Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Hepatol ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38759889

ABSTRACT

BACKGROUND & AIMS: The liver is the main organ of ketogenesis, while ketones are mainly metabolized in peripheral tissues via the critical enzyme OXCT1. We previously found that ketolysis is reactivated in hepatocellular carcinoma (HCC) cells through OXCT1 expression to promote tumor progression; however, whether OXCT1 regulates antitumor immunity remains unclear. METHODS: To investigate the expression pattern of OXCT1 in hepatocellular carcinoma in vivo, we conducted multiplex immunohistochemistry (mIHC) experiments on human HCC specimens. To explore the role of OXCT1 in mouse hepatocellular carcinoma tumor-associated macrophages (TAMs), we generated LysMcreOXCT1f/f (OXCT1 conditional knockout in macrophages) mice. RESULTS: Here, we found that inhibiting OXCT1 expression in tumor-associated macrophages reduced CD8+ T-cell exhaustion through the succinate-H3K4me3-Arg1 axis. Initially, we found that OXCT1 was highly expressed in liver macrophages under steady state and that OXCT expression was further increased in TAMs. OXCT1 deficiency in macrophages suppressed tumor growth by reprogramming TAMs toward an antitumor phenotype, reducing CD8+ T-cell exhaustion and increasing CD8+ T-cell cytotoxicity. Mechanistically, high OXCT1 expression induced the accumulation of succinate, a byproduct of ketolysis, in TAMs, which promoted Arg1 transcription by increasing the H3K4 trimethylation (H3K4me3) level in the Arg1 promoter. In addition, Pimozide, an inhibitor of OXCT1, suppressed Arg1 expression as well as TAM polarization toward the protumor phenotype, leading to decreasing CD8+ T-cell exhaustion and deceleration of tumor growth. Finally, high expression of OXCT1 in macrophages was positively associated with poor survival in HCC patients. CONCLUSIONS: In conclusion, our results demonstrate that OXCT1 epigenetically suppresses antitumor immunity, suggesting that suppressing OXCT1 activity in TAMs is an effective approach for treating liver cancer. IMPACT AND IMPLICATIONS: The intricate metabolism of liver macrophages plays a critical role in shaping HCC progression and immune modulation. Targeting macrophage metabolism to counteract immune suppression presents a promising avenue for HCC. Here, we found that ketogenesis gene OXCT1 was highly expressed in tumor-associated macrophages and promoted tumor growth by reprogramming TAMs toward a protumor phenotype. And the strategic pharmacological intervention or genetic downregulation of OXCT1 in TAMs enhances the antitumor immunity and decelerated tumor growth. Our results suggest that suppressing OXCT1 activity in TAMs is an effective approach for treating liver cancer.

2.
Nat Immunol ; 25(3): 483-495, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38177283

ABSTRACT

Tumor cells and surrounding immune cells undergo metabolic reprogramming, leading to an acidic tumor microenvironment. However, it is unclear how tumor cells adapt to this acidic stress during tumor progression. Here we show that carnosine, a mobile buffering metabolite that accumulates under hypoxia in tumor cells, regulates intracellular pH homeostasis and drives lysosome-dependent tumor immune evasion. A previously unrecognized isoform of carnosine synthase, CARNS2, promotes carnosine synthesis under hypoxia. Carnosine maintains intracellular pH (pHi) homeostasis by functioning as a mobile proton carrier to accelerate cytosolic H+ mobility and release, which in turn controls lysosomal subcellular distribution, acidification and activity. Furthermore, by maintaining lysosomal activity, carnosine facilitates nuclear transcription factor X-box binding 1 (NFX1) degradation, triggering galectin-9 and T-cell-mediated immune escape and tumorigenesis. These findings indicate an unconventional mechanism for pHi regulation in cancer cells and demonstrate how lysosome contributes to immune evasion, thus providing a basis for development of combined therapeutic strategies against hepatocellular carcinoma that exploit disrupted pHi homeostasis with immune checkpoint blockade.


Subject(s)
Carcinoma, Hepatocellular , Carnosine , Liver Neoplasms , Humans , Homeostasis , Lysosomes , Hypoxia , Hydrogen-Ion Concentration , Tumor Microenvironment
3.
Nat Commun ; 14(1): 8154, 2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38071226

ABSTRACT

Itaconate is a well-known immunomodulatory metabolite; however, its role in hepatocellular carcinoma (HCC) remains unclear. Here, we find that macrophage-derived itaconate promotes HCC by epigenetic induction of Eomesodermin (EOMES)-mediated CD8+ T-cell exhaustion. Our results show that the knockout of immune-responsive gene 1 (IRG1), responsible for itaconate production, suppresses HCC progression. Irg1 knockout leads to a decreased proportion of PD-1+ and TIM-3+ CD8+ T cells. Deletion or adoptive transfer of CD8+ T cells shows that IRG1-promoted tumorigenesis depends on CD8+ T-cell exhaustion. Mechanistically, itaconate upregulates PD-1 and TIM-3 expression levels by promoting succinate-dependent H3K4me3 of the Eomes promoter. Finally, ibuprofen is found to inhibit HCC progression by targeting IRG1/itaconate-dependent tumor immunoevasion, and high IRG1 expression in macrophages predicts poor prognosis in HCC patients. Taken together, our results uncover an epigenetic link between itaconate and HCC and suggest that targeting IRG1 or itaconate might be a promising strategy for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , CD8-Positive T-Lymphocytes/metabolism , Liver Neoplasms/metabolism , Hepatitis A Virus Cellular Receptor 2/genetics , Programmed Cell Death 1 Receptor/metabolism , T-Cell Exhaustion , Succinates/pharmacology , Succinates/metabolism , Epigenesis, Genetic
4.
Nat Chem Biol ; 19(12): 1492-1503, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37500770

ABSTRACT

Enolase 1 (ENO1) is a glycolytic enzyme that plays essential roles in various pathological activities including cancer development. However, the mechanisms underlying ENO1-contributed tumorigenesis are not well explained. Here, we uncover that ENO1, as an RNA-binding protein, binds to the cytosine-uracil-guanine-rich elements of YAP1 messenger RNA to promote its translation. ENO1 and YAP1 positively regulate alternative arachidonic acid (AA) metabolism by inverse regulation of PLCB1 and HPGD (15-hydroxyprostaglandin dehydrogenase). The YAP1/PLCB1/HPGD axis-mediated activation of AA metabolism and subsequent accumulation of prostaglandin E2 (PGE2) are responsible for ENO1-mediated cancer progression, which can be retarded by aspirin. Finally, aberrant activation of ENO1/YAP1/PLCB1 and decreased HPGD expression in clinical hepatocellular carcinoma samples indicate a potential correlation between ENO1-regulated AA metabolism and cancer development. These findings underline a new function of ENO1 in regulating AA metabolism and tumorigenesis, suggesting a therapeutic potential for aspirin in patients with liver cancer with aberrant expression of ENO1 or YAP1.


Subject(s)
Carcinogenesis , Liver Neoplasms , Humans , Arachidonic Acid , Cell Line, Tumor , Cell Proliferation , Carcinogenesis/genetics , Cell Transformation, Neoplastic , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Liver Neoplasms/genetics , Aspirin/pharmacology , DNA-Binding Proteins/genetics , Biomarkers, Tumor , Tumor Suppressor Proteins/genetics
5.
Front Oncol ; 12: 857968, 2022.
Article in English | MEDLINE | ID: mdl-35433434

ABSTRACT

Staphylococcal nuclease domain-containing protein 1 (SND1) is an evolutionarily conserved multifunctional protein that functions mainly in the nucleus and cytoplasm. However, whether SND1 regulates cellular activity through mitochondrial-related functions remains unclear. Herein, we demonstrate that SND1 is localized to mitochondria to promote phosphoglycerate mutase 5 (PGAM5)-mediated mitophagy. We find that SND1 is present in mitochondria based on mass spectrometry data and verified this phenomenon in different liver cancer cell types by performing organelle subcellular isolation. Specifically, The N-terminal amino acids 1-63 of SND1 serve as a mitochondrial targeting sequence (MTS), and the translocase of outer membrane 70 (TOM 70) promotes the import of SND1 into mitochondria. By immunoprecipitation-mass spectrometry (IP-MS), we find that SND1 interacts with PGAM5 in mitochondria and is crucial for the binding of PGAM5 to dynamin-related protein 1 (DRP1). Importantly, we demonstrate that PGAM5 and SND1-MTS are required for SND1-mediated mitophagy under FCCP and glucose deprivation treatment as well as for SND1-mediated cell proliferation and tumor growth both in vitro and in vivo. Aberrant expression of SND1 and PGAM5 predicts poor outcomes in hepatocellular carcinoma (HCC) patients. Taken together, these findings establish a previously unappreciated role of SND1 and the association of mitochondrion-localized SND1 with PGAM5 in mitophagy and tumor progression.

6.
Nat Cancer ; 3(1): 75-89, 2022 01.
Article in English | MEDLINE | ID: mdl-35121990

ABSTRACT

α-Enolase 1 (ENO1) is a critical glycolytic enzyme whose aberrant expression drives the pathogenesis of various cancers. ENO1 has been indicated as having additional roles beyond its conventional metabolic activity, but the underlying mechanisms and biological consequences remain elusive. Here, we show that ENO1 suppresses iron regulatory protein 1 (IRP1) expression to regulate iron homeostasis and survival of hepatocellular carcinoma (HCC) cells. Mechanistically, we demonstrate that ENO1, as an RNA-binding protein, recruits CNOT6 to accelerate the messenger RNA decay of IRP1 in cancer cells, leading to inhibition of mitoferrin-1 (Mfrn1) expression and subsequent repression of mitochondrial iron-induced ferroptosis. Moreover, through in vitro and in vivo experiments and clinical sample analysis, we identified IRP1 and Mfrn1 as tumor suppressors by inducing ferroptosis in HCC cells. Taken together, this study establishes an important role for the ENO1-IRP1-Mfrn1 pathway in the pathogenesis of HCC and reveals a previously unknown connection between this pathway and ferroptosis, suggesting a potential innovative cancer therapy.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Iron Regulatory Protein 1/metabolism , Liver Neoplasms , Biomarkers, Tumor , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Ferroptosis/genetics , Humans , Iron/metabolism , Iron Regulatory Protein 1/genetics , Liver Neoplasms/genetics , Phosphopyruvate Hydratase/genetics , RNA, Messenger/genetics , Tumor Suppressor Proteins/metabolism
7.
Cell Death Dis ; 12(10): 902, 2021 10 02.
Article in English | MEDLINE | ID: mdl-34601503

ABSTRACT

Metformin, the first-line drug for type II diabetes, has recently been considered an anticancer agent. However, the molecular target and underlying mechanism of metformin's anti-cancer effects remain largely unclear. Herein, we report that metformin treatment increases the sensitivity of hepatocarcinoma cells to methotrexate (MTX) by suppressing the expression of the one-carbon metabolism enzyme DHFR. We show that the combination of metformin and MTX blocks nucleotide metabolism and thus effectively inhibits cell cycle progression and tumorigenesis. Mechanistically, metformin not only transcriptionally represses DHFR via E2F4 but also promotes lysosomal degradation of the DHFR protein. Notably, metformin dramatically increases the response of patient-derived hepatocarcinoma organoids to MTX without obvious toxicity to organoids derived from normal liver tissue. Taken together, our findings identify an important role for DHFR in the suppressive effects of metformin on therapeutic resistance, thus revealing a therapeutically targetable potential vulnerability in hepatocarcinoma.


Subject(s)
Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Metformin/pharmacology , Methotrexate/pharmacology , Tetrahydrofolate Dehydrogenase/metabolism , Animals , Cell Count , Drug Resistance, Neoplasm/drug effects , E2F4 Transcription Factor/metabolism , Folic Acid Antagonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Organoids/drug effects , Organoids/pathology , Proteolysis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic/drug effects
8.
EMBO Rep ; 22(3): e51519, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33426808

ABSTRACT

The MYC oncoprotein activates and represses gene expression in a transcription-dependent or transcription-independent manner. Modification of mRNA emerges as a key gene expression regulatory nexus. We sought to determine whether MYC alters mRNA modifications and report here that MYC promotes cancer progression by down-regulating N6-methyladenosine (m6 A) preferentially in transcripts of a subset of MYC-repressed genes (MRGs). We find that MYC activates the expression of ALKBH5 and reduces m6 A levels in the mRNA of the selected MRGs SPI1 and PHF12. We also show that MYC-regulated m6 A controls the translation of MRG mRNA via the specific m6 A reader YTHDF3. Finally, we find that inhibition of ALKBH5, or overexpression of SPI1 or PHF12, effectively suppresses the growth of MYC-deregulated B-cell lymphomas, both in vitro and in vivo. Our findings uncover a novel mechanism by which MYC suppresses gene expression by altering m6 A modifications in selected MRG transcripts promotes cancer progression.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Neoplasms , Adenosine , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , RNA, Messenger/genetics
9.
Cancer Res ; 81(5): 1265-1278, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33402389

ABSTRACT

Metastasis is responsible for the majority of breast cancer-related deaths, however, the mechanisms underlying metastasis in this disease remain largely elusive. Here we report that under hypoxic conditions, alternative splicing of MBD2 is suppressed, favoring the production of MBD2a, which facilitates breast cancer metastasis. Specifically, MBD2a promoted, whereas its lesser known short form MBD2c suppressed metastasis. Activation of HIF1 under hypoxia facilitated MBD2a production via repression of SRSF2-mediated alternative splicing. As a result, elevated MBD2a outcompeted MBD2c for binding to promoter CpG islands to activate expression of FZD1, thereby promoting epithelial-to-mesenchymal transition and metastasis. Strikingly, clinical data reveal significantly correlated expression of MBD2a and MBD2c with the invasiveness of malignancy, indicating opposing roles for MBD2 splicing variants in regulating human breast cancer metastasis. Collectively, our findings establish a novel link between MBD2 switching and tumor metastasis and provide a promising therapeutic strategy and predictive biomarkers for hypoxia-driven breast cancer metastasis. SIGNIFICANCE: This study defines the opposing roles and clinical relevance of MBD2a and MBD2c, two MBD2 alternative splicing products, in hypoxia-driven breast cancer metastasis. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/5/1265/F1.large.jpg.


Subject(s)
Alternative Splicing , Breast Neoplasms/pathology , DNA-Binding Proteins/genetics , Frizzled Receptors/genetics , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , CpG Islands , Epithelial-Mesenchymal Transition/genetics , Female , Frizzled Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , MicroRNAs/genetics , Promoter Regions, Genetic , Serine-Arginine Splicing Factors/genetics , Tumor Hypoxia/genetics , Xenograft Model Antitumor Assays
10.
Cell Mol Immunol ; 17(2): 163-177, 2020 02.
Article in English | MEDLINE | ID: mdl-30760919

ABSTRACT

OX40L is one of the co-stimulatory molecules that can be expressed by splenic lymphoid tissue inducer (Lti) cells, a subset of group 3 innate lymphoid cells (ILC3s). OX40L expression in subsets of intestinal ILC3s and the molecular regulation of OX40L expression in ILC3s are unknown. Here, we showed intestinal ILC3s marked as an OX40Lhigh population among all the intestinal leukocytes and were the dominant source of OX40L in Rag1-/- mice. All ILC3 subsets expressed OX40L, and NCR-ILC3s were the most abundant source of OX40L. The expression of OX40L in ILC3s could be upregulated during inflammation. In addition to tumor necrosis factor (TNF)-like cytokine 1A (TL1A), which has been known as a trigger for OX40L, we found that Poly (I:C) representing viral stimulus promoted OX40L expression in ILC3s via a cell-autonomous manner. Furthermore, we demonstrated that IL-7-STAT5 signaling sustained OX40L expression by ILC3s. Intestinal regulatory T cells (Tregs), most of which expressed OX40, had defective expansion in chimeric mice, in which ILC3s were specifically deficient for OX40L expression. Consistently, co-localization of Tregs and ILC3s was found in the cryptopatches of the intestine, which suggests the close interaction between ILC3s and Tregs. Our study has unveiled the crosstalk between Tregs and ILC3s in mucosal tissues through OX40-OX40L signaling, which is crucial for the homeostasis of intestinal Tregs.


Subject(s)
Homeostasis/genetics , Homeostasis/immunology , Immunocompromised Host/genetics , Intestinal Mucosa/immunology , OX40 Ligand/deficiency , Signal Transduction/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Cell Communication/immunology , Cells, Cultured , Coculture Techniques , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunity, Innate , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , OX40 Ligand/genetics , Receptors, OX40/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism
11.
Adv Exp Med Biol ; 1136: 87-95, 2019.
Article in English | MEDLINE | ID: mdl-31201718

ABSTRACT

The hypoxic microenvironment is one of the major features of solid tumors, which regulates cell malignancy in multiple ways. As a response to hypoxia, a large number of target genes involved in cell growth, metabolism, metastasis and immunity are activated in cancer cells. Hypoxia-inducible factor 1 (HIF-1), as a heterodimeric DNA-binding complex, is comprised of a constitutively expressed HIF-1ß subunit and an oxygen sensitive HIF-1α subunit, thus, adapts to decreased oxygen availability as a transcriptional factor. HIF-1 regulates many genes involved in tumorigenesis. Here, we focus on cancer cell metabolism and metastasis regulated by hypoxia.


Subject(s)
Neoplasm Metastasis/pathology , Neoplasms/metabolism , Neoplasms/pathology , Tumor Hypoxia , Cell Hypoxia , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Oxygen , Tumor Microenvironment
12.
Cells ; 8(5)2019 05 10.
Article in English | MEDLINE | ID: mdl-31083403

ABSTRACT

HIF-1 serves as an important regulator in cell response to hypoxia. Due to its key role in promoting tumor survival and progression under hypoxia, HIF-1 has become a promising target of cancer therapy. Thus far, several HIF-1 inhibitors have been identified, most of which are from synthesized chemical compounds. Here, we report that ALM (ActinoLactoMycin), a compound extracted from metabolites of Streptomyces flavoretus, exhibits inhibitory effect on HIF-1α. Mechanistically, we found that ALM inhibited the translation of HIF-1α protein by suppressing mTOR signaling activity. Treatment with ALM induced cell apoptosis and growth inhibition of cancer cells both in vitro and in vivo in a HIF-1 dependent manner. More interestingly, low dose of ALM treatment enhanced the anti-tumor effect of Everolimus, an inhibitor of mTOR, suggesting its potential use in combination therapy of tumors, especially solid tumor patients. Thus, we identified a novel HIF-1α inhibitor from the metabolites of Streptomyces flavoretus, which shows promising anti-cancer potential.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Fatty Acids/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactones/pharmacology , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/therapeutic use , Everolimus/pharmacology , Everolimus/therapeutic use , Fatty Acids/therapeutic use , Humans , Lactones/therapeutic use , Mice, Inbred BALB C , Mice, Nude , Neoplastic Processes , PC-3 Cells , Signal Transduction/drug effects , Streptomyces/metabolism , TOR Serine-Threonine Kinases/metabolism
13.
J Allergy Clin Immunol ; 143(1): 229-244.e9, 2019 01.
Article in English | MEDLINE | ID: mdl-29625134

ABSTRACT

BACKGROUND: IL-17 plays a pathogenic role in asthma. ST2- inflammatory group 2 innate lymphoid cells (ILC2s) driven by IL-25 can produce IL-17, whereas ST2+ natural ILC2s produce little IL-17. OBJECTIVE: We characterized ST2+IL-17+ ILC2s during lung inflammation and determined the pathogenesis and molecular regulation of ST2+IL-17+ ILC2s. METHODS: Lung inflammation was induced by papain or IL-33. IL-17 production by lung ILC2s from wild-type, Rag1-/-, Rorcgfp/gfp, and aryl hydrocarbon receptor (Ahr)-/- mice was examined by using flow cytometry. Bone marrow transfer experiments were performed to evaluate hematopoietic myeloid differentiation primary response gene-88 (MyD88) signaling in regulating IL-17 production by ILC2s. mRNA expression of IL-17 was analyzed in purified naive ILC2s treated with IL-33, leukotrienes, and inhibitors for nuclear factor of activated T cells, p38, c-Jun N-terminal kinase, or nuclear factor κ light-chain enhancer of activated B cells. The pathogenesis of IL-17+ ILC2s was determined by transferring wild-type or Il17-/- ILC2s to Rag2-/-Il2rg-/- mice, which further induced lung inflammation. Finally, expression of 106 ILC2 signature genes was compared between ST2+IL-17+ ILC2s and ST2+IL-17- ILC2s. RESULTS: Papain or IL-33 treatment boosted IL-17 production from ST2+ ILC2s (referred to by us as ILC217s) but not ST2- ILC2s. Ahr, but not retinoic acid receptor-related orphan receptor γt, facilitated the production of IL-17 by ILC217s. The hematopoietic compartment of MyD88 signaling is essential for ILC217 induction. IL-33 works in synergy with leukotrienes, which signal through nuclear factor of activated T-cell activation to promote IL-17 in ILC217s. Il17-/- ILC2s were less pathogenic in lung inflammation. ILC217s concomitantly expressed IL-5 and IL-13 but expressed little GM-CSF. CONCLUSION: During lung inflammation, IL-33 and leukotrienes synergistically induce ILC217s. ILC217s are a highly pathogenic and unexpected source for IL-17 in lung inflammation.


Subject(s)
Immunity, Innate , Interleukin-17/immunology , Lung/immunology , Lymphocytes/immunology , Pneumonia/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Gene Expression Regulation/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-17/genetics , Interleukin-33/genetics , Interleukin-33/immunology , Interleukins/genetics , Interleukins/immunology , Leukotrienes/genetics , Leukotrienes/immunology , Lung/pathology , Lymphocytes/pathology , Mice , Mice, Knockout , Papain/pharmacology , Pneumonia/genetics , Pneumonia/pathology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology
14.
Biochim Biophys Acta Rev Cancer ; 1870(1): 51-66, 2018 08.
Article in English | MEDLINE | ID: mdl-29959989

ABSTRACT

While metabolic reprogramming of cancer cells has long been considered from the standpoint of how and why cancer cells preferentially utilize glucose via aerobic glycolysis, the so-called Warburg Effect, the progress in the following areas during the past several years has substantially advanced our understanding of the rewired metabolic network in cancer cells that is intertwined with oncogenic signaling. First, in addition to the major nutrient substrates glucose and glutamine, cancer cells have been discovered to utilize a variety of unconventional nutrient sources for survival. Second, the deregulated biomass synthesis is intertwined with cell cycle progression to coordinate the accelerated progression of cancer cells. Third, the reciprocal regulation of cancer cell's metabolic alterations and the microenvironment, involving extensive host immune cells and microbiota, have come into view as critical mechanisms to regulate cancer progression. These and other advances are shaping the current and future paradigm of cancer metabolism.


Subject(s)
Neoplasms/metabolism , Tumor Microenvironment , Amino Acids, Branched-Chain/metabolism , Cell Proliferation , Humans , Nutrients , Pentose Phosphate Pathway , Serine/biosynthesis
15.
Sci Rep ; 7(1): 5166, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701769

ABSTRACT

Perfluorooctane sulfonate (PFOS) is an environmental contaminant that has been manufactured to be used as surfactants and repellents in industry. Due to long half-life for clearance and degradation, PFOS is accumulative in human body and has potential threat to human health. Previous studies have shown the development and function of immune cells can be affected by PFOS. Although PFOS has a high chance of being absorbed through the oral route, whether and how PFOS affects immune cells in the gut is unknown. Using mouse model of Citrobacter rodentium infection, we investigated the role of PFOS on intestinal immunity. We found at early phase of the infection, PFOS inhibited the expansion of the pathogen by promoting IL-22 production from the group 3 innate lymphoid cell (ILC3) in an aryl hydrocarbon receptor dependent manner. Nevertheless, persistent PFOS treatment in mice finally led to a failure to clear the pathogen completely. At late phase of infection, enhanced bacterial counts in PFOS treated mice were accompanied by increased inflammatory cytokines, reduced mucin production and dysbiosis, featured by decreased level of Lactobacillus casei, Lactobacillus johnsonii and increased E. coli. Our study reveals a deleterious consequence in intestinal bacterial infection caused by PFOS accumulation.


Subject(s)
Alkanesulfonic Acids/pharmacology , Bacterial Infections/immunology , Bacterial Infections/microbiology , Fluorocarbons/pharmacology , Intestines/drug effects , Intestines/immunology , Intestines/microbiology , Animals , Bacterial Infections/drug therapy , Bacterial Infections/pathology , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Mice , Mucins/metabolism , Severity of Illness Index , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...