Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Clin Invest ; 134(6)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38488001

ABSTRACT

Breast cancer stem cells (BCSCs) mitigate oxidative stress to maintain their viability and plasticity. However, the regulatory mechanism of oxidative stress in BCSCs remains unclear. We recently found that the histone reader ZMYND8 was upregulated in BCSCs. Here, we showed that ZMYND8 reduced ROS and iron to inhibit ferroptosis in aldehyde dehydrogenase-high (ALDHhi) BCSCs, leading to BCSC expansion and tumor initiation in mice. The underlying mechanism involved a two-fold posttranslational regulation of nuclear factor erythroid 2-related factor 2 (NRF2). ZMYND8 increased stability of NRF2 protein through KEAP1 silencing. On the other hand, ZMYND8 interacted with and recruited NRF2 to the promoters of antioxidant genes to enhance gene transcription in mammospheres. NRF2 phenocopied ZMYND8 to enhance BCSC stemness and tumor initiation by inhibiting ROS and ferroptosis. Loss of NRF2 counteracted ZMYND8's effects on antioxidant genes and ROS in mammospheres. Interestingly, ZMYND8 expression was directly controlled by NRF2 in mammospheres. Collectively, these findings uncover a positive feedback loop that amplifies the antioxidant defense mechanism sustaining BCSC survival and stemness.


Subject(s)
Breast Neoplasms , Ferroptosis , NF-E2-Related Factor 2 , Neoplastic Stem Cells , Trans-Activators , Animals , Mice , Antioxidants , Ferroptosis/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Trans-Activators/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology
2.
J Clin Invest ; 133(17)2023 09 01.
Article in English | MEDLINE | ID: mdl-37655663

ABSTRACT

SAP30 is a core subunit of the transcriptional corepressor SIN3 complex, but little is known about its role in gene regulation and human cancer. Here, we show that SAP30 was a nonmutational oncoprotein upregulated in more than 50% of human breast tumors and correlated with unfavorable outcomes in patients with breast cancer. In various breast cancer mouse models, we found that SAP30 promoted tumor growth and metastasis through its interaction with SIN3A/3B. Surprisingly, the canonical gene silencing role was not essential for SAP30's tumor-promoting actions. SAP30 enhanced chromatin accessibility and RNA polymerase II occupancy at promoters in breast cancer cells, acting as a coactivator for genes involved in cell motility, angiogenesis, and lymphangiogenesis, thereby driving tumor progression. Notably, SAP30 formed a homodimer with 1 subunit binding to SIN3A and another subunit recruiting MLL1 through specific Phe186/200 residues within its transactivation domain. MLL1 was required for SAP30-mediated transcriptional coactivation and breast tumor progression. Collectively, our findings reveal that SAP30 represents a transcriptional dependency in breast cancer.


Subject(s)
Breast Neoplasms , Mammary Neoplasms, Animal , Sin3 Histone Deacetylase and Corepressor Complex , Animals , Female , Humans , Mice , Breast Neoplasms/genetics , Cell Nucleus , Chromatin , Histone Deacetylases/genetics , Sin3 Histone Deacetylase and Corepressor Complex/genetics
3.
Mol Psychiatry ; 27(12): 5213-5226, 2022 12.
Article in English | MEDLINE | ID: mdl-36028572

ABSTRACT

The excitatory neurotransmitter glutamate shapes learning and memory, but the underlying epigenetic mechanism of glutamate regulation in neuron remains poorly understood. Here, we showed that lysine demethylase KDM6B was expressed in excitatory neurons and declined in hippocampus with age. Conditional knockout of KDM6B in excitatory neurons reduced spine density, synaptic vesicle number and synaptic activity, and impaired learning and memory without obvious effect on brain morphology in mice. Mechanistically, KDM6B upregulated vesicular glutamate transporter 1 and 2 (VGLUT1/2) in neurons through demethylating H3K27me3 at their promoters. Tau interacted and recruited KDM6B to the promoters of Slc17a7 and Slc17a6, leading to a decrease in local H3K27me3 levels and induction of VGLUT1/2 expression in neurons, which could be prevented by loss of Tau. Ectopic expression of KDM6B, VGLUT1, or VGLUT2 restored spine density and synaptic activity in KDM6B-deficient cortical neurons. Collectively, these findings unravel a fundamental mechanism underlying epigenetic regulation of synaptic plasticity and cognition.


Subject(s)
Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases , Neuronal Plasticity , tau Proteins , Animals , Mice , Cognition/physiology , Glutamic Acid/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Synapses/metabolism , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism , tau Proteins/metabolism
4.
Sci Adv ; 8(28): eabn5295, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35857506

ABSTRACT

27-Hydroxycholesterol (27-HC) is the most abundant oxysterol that increases the risk of breast cancer progression. However, little is known about epigenetic regulation of 27-HC metabolism and its role in breast tumor initiation. Using genetic mouse mammary tumor and human breast cancer models, we showed here that the histone reader ZMYND8 was selectively expressed in breast cancer stem cells (BCSCs) and promoted epithelial-mesenchymal transition (EMT), BCSC maintenance and self-renewal, and oncogenic transformation through its epigenetic functions, leading to breast tumor initiation. Mechanistically, ZMYND8 was a master transcriptional regulator of 27-HC metabolism. It increased cholesterol biosynthesis and oxidation but blocked cholesterol efflux and 27-HC catabolism, leading to accumulation of 27-HC in BCSCs. Consequently, 27-HC promoted EMT, oncogenic transformation, and tumor initiation through activation of liver X receptor. These findings reveal that ZMYND8 is an epigenetic booster that drives breast tumor initiation through metabolic reprogramming.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/pathology , Carcinogenesis/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cholesterol/metabolism , Epigenesis, Genetic , Female , Humans , Hydroxycholesterols , Mice , Neoplastic Stem Cells/metabolism
5.
Cancer Res ; 82(13): 2388-2402, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35499760

ABSTRACT

Branched-chain amino acid transaminase 1 (BCAT1) is upregulated selectively in human isocitrate dehydrogenase (IDH) wildtype (WT) but not mutant glioblastoma multiforme (GBM) and promotes IDHWT GBM growth. Through a metabolic synthetic lethal screen, we report here that α-ketoglutarate (AKG) kills IDHWT GBM cells when BCAT1 protein is lost, which is reversed by reexpression of BCAT1 or supplementation with branched-chain α-ketoacids (BCKA), downstream metabolic products of BCAT1. In patient-derived IDHWT GBM tumors in vitro and in vivo, cotreatment of BCAT1 inhibitor gabapentin and AKG resulted in synthetic lethality. However, AKG failed to evoke a synthetic lethal effect with loss of BCAT2, BCKDHA, or GPT2 in IDHWT GBM cells. Mechanistically, loss of BCAT1 increased the NAD+/NADH ratio but impaired oxidative phosphorylation, mTORC1 activity, and nucleotide biosynthesis. These metabolic alterations were synergistically augmented by AKG treatment, thereby causing mitochondrial dysfunction and depletion of cellular building blocks, including ATP, nucleotides, and proteins. Partial restoration of ATP, nucleotides, proteins, and mTORC1 activity by BCKA supplementation prevented IDHWT GBM cell death conferred by the combination of BCAT1 loss and AKG. These findings define a targetable metabolic vulnerability in the most common subset of GBM that is currently incurable. SIGNIFICANCE: Metabolic synthetic lethal screening in IDHWT glioblastoma defines a vulnerability to ΑΚG following BCAT1 loss, uncovering a therapeutic strategy to improve glioblastoma treatment. See related commentary by Meurs and Nagrath, p. 2354.


Subject(s)
Glioblastoma , Adenosine Triphosphate , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Ketoglutaric Acids/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Nucleotides , Synthetic Lethal Mutations , Transaminases/genetics , Transaminases/metabolism
6.
Cell Mol Life Sci ; 79(1): 39, 2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34921640

ABSTRACT

Traumatic brain injury (TBI), often induced by sports, car accidents, falls, or other daily occurrences, is a primary non-genetically related risk factor for the development of subsequent neurodegeneration and neuronal cell death. However, the molecular mechanisms underlying neurodegeneration, cell death, and neurobehavioral dysfunction following TBI remain unclear. Here, we found that poly(ADP-ribose) polymerase-1 (PARP-1) was hyperactivated following TBI and its inhibition reduced TBI-induced brain injury. Macrophage migration inhibitory factor (MIF), a newly identified nuclease involved in PARP-1-dependent cell death, was translocated from the cytosol to the nucleus in cortical neurons following TBI and promoted neuronal cell death in vivo. Genetic deletion of MIF protected neurons from TBI-induced dendritic spine loss, morphological complexity degeneration, and subsequent neuronal cell death in mice. Moreover, MIF knockout reduced the brain injury volume and improved long-term animal behavioral rehabilitation. These neuroprotective effects in MIF knockout mice were reversed by the expression of wild-type MIF but not nuclease-deficient MIF mutant. In contrast, genetic deletion of MIF did not alter TBI-induced neuroinflammation. These findings reveal that MIF mediates TBI-induced neurodegeneration, neuronal cell death and neurobehavioral dysfunction through its nuclease activity, but not its pro-inflammatory role. Targeting MIF's nuclease activity may offer a novel strategy to protect neurons from TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Nerve Degeneration/metabolism , Poly (ADP-Ribose) Polymerase-1/physiology , Animals , Cell Death , Male , Mice , Mice, Knockout
7.
Nat Commun ; 12(1): 2954, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34012010

ABSTRACT

How cancer cells cope with high levels of replication stress during rapid proliferation is currently unclear. Here, we show that macrophage migration inhibitory factor (MIF) is a 3' flap nuclease that translocates to the nucleus in S phase. Poly(ADP-ribose) polymerase 1 co-localizes with MIF to the DNA replication fork, where MIF nuclease activity is required to resolve replication stress and facilitates tumor growth. MIF loss in cancer cells leads to mutation frequency increases, cell cycle delays and DNA synthesis and cell growth inhibition, which can be rescued by restoring MIF, but not nuclease-deficient MIF mutant. MIF is significantly upregulated in breast tumors and correlates with poor overall survival in patients. We propose that MIF is a unique 3' nuclease, excises flaps at the immediate 3' end during DNA synthesis and favors cancer cells evading replication stress-induced threat for their growth.


Subject(s)
Breast Neoplasms/metabolism , DNA Replication/physiology , Flap Endonucleases/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , DNA/chemistry , DNA/metabolism , DNA Damage , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , DNA Replication/genetics , Female , Flap Endonucleases/deficiency , Flap Endonucleases/genetics , Gene Knockout Techniques , Genomic Instability , HCT116 Cells , Humans , Intramolecular Oxidoreductases/deficiency , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/deficiency , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nucleic Acid Conformation , Poly (ADP-Ribose) Polymerase-1/metabolism , S Phase , Substrate Specificity
8.
Mol Neurodegener ; 16(1): 25, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33853653

ABSTRACT

BACKGROUND: Apoptosis-inducing factor (AIF), as a mitochondrial flavoprotein, plays a fundamental role in mitochondrial bioenergetics that is critical for cell survival and also mediates caspase-independent cell death once it is released from mitochondria and translocated to the nucleus under ischemic stroke or neurodegenerative diseases. Although alternative splicing regulation of AIF has been implicated, it remains unknown which AIF splicing isoform will be induced under pathological conditions and how it impacts mitochondrial functions and neurodegeneration in adult brain. METHODS: AIF splicing induction in brain was determined by multiple approaches including 5' RACE, Sanger sequencing, splicing-specific PCR assay and bottom-up proteomic analysis. The role of AIF splicing in mitochondria and neurodegeneration was determined by its biochemical properties, cell death analysis, morphological and functional alterations and animal behavior. Three animal models, including loss-of-function harlequin model, gain-of-function AIF3 knockin model and conditional inducible AIF splicing model established using either Cre-loxp recombination or CRISPR/Cas9 techniques, were applied to explore underlying mechanisms of AIF splicing-induced neurodegeneration. RESULTS: We identified a nature splicing AIF isoform lacking exons 2 and 3 named as AIF3. AIF3 was undetectable under physiological conditions but its expression was increased in mouse and human postmortem brain after stroke. AIF3 splicing in mouse brain caused enlarged ventricles and severe neurodegeneration in the forebrain regions. These AIF3 splicing mice died 2-4 months after birth. AIF3 splicing-triggered neurodegeneration involves both mitochondrial dysfunction and AIF3 nuclear translocation. We showed that AIF3 inhibited NADH oxidase activity, ATP production, oxygen consumption, and mitochondrial biogenesis. In addition, expression of AIF3 significantly increased chromatin condensation and nuclear shrinkage leading to neuronal cell death. However, loss-of-AIF alone in harlequin or gain-of-AIF3 alone in AIF3 knockin mice did not cause robust neurodegeneration as that observed in AIF3 splicing mice. CONCLUSIONS: We identified AIF3 as a disease-inducible isoform and established AIF3 splicing mouse model. The molecular mechanism underlying AIF3 splicing-induced neurodegeneration involves mitochondrial dysfunction and AIF3 nuclear translocation resulting from the synergistic effect of loss-of-AIF and gain-of-AIF3. Our study provides a valuable tool to understand the role of AIF3 splicing in brain and a potential therapeutic target to prevent/delay the progress of neurodegenerative diseases.


Subject(s)
Alternative Splicing , Apoptosis Inducing Factor/physiology , Mitochondria/metabolism , Nerve Degeneration/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , Animals , Apoptosis Inducing Factor/deficiency , Apoptosis Inducing Factor/genetics , Cells, Cultured , Child , Disease Models, Animal , Exons/genetics , Female , Frontal Lobe/chemistry , Gain of Function Mutation , Gene Editing , Gene Knock-In Techniques , Humans , Infant , Infant, Newborn , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Loss of Function Mutation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Middle Aged , Neurons/metabolism , Oxidation-Reduction , Oxygen Consumption , Protein Isoforms/genetics , Protein Isoforms/physiology
9.
Mol Cancer Res ; 19(4): 678-687, 2021 04.
Article in English | MEDLINE | ID: mdl-33380467

ABSTRACT

Hypoxia induces thousands of mRNAs and miRNAs to mediate tumor malignancy. However, hypoxia-induced long noncoding RNA (lncRNA) transcriptome and their role in triple-negative breast cancer (TNBC) have not been defined. Here we identified hypoxia-induced lncRNA transcriptome in two human TNBC cell lines by whole transcriptome sequencing. AC093818.1 was one of 26 validated lncRNAs and abundantly expressed in TNBC in vitro and in vivo. 5'- and 3'-rapid amplification of cDNA ends assays revealed that the isoform 2 was a dominant AC093818.1 transcript in TNBC cells and thus referred to as lncIHAT (lncRNA induced by hypoxia and abundant in TNBC). Hypoxia-inducible factor 1 (HIF1) but not HIF2 bound to the hypoxia response element at the promoter of lncIHAT to activate its transcription in hypoxic TNBC cells. LncIHAT promoted TNBC cell survival in vitro and tumor growth and lung metastasis in mice. Mechanistically, lncIHAT was required for the expression of its proximal neighboring oncogenic genes PDK1 and ITGA6 in TNBC cells and tumors. Reexpression of PDK1 and ITGA6 rescued survival and growth of lncIHAT knockdown TNBC cells in vitro. Collectively, these findings uncovered lncIHAT as a new hypoxia-induced oncogenic cis-acting lncRNA in TNBC. IMPLICATIONS: This study systematically identified hypoxia-induced lncRNA transcriptome in TNBC and sheds light on multiple layers of regulatory mechanisms of gene expression under hypoxia.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , RNA, Long Noncoding/biosynthesis , Triple Negative Breast Neoplasms/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Disease Progression , Female , HEK293 Cells , Heterografts , Humans , Hypoxia-Inducible Factor 1/genetics , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Long Noncoding/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
10.
Cancer Res ; 81(1): 174-186, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33148660

ABSTRACT

Emerging studies indicate that DNA damage in cancer cells triggers antitumor immunity, but its intrinsic regulatory mechanism in breast cancer cells remains poorly understood. Here, we show that ZMYND8 is upregulated and inhibits micronucleus formation and DNA damage in breast cancer cells. Loss of ZMYND8 triggered activation of the DNA sensor cyclic guanosine monophosphate-adenosine monophosphate synthase in micronuclei, leading to further activation of the downstream signaling effectors stimulator of IFN genes and NF-κB, but not TANK-binding kinase 1 and IFN regulatory factor 3, thereby inducing the expression of IFNß and IFN-stimulated genes (ISG) in breast cancer cells in vitro and tumors in vivo. ZMYND8 knockout (KO) in breast cancer cells promoted infiltration of CD4+ and CD8+ T cells, leading to tumor inhibition in syngeneic mouse models, which was significantly attenuated by treatment of anti-CD4/CD8-depleting antibodies or anti-IFNAR1 antibody and in immunodeficient Rag1 KO mice. In human breast tumors, ZMYND8 was negatively correlated with ISGs, CD4, CD8A, CD8B, and the tumor-lymphocyte infiltration phenotype. Collectively, these findings demonstrate that maintenance of genome stability by ZMYND8 causes breast cancer cells to evade cytotoxic T-lymphocyte surveillance, which leads to tumor growth. SIGNIFICANCE: These findings show that ZMYND8 is a new negative and intrinsic regulator of the innate immune response in breast tumor cells, and ZMYND8 may be a possible target for antitumor immunotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
11.
Cancer Res ; 80(18): 3880-3891, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32699137

ABSTRACT

Recruitment of RNA polymerase II to hypoxia-inducible factor (HIF) target genes under normoxia is a prerequisite for HIF-mediated transactivation. However, the underlying mechanism of this recruitment remains unknown. Here we report that chromodomain helicase DNA-binding protein 4 (CHD4) physically interacts with α and ß subunits of HIF1 and HIF2 and enhances HIF-driven transcriptional programs to promote breast cancer progression. Loss of HIF1/2α abolished CHD4-mediated breast tumor growth in mice. In breast cancer cells under normoxia, CHD4 enrichment at HIF target gene promoters increased RNA polymerase II loading through p300. Hypoxia further promoted CHD4 binding to the chromatin via HIF1/2α, where CHD4 in turn enhanced recruitment of HIF1α, leading to HIF target gene transcription. CHD4 was upregulated and correlated with HIF target gene expression in human breast tumors; upregulation of CHD4 and other known HIF coactivators in human breast tumors was mutually exclusive. Furthermore, CHD4 was associated with poor overall survival of patients with breast cancer. Collectively, these findings reveal a new fundamental mechanism of HIF regulation in breast cancer, which has clinical relevance. SIGNIFICANCE: This study identifies CHD4 as a HIF coactivator and elucidates the fundamental mechanism underlying CHD4-mediated HIF transactivation in breast tumors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/metabolism , Disease Progression , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Breast Neoplasms/pathology , Chromatin Immunoprecipitation/methods , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , RNA Polymerase II/metabolism , Transcriptional Activation , Tumor Hypoxia
12.
Cancer Res ; 80(5): 964-975, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31900259

ABSTRACT

Hypoxia induces a vast array of long noncoding RNAs (lncRNA) in breast cancer cells, but their biological functions remain largely unknown. Here, we identified a hitherto uncharacterized hypoxia-induced lncRNA RAB11B-AS1 in breast cancer cells. RAB11B-AS1 is a natural lncRNA upregulated in human breast cancer and its expression is induced by hypoxia-inducible factor 2 (HIF2), but not HIF1, in response to hypoxia. RAB11B-AS1 enhanced the expression of angiogenic factors including VEGFA and ANGPTL4 in hypoxic breast cancer cells by increasing recruitment of RNA polymerase II. In line with increased angiogenic factors, conditioned media from RAB11B-AS1-overexpressing breast cancer cells promoted tube formation of human umbilical vein endothelial cells in vitro. Gain- and loss-of-function studies revealed that RAB11B-AS1 increased breast cancer cell migration and invasion in vitro and promoted tumor angiogenesis and breast cancer distant metastasis without affecting primary tumor growth in mice. Taken together, these findings uncover a fundamental mechanism of hypoxia-induced tumor angiogenesis and breast cancer metastasis. SIGNIFICANCE: This study reveals the molecular mechanism by which the lncRNA RAB11B-AS1 regulates hypoxia-induced angiogenesis and breast cancer metastasis, and provides new insights into the functional interaction between a lncRNA and tumor microenvironment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/5/964/F1.large.jpg.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/genetics , RNA, Long Noncoding/metabolism , Angiopoietin-Like Protein 4/genetics , Animals , Breast Neoplasms/blood supply , Cell Hypoxia/genetics , Cell Line, Tumor , Endothelial Cells , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Neoplasm Metastasis/genetics , Neovascularization, Pathologic/pathology , RNA Polymerase II/metabolism , Tumor Microenvironment/genetics , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays
13.
Nucleic Acids Res ; 46(13): 6576-6591, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29860315

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a master transcriptional regulator in response to hypoxia and its transcriptional activity is crucial for cancer cell mobility. Here we present evidence for a novel epigenetic mechanism that regulates HIF-1 transcriptional activity and HIF-1-dependent migration of glioblastoma cells. The lysine methyltransferases G9a and GLP directly bound to the α subunit of HIF-1 (HIF-1α) and catalyzed mono- and di-methylation of HIF-1α at lysine (K) 674 in vitro and in vivo. K674 methylation suppressed HIF-1 transcriptional activity and expression of its downstream target genes PTGS1, NDNF, SLC6A3, and Linc01132 in human glioblastoma U251MG cells. Inhibition of HIF-1 by K674 methylation is due to reduced HIF-1α transactivation domain function but not increased HIF-1α protein degradation or impaired binding of HIF-1 to hypoxia response elements. K674 methylation significantly decreased HIF-1-dependent migration of U251MG cells under hypoxia. Importantly, we found that G9a was downregulated by hypoxia in glioblastoma, which was inversely correlated with PTGS1 expression and survival of patients with glioblastoma. Therefore, our findings uncover a hypoxia-induced negative feedback mechanism that maintains high activity of HIF-1 and cell mobility in human glioblastoma.


Subject(s)
Autoantigens/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Golgi Matrix Proteins/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transcription, Genetic , Cell Hypoxia , Cell Line , Cell Movement , Glioblastoma/metabolism , Glioblastoma/physiopathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Lysine/metabolism , Methylation , Response Elements
14.
J Clin Invest ; 128(5): 1937-1955, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29629903

ABSTRACT

Altered epigenetic reprogramming contributes to breast cancer progression and metastasis. How the epigenetic reader mediates breast cancer progression remains poorly understood. Here, we showed that the epigenetic reader zinc finger MYND-type containing 8 (ZMYND8) is induced by HIF-1 and HIF-2 in breast cancer cells and also upregulated in human breast tumors, and is correlated with poor survival of patients with breast cancer. Genetic deletion of ZMYND8 decreases breast cancer cell colony formation, migration, and invasion in vitro, and inhibits breast tumor growth and metastasis to the lungs in mice. The ZMYND8's oncogenic effect in breast cancer requires HIF-1 and HIF-2. We further showed that ZMYND8 interacts with HIF-1α and HIF-2α and enhances elongation of the global HIF-induced oncogenic genes by increasing recruitment of BRD4 and subsequent release of paused RNA polymerase II in breast cancer cells. ZMYND8 acetylation at lysines 1007 and 1034 by p300 is required for HIF activation and breast cancer progression and metastasis. These findings uncover a primary epigenetic mechanism of HIF activation and HIF-mediated breast cancer progression, and discover a possible molecular target for the diagnosis and treatment of breast cancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Tumor Suppressor Proteins/metabolism , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins , Female , HEK293 Cells , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MCF-7 Cells , Mice, SCID , Neoplasm Metastasis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics
15.
Science ; 354(6308)2016 10 07.
Article in English | MEDLINE | ID: mdl-27846469

ABSTRACT

Inhibition or genetic deletion of poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) is protective against toxic insults in many organ systems. The molecular mechanisms underlying PARP-1-dependent cell death involve release of mitochondrial apoptosis-inducing factor (AIF) and its translocation to the nucleus, which results in chromatinolysis. We identified macrophage migration inhibitory factor (MIF) as a PARP-1-dependent AIF-associated nuclease (PAAN). AIF was required for recruitment of MIF to the nucleus, where MIF cleaves genomic DNA into large fragments. Depletion of MIF, disruption of the AIF-MIF interaction, or mutation of glutamic acid at position 22 in the catalytic nuclease domain blocked MIF nuclease activity and inhibited chromatinolysis, cell death induced by glutamate excitotoxicity, and focal stroke. Inhibition of MIF's nuclease activity is a potential therapeutic target for diseases caused by excessive PARP-1 activation.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis , DNA Cleavage , DNA Damage , DNA, Single-Stranded/metabolism , Deoxyribonucleases/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Apoptosis/genetics , Apoptosis Inducing Factor/genetics , Base Sequence , Catalytic Domain , Cell Nucleus/enzymology , Chromatin/metabolism , DNA Damage/genetics , DNA Fragmentation , Deoxyribonucleases/chemistry , Deoxyribonucleases/genetics , Glutamic Acid/chemistry , Glutamic Acid/genetics , Glutamic Acid/toxicity , HeLa Cells , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Knockout , Mitochondria/enzymology , Mutation , Neurons/enzymology , Nucleic Acid Conformation , Oxidative Stress , Stroke/enzymology , Stroke/genetics
16.
PLoS One ; 8(4): e61270, 2013.
Article in English | MEDLINE | ID: mdl-23620738

ABSTRACT

Innate behaviors have their origins in the specification of neural fates during development. Within Drosophila, BTB (Bric-a-brac,Tramtrack, Broad) domain proteins such as Fruitless are known to play key roles in the neural differentiation underlying such responses. We previously identified a gene, which we have termed jim lovell (lov), encoding a BTB protein with a role in gravity responses. To understand more fully the behavioral roles of this gene we have investigated its function through several approaches. Transcript and protein expression patterns have been examined and behavioral phenotypes of new lov mutations have been characterized. Lov is a nuclear protein, suggesting a role as a transcriptional regulator, as for other BTB proteins. In late embryogenesis, Lov is expressed in many CNS and PNS neurons. An examination of the PNS expression indicates that lov functions in the late specification of several classes of sensory neurons. In particular, only two of the five abdominal lateral chordotonal neurons express Lov, predicting functional variation within this highly similar group. Surprisingly, Lov is also expressed very early in embryogenesis in ways that suggests roles in morphogenetic movements, amnioserosa function and head neurogenesis. The phenotypes of two new lov mutations that delete adjacent non-coding DNA regions are strikingly different suggesting removal of different regulatory elements. In lov(47) , Lov expression is lost in many embryonic neurons including the two lateral chordotonal neurons. lov(47) mutant larvae show feeding and locomotor defects including spontaneous backward movement. Adult lov(47) males perform aberrant courtship behavior distinguished by courtship displays that are not directed at the female. lov(47) adults also show more defective negative gravitaxis than the previously isolated lov(91Y) mutant. In contrast, lov(66) produces largely normal behavior but severe female sterility associated with ectopic lov expression in the ovary. We propose a negative regulatory role for the DNA deleted in lov(66) .


Subject(s)
Aging/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Behavior, Animal , Cell Differentiation/genetics , Courtship , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental , Genetic Loci/genetics , Genotype , Larva/metabolism , Male , Mutation/genetics , Neurons/cytology , Neurons/metabolism , Organ Specificity/genetics , Ovum/metabolism , Phenotype , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Mol Pharmacol ; 83(3): 587-93, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23229509

ABSTRACT

Regulation of multiple adenylyl cyclases (AC) provides unique inputs to mediate the synthesis of cAMP, a ubiquitous second messenger that controls many aspects of cellular function. On stimulation by G(s), the activities of ACs can be further selectively modulated by other pathways to ensure precise control of intracellular cAMP responses to specific stimuli. Recently, we reported that one of the AC isoforms, AC7, is uniquely regulated by the G(13) pathway. To understand more fully the molecular mechanism of this regulation, we compared the regulation of AC7 with that of AC2 in bone marrow-derived macrophages devoid of AC7. Although both enzymes could fully restore regulation of cAMP by Gßγ, activation of the G(13) pathway preferentially synergized with AC7. Exchange of domains between the two isoforms indicates that the C1b domain and the N-terminus of the C1a domain are important for directing selective regulation of AC7 by the G(13) pathway. A mutagenesis screen identified more specific regions of AC7 that differentially mediate its regulation by distinct pathways.


Subject(s)
Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Animals , Cyclic AMP/metabolism , Female , Humans , Isoenzymes , Lysophospholipids/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis , Protein Structure, Tertiary , Sphingosine/analogs & derivatives , Sphingosine/metabolism
18.
Mol Immunol ; 54(1): 14-22, 2013 May.
Article in English | MEDLINE | ID: mdl-23178822

ABSTRACT

Pathogens use a variety of strategies to evade host immune defenses. A powerful way to suppress immune function is to increase intracellular concentrations of cAMP in host immune cells, which dampens inflammatory responses and prevents microbial killing. We found that the yeast cell wall extract, zymosan, is capable of increasing intracellular cAMP and activates the protein kinase A pathway in bone marrow derived macrophage (BMDM) cells from mice. This response is dependent on adenylyl cyclase type VII (AC7) and heterotrimeric G proteins, primarily G(12/13). Consequently, zymosan induced production of the inflammatory cytokine, TNFα, was much stronger in BMDMs from AC7 deficient mice compared to the response in wild type cells. In a model of zymosan induced peritonitis, mice deficient of AC7 in the myeloid lineage displayed prolonged inflammation. We propose that zymosan induced increases in cAMP and activation of PKA serve as a mechanism to dampen inflammatory responses in host cells, which consequently favors the survival of microbes. This would also help explain a well documented phenomenon, that the ability of zymosan to stimulate inflammatory cytokine responses via TLR2 appears to be weaker than other stimuli of TLR2.


Subject(s)
Adenylyl Cyclases/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Immunity, Innate/drug effects , Inflammation/immunology , Zymosan/pharmacology , Adenylyl Cyclases/metabolism , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Humans , Immunity, Innate/physiology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/physiology , Zymosan/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...