression. The KMT5A inhibitor UNC0379 was used to verify that DTX resistance induced by KMT5A through the inhibition of FBP1 depended on the methylase activity of KMT5A. According to previous literature and interaction network structure, it was revealed that KMT5A acts on the transcription factor twist family BHLH transcription factor 1 (TWIST1). Then, it was verified that TWSIT1 promoted the expression of FBP1 by using a dual­luciferase reporter gene experiment. KMT5A induces chemotherapy resistance in BRCA cells by promoting cell proliferation and glycolysis. After the knockdown of the KMT5A gene, the FBP1 related to glucose metabolism in BRCA was upregulated. KMT5A knockdown expression and FBP1 overexpression synergistically inhibit cell proliferation and block cells in the G2/M phase. KMT5A inhibits the expression of FBP1 by methylating TWIST1 and weakening its promotion of FBP1 transcription. In conclusion, KMT5A was shown to affect chemotherapy resistance by regulating the cell cycle and positively regulate glycolysis­mediated chemotherapy resistance by inhibiting the transcription of FBP1 in collaboration with TWIST1. KMT5A may be a potential therapeutic target for chemotherapy resistance in BRCA.


Subject(s)
Breast Neoplasms , Docetaxel , Drug Resistance, Neoplasm , Fructose-Bisphosphatase , Gene Expression Regulation, Neoplastic , Nuclear Proteins , Twist-Related Protein 1 , Humans , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Female , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Fructose-Bisphosphatase/genetics , Fructose-Bisphosphatase/metabolism , Docetaxel/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Cell Proliferation/drug effects , DNA Methylation
5.
Nat Commun ; 15(1): 5964, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013853

ABSTRACT

Meiotic rapid prophase chromosome movements (RPMs) require connections between the chromosomes and the cytoskeleton, involving SUN (Sad1/UNC-84)-domain-containing proteins at the inner nuclear envelope (NE). RPMs remain significantly understudied in plants, with respect to their importance in the regulation of meiosis. Here, we demonstrate that Arabidopsis thaliana meiotic centromeres undergo rapid (up to 500 nm/s) and uncoordinated movements during the zygotene and pachytene stages. These centromere movements are not affected by altered chromosome organization and recombination but are abolished in the double mutant sun1 sun2. We also document the changes in chromosome dynamics and nucleus organization during the transition from leptotene to zygotene, including telomere attachment to SUN-enriched NE domains, bouquet formation, and nucleolus displacement, all of which were defective in sun1 sun2. These results establish A. thaliana as a model species for studying the functional implications of meiotic RPMs and demonstrate the mechanistic conservation of telomere-led RPMs in plants.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Chromosomes, Plant , Meiosis , Nuclear Envelope , Telomere , Arabidopsis/genetics , Arabidopsis/metabolism , Nuclear Envelope/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Chromosomes, Plant/genetics , Telomere/metabolism , Centromere/metabolism , Prophase , Meiotic Prophase I , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
6.
Cell Death Dis ; 15(7): 506, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013911

ABSTRACT

NOP2, a member of the NOL1/NOP2/SUN domain (NSUN) family, is responsible for catalyzing the posttranscriptional modification of RNA through 5-methylcytosine (m5C). Dysregulation of m5C modification has been linked to the pathogenesis of various malignant tumors. Herein, we investigated the expression of NOP2 in lung adenocarcinoma (LUAD) tissues and cells, and found that it was significantly upregulated. Moreover, lentivirus-mediated overexpression of NOP2 in vitro resulted in enhanced migration and invasion capabilities of lung cancer cells, while in vivo experiments demonstrated its ability to promote the growth and metastasis of xenograft tumors. In contrast, knockdown of NOP2 effectively inhibited the growth and metastasis of lung cancer cells. RNA-sequencing was conducted to ascertain the downstream targets of NOP2, and the findings revealed a significant upregulation in EZH2 mRNA expression upon overexpression of NOP2. Subsequent validation experiments demonstrated that NOP2 exerted an m5C-dependent influence on the stability of EZH2 mRNA. Additionally, our investigations revealed a co-regulatory relationship between NOP2 and the m5C reader protein ALYREF in modulating the stability of EZH2 mRNA. Notably, the NOP2/EZH2 axis facilitated the malignant phenotype of lung cancer cells by inducing epithelial-mesenchymal transition (EMT) both in vitro and in vivo. Mechanistically, ChIP analysis proved that EZH2 counteracted the impact of NOP2 on the occupancy capacity of EZH2 and H3K27me3 in the promoter regions of E-cadherin, a gene crucial for regulating EMT. In a word, our research highlights the significant role of NOP2 in LUAD and offers novel mechanistic insights into the NOP2/ALYREF/EZH2 axis, which holds promise as a potential target for lung cancer therapy.


Subject(s)
Enhancer of Zeste Homolog 2 Protein , Epithelial-Mesenchymal Transition , Lung Neoplasms , RNA Stability , Humans , Epithelial-Mesenchymal Transition/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , RNA Stability/genetics , Mice , Mice, Nude , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Disease Progression , Methylation , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Mice, Inbred BALB C , Female , Cell Movement/genetics , Male , RNA, Messenger/metabolism , RNA, Messenger/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , A549 Cells , Cell Proliferation/genetics
7.
Cell Death Dis ; 15(7): 520, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039052

ABSTRACT

Aldehyde dehydrogenases superfamily (ALDHs), which are ubiquitously present in various organisms with diverse subcellular localizations, play a crucial role in regulating malignant tumor progression; Nevertheless, their involvement in clear cell renal cell carcinoma (ccRCC) has not been elucidated. In this study, we performed comprehensive bioinformatics analyses on the 19 ALDHs genes, and identified ALDH9A1 as a key contributor in ccRCC. Expression patterns and clinical relevance of ALDH9A1 were determined using bioinformatics analyses, real-time PCR, western blotting, and immunohistochemistry. To explore the underlying mechanism behind the tumor suppressor role of ALDH9A1, RNA sequencing, methylated RNA immunoprecipitation, luciferase reporter assay, mass spectroscopy, immunoprecipitation, mutational studies and immunofluorescence were employed. The impact of ALDH9A1 in ccRCC progression and metabolic programming was assessed through both in vitro and in vivo. Here, this study revealed ALDH9A1 as a tumor suppressor gene in ccRCC. The fat mass and obesity associated protein (FTO) was identified as a demethylase for ALDH9A1 mRNA, resulting in its reduced stability and expression levels in ccRCC. Functional experiments demonstrated that the deficiency of ALDH9A1 in ccRCC promoted tumor proliferation, invasion, migration and lipid accumulation. Mechanistic insights illustrated that the diminished levels of ALDH9A1 resulted in the failure to sequester nucleophosmin 1 (NPM1) within cytoplasm, thereby suppressing the transcription of IQ motif containing the GTPase-activating protein 2 (IQGAP2), subsequently activating the AKT-mTOR signaling, ultimately fostering tumor progression and lipid accumulation. In conclusion, the present study highlights the robust prognostic significance of ALDH9A1 and delivers a comprehensive understanding of ALDH9A1-NPM1-IQGAP2-AKT axis in ccRCC. These findings established a solid research foundation for novel therapeutic strategies for ccRCC patients.


Subject(s)
Adenosine , Carcinoma, Renal Cell , Kidney Neoplasms , Nucleophosmin , Proto-Oncogene Proteins c-akt , Signal Transduction , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Proto-Oncogene Proteins c-akt/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Disease Progression , Cell Line, Tumor , Mice , Mice, Nude , Cell Proliferation , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Gene Expression Regulation, Neoplastic , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Male , Female
8.
PLoS Biol ; 22(4): e3001767, 2024 Apr.
Article in English | MEDLINE | ID: mdl-39038273

ABSTRACT

The 18S rRNA sequence is highly conserved, particularly at its 3'-end, which is formed by the endonuclease Nob1. How Nob1 identifies its target sequence is not known, and in vitro experiments have shown Nob1 to be error-prone. Moreover, the sequence around the 3'-end is degenerate with similar sites nearby. Here, we used yeast genetics, biochemistry, and next-generation sequencing to investigate a role for the ATPase Rio1 in monitoring the accuracy of the 18S rRNA 3'-end. We demonstrate that Nob1 can miscleave its rRNA substrate and that miscleaved rRNA accumulates upon bypassing the Rio1-mediated quality control (QC) step, but not in healthy cells with intact QC mechanisms. Mechanistically, we show that Rio1 binding to miscleaved rRNA is weaker than its binding to accurately processed 18S rRNA. Accordingly, excess Rio1 results in accumulation of miscleaved rRNA. Ribosomes containing miscleaved rRNA can translate, albeit more slowly, thereby inviting collisions with trailing ribosomes. These collisions result in degradation of the defective ribosomes utilizing parts of the machinery for mRNA QC. Altogether, the data support a model in which Rio1 inspects the 3'-end of the nascent 18S rRNA to prevent miscleaved 18S rRNA-containing ribosomes from erroneously engaging in translation, where they induce ribosome collisions. The data also demonstrate how ribosome collisions purify cells of altered ribosomes with different functionalities, with important implications for the concept of ribosome heterogeneity.


Subject(s)
RNA, Ribosomal, 18S , Ribosomes , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , RNA, Ribosomal, 18S/metabolism , RNA, Ribosomal, 18S/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Ribosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , RNA Stability/genetics , RNA Cleavage , RNA, Fungal/metabolism , RNA, Fungal/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
9.
Genome Med ; 16(1): 91, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39034402

ABSTRACT

BACKGROUND: The identification of cancer driver genes from sequencing data has been crucial in deepening our understanding of tumor biology and expanding targeted therapy options. However, apart from the most commonly altered genes, the mechanisms underlying the contribution of other mutations to cancer acquisition remain understudied. Leveraging on our whole-exome sequencing of the largest Asian lung adenocarcinoma (LUAD) cohort (n = 302), we now functionally assess the mechanistic role of a novel driver, PARP4. METHODS: In vitro and in vivo tumorigenicity assays were used to study the functional effects of PARP4 loss and mutation in multiple lung cancer cell lines. Interactomics analysis by quantitative mass spectrometry was conducted to identify PARP4's interaction partners. Transcriptomic data from cell lines and patient tumors were used to investigate splicing alterations. RESULTS: PARP4 depletion or mutation (I1039T) promotes the tumorigenicity of KRAS- or EGFR-driven lung cancer cells. Disruption of the vault complex, with which PARP4 is commonly associated, did not alter tumorigenicity, indicating that PARP4's tumor suppressive activity is mediated independently. The splicing regulator hnRNPM is a potentially novel PARP4 interaction partner, the loss of which likewise promotes tumor formation. hnRNPM loss results in splicing perturbations, with a propensity for dysregulated intronic splicing that was similarly observed in PARP4 knockdown cells and in LUAD cohort patients with PARP4 copy number loss. CONCLUSIONS: PARP4 is a novel modulator of lung adenocarcinoma, where its tumor suppressive activity is mediated not through the vault complex-unlike conventionally thought, but in association with its novel interaction partner hnRNPM, thus suggesting a role for splicing dysregulation in LUAD tumorigenesis.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group M , Lung Neoplasms , Nuclear Proteins , Animals , Humans , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein Group M/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group M/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Protein Binding , RNA Splicing , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
10.
Mol Biol Rep ; 51(1): 842, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042261

ABSTRACT

BACKGROUND: Although Huaier granules can be used as prospective anti-cholangiocarcinoma drugs, the mechanism of action of Huaier granules in cholangiocarcinoma is not clear. The anti-cholangiocarcinoma effect of Huaier granules was validated in cell line research. In vitro experiments were conducted to investigate the signalling pathways affected by Huaier in CCA cells. METHODS AND RESULTS: Real-time quantitative PCR (RT‒qPCR) and Western blot analysis were performed to analyse gene expression in CCA cells. MTT assays, scratch tests, and Transwell assays were used to explore the effects on the proliferation and metastasis of CCA cells. Chromatin immunoprecipitation assays were performed to reveal the potential underlying mechanisms involved. Twist1 was upregulated in human CCA tissues. In addition, its expression levels were negatively related to FBP1 expression levels. Mechanistically, Twist1 can bind to the region of the FBP1 promoter to reduce its expression. Huaier plays an indispensable role in suppressing Twist1 expression to inhibit the Twist1/FBP1/Wnt/ß-catenin axis. Then, we verified the effect of Huaier in vitro. CONCLUSIONS: These findings suggested that Huaier granules were capable of inhibiting CCA development through regulating the Twist1/FBP1/Wnt/ß-catenin signalling axis and provided a novel orientation for the development of novel anti-CCA drugs.


