Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(9): 113047, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37651234

ABSTRACT

CD4 T cells are central effectors of anti-cancer immunity and immunotherapy, yet the regulation of CD4 tumor-specific T (TTS) cells is unclear. We demonstrate that CD4 TTS cells are quickly primed and begin to divide following tumor initiation. However, unlike CD8 TTS cells or exhaustion programming, CD4 TTS cell proliferation is rapidly frozen in place by a functional interplay of regulatory T cells and CTLA4. Together these mechanisms paralyze CD4 TTS cell differentiation, redirecting metabolic circuits, and reducing their accumulation in the tumor. The paralyzed state is actively maintained throughout cancer progression and CD4 TTS cells rapidly resume proliferation and functional differentiation when the suppressive constraints are alleviated. Overcoming their paralysis established long-term tumor control, demonstrating the importance of rapidly crippling CD4 TTS cells for tumor progression and their potential restoration as therapeutic targets.


Subject(s)
CD4-Positive T-Lymphocytes , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Neoplasms/metabolism , T-Lymphocytes, Regulatory , Lymph Nodes
2.
bioRxiv ; 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37131587

ABSTRACT

CD4 T cells are important effectors of anti-tumor immunity, yet the regulation of CD4 tumor-specific T (T TS ) cells during cancer development is still unclear. We demonstrate that CD4 T TS cells are initially primed in the tumor draining lymph node and begin to divide following tumor initiation. Distinct from CD8 T TS cells and previously defined exhaustion programs, CD4 T TS cell proliferation is rapidly frozen in place and differentiation stunted by a functional interplay of T regulatory cells and both intrinsic and extrinsic CTLA4 signaling. Together these mechanisms paralyze CD4 T TS cell differentiation, redirecting metabolic and cytokine production circuits, and reducing CD4 T TS cell accumulation in the tumor. Paralysis is actively maintained throughout cancer progression and CD4 T TS cells rapidly resume proliferation and functional differentiation when both suppressive reactions are alleviated. Strikingly, Treg depletion alone reciprocally induced CD4 T TS cells to themselves become tumor-specific Tregs, whereas CTLA4 blockade alone failed to promote T helper differentiation. Overcoming their paralysis established long-term tumor control, demonstrating a novel immune evasion mechanism that specifically cripples CD4 T TS cells to favor tumor progression.

3.
Immunity ; 55(2): 324-340.e8, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139353

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a sensor of products of tryptophan metabolism and a potent modulator of immunity. Here, we examined the impact of AhR in tumor-associated macrophage (TAM) function in pancreatic ductal adenocarcinoma (PDAC). TAMs exhibited high AhR activity and Ahr-deficient macrophages developed an inflammatory phenotype. Deletion of Ahr in myeloid cells or pharmacologic inhibition of AhR reduced PDAC growth, improved efficacy of immune checkpoint blockade, and increased intra-tumoral frequencies of IFNγ+CD8+ T cells. Macrophage tryptophan metabolism was not required for this effect. Rather, macrophage AhR activity was dependent on Lactobacillus metabolization of dietary tryptophan to indoles. Removal of dietary tryptophan reduced TAM AhR activity and promoted intra-tumoral accumulation of TNFα+IFNγ+CD8+ T cells; provision of dietary indoles blocked this effect. In patients with PDAC, high AHR expression associated with rapid disease progression and mortality, as well as with an immune-suppressive TAM phenotype, suggesting conservation of this regulatory axis in human disease.


Subject(s)
Immune Tolerance/immunology , Receptors, Aryl Hydrocarbon/immunology , Tryptophan/immunology , Tumor-Associated Macrophages/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Humans , Indoles/immunology , Indoles/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Microbiota/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/metabolism
4.
Front Immunol ; 12: 695238, 2021.
Article in English | MEDLINE | ID: mdl-34456909

ABSTRACT

Regulation of amino acid availability and metabolism in immune cells is essential for immune system homeostasis and responses to exogenous and endogenous challenges including microbial infection, tumorigenesis and autoimmunity. In myeloid cells the consumption of amino acids such as arginine and tryptophan and availability of their metabolites are key drivers of cellular identity impacting development, functional polarization to an inflammatory or regulatory phenotype, and interaction with other immune cells. In this review, we discuss recent developments and emerging concepts in our understanding of the impact amino acid availability and consumption has on cellular phenotype focusing on two key myeloid cell populations, macrophages and myeloid derived suppressor cells (MDSCs). We also highlight the potential of myeloid-specific of amino acid transporters and catabolic enzymes as immunotherapy targets in a variety of conditions such as cancer and autoimmune disease discussing the opportunities and limitations in targeting these pathways for clinical therapy.


