Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.500
Filter
2.
Clin Exp Med ; 24(1): 212, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249558

ABSTRACT

Emerging evidence suggests that the APOBEC family is implicated in multiple cancers and might be utilized as a new target for cancer detection and treatment. However, the dysregulation and clinical implication of the APOBEC family in clear cell renal cell cancer (ccRCC) remain elusive. TCGA multiomics data facilitated a comprehensive exploration of the APOBEC family across cancers, including ccRCC. Remodeling analysis classified ccRCC patients into two distinct subgroups: APOBEC family pattern cancer subtype 1 (APCS1) and subtype 2 (APCS2). The study investigated differences in clinical parameters, tumor immune microenvironment, therapeutic responsiveness, and genomic mutation landscapes between these subtypes. An APOBEC family-related risk model was developed and validated for predicting ccRCC patient prognosis, demonstrating good sensitivity and specificity. Finally, the overview of APOBEC3B function was investigated in multiple cancers and verified in clinical samples. APCS1 and APCS2 demonstrated considerably distinct clinical features and biological processes in ccRCC. APCS1, an aggressive subtype, has advanced clinical stage and a poor prognosis. APCS1 exhibited an oncogenic and metabolically active phenotype. APCS1 also exhibited a greater tumor mutation load and immunocompromised condition, resulting in immunological dysfunction and immune checkpoint treatment resistance. The genomic copy number variation of APCS1, including arm gain and loss, was much more than that of APCS2, which may help explain the tired immune system. Furthermore, the two subtypes have distinct drug sensitivity patterns in clinical specimens and matching cell lines. Finally, we developed a predictive risk model based on subtype biomarkers that performed well for ccRCC patients and validated the clinical impact of APOBEC3B. Aberrant APOBEC family expression patterns might modify the tumor immune microenvironment by increasing the genome mutation frequency, thus inducing an immune-exhausted phenotype. APOBEC family-based molecular subtypes could strengthen the understanding of ccRCC characterization and guide clinical treatment. Targeting APOBEC3B may be regarded as a new therapeutic target for ccRCC.


Subject(s)
APOBEC Deaminases , Carcinoma, Renal Cell , Kidney Neoplasms , Tumor Microenvironment , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , APOBEC Deaminases/genetics , Prognosis , Mutation , Minor Histocompatibility Antigens/genetics , Biomarkers, Tumor/genetics
3.
JCI Insight ; 9(17)2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39253973

ABSTRACT

Elevated numbers of antibody-secreting cells (ASCs) and anti-double-stranded DNA (anti-dsDNA) antibodies are found in nasal polyp (NP) tissue. The presence of anti-dsDNA IgG in tissue prospectively predicts recurrent NP but the characteristics of the source ASCs are unknown. Here, we investigated whether NP B cells expressing the extrafollicular marker EBI2 have increased propensity for autoantibody production and evaluated the molecular characteristics of NP ASCs. NPs showed increased frequencies of anti-dsDNA IgG and total IgG ASCs compared with tonsils, with more pronounced differences among EBI2+ cells. In NPs, EBI2+ cells were frequently double negative (IgD-CD27-) and ASCs. Single-cell RNA-Seq analysis of tonsils and NPs revealed substantial differences in B lineage composition, including differences in percentages of ASCs, germinal centers, proliferative cells, and non-ASCs. NPs exhibited higher expression of specific isotypes (IGHE, IGHA1, IGHA2, and IGHG4) and mature plasma genes, including SDC1 and XBP1, than tonsils. Gene Ontology biological processes indicated upregulated NF-κB and downregulated apoptosis pathways in NP ASCs. Together, these data indicate that NP EBI2+ ASCs secret increased total and anti-dsDNA IgG compared with those from tonsils and had molecular features of mature plasma cell differentiation.


Subject(s)
Antibody-Producing Cells , Immunoglobulin G , Nasal Polyps , Humans , Nasal Polyps/immunology , Nasal Polyps/pathology , Nasal Polyps/metabolism , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , Male , Female , Adult , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Middle Aged , Palatine Tonsil/immunology , Palatine Tonsil/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/immunology , Antibodies, Antinuclear/immunology , Aged , Young Adult
4.
Sci Rep ; 14(1): 21073, 2024 09 10.
Article in English | MEDLINE | ID: mdl-39256448

ABSTRACT

The mitochondrial enzyme methylenetetrahydrofolate dehydrogenase (MTHFD2) is involved in purine and thymidine synthesis via 1C metabolism. MTHFD2 is exclusively overexpressed in cancer cells but absent in most healthy adult human tissues. However, the two close homologs of MTHFD2 known as MTHFD1 and MTHFD2L are expressed in healthy adult human tissues and share a great structural resemblance to MTHFD2 with 54% and 89% sequence similarity, respectively. It is therefore notably challenging to find selective inhibitors of MTHFD2 due to the structural similarity, in particular protein binding site similarity with MTHFD1 and MTHFD2L. Tricyclic coumarin-based compounds (substrate site binders) and xanthine derivatives (allosteric site binders) are the only selective inhibitors of MTHFD2 reported till date. Nanomolar potent diaminopyrimidine-based inhibitors of MTHFD2 have been reported recently, however, they also demonstrate significant inhibitory activities against MTHFD1 and MTHFD2L. In this study, we have employed extensive computational modeling involving molecular docking and molecular dynamics simulations in order to investigate the binding modes and key interactions of diaminopyrimidine-based inhibitors at the substrate binding sites of MTHFD1, MTHFD2 and MTHFD2L, and compare with the tricyclic coumarin-based selective MTHFD2 inhibitor. The outcomes of our study provide significant insights into desirable and undesirable structural elements for rational structure-based design of new and selective inhibitors of MTHFD2 against cancer.


Subject(s)
Aminohydrolases , Enzyme Inhibitors , Methylenetetrahydrofolate Dehydrogenase (NADP) , Minor Histocompatibility Antigens , Multifunctional Enzymes , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/antagonists & inhibitors , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Humans , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/chemistry , Multifunctional Enzymes/genetics , Multifunctional Enzymes/antagonists & inhibitors , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/chemistry , Aminohydrolases/genetics , Aminohydrolases/metabolism , Aminohydrolases/antagonists & inhibitors , Aminohydrolases/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemistry , Molecular Docking Simulation , Mitochondrial Proteins/genetics , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Binding Sites , Protein Binding
5.
Theranostics ; 14(15): 6036-6052, 2024.
Article in English | MEDLINE | ID: mdl-39346550

ABSTRACT

Background: Clinically, the persistence of HBV cccDNA is the major obstacle in anti-HBV therapy. However, the underlying mechanism of HBV cccDNA is poorly understood. The transcriptional factor STAT3 is able to activate HBV replication in liver. Approach & Results: RNA-Seq analysis demonstrated that cucurbitacin I targeting STAT3 was associated with virus replication in liver. HBV-infected human liver chimeric mouse model and HBV hydrodynamic injection mouse model were established. Then, we validated that cucurbitacin I effectively limited the stability of HBV cccDNA and HBV replication in cells, in which cucurbitacin I enhanced the sensitivity of pegylated interferon α (PEG-IFN α) against HBV via combination in vitro and in vivo. Mechanistically, we identified that cucurbitacin I increased the levels of APOBEC3B to control HBV cccDNA by inhibiting p-STAT3 in cells, resulting in the inhibition of HBV replication. Moreover, RNA-Seq data showed that E3 ubiquitin ligase DTX4 might be involved in the events. Then, we observed that HBV particles could upregulate DTX4 by increasing the levels of p-STAT3 in vitro and in vivo. The p-STAT3-elevated DTX4/male-specific lethal 2 (MSL2) independently and synergistically enhanced the stability of HBV cccDNA by facilitating the ubiquitination degradation of APOBEC3B in cells, leading to the HBV replication. Conclusions: p-STAT3-elevated DTX4 confers the stability of HBV cccDNA and HBV replication by facilitating the ubiquitination degradation of APOBEC3B. Cucurbitacin Ⅰ effectively enhances the sensitivity of PEG-IFN α in anti-HBV therapy by inhibiting the p-STAT3/DTX4/MSL2/APOBEC3B signalling. Our finding provides new insights into the mechanism of HBV cccDNA. The p-STAT3 and DTX4/MSL2 might serve as the therapeutical targets of HBV cccDNA.


Subject(s)
Cytidine Deaminase , Hepatitis B virus , Liver , Minor Histocompatibility Antigens , STAT3 Transcription Factor , Ubiquitin-Protein Ligases , Virus Replication , STAT3 Transcription Factor/metabolism , Humans , Animals , Hepatitis B virus/drug effects , Mice , Virus Replication/drug effects , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics , Liver/virology , Liver/metabolism , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Male , Ubiquitin-Protein Ligases/metabolism , DNA, Circular/metabolism , DNA, Circular/genetics , Ubiquitination/drug effects , DNA, Viral/metabolism , DNA, Viral/genetics , Hepatitis B/metabolism , Hepatitis B/virology , Hepatitis B/drug therapy , Antiviral Agents/pharmacology , Triterpenes/pharmacology , Triterpenes/metabolism , Hep G2 Cells , Disease Models, Animal , Interferon-alpha/metabolism , Interferon-alpha/pharmacology
6.
Cell Death Dis ; 15(9): 677, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285161

ABSTRACT

Myeloid cells are the first line of defence against pathogens. Mitochondrial apoptosis signalling is a crucial regulator of myeloid cell lifespan and modulates the function of myeloid cells. The anti-apoptotic protein BCL-2-family protein BCL2A1/A1/BFL-1 is strongly upregulated in inflammation in macrophages. We analysed the contribution of A1 to apoptosis regulation in a conditional system of in vitro differentiation of murine macrophages from immortalised progenitors. We disabled the expression of A1 by targeting all murine A1 isoforms in the genome. Specific inhibitors were used to inactivate other anti-apoptotic proteins. Macrophage progenitor survival mainly depended on the anti-apoptotic proteins MCL-1, BCL-XL and A1 but not BCL-2. Deletion of A1 on its own had little effect on progenitor cell survival but was sensitised to cell death induction when BCL-XL or MCL-1 was neutralised. In progenitors, A1 was required for survival in the presence of the inflammatory stimulus LPS. Differentiated macrophages were resistant to inhibition of single anti-apoptotic proteins, but A1 was required to protect macrophages against inhibition of either BCL-XL or MCL-1; BCL-2 only had a minor role in these cells. Cell death by neutralisation of anti-apoptotic proteins completely depended on BAX with a small contribution of BAK only in progenitors in the presence of LPS. A1 and NOXA appeared to stabilise each other at the posttranscriptional level suggesting direct binding. Co-immunoprecipitation experiments showed the binding of A1 to NOXA and BIM. Interaction between A1 and Noxa may indirectly prevent neutralisation and destabilization of MCL-1. Our findings suggest a unique role for A1 as a modulator of survival in the macrophage lineage in concert with MCL-1 and BCL-XL, especially in a pro-inflammatory environment.


Subject(s)
Apoptosis , Cell Differentiation , Cell Survival , Macrophages , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2 , bcl-X Protein , Animals , bcl-X Protein/metabolism , Macrophages/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics , Lipopolysaccharides/pharmacology , Myeloid Cells/metabolism
7.
Redox Rep ; 29(1): 2404794, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39314036

ABSTRACT

OBJECTIVES: Diabetic nephropathy (DN) is one of the most serious microvascular complications of diabetes and is the most common cause of end-stage renal disease. Tripartite motif-containing (TRIM) proteins are a large family of E3 ubiquitin ligases that contribute to protein quality control by regulating the ubiquitin - proteasome system. However, the detailed mechanisms through which various TRIM proteins regulate downstream events have not yet been fully elucidated. The current research aimed to determine the function and mechanism of TRIM22 in DN. METHODS: DN models were established by inducing HK-2 cells using high glucose (HG) and diabetic mice (db/db mice). Cell viability, apoptosis, mitochondrial reactive oxygen species, and mitochondrial membrane potential were detected by Cell Counting Kit-8 and flow cytometry, respectively. Pathological changes were evaluated using hematoxylin and eosin, periodic acid schiff and Masson staining. The binding between TRIM22 and optic atrophy 1 (OPA1) was analyzed using co-immunoprecipitation. The m6A level of TRIM22 5'UTR was detected using RNA immunoprecipitation. RESULTS: TRIM22 was highly expressed in patients with DN. TRIM22 silencing inhibited HG-induced apoptosis and mitochondrial dysfunction in HK-2 cells. Promoting mitochondrial fusion alleviated TRIM22 overexpression-induced cell apoptosis, mitochondrial dysfunction in HK-2 cells, and kidney damage in mice. Mechanistically, TRIM22 interacted with OPA1 and induced its ubiquitination. Wilms tumor 1-associating protein (WTAP) promoted m6A modification of TRIM22 through the m6A reader insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1). DISCUSSION: TRIM22 silencing inhibited the progression of DN by interacting with OPA1 and inducing its ubiquitination. Furthermore, WTAP promoted m6A modification of TRIM22 via IGF2BP1.


Subject(s)
Diabetic Nephropathies , GTP Phosphohydrolases , Minor Histocompatibility Antigens , Mitochondria , Tripartite Motif Proteins , Ubiquitination , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Animals , Humans , Mice , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Mitochondria/metabolism , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics , Male , Repressor Proteins/metabolism , Repressor Proteins/genetics , Apoptosis , Diabetes Mellitus, Experimental/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
8.
J Immunol ; 213(8): 1115-1124, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39240167

ABSTRACT

EBV-induced gene 3 (Ebi3) is a ß subunit within the IL-12 cytokine family that canonically binds to α subunits p19, p28, or p35 to form the heterodimeric cytokines IL-39, IL-27, and IL-35, respectively. In the last decade, the binding partners for Ebi3 have continued to expand to include IL-6 and the other IL-12 family ß subunit p40, revealing the possibility that Ebi3 may be able to bind to other cytokines and have distinct functions. We first explored this possibility utilizing an in vivo mouse model of regulatory T cell-restricted deletions of the subunits composing the cytokine IL-35, p35, and Ebi3, and we observed a differential impact on CD8+ T cell inhibitory receptor expression despite comparable reduction in tumor growth. We then screened the ability of Ebi3 to bind to different cytokines with varying structural resemblance to the IL-12 family α subunits. These in vitro screens revealed extracellular binding of Ebi3 to both IFN-γ and IL-10. Ebi3 bound to IFN-γ and IL-10 abrogated signal transduction and downstream functions of both cytokines. Lastly, we validated that extracellular complex formation after mixing native proteins resulted in loss of function. These data suggest that secreted partnerless Ebi3 may bind to cytokines within the extracellular microenvironment and act as a cytokine sink, further expanding the potential immunological impact of Ebi3.


Subject(s)
Interferon-gamma , Interleukin-10 , Minor Histocompatibility Antigens , Animals , Mice , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Protein Binding , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , Signal Transduction/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Interleukins/metabolism , Interleukins/immunology , Receptors, Cytokine
9.
J Med Chem ; 67(18): 16455-16479, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39291659

ABSTRACT

Bfl-1, a member of the Bcl-2 family of proteins, plays a crucial role in apoptosis regulation and has been implicated in cancer cell survival and resistance to venetoclax therapy. Due to the unique cysteine residue in the BH3 binding site, the development of covalent inhibitors targeting Bfl-1 represents a promising strategy for cancer treatment. Herein, the optimization of a covalent cellular tool from a lead-like hit using structure based design is described. Informed by a reversible X-ray fragment screen, the strategy to establish interactions with a key glutamic acid residue (Glu78) and optimize binding in a cryptic pocket led to a 1000-fold improvement in biochemical potency without increasing reactivity of the warhead. Compound (R,R,S)-26 has a kinact/KI of 4600 M-1 s-1, shows <1 µM caspase activation in a cellular assay and cellular target engagement, and has good physicochemical properties and a promising in vivo profile.


Subject(s)
Proto-Oncogene Proteins c-bcl-2 , Humans , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Structure-Activity Relationship , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Models, Molecular , Crystallography, X-Ray , Mice , Molecular Structure , Apoptosis/drug effects , Minor Histocompatibility Antigens
10.
Cell Rep Med ; 5(9): 101706, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39236712

ABSTRACT

Antipsychotic drugs have been shown to have antitumor effects but have had limited potency in the clinic. Here, we unveil that pimozide inhibits lysosome hydrolytic function to suppress fatty acid and cholesterol release in glioblastoma (GBM), the most lethal brain tumor. Unexpectedly, GBM develops resistance to pimozide by boosting glutamine consumption and lipogenesis. These elevations are driven by SREBP-1, which we find upregulates the expression of ASCT2, a key glutamine transporter. Glutamine, in turn, intensifies SREBP-1 activation through the release of ammonia, creating a feedforward loop that amplifies both glutamine metabolism and lipid synthesis, leading to drug resistance. Disrupting this loop via pharmacological targeting of ASCT2 or glutaminase, in combination with pimozide, induces remarkable mitochondrial damage and oxidative stress, leading to GBM cell death in vitro and in vivo. Our findings underscore the promising therapeutic potential of effectively targeting GBM by combining glutamine metabolism inhibition with lysosome suppression.


Subject(s)
Glioblastoma , Glutamine , Lipid Metabolism , Lysosomes , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , Glutamine/metabolism , Humans , Lysosomes/metabolism , Lysosomes/drug effects , Lipid Metabolism/drug effects , Cell Line, Tumor , Animals , Amino Acid Transport System ASC/metabolism , Amino Acid Transport System ASC/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Mice , Glutaminase/metabolism , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Lipogenesis/drug effects , Oxidative Stress/drug effects , Minor Histocompatibility Antigens
11.
Cancer Lett ; 600: 217157, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39127340

ABSTRACT

Deregulation of E3 ubiquitin ligases drives the proliferation and metastasis of various cancers; however, the underlying mechanisms remain unknown. This study aimed to investigate the role of tripartite motif-containing 22 (TRIM22), a poorly investigated E3 ubiquitin ligase in the TRIM family, as a tumor suppressor in breast cancer. High expression of TRIM22 in breast cancer correlated with better prognosis. Functional experiments demonstrated that TRIM22 significantly inhibited the proliferation and invasion of breast cancer cells. Label-free proteomics and biochemical analyses revealed that the copper chaperone for superoxide dismutase (CCS), an oncoprotein that is upregulated in breast cancer and promotes the growth and invasion of breast cancer cells, was a target of TRIM22 for degradation via K27-linked ubiquitination. Notably, the ability of the coiled-coil domain-defective mutants of TRIM22 to induce CCS ubiquitination and degradation diminished, with lysine 76 of the CCS serving as the ubiquitination site. Moreover, the TRIM22-mediated inhibition of the proliferation and invasion of breast cancer cells was restored by ectopic CCS expression. RNA-sequencing experiments using Gene Set Enrichment Analysis demonstrated that TRIM22 is involved in the JAK-STAT signaling pathway. TRIM22 overexpression also improved reactive oxygen species levels in breast cancer cells and inhibited STAT3 phosphorylation, which was restored via CCS overexpression or N-acetyl-l-cysteine treatment. Chromatin immunoprecipitation-quantitative polymerase chain reaction results showed that TRIM22 overexpression decreased the enrichment of phosphorylated STAT3 in FN1, VIM and JARID2 promoters. Clinically, low TRIM22 expression correlated with high CCS expression and decreased survival rates in patients with breast cancer. Moreover, TRIM22 upregulation was associated with a better prognosis in patients with breast cancer who received classical therapy. TRIM22 expression was downregulated in many cancer types, including colon, kidney, lung, and prostate cancers. To the best of our knowledge, the E3 ubiquitin ligase TRIM22 was first reported as a tumor suppressor that inhibits the proliferation and invasion of breast cancer cells through CCS ubiquitination and degradation. TRIM22 is a potential prognostic biomarker in patients with breast cancer.