Subject(s)
Bile Duct Neoplasms , Cell Proliferation , Cholangiocarcinoma , Gene Expression Regulation, Neoplastic , Nuclear Proteins , Twist-Related Protein 1 , Wnt Signaling Pathway , beta Catenin , Humans , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Cholangiocarcinoma/drug therapy , Wnt Signaling Pathway/drug effects , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cell Line, Tumor , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Cell Proliferation/drug effects , beta Catenin/metabolism , Cell Movement/drug effects , Cell Movement/genetics
11.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999944

ABSTRACT

Bri1-EMS Suppressor 1 (BES1) and Brassinazole Resistant 1 (BZR1) are two key transcription factors in the brassinosteroid (BR) signaling pathway, serving as crucial integrators that connect various signaling pathways in plants. Extensive genetic and biochemical studies have revealed that BES1 and BZR1, along with other protein factors, form a complex interaction network that governs plant growth, development, and stress tolerance. Among the interactome of BES1 and BZR1, several proteins involved in posttranslational modifications play a key role in modifying the stability, abundance, and transcriptional activity of BES1 and BZR1. This review specifically focuses on the functions and regulatory mechanisms of BES1 and BZR1 protein interactors that are not involved in the posttranslational modifications but are crucial in specific growth and development stages and stress responses. By highlighting the significance of the BZR1 and BES1 interactome, this review sheds light on how it optimizes plant growth, development, and stress responses.


Subject(s)
Arabidopsis Proteins , DNA-Binding Proteins , Gene Expression Regulation, Plant , Nuclear Proteins , Plant Development , Stress, Physiological , Plant Development/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Transcription Factors/metabolism , Brassinosteroids/metabolism , Signal Transduction , Protein Processing, Post-Translational , Protein Binding
12.
Cancer Biol Ther ; 25(1): 2375440, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38978225

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most lethal malignant tumors worldwide. Brahma-related gene 1 (BRG1), as a catalytic ATPase, is a major regulator of gene expression and is known to mutate and overexpress in HCC. The purpose of this study was to investigate the mechanism of action of BRG1 in HCC cells. In our study, BRG1 was silenced or overexpressed in human HCC cell lines. Transwell and wound healing assays were used to analyze cell invasiveness and migration. Mitochondrial membrane potential (MMP) and mitochondrial permeability transition pore (mPTP) detection were used to evaluate mitochondrial function in HCC cells. Colony formation and cell apoptosis assays were used to evaluate the effect of BRG1/TOMM40/ATP5A1 on HCC cell proliferation and apoptosis/death. Immunocytochemistry (ICC), immunofluorescence (IF) staining and western blot analysis were used to determine the effect of BRG1 on TOMM40, ATP5A1 pathway in HCC cells. As a result, knockdown of BRG1 significantly inhibited cell proliferation and invasion, promoted apoptosis in HCC cells, whereas BRG1 overexpression reversed the above effects. Overexpression of BRG1 can up-regulate MMP level, inhibit mPTP opening and activate TOMM40, ATP5A1 expression. Our results suggest that BRG1, as an oncogene, promotes HCC progression by regulating TOMM40 affecting mitochondrial function and ATP5A1 synthesis. Targeting BRG1 may represent a new and effective way to prevent HCC development.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular , Cell Proliferation , DNA Helicases , Liver Neoplasms , Mitochondria , Mitochondrial Precursor Protein Import Complex Proteins , Nuclear Proteins , Transcription Factors , Humans , DNA Helicases/metabolism , DNA Helicases/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Mitochondria/metabolism , Membrane Potential, Mitochondrial , Neoplasm Metastasis , Cell Movement , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Gene Expression Regulation, Neoplastic , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/genetics
13.
Front Immunol ; 15: 1426620, 2024.
Article in English | MEDLINE | ID: mdl-39035010

ABSTRACT

The nucleotide-binding and oligomerization domain-like receptors (NLRs) NLR family CARD domain-containing protein 5 (NLRC5) and Class II Major Histocompatibility Complex Transactivator (CIITA) are transcriptional regulators of major histocompatibility complex (MHC) class I and class II genes, respectively. MHC molecules are central players in our immune system, allowing the detection of hazardous 'non-self' antigens and, thus, the recognition and elimination of infected or transformed cells from the organism. Recently, CIITA and NLRC5 have emerged as regulators of selected genes of the butyrophilin (BTN) family that interestingly are located in the extended MHC locus. BTNs are transmembrane proteins exhibiting structural similarities to B7 family co-modulatory molecules. The family member BTN2A2, which indeed contributes to the control of T cell activation, was found to be transcriptionally regulated by CIITA. NLRC5 emerged instead as an important regulator of the BTN3A1, BTN3A2, and BTN3A3 genes. Together with BTN2A1, BTN3As regulate non-conventional Vγ9Vδ2 T cell responses triggered by selected metabolites of microbial origin or accumulating in hematologic cancer cells. Even if endogenous metabolites conform to the canonical definition of 'self', metabolically abnormal cells can represent a danger for the organism and should be recognized and controlled by immune system cells. Collectively, new data on the role of NLRC5 in the expression of BTN3As link the mechanisms regulating canonical 'non-self' presentation and those marking cells with abnormal metabolic configurations for immune recognition, an evolutionary parallel that we discuss in this perspective review.


Subject(s)
Butyrophilins , Intracellular Signaling Peptides and Proteins , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Butyrophilins/metabolism , Butyrophilins/genetics , Butyrophilins/immunology , Animals , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Gene Expression Regulation , Lymphocyte Activation/immunology , Antigens, CD
14.
Pharmacol Res ; 206: 107283, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964523

ABSTRACT

The maintenance of nuclear shape is essential for cellular homeostasis and disruptions in this process have been linked to various pathological conditions, including cancer, laminopathies, and aging. Despite the significance of nuclear shape, the precise molecular mechanisms controlling it are not fully understood. In this study, we have identified the YEATS domain-containing protein 4 (GAS41) as a previously unidentified factor involved in regulating nuclear morphology. Genetic ablation of GAS41 in colorectal cancer cells resulted in significant abnormalities in nuclear shape and inhibited cancer cell proliferation both in vitro and in vivo. Restoration experiments revealed that wild-type GAS41, but not a YEATS domain mutant devoid of histone H3 lysine 27 acetylation or crotonylation (H3K27ac/cr) binding, rescued the aberrant nuclear phenotypes in GAS41-deficient cells, highlighting the importance of GAS41's binding to H3K27ac/cr in nuclear shape regulation. Further experiments showed that GAS41 interacts with H3K27ac/cr to regulate the expression of key nuclear shape regulators, including LMNB1, LMNB2, SYNE4, and LEMD2. Mechanistically, GAS41 recruited BRD2 and the Mediator complex to gene loci of these regulators, promoting their transcriptional activation. Disruption of GAS41-H3K27ac/cr binding caused BRD2, MED14 and MED23 to dissociate from gene loci, leading to nuclear shape abnormalities. Overall, our findings demonstrate that GAS41 collaborates with BRD2 and the Mediator complex to control the expression of crucial nuclear shape regulators.


Subject(s)
Cell Nucleus , Colorectal Neoplasms , Transcription Factors , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Animals , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Mice , Histones/metabolism , Mice, Nude , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , HCT116 Cells , Bromodomain Containing Proteins
15.
Nature ; 631(8021): 663-669, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961290

ABSTRACT

The Warburg effect is a hallmark of cancer that refers to the preference of cancer cells to metabolize glucose anaerobically rather than aerobically1,2. This results in substantial accumulation of lacate, the end product of anaerobic glycolysis, in cancer cells3. However, how cancer metabolism affects chemotherapy response and DNA repair in general remains incompletely understood. Here we report that lactate-driven lactylation of NBS1 promotes homologous recombination (HR)-mediated DNA repair. Lactylation of NBS1 at lysine 388 (K388) is essential for MRE11-RAD50-NBS1 (MRN) complex formation and the accumulation of HR repair proteins at the sites of DNA double-strand breaks. Furthermore, we identify TIP60 as the NBS1 lysine lactyltransferase and the 'writer' of NBS1 K388 lactylation, and HDAC3 as the NBS1 de-lactylase. High levels of NBS1 K388 lactylation predict poor patient outcome of neoadjuvant chemotherapy, and lactate reduction using either genetic depletion of lactate dehydrogenase A (LDHA) or stiripentol, a lactate dehydrogenase A inhibitor used clinically for anti-epileptic treatment, inhibited NBS1 K388 lactylation, decreased DNA repair efficacy and overcame resistance to chemotherapy. In summary, our work identifies NBS1 lactylation as a critical mechanism for genome stability that contributes to chemotherapy resistance and identifies inhibition of lactate production as a promising therapeutic cancer strategy.


Subject(s)
Cell Cycle Proteins , DNA Breaks, Double-Stranded , Drug Resistance, Neoplasm , MRE11 Homologue Protein , Nuclear Proteins , Humans , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Cell Cycle Proteins/metabolism , Animals , MRE11 Homologue Protein/metabolism , Mice , Nuclear Proteins/metabolism , Cell Line, Tumor , Lysine Acetyltransferase 5/metabolism , Lysine Acetyltransferase 5/genetics , Recombinational DNA Repair , DNA-Binding Proteins/metabolism , Lactic Acid/metabolism , Lysine/metabolism , Female , DNA Repair Enzymes/metabolism , DNA Repair/drug effects , Male , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/genetics , Acid Anhydride Hydrolases
16.
CNS Neurosci Ther ; 30(7): e14850, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39021287

ABSTRACT

INTRODUCTION: Glioma is the most frequent and lethal form of primary brain tumor. The molecular mechanism of oncogenesis and progression of glioma still remains unclear, rendering the therapeutic effect of conventional radiotherapy, chemotherapy, and surgical resection insufficient. In this study, we sought to explore the function of HEC1 (highly expressed in cancer 1) in glioma; a component of the NDC80 complex in glioma is crucial in the regulation of kinetochore. METHODS: Bulk RNA and scRNA-seq analyses were used to infer HEC1 function, and in vitro experiments validated its function. RESULTS: HEC1 overexpression was observed in glioma and was indicative of poor prognosis and malignant clinical features, which was confirmed in human glioma tissues. High HEC1 expression was correlated with more active cell cycle, DNA-associated activities, and the formation of immunosuppressive tumor microenvironment, including interaction with immune cells, and correlated strongly with infiltrating immune cells and enhanced expression of immune checkpoints. In vitro experiments and RNA-seq further confirmed the role of HEC1 in promoting cell proliferation, and the expression of DNA replication and repair pathways in glioma. Coculture assay confirmed that HEC1 promotes microglial migration and the transformation of M1 phenotype macrophage to M2 phenotype. CONCLUSION: Altogether, these findings demonstrate that HEC1 may be a potential prognostic marker and an immunotherapeutic target in glioma.


Subject(s)
Brain Neoplasms , Glioma , Macrophages , RNA-Seq , Humans , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Prognosis , Macrophages/metabolism , Single-Cell Analysis , Male , Female , Tumor Microenvironment/genetics , Cell Line, Tumor , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Middle Aged , Cell Proliferation , Single-Cell Gene Expression Analysis , Cytoskeletal Proteins
17.
Mol Cell Probes ; 76: 101971, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38977039

ABSTRACT

OBJECTIVE: This study aimed to conclude the effect and mechanism of ZIC2 on immune infiltration in lung adenocarcinoma (LUAD). METHODS: Expression of ZIC2 in several kinds of normal tissues of TCGA data was analyzed and its correlation with the baseline characteristic of LUAD patients were analyzed. The immune infiltration analysis of LUAD patients was performed by CIBERSORT algorithm. The correlation analysis between ZIC2 and immune cell composition was performed. Additionally, the potential upstream regulatory mechanisms of ZIC2 were predicted to identify the possible miRNAs and lncRNAs that regulated ZIC2 in LUAD. In vitro and in vivo experiments were also conducted to confirm the potential effect of ZIC2 on cell proliferation and invasion ability of LUAD cells. RESULTS: ZIC2 expression was decreased in various normal tissues, but increased in multiple tumors, including LUAD, and correlated with the prognosis of LUAD patients. Enrichment by GO and KEGG suggested the possible association of ZIC2 with cell cycle and p53 signal pathway. ZIC2 expression was significantly correlated with T cells CD4 memory resting, Macrophages M1, and plasma cells, indicating that dysregulated ZIC2 expression in LUAD may directly influence immune infiltration. ZIC2 might be regulated by several different lncRNA-mediated ceRNA mechanisms. In vitro experiments validated the promotive effect of ZIC2 on cell viability and invasion ability of LUAD cells. In vivo experiments validated ZIC2 can accelerate tumor growth in nude mouse. CONCLUSION: ZIC2 regulated by different lncRNA-mediated ceRNA mechanisms may play a critical regulatory role in LUAD through mediating the composition of immune cells in tumor microenvironment.


Subject(s)
Adenocarcinoma of Lung , Cell Proliferation , Computational Biology , Gene Expression Regulation, Neoplastic , Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , Mice , Mice, Nude , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Competitive Endogenous
18.
Nat Commun ; 15(1): 6059, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025847

ABSTRACT

Synthetic lethality provides an attractive strategy for developing targeted cancer therapies. For example, cancer cells with high levels of microsatellite instability (MSI-H) are dependent on the Werner (WRN) helicase for survival. However, the mechanisms that regulate WRN spatiotemporal dynamics remain poorly understood. Here, we used single-molecule tracking (SMT) in combination with a WRN inhibitor to examine WRN dynamics within the nuclei of living cancer cells. WRN inhibition traps the helicase on chromatin, requiring p97/VCP for extraction and proteasomal degradation in a MSI-H dependent manner. Using a phenotypic screen, we identify the PIAS4-RNF4 axis as the pathway responsible for WRN degradation. Finally, we show that co-inhibition of WRN and SUMOylation has an additive toxic effect in MSI-H cells and confirm the in vivo activity of WRN inhibition using an MSI-H mouse xenograft model. This work elucidates a regulatory mechanism for WRN that may facilitate identification of new therapeutic modalities, and highlights the use of SMT as a tool for drug discovery and mechanism-of-action studies.


Subject(s)
Chromatin , Protein Inhibitors of Activated STAT , Valosin Containing Protein , Werner Syndrome Helicase , Werner Syndrome Helicase/metabolism , Werner Syndrome Helicase/genetics , Humans , Animals , Chromatin/metabolism , Valosin Containing Protein/metabolism , Valosin Containing Protein/genetics , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/genetics , Mice , Cell Line, Tumor , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Microsatellite Instability , Proteolysis/drug effects , Sumoylation/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Xenograft Model Antitumor Assays , Female
19.
Zhongguo Fei Ai Za Zhi ; 27(6): 466-470, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-39026498

ABSTRACT

NUT carcinoma (nuclear protein in testis carcinoma) is a rare and highly invasive malignant tumor, which is most common in midline organs and lungs. The characteristic genetic change of NUT carcinoma is the rearrangement of NUT middle carcinoma family member 1 (NUTM1) gene. In this article, we will review the pathogenic mechanism of its most common fusion form, bromodomaincontaining protein 4 (BRD4)-NUTM1 fusion gene, and the progress in the research and development of targeting drugs.
.


Subject(s)
Nuclear Proteins , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Animals , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/therapy , Carcinoma/drug therapy , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Nat Commun ; 15(1): 5379, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956052

ABSTRACT

Targeted protein degradation is a groundbreaking modality in drug discovery; however, the regulatory mechanisms are still not fully understood. Here, we identify cellular signaling pathways that modulate the targeted degradation of the anticancer target BRD4 and related neosubstrates BRD2/3 and CDK9 induced by CRL2VHL- or CRL4CRBN -based PROTACs. The chemicals identified as degradation enhancers include inhibitors of cellular signaling pathways such as poly-ADP ribosylation (PARG inhibitor PDD00017273), unfolded protein response (PERK inhibitor GSK2606414), and protein stabilization (HSP90 inhibitor luminespib). Mechanistically, PARG inhibition promotes TRIP12-mediated K29/K48-linked branched ubiquitylation of BRD4 by facilitating chromatin dissociation of BRD4 and formation of the BRD4-PROTAC-CRL2VHL ternary complex; by contrast, HSP90 inhibition promotes BRD4 degradation after the ubiquitylation step. Consequently, these signal inhibitors sensitize cells to the PROTAC-induced apoptosis. These results suggest that various cell-intrinsic signaling pathways spontaneously counteract chemically induced target degradation at multiple steps, which could be liberated by specific inhibitors.


Subject(s)
Cell Cycle Proteins , Proteolysis , Signal Transduction , Transcription Factors , Ubiquitination , Humans , Signal Transduction/drug effects , Proteolysis/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Cycle Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Cell Line, Tumor , Apoptosis/drug effects , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Bromodomain Containing Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...