Subject(s)
Amino Acid Transport Systems/metabolism , Amino Acids/metabolism , Macrophages/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Animals , Humans , Immunity, Innate , Macrophages/immunology , Myeloid-Derived Suppressor Cells/immunology , Phenotype
5.
J Clin Invest ; 131(3)2021 02 01.
Article in English | MEDLINE | ID: mdl-33529165

ABSTRACT

Germline mutations in BRCA1 and BRCA2 (BRCA1/2) genes considerably increase breast and ovarian cancer risk. Given that tumors with these mutations have elevated genomic instability, they exhibit relative vulnerability to certain chemotherapies and targeted treatments based on poly (ADP-ribose) polymerase (PARP) inhibition. However, the molecular mechanisms that influence cancer risk and therapeutic benefit or resistance remain only partially understood. BRCA1 and BRCA2 have also been implicated in the suppression of R-loops, triple-stranded nucleic acid structures composed of a DNA:RNA hybrid and a displaced ssDNA strand. Here, we report that loss of RNF168, an E3 ubiquitin ligase and DNA double-strand break (DSB) responder, remarkably protected Brca1-mutant mice against mammary tumorigenesis. We demonstrate that RNF168 deficiency resulted in accumulation of R-loops in BRCA1/2-mutant breast and ovarian cancer cells, leading to DSBs, senescence, and subsequent cell death. Using interactome assays, we identified RNF168 interaction with DHX9, a helicase involved in the resolution and removal of R-loops. Mechanistically, RNF168 directly ubiquitylated DHX9 to facilitate its recruitment to R-loop-prone genomic loci. Consequently, loss of RNF168 impaired DHX9 recruitment to R-loops, thereby abrogating its ability to resolve R-loops. The data presented in this study highlight a dependence of BRCA1/2-defective tumors on factors that suppress R-loops and reveal a fundamental RNF168-mediated molecular mechanism that governs cancer development and vulnerability.


Subject(s)
BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , DNA, Neoplasm/metabolism , Genomic Instability , Mammary Neoplasms, Animal/metabolism , Ovarian Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA, Neoplasm/genetics , Female , Genetic Loci , Humans , Mammary Neoplasms, Animal/genetics , Mice , Mice, Knockout , Ovarian Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics
6.
7.
Sci Immunol ; 4(42)2019 12 13.
Article in English | MEDLINE | ID: mdl-31836669

ABSTRACT

General control nonderepressible 2 (GCN2) is an environmental sensor controlling transcription and translation in response to nutrient availability. Although GCN2 is a putative therapeutic target for immuno-oncology, its role in shaping the immune response to tumors is poorly understood. Here, we used mass cytometry, transcriptomics, and transcription factor-binding analysis to determine the functional impact of GCN2 on the myeloid phenotype and immune responses in melanoma. We found that myeloid-lineage deletion of GCN2 drives a shift in the phenotype of tumor-associated macrophages and myeloid-derived suppressor cells (MDSCs) that promotes antitumor immunity. Time-of-flight mass cytometry (CyTOF) and single-cell RNA sequencing showed that this was due to changes in the immune microenvironment with increased proinflammatory activation of macrophages and MDSCs and interferon-γ expression in intratumoral CD8+ T cells. Mechanistically, GCN2 altered myeloid function by promoting increased translation of the transcription factor CREB-2/ATF4, which was required for maturation and polarization of macrophages and MDSCs in both mice and humans, whereas targeting Atf4 by small interfering RNA knockdown reduced tumor growth