Subject(s)
Breast Neoplasms , Cell Proliferation , Minor Histocompatibility Antigens , STAT3 Transcription Factor , Signal Transduction , Tripartite Motif Proteins , Ubiquitination , Female , Humans , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , MCF-7 Cells , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Neoplasm Invasiveness , Prognosis , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
12.
J Transl Med ; 22(1): 750, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123229

ABSTRACT

BACKGROUND: ERAP2 is an aminopeptidase involved in antigen processing and presentation, and harbor genetic variants linked to several inflammatory diseases such as Inflammatory Bowel Disease (IBD). The lack of an ERAP2 gene homologue in mice has hampered functional studies, and most human studies have focused on cells of hematopoietic origin. Using an IBD biobank as vantage point, this study explores how genetic variation in ERAP2 affects gene expression in human-derived epithelial organoids upon proinflammatory stimulation. METHODS: An IBD patient cohort was genotyped with regards to two single nucleotide polymorphisms (SNP) (rs2910686/rs2248374) associated with ERAP2 expression levels, and we examined the correlation between colon gene expression and genotype, specifically aiming to establish a relationship with ERAP2 expression proficiency. Human-derived colon organoids (colonoids) with known ERAP2 genotype were established and used to explore differences in whole genome gene expression between ERAP2-deficient (n = 4) and -proficient (n = 4) donors upon pro-inflammatory encounter. RESULTS: When taking rs2910686 genotype into account, ERAP2 gene expression is upregulated in the inflamed colon of IBD patients. Colonoids upregulate ERAP2 upon IFNÉ£ stimulation, and ERAP2 expression proficiency is dependent on rs2910686 genotype. Colonoid genotyping confirms that mechanisms independent of the frequently studied SNP rs2248374 can cause ERAP2-deficiency. A total of 586 genes involved in various molecular mechanisms are differentially expressed between ERAP2 proficient- and deficient colonoids upon proinflammatory stimulation, including genes encoding proteins with the following molecular function: catalytic activity (AOC1, CPE, ANPEP and MEP1A), regulator activity (TNFSF9, MDK, GDF15, ILR6A, LGALS3 and FLNA), transmembrane transporter activity (SLC40A1 and SLC5A1), and extracellular matrix structural constituents (FGL2, HMCN2, and MUC17). CONCLUSIONS: ERAP2 is upregulated in the inflamed IBD colon mucosa, and expression proficiency is highly correlated with genotype of rs2910686. While the SNP rs2248374 is commonly used to determine ERAP2 expressional proficiency, our data confirms that mechanisms independent of this SNP can lead to ERAP2 deficiency. Our data demonstrates that epithelial ERAP2 presence affects the inflammatory response in colonoids, suggesting a pleiotropic role of ERAP2 beyond MHC class I antigen processing.


Subject(s)
Aminopeptidases , Colon , Inflammation , Inflammatory Bowel Diseases , Polymorphism, Single Nucleotide , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/metabolism , Polymorphism, Single Nucleotide/genetics , Aminopeptidases/genetics , Aminopeptidases/metabolism , Inflammation/genetics , Inflammation/pathology , Colon/pathology , Colon/metabolism , Organoids/metabolism , Genotype , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Female , Male , Cohort Studies , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Epithelial Cells/metabolism
13.
Nat Commun ; 15(1): 6570, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39095408

ABSTRACT

ASCT2 is an obligate exchanger of neutral amino acids, contributing to cellular amino acid homeostasis. ASCT2 belongs to the same family (SLC1) as Excitatory Amino Acid Transporters (EAATs) that concentrate glutamate in the cytosol. The mechanism that makes ASCT2 an exchanger rather than a concentrator remains enigmatic. Here, we employ cryo-electron microscopy and molecular dynamics simulations to elucidate the structural basis of the exchange mechanism of ASCT2. We establish that ASCT2 binds three Na+ ions per transported substrate and visits a state that likely acts as checkpoint in preventing Na+ ion leakage, both features shared with EAATs. However, in contrast to EAATs, ASCT2 retains one Na+ ion even under Na+-depleted conditions. We demonstrate that ASCT2 cannot undergo the structural transition in TM7 that is essential for the concentrative transport cycle of EAATs. This structural rigidity and the high-affinity Na+ binding site effectively confine ASCT2 to an exchange mode.


Subject(s)
Amino Acid Transport System ASC , Cryoelectron Microscopy , Minor Histocompatibility Antigens , Molecular Dynamics Simulation , Sodium , Humans , Amino Acid Transport System ASC/metabolism , Amino Acid Transport System ASC/chemistry , Amino Acid Transport System ASC/genetics , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/chemistry , Sodium/metabolism , Binding Sites , HEK293 Cells , Protein Binding
14.
Immunobiology ; 229(5): 152841, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39096658

ABSTRACT

Hepatocellular carcinoma (HCC) stands as one of the most prevalent malignancies. While PD-1 immune checkpoint inhibitors have demonstrated promising therapeutic efficacy in HCC, not all patients exhibit a favorable response to these treatments. Glutamine is a crucial immune cell regulatory factor, and tumor cells exhibit glutamine dependence. In this study, HCC patients were divided into two subtypes (C1 and C2) based on glutamine metabolism-related genes via consensus clustering. The C1 pattern, in contrast to C2, was associated with a lower survival probability among HCC patients. Additionally, the C1 pattern exhibited higher proportions of patients with advanced tumor stages. The activity of C1 in glutamine metabolism and transport is significantly enhanced, while its oxidative phosphorylation activity is reduced. And, C1 was mainly involved in the progression-related pathway of HCC. Furthermore, C1 exhibited high levels of immunosuppressive cells, cytokine-receptor interactions and immune checkpoint genes, suggesting C1 as an immunosuppressive subtype. After stepwise selection based on integrated four machine learning methods, SLC1A5 was finally identified as the pivotal gene that distinguishes the subtypes. The expression of SLC1A5 was significantly positively correlated with immunosuppressive status. SLC1A5 showed the most significant correlation with macrophage infiltration, and this correlation was confirmed through the RNA-seq data of CLCA project and our cohort. Low-SLC1A5-expression samples had better immunogenicity and responsiveness to immunotherapy. As expected, SubMap and survival analysis indicated that individuals with low SLC1A5 expression were more responsive to anti-PD1 therapy. Collectively, this study categorized HCC patients based on glutamine metabolism-related genes and proposed two subclasses with different clinical traits, biological behavior, and immune status. Machine learning was utilized to identify the hub gene SLC1A5 for HCC classification, which also could predict immunotherapy response.


Subject(s)
Amino Acid Transport System ASC , Biomarkers, Tumor , Carcinoma, Hepatocellular , Glutamine , Immunotherapy , Liver Neoplasms , Machine Learning , Humans , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Glutamine/metabolism , Amino Acid Transport System ASC/genetics , Amino Acid Transport System ASC/metabolism , Immunotherapy/methods , Gene Expression Regulation, Neoplastic , Prognosis , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Female , Male , Minor Histocompatibility Antigens
15.
Microbiol Spectr ; 12(10): e0010124, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39162523

ABSTRACT

Understanding the functions of human transcriptional regulatory genes SP110 and SP140 during Mycobacterium tuberculosis infection is crucial; in a mouse model, homologous genes Sp110 and Sp140 have been shown to negatively regulate inflammatory response genes, including the type I interferon (IFN) response. The reduction of these genes in mice is associated with susceptibility to M. tuberculosis infection and the development of necrotizing granulomatous lesions. To investigate the involvement of SP110 and SP140 in human inflammatory response, we analyzed their regulatory manner in THP-1 macrophages infected with M. tuberculosis. Genome-wide transcriptional profiling revealed that the depletion of SP110 and/or SP140 impaired the induction of gene expression associated with inflammatory responses, including IFN response genes, although it had little effect on the intracellular proliferation of M. tuberculosis. By contrast, genes related to phosphorylation were upregulated in infected macrophages with SP110 and/or SP140 knockdown, but downregulated in infected control macrophages without their knockdown. Reverse transcription-quantitative PCR and ELISA further confirmed the impairment of the induction of IFN response genes by the depletion of SP110 and/or SP140 in M. tuberculosis-infected macrophages. These findings suggest that human SP110 and SP140 act as positive regulators for genes associated with inflammatory responses in M. tuberculosis-infected macrophages. IMPORTANCE: Tuberculosis (TB) is one of the most serious infectious diseases, with high morbidity and mortality worldwide. C3HeB/FeJ mice are widely utilized for evaluating anti-TB drugs because their drug sensitivity and pathology during M. tuberculosis infection resemble those of human TB, including the development of necrotizing granulomas. Downregulation of the transcriptional regulatory genes Sp110 and Sp140 in C3HeB/FeJ mice has been demonstrated to activate gene expression associated with inflammatory responses during M. tuberculosis infection, resulting in susceptibility to the infection. Here, we examined the regulatory manner of SP110 and SP140 using transcriptomic analysis in M. tuberculosis-infected human macrophages. Depletion of SP110 and/or SP140 in M. tuberculosis-infected THP-1 macrophages impaired the induction of gene expression associated with inflammatory responses, including interferon response genes, compared with that in control macrophages. These results suggest that human SP110 and SP140 act as positive regulators for genes associated with inflammatory responses upon M. tuberculosis infection.


Subject(s)
Macrophages , Mycobacterium tuberculosis , Tuberculosis , Animals , Humans , Mice , Butyrate Response Factor 1/genetics , Gene Expression Profiling , Gene Expression Regulation , Inflammation/genetics , Interferon Type I/metabolism , Interferon Type I/genetics , Macrophages/immunology , Macrophages/microbiology , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/genetics , Nuclear Proteins , THP-1 Cells , Tuberculosis/microbiology , Tuberculosis/immunology , Tuberculosis/genetics , Mice, Inbred C3H
16.
Best Pract Res Clin Haematol ; 37(2): 101555, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39098803

ABSTRACT

Allogeneic hematopoietic cell transplantation (alloHCT) provides a potential curative treatment for haematological malignancies. The therapeutic Graft-versus-Leukaemia (GvL) effect is induced by donor T cells attacking patient hematopoietic (malignant) cells. However, if healthy non-hematopoietic tissues are targeted, Graft-versus-Disease (GvHD) may develop. After HLA-matched alloHCT, GvL and GvHD are induced by donor T cells recognizing polymorphic peptides presented by HLA on patient cells, so-called minor histocompatibility antigens (MiHAs). The balance between GvL and GvHD depends on the tissue distribution of MiHAs and T-cell frequencies targeting these MiHAs. T cells against broadly expressed MiHAs induce GvL and GvHD, whereas those targeting MiHAs with hematopoietic-restricted expression induce GvL without GvHD. Recently, the MiHA repertoire identified in natural immune responses after alloHCT was expanded to 159 total HLA-I-restricted MiHAs, including 14 hematopoietic-restricted MiHAs. This review explores their potential relevance to predict, monitor, and manipulate GvL and GvHD for improving clinical outcome after HLA-matched alloHCT.


Subject(s)
Graft vs Host Disease , Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Minor Histocompatibility Antigens , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Graft vs Host Disease/immunology , Minor Histocompatibility Antigens/immunology , Minor Histocompatibility Antigens/genetics , Graft vs Leukemia Effect/immunology , Transplantation, Homologous , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , T-Lymphocytes/immunology , Allografts
17.
Viruses ; 16(8)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39205150

ABSTRACT

The different susceptibility to HIV-1 infection in U937 cells-permissive (Plus) or nonpermissive (Minus)-is linked to the expression in Minus cells of interferon (IFN)-γ inducible antiviral factors such as tripartite motif-containing protein 22 (TRIM22) and class II transactivator (CIITA). CIITA interacts with Cyclin T1, a key component of the Positive-Transcription Elongation Factor b (P-TEFb) complex needed for the efficient transcription of HIV-1 upon interaction with the viral transactivator Tat. TRIM22 interacts with CIITA, recruiting it into nuclear bodies together with Cyclin T1. A 50 kDa Cyclin T1 was found only in Minus cells, alongside the canonical 80 kDa protein. The expression of this truncated form remained unaffected by proteasome inhibitors but was reduced by IFNγ treatment. Unlike the nuclear full-length protein, truncated Cyclin T1 was also present in the cytoplasm, and this subcellular localization correlated with its capacity to inhibit Tat-mediated HIV-1 transcription. The 50 kDa Cyclin T1 in Minus cells likely contributes to their non-permissive phenotype by acting as a dominant negative factor, disrupting P-TEFb complex formation and function. Its reduction upon IFNγ treatment suggests a regulatory loop by which its inhibitory role on HIV-1 replication is then exerted by the IFNγ-induced CIITA, which binds to the canonical Cyclin T1, displacing it from the P-TEFb complex.


Subject(s)
Cyclin T , HIV-1 , Humans , Cyclin T/metabolism , HIV-1/physiology , U937 Cells , HIV Infections/virology , HIV Infections/metabolism , Protein Isoforms/metabolism , Protein Isoforms/genetics , Virus Replication , Phenotype , Interferon-gamma/pharmacology , Interferon-gamma/metabolism , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Minor Histocompatibility Antigens
18.
Cell Commun Signal ; 22(1): 415, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39192247

ABSTRACT

The antiapoptotic protein BCL2A1 is highly, but very heterogeneously expressed in Diffuse Large B-cell Lymphoma (DLBCL). Particularly in the context of resistance to current therapies, BCL2A1 appears to play an important role in protecting cancer cells from the induction of cell death. Reducing BCL2A1 levels may have therapeutic potential, however, no specific inhibitor is currently available. In this study, we hypothesized that the signaling network regulated by epigenetic readers may regulate the transcription of BCL2A1 and hence that inhibition of Bromodomain and Extra-Terminal (BET) proteins may reduce BCL2A1 expression thus leading to cell death in DLBCL cell lines. We found that the mechanisms of action of acetyl-lysine competitive BET inhibitors are different from those of proteolysis targeting chimeras (PROTACs) that induce the degradation of BET proteins. Both classes of BETi reduced the expression of BCL2A1 which coincided with a marked downregulation of c-MYC. Mechanistically, BET inhibition attenuated the constitutively active canonical nuclear factor kappa-light-chain-enhancer of activated B-cells (NFκB) signaling pathway and inhibited p65 activation. Furthermore, signal transducer of activated transcription (STAT) signaling was reduced by inhibiting BET proteins, targeting another pathway that is often constitutively active in DLBCL. Both pathways were also inhibited by the IκB kinase inhibitor TPCA-1, resulting in decreased BCL2A1 and c-MYC expression. Taken together, our study highlights a novel complex regulatory network that links BET proteins to both NFκB and STAT survival signaling pathways controlling both BCL2A1 and c-MYC expression in DLBCL.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , NF-kappa B , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins c-myc , Signal Transduction , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/drug effects , NF-kappa B/metabolism , Cell Line, Tumor , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Gene Expression Regulation, Neoplastic/drug effects , Apoptosis/drug effects , Bromodomain Containing Proteins , Proteins , Minor Histocompatibility Antigens
19.
Saudi Med J ; 45(8): 783-790, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39074897

ABSTRACT

OBJECTIVES: To identify biomarkers that can discriminated small cell lung cancer (SCLC) from non-SCLC (NSCLC), and explore their association with the prognosis of SCLC under chemoradiotherapy. METHODS: The GSE40275 dataset was used to identify potential targets in SCLC. There were 196 patients of lung cancer (LC) in cohort 1 of this study. MTHFD1 levels in tissues were determined by immunohistochemistry assay in cohort 1. Lung cancer patients who were all underwent local chemoradiotherapy (CRT) were included in cohort 2, and the association of MTHFD1 levels with CRT treatment outcome were determined in cohort 2. Cell experiments were used to determine the function of MTHFD1 on the radio-sensitivity of SCLC and NSCLC cells. RESULTS: The MTHFD1 levels in LC tissues were increased, and could discriminate SCLC from both lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD). Small cell lung cancer patients with MTHFD1 high phenotype had a poorer prognosis after CRT treatment, whereas no significant correlation was found between MTHFD1 levels and prognosis in LUSC and LUAD group. Cell experiments demonstrated that overexpression of MTHFD1 increases radio-resistance in both SCLC and NSCLC in vitro. CONCLUSION: MTHFD1 expressions might be a novel specifically prognostic biomarker for SCLC and the CRT treatment outcome.


Subject(s)
Chemoradiotherapy , Lung Neoplasms , Methylenetetrahydrofolate Dehydrogenase (NADP) , Small Cell Lung Carcinoma , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Small Cell Lung Carcinoma/therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Lung Neoplasms/therapy , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Female , Male , Middle Aged , Prognosis , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Aged , Treatment Outcome , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Cohort Studies , Carcinoma, Squamous Cell/therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/therapy , Adenocarcinoma of Lung/pathology
20.
Genetics ; 227(4)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38946641

ABSTRACT

APOBEC proteins are cytidine deaminases that restrict the replication of viruses and transposable elements. Several members of the APOBEC3 family, APOBEC3A, APOBEC3B, and APOBEC3H-I, can access the nucleus and cause what is thought to be indiscriminate deamination of the genome, resulting in mutagenesis and genome instability. Although APOBEC3C is also present in the nucleus, the full scope of its deamination target preferences is unknown. By expressing human APOBEC3C in a yeast model system, I have defined the APOBEC3C mutation signature, as well as the preferred genome features of APOBEC3C targets. The APOBEC3C mutation signature is distinct from those of the known cancer genome mutators APOBEC3A and APOBEC3B. APOBEC3C produces DNA strand-coordinated mutation clusters, and APOBEC3C mutations are enriched near the transcription start sites of active genes. Surprisingly, APOBEC3C lacks the bias for the lagging strand of DNA replication that is seen for APOBEC3A and APOBEC3B. The unique preferences of APOBEC3C constitute a mutation profile that will be useful in defining sites of APOBEC3C mutagenesis in human genomes.


Subject(s)
Cytidine Deaminase , Mutation , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Humans , Genome, Human , DNA Replication , Proteins/genetics , Proteins/metabolism , Mutagenesis , Saccharomyces cerevisiae/genetics , Minor Histocompatibility Antigens
SELECTION OF CITATIONS
SEARCH DETAIL