Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Nat Cancer ; 4(10): 1437-1454, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37640929

ABSTRACT

Cholinergic nerves are involved in tumor progression and dissemination. In contrast to other visceral tissues, cholinergic innervation in the hepatic parenchyma is poorly detected. It remains unclear whether there is any form of cholinergic regulation of liver cancer. Here, we show that cholinergic T cells curtail the development of liver cancer by supporting antitumor immune responses. In a mouse multihit model of hepatocellular carcinoma (HCC), we observed activation of the adaptive immune response and induction of two populations of CD4+ T cells expressing choline acetyltransferase (ChAT), including regulatory T cells and dysfunctional PD-1+ T cells. Tumor antigens drove the clonal expansion of these cholinergic T cells in HCC. Genetic ablation of Chat in T cells led to an increased prevalence of preneoplastic cells and exacerbated liver cancer due to compromised antitumor immunity. Mechanistically, the cholinergic activity intrinsic in T cells constrained Ca2+-NFAT signaling induced by T cell antigen receptor engagement. Without this cholinergic modulation, hyperactivated CD25+ T regulatory cells and dysregulated PD-1+ T cells impaired HCC immunosurveillance. Our results unveil a previously unappreciated role for cholinergic T cells in liver cancer immunobiology.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Monitoring, Immunologic , T-Lymphocytes, Regulatory/pathology
2.
Cancer Cell ; 41(2): 323-339.e10, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36736318

ABSTRACT

Angioimmunoblastic T cell lymphoma (AITL) is a peripheral T cell lymphoma that originates from T follicular helper (Tfh) cells and exhibits a prominent tumor microenvironment (TME). IDH2 and TET2 mutations co-occur frequently in AITL, but their contribution to tumorigenesis is poorly understood. We developed an AITL mouse model that is driven by Idh2 and Tet2 mutations. Malignant Tfh cells display aberrant transcriptomic and epigenetic programs that impair TCR signaling. Neoplastic Tfh cells bearing combined Idh2 and Tet2 mutations show altered cross-talk with germinal center B cells that promotes B cell clonal expansion while decreasing Fas-FasL interaction and reducing B cell apoptosis. The plasma cell count and angiogenesis are also increased in the Idh2-mutated tumors, implying a major relationship between Idh2 mutation and the characteristic AITL TME. Our mouse model recapitulates several features of human IDH2-mutated AITL and provides a rationale for exploring therapeutic targeting of Tfh-TME cross-talk for AITL patients.


Subject(s)
Dioxygenases , Immunoblastic Lymphadenopathy , Lymphoma, T-Cell , Animals , Humans , Mice , Dioxygenases/genetics , DNA-Binding Proteins/genetics , Immunoblastic Lymphadenopathy/genetics , Isocitrate Dehydrogenase/genetics , Lymphoma, T-Cell/genetics , Mutation , T Follicular Helper Cells/pathology , T-Lymphocytes, Helper-Inducer , Tumor Microenvironment/genetics
3.
Exp Neurol ; 351: 114010, 2022 05.
Article in English | MEDLINE | ID: mdl-35167826

ABSTRACT

Cisplatin is a member of a widely utilized class of chemotherapeutic agent that initiates DNA damage response, cell cycle arrest, and p53-dependent apoptotic cell death in concert with DNA­platinum adduct formation. While normal programmed cell death (PCD) can occur in the developing neuroepithelium in the absence of caspase-3 within certain genetic backgrounds, we observed an absolute dependency upon this executioner caspase with respect to cisplatin-induced PCD in the developing central nervous system (CNS). We therefore examined the nature of this genotoxic injury in the CNS in vivo, in which cisplatin treatment causes widespread cellular injury consistent with hallmarks of apoptosis which are averted upon caspase-3 inhibition. Examination of cisplatin-mediated injury as a function of time revealed the presence of an alternative, delayed form of necroptosis-like cell death which manifests in Casp3-/- neuroepithelia for several days following the normal pattern of apoptosis. Together, these findings suggest a coordinated regulation of these disparate PCD pathways in response to genotoxic stress in vivo and highlight the unique and critical role which caspase-3 plays among executioner caspases in coordinating apoptotic versus necroptotic responsiveness of the developing CNS to genotoxic injury.


Subject(s)
Caspases , Cisplatin , Apoptosis/physiology , Brain/metabolism , Caspase 3/metabolism , Caspases/metabolism , Cisplatin/toxicity
4.
Cell Mol Neurobiol ; 41(8): 1727-1742, 2021 Nov.
Article in English | MEDLINE | ID: mdl-32844322

ABSTRACT

The primary forms of cell death seen in ischemic stroke are of two major types: a necrotic/necroptotic form, and an apoptotic form that is frequently seen in penumbral regions of injury. Typically apoptotic versus necroptotic programmed cell death is described as competitive in nature, where necroptosis is often described as playing a backup role to apoptosis. In the present study, we examined the relationship between these two forms of cell death in a murine endothelin-1 model of ischemia-reperfusion injury in wildtype and caspase-3 null mice with and without addition of the pharmacologic RIPK1 phosphorylation inhibitor necrostatin-1. Analyses of ischemic brain injury were performed via both cellular and volumetric assessments, electron microscopy, TUNEL staining, activated caspase-3 and caspase-7 staining, as well as CD11b and F4/80 staining. Inhibition of caspase-3 or RIPK1 phosphorylation demonstrates significant neural protective effects which are non-additive and exhibit significant overlap in protected regions. Interestingly, morphologic analysis of the cortex demonstrates reduced apoptosis following RIPK1 inhibition. Consistent with this, RIPK1 inhibition reduces the levels of both caspase-3 and caspase-7 activation. Additionally, this protection appears independent of secondary inflammatory mediators. Together, these observations demonstrate that the necroptotic protein RIPK1 modifies caspase-3/-7 activity, ultimately resulting in decreased neuronal apoptosis. These findings thus modify the traditional exclusionary view of apoptotic/necroptotic signaling, revealing a new form of interaction between these dominant forms of cell death.


Subject(s)
Apoptosis/drug effects , Brain Ischemia/chemically induced , Brain Ischemia/pathology , Endothelin-1/toxicity , Animals , Apoptosis/physiology , Brain Ischemia/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/chemically induced , Necrosis/metabolism , Necrosis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
5.
PLoS One ; 15(12): e0244366, 2020.
Article in English | MEDLINE | ID: mdl-33382742

ABSTRACT

Dendritic cells are sentinels of the immune system and represent a key cell in the activation of the adaptive immune response. Hypoxia-inducible factor 1 alpha (HIF-1α)-a crucial oxygen sensor stabilized during hypoxic conditions-has been shown to have both activating and inhibitory effects in immune cells in a context- and cell-dependent manner. Previous studies have demonstrated that in some immune cell types, HIF-1α serves a pro-inflammatory role. Genetic deletion of HIF-1α in macrophages has been reported to reduce their pro-inflammatory function. In contrast, loss of HIF-1α enhanced the pro-inflammatory activity of dendritic cells in a bacterial infection model. In this study, we aimed to further clarify the effects of HIF-1α in dendritic cells. Constitutive expression of HIF-1α resulted in diminished immunostimulatory capacity of dendritic cells in vivo, while conditional deletion of HIF-1α in dendritic cells enhanced their ability to induce a cytotoxic T cell response. HIF-1α-expressing dendritic cells demonstrated increased production of inhibitory mediators including IL-10, iNOS and VEGF, which correlated with their reduced capacity to drive effector CD8+ T cell function. Altogether, these data reveal that HIF-1α can promote the anti-inflammatory functions of dendritic cells and provides insight into dysfunctional immune responses in the context of HIF-1α activation.


Subject(s)
Biomarkers/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Animals , Cells, Cultured , Dendritic Cells/metabolism , Gene Knockout Techniques , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-10/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Vascular Endothelial Growth Factor A/metabolism
6.
Apoptosis ; 24(7-8): 578-595, 2019 08.
Article in English | MEDLINE | ID: mdl-31073782

ABSTRACT

Over the past 30 years a number of animal models of cerebral ischemic injury have been developed. Middle cerebral artery occlusion (MCAO) in particular reproduces both ischemic and reperfusion elements and is widely utilized as a model of ischemic stroke in rodents. However substantial variability exists in this model even in clonal inbred mice due to stochastic elements of the cerebral vasculature. Models such as MCAO thus exhibit significant irreducible variabilities with respect to their zone of injury as well as inducing a sizable volume of injury to the cerebrum with damage to sub-cortical structures, conditions not typically seen for the majority of human clinical strokes. An alternative model utilizes endothelin-1 application focally to cerebral vasculature, resulting in an ischemic reperfusion injury which more closely mimics that seen in human clinical stroke. In order to further define this model we demonstrate that intra-cortical administration of ET-1 results in a highly reproducible pattern of tissue injury which is limited to the cerebral cortex, characterizing the early cellular and molecular events which occur during the first 24 h post-injury. In addition we demonstrate that caspase-3 is both necessary and sufficient to regulate a majority of cortical cell death observed during this period. The enhanced survival effects seen upon genetic deletion of caspase-3 appear to arise as a result of direct modification of cell autonomous PCD signaling as opposed to secondary effectors such as granulocyte infiltration or microglia activation. Taken together these findings detail the early mechanistic features regulating endothelin-1-mediated ischemic injury.


Subject(s)
Brain Ischemia/chemically induced , Caspase 3/metabolism , Cerebral Cortex/drug effects , Endothelin-1/toxicity , Animals , Brain Ischemia/pathology , Caspase 3/genetics , Cell Death/drug effects , Cerebral Cortex/injuries , Cerebral Cortex/pathology , Disease Models, Animal , Endothelin-1/administration & dosage , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/enzymology , Neurons/pathology
7.
Science ; 363(6427): 639-644, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30733420

ABSTRACT

Although widely studied as a neurotransmitter, T cell-derived acetylcholine (ACh) has recently been reported to play an important role in regulating immunity. However, the role of lymphocyte-derived ACh in viral infection is unknown. Here, we show that the enzyme choline acetyltransferase (ChAT), which catalyzes the rate-limiting step of ACh production, is robustly induced in both CD4+ and CD8+ T cells during lymphocytic choriomeningitis virus (LCMV) infection in an IL-21-dependent manner. Deletion of Chat within the T cell compartment in mice ablated vasodilation in response to infection, impaired the migration of antiviral T cells into infected tissues, and ultimately compromised the control of chronic LCMV clone 13 infection. Our results reveal a genetic proof of function for ChAT in T cells during viral infection and identify a pathway of T cell migration that sustains antiviral immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Choline O-Acetyltransferase/immunology , Interleukins/immunology , Lymphocytic Choriomeningitis/immunology , Animals , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/enzymology , Cell Movement , Choline O-Acetyltransferase/genetics , Female , Lymphocyte Activation , Lymphocytic choriomeningitis virus , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vasodilation
8.
Proc Natl Acad Sci U S A ; 114(7): E1148-E1157, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137882

ABSTRACT

Wnt signaling, named after the secreted proteins that bind to cell surface receptors to activate the pathway, plays critical roles both in embryonic development and the maintenance of homeostasis in many adult tissues. Two particularly important cellular programs orchestrated by Wnt signaling are proliferation and stem cell self-renewal. Constitutive activation of the Wnt pathway resulting from mutation or improper modulation of pathway components contributes to cancer development in various tissues. Colon cancers frequently bear inactivating mutations of the adenomatous polyposis coli (APC) gene, whose product is an important component of the destruction complex that regulates ß-catenin levels. Stabilization and nuclear localization of ß-catenin result in the expression of a panel of Wnt target genes. We previously showed that Mule/Huwe1/Arf-BP1 (Mule) controls murine intestinal stem and progenitor cell proliferation by modulating the Wnt pathway via c-Myc. Here we extend our investigation of Mule's influence on oncogenesis by showing that Mule interacts directly with ß-catenin and targets it for degradation under conditions of hyperactive Wnt signaling. Our findings suggest that Mule uses various mechanisms to fine-tune the Wnt pathway and provides multiple safeguards against tumorigenesis.


Subject(s)
Tumor Suppressor Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/deficiency , Animals , Axin Protein/biosynthesis , Axin Protein/genetics , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Colonic Neoplasms/metabolism , Cyclin D1/biosynthesis , Cyclin D1/genetics , Down-Regulation , Genes, APC , Genes, Tumor Suppressor , HEK293 Cells , Humans , Mice , Mice, Knockout , Neoplasm Proteins/physiology , Organoids/metabolism , Organoids/ultrastructure , Protein Binding , Protein Processing, Post-Translational , Proteolysis , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
Cancer Cell ; 30(2): 337-348, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27424808

ABSTRACT

Mutations in the isocitrate dehydrogenase-1 gene (IDH1) are common drivers of acute myeloid leukemia (AML) but their mechanism is not fully understood. It is thought that IDH1 mutants act by inhibiting TET2 to alter DNA methylation, but there are significant unexplained clinical differences between IDH1- and TET2-mutant diseases. We have discovered that mice expressing endogenous mutant IDH1 have reduced numbers of hematopoietic stem cells (HSCs), in contrast to Tet2 knockout (TET2-KO) mice. Mutant IDH1 downregulates the DNA damage (DD) sensor ATM by altering histone methylation, leading to impaired DNA repair, increased sensitivity to DD, and reduced HSC self-renewal, independent of TET2. ATM expression is also decreased in human IDH1-mutated AML. These findings may have implications for treatment of IDH-mutant leukemia.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/enzymology , Isocitrate Dehydrogenase/genetics , Proto-Oncogene Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases , Down-Regulation , Hematopoietic Stem Cells/cytology , Humans , Isocitrate Dehydrogenase/metabolism , Mice , Mutation , Proto-Oncogene Proteins/metabolism
10.
Cell Stem Cell ; 19(2): 205-216, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27184401

ABSTRACT

The E3 ubiquitin ligase Mule is often overexpressed in human colorectal cancers, but its role in gut tumorigenesis is unknown. Here, we show in vivo that Mule controls murine intestinal stem and progenitor cell proliferation by modulating Wnt signaling via c-Myc. Mule also regulates protein levels of the receptor tyrosine kinase EphB3 by targeting it for proteasomal and lysosomal degradation. In the intestine, EphB/ephrinB interactions position cells along the crypt-villus axis and compartmentalize incipient colorectal tumors. Our study thus unveils an important new avenue by which Mule acts as an intestinal tumor suppressor by regulation of the intestinal stem cell niche.


Subject(s)
Ephrin-B3/metabolism , Intestines/cytology , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Stem Cell Niche , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway , Adenomatous Polyposis Coli/pathology , Alleles , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Endocytosis , HEK293 Cells , Humans , Mice, Knockout , Models, Biological , Mutation/genetics , Paneth Cells/pathology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/deficiency
11.
Nat Med ; 20(5): 484-92, 2014 May.
Article in English | MEDLINE | ID: mdl-24747746

ABSTRACT

Inflammation has a critical role in the development of insulin resistance. Recent evidence points to a contribution by the central nervous system in the modulation of peripheral inflammation through the anti-inflammatory reflex. However, the importance of this phenomenon remains elusive in type 2 diabetes pathogenesis. Here we show that rat insulin-2 promoter (Rip)-mediated deletion of Pten, a gene encoding a negative regulator of PI3K signaling, led to activation of the cholinergic anti-inflammatory pathway that is mediated by M2 activated macrophages in peripheral tissues. As such, Rip-cre(+) Pten(flox/flox) mice showed lower systemic inflammation and greater insulin sensitivity under basal conditions compared to littermate controls, which were abolished when the mice were treated with an acetylcholine receptor antagonist or when macrophages were depleted. After feeding with a high-fat diet, the Pten-deleted mice remained markedly insulin sensitive, which correlated with massive subcutaneous fat expansion. They also exhibited more adipogenesis with M2 macrophage infiltration, both of which were abolished after disruption of the anti-inflammatory efferent pathway by left vagotomy. In summary, we show that Pten expression in Rip(+) neurons has a critical role in diabetes pathogenesis through mediating the anti-inflammatory reflex.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Inflammation/metabolism , Insulin/genetics , PTEN Phosphohydrolase/genetics , Animals , Anti-Inflammatory Agents/administration & dosage , Central Nervous System/metabolism , Diabetes Mellitus, Type 2/complications , Diet, High-Fat , Humans , Inflammation/complications , Inflammation/drug therapy , Insulin/metabolism , Insulin Resistance/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , PTEN Phosphohydrolase/metabolism , Promoter Regions, Genetic , Rats , Receptors, Muscarinic/administration & dosage , Sequence Deletion , Signal Transduction
12.
Mol Cell ; 53(6): 904-15, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24656129

ABSTRACT

Little is known about how mammalian cells maintain cell size homeostasis. We conducted a novel genetic screen to identify cell-size-controlling genes and isolated Largen, the product of a gene (PRR16) that increased cell size upon overexpression in human cells. In vitro evidence indicated that Largen preferentially stimulates the translation of specific subsets of mRNAs, including those encoding proteins affecting mitochondrial functions. The involvement of Largen in mitochondrial respiration was consistent with the increased mitochondrial mass and greater ATP production in Largen-overexpressing cells. Furthermore, Largen overexpression led to increased cell size in vivo, as revealed by analyses of conditional Largen transgenic mice. Our results establish Largen as an important link between mRNA translation, mitochondrial functions, and the control of mammalian cell size.


Subject(s)
Cell Size/drug effects , Gene Expression Regulation , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/genetics , Animals , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Vectors , High-Throughput Screening Assays , Humans , Jurkat Cells , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Proteins/metabolism , RNA, Messenger/metabolism , Retroviridae/genetics , Retroviridae/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology
13.
Proc Natl Acad Sci U S A ; 111(5): 1843-8, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449892

ABSTRACT

The generation of viable sperm proceeds through a series of coordinated steps, including germ cell self-renewal, meiotic recombination, and terminal differentiation into functional spermatozoa. The p53 family of transcription factors, including p53, p63, and p73, are critical for many physiological processes, including female fertility, but little is known about their functions in spermatogenesis. Here, we report that deficiency of the TAp73 isoform, but not p53 or ΔNp73, results in male infertility because of severe impairment of spermatogenesis. Mice lacking TAp73 exhibited increased DNA damage and cell death in spermatogonia, disorganized apical ectoplasmic specialization, malformed spermatids, and marked hyperspermia. We demonstrated that TAp73 regulates the mRNA levels of crucial genes involved in germ stem/progenitor cells (CDKN2B), spermatid maturation/spermiogenesis (metalloproteinase and serine proteinase inhibitors), and steroidogenesis (CYP21A2 and progesterone receptor). These alterations of testicular histology and gene expression patterns were specific to TAp73 null mice and not features of mice lacking p53. Our work provides previously unidentified in vivo evidence that TAp73 has a unique role in spermatogenesis that ensures the maintenance of mitotic cells and normal spermiogenesis. These results may have implications for the diagnosis and management of human male infertility.


Subject(s)
DNA-Binding Proteins/metabolism , Fertility , Nuclear Proteins/metabolism , Spermatogenesis , Tumor Suppressor Proteins/metabolism , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Aging/pathology , Animals , Apoptosis/genetics , Cell Count , Cell Proliferation , DNA Damage/genetics , DNA-Binding Proteins/deficiency , Female , Fertility/genetics , Gene Expression Regulation , Humans , Infertility, Male/blood , Infertility, Male/genetics , Infertility, Male/pathology , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Oxidative Stress/genetics , Progesterone/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatogenesis/genetics , Spermatozoa/metabolism , Spermatozoa/pathology , Testis/metabolism , Testis/pathology , Tumor Protein p73 , Tumor Suppressor Proteins/deficiency
14.
Proc Natl Acad Sci U S A ; 110(36): 14723-8, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23946427

ABSTRACT

Pancreatic endocrine cells expand rapidly during embryogenesis by neogenesis and proliferation, but during adulthood, islet cells have a very slow turnover. Disruption of murine retinoblastoma tumor suppressor protein (Rb) in mature pancreatic ß-cells has a limited effect on cell proliferation. Here we show that deletion of Rb during embryogenesis in islet progenitors leads to an increase in the neurogenin 3-expressing precursor cell population, which persists in the postnatal period and is associated with increased ß-cell mass in adults. In contrast, Rb-deficient islet precursors, through repression of the cell fate factor aristaless related homeobox, result in decreased α-cell mass. The opposing effect on survival of Rb-deficient α- and ß-cells was a result of opposing effects on p53 in these cell types. As a consequence, loss of Rb in islet precursors led to a reduced α- to ß-cell ratio, leading to improved glucose homeostasis and protection against diabetes.


Subject(s)
Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Retinoblastoma Protein/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Base Sequence , Cell Differentiation/genetics , Cell Line, Tumor , Cell Survival/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Female , Glucagon-Secreting Cells/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , RNA Interference , Retinoblastoma Protein/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
J Exp Med ; 210(8): 1529-44, 2013 Jul 29.
Article in English | MEDLINE | ID: mdl-23857982

ABSTRACT

Oxidative stress plays an important role in cancer development and treatment. Recent data implicate the tumor suppressor BRCA1 in regulating oxidative stress, but the molecular mechanism and the impact in BRCA1-associated tumorigenesis remain unclear. Here, we show that BRCA1 regulates Nrf2-dependent antioxidant signaling by physically interacting with Nrf2 and promoting its stability and activation. BRCA1-deficient mouse primary mammary epithelial cells show low expression of Nrf2-regulated antioxidant enzymes and accumulate reactive oxygen species (ROS) that impair survival in vivo. Increased Nrf2 activation rescues survival and ROS levels in BRCA1-null cells. Interestingly, 53BP1 inactivation, which has been shown to alleviate several defects associated with BRCA1 loss, rescues survival of BRCA1-null cells without restoring ROS levels. We demonstrate that estrogen treatment partially restores Nrf2 levels in the absence of BRCA1. Our data suggest that Nrf2-regulated antioxidant response plays a crucial role in controlling survival downstream of BRCA1 loss. The ability of estrogen to induce Nrf2 posits an involvement of an estrogen-Nrf2 connection in BRCA1 tumor suppression. Lastly, BRCA1-mutated tumors retain a defective antioxidant response that increases the sensitivity to oxidative stress. In conclusion, the role of BRCA1 in regulating Nrf2 activity suggests important implications for both the etiology and treatment of BRCA1-related cancers.


Subject(s)
Antioxidants/metabolism , BRCA1 Protein/metabolism , Cell Survival , NF-E2-Related Factor 2/metabolism , Signal Transduction , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Mutation , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/genetics , Oxidative Stress , Protein Binding , Protein Stability , Reactive Oxygen Species/metabolism , Ubiquitination
16.
Genes Dev ; 27(10): 1101-14, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23699408

ABSTRACT

Tumorigenesis results from dysregulation of oncogenes and tumor suppressors that influence cellular proliferation, differentiation, apoptosis, and/or senescence. Many gene products involved in these processes are substrates of the E3 ubiquitin ligase Mule/Huwe1/Arf-BP1 (Mule), but whether Mule acts as an oncogene or tumor suppressor in vivo remains controversial. We generated K14Cre;Mule(flox/flox(y)) (Mule kKO) mice and subjected them to DMBA/PMA-induced skin carcinogenesis, which depends on oncogenic Ras signaling. Mule deficiency resulted in increased penetrance, number, and severity of skin tumors, which could be reversed by concomitant genetic knockout of c-Myc but not by knockout of p53 or p19Arf. Notably, in the absence of Mule, c-Myc/Miz1 transcriptional complexes accumulated, and levels of p21CDKN1A (p21) and p15INK4B (p15) were down-regulated. In vitro, Mule-deficient primary keratinocytes exhibited increased proliferation that could be reversed by Miz1 knockdown. Transfer of Mule-deficient transformed cells to nude mice resulted in enhanced tumor growth that again could be abrogated by Miz1 knockdown. Our data demonstrate in vivo that Mule suppresses Ras-mediated tumorigenesis by preventing an accumulation of c-Myc/Miz1 complexes that mediates p21 and p15 down-regulation.


Subject(s)
Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Nuclear Proteins/antagonists & inhibitors , Oncogene Protein p21(ras)/metabolism , Protein Inhibitors of Activated STAT/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Genes, ras , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Protein p21(ras)/antagonists & inhibitors , Oncogene Protein p21(ras)/genetics , Protein Inhibitors of Activated STAT/deficiency , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Suppressor Protein p53 , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
17.
Proc Natl Acad Sci U S A ; 109(51): 21034-9, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23213242

ABSTRACT

The endogenous metabolite of estradiol, 2-Methoxyestradiol (2ME2), is an antimitotic and antiangiogenic cancer drug candidate that also exhibits disease-modifying activity in animal models of rheumatoid arthritis (RA). We found that 2ME2 dramatically suppresses development of mouse experimental autoimmune encephalomyelitis (EAE), a rodent model of multiple sclerosis (MS). 2ME2 inhibits in vitro lymphocyte activation, cytokine production, and proliferation in a dose-dependent fashion. 2ME2 treatment of lymphocytes specifically reduced the nuclear translocation and transcriptional activity of nuclear factor of activated T-cells (NFAT) c1, whereas NF-κB and activator protein 1 (AP-1) activation were not adversely affected. We therefore propose that 2ME2 attenuates EAE through disruption of the NFAT pathway and subsequent lymphocyte activation. By extension, our findings provide a molecular rationale for the use of 2ME2 as a tolerable oral immunomodulatory agent for the treatment of autoimmune disorders such as MS in humans.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Estradiol/analogs & derivatives , 2-Methoxyestradiol , Animals , Autoimmunity , CD4-Positive T-Lymphocytes/cytology , Cytokines/biosynthesis , Estradiol/pharmacology , Humans , Lymphocyte Activation , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Tubulin Modulators/pharmacology
18.
Genes Dev ; 26(18): 2038-49, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22925884

ABSTRACT

Isocitrate dehydrogenase-1 (IDH1) R132 mutations occur in glioma, but their physiological significance is unknown. Here we describe the generation and characterization of brain-specific Idh1 R132H conditional knock-in (KI) mice. Idh1 mutation results in hemorrhage and perinatal lethality. Surprisingly, intracellular reactive oxygen species (ROS) are attenuated in Idh1-KI brain cells despite an apparent increase in the NADP(+)/NADPH ratio. Idh1-KI cells also show high levels of D-2-hydroxyglutarate (D2HG) that are associated with inhibited prolyl-hydroxylation of hypoxia-inducible transcription factor-1α (Hif1α) and up-regulated Hif1α target gene transcription. Intriguingly, D2HG also blocks prolyl-hydroxylation of collagen, causing a defect in collagen protein maturation. An endoplasmic reticulum (ER) stress response induced by the accumulation of immature collagens may account for the embryonic lethality of these mutants. Importantly, D2HG-mediated impairment of collagen maturation also led to basement membrane (BM) aberrations that could play a part in glioma progression. Our study presents strong in vivo evidence that the D2HG produced by the mutant Idh1 enzyme is responsible for the above effects.


Subject(s)
Basement Membrane/pathology , Collagen/metabolism , Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Animals , Basement Membrane/metabolism , Brain/cytology , Brain/pathology , Gene Knock-In Techniques , Genotype , Glioma/pathology , Mice , Mutation , Protein Stability , Reactive Oxygen Species/metabolism , Stress, Physiological
19.
Proc Natl Acad Sci U S A ; 108(25): 10243-8, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21636789

ABSTRACT

The highly conserved ANP32 proteins are proposed to function in a broad array of physiological activities through molecular mechanisms as diverse as phosphatase inhibition, chromatin regulation, caspase activation, and intracellular transport. On the basis of previous analyses of mice bearing targeted mutations of Anp32a or Anp32e, there has been speculation that all ANP32 proteins play redundant roles and are dispensable for normal development. However, more recent work has suggested that ANP32B may in fact have functions that are not shared by other ANP32 family members. Here we report that ANP32B expression is associated with a poor prognosis in human breast cancer, consistent with the increased levels of Anp32b mRNA present in proliferating wild-type (WT) murine embryonic fibroblasts and stimulated WT B and T lymphocytes. Moreover, we show that, contrary to previous assumptions, Anp32b is very important for murine embryogenesis. In a mixed genetic background, ANP32B-deficient mice displayed a partially penetrant perinatal lethality that became fully penetrant in a pure C57BL/6 background. Surviving ANP32B-deficient mice showed reduced viability due to variable defects in various organ systems. Study of compound mutants lacking ANP32A, ANP32B, and/or ANP32E revealed previously hidden roles for ANP32A in mouse development that became apparent only in the complete absence of ANP32B. Our data demonstrate a hierarchy of importance for the mammalian Anp32 genes, with Anp32b being the most critical for normal development.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Embryo, Mammalian/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/anatomy & histology , Female , Fibroblasts/cytology , Fibroblasts/physiology , Gene Targeting , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Rate
20.
Proc Natl Acad Sci U S A ; 107(32): 13984-90, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20679220

ABSTRACT

MAP kinase-interacting kinase 1 and 2 (Mnk1 and Mnk2) are protein-serine/threonine kinases that are activated by ERK or p38 and phosphorylate eIF4E, which is involved in cap-dependent translation initiation. However, Mnk1/2 double knockout (Mnk-DKO) mice show normal cell growth and development despite an absence of eIF4E phosphorylation. Here we show that the tumorigenesis occurring in the Lck-Pten mouse model (referred to here as tPten(-/-) mice) can be suppressed by the loss of Mnk1/2. Phosphorylation of eIF4E was greatly enhanced in lymphomas of parental tPten(-/-) mice compared with lymphoid tissues of wild-type mice, but was totally absent in lymphomas of tPten(-/-); Mnk-DKO mice. Notably, stable knockdown of Mnk1 in the human glioma cell line U87MG resulted in dramatically decreased tumor formation when these cells were injected into athymic nude mice. Our data demonstrate an oncogenic role for Mnk1/2 in tumor development, and highlight these molecules as potential anticancer drug targets that could be inactivated with minimal side effects.


Subject(s)
Neoplasms/etiology , Protein Serine-Threonine Kinases/deficiency , Animals , Cell Line, Tumor , Eukaryotic Initiation Factor-4E/metabolism , Glioma/pathology , Lymphoma/etiology , Mice , Mice, Knockout , Mice, Nude , Neoplasm Transplantation , PTEN Phosphohydrolase/deficiency , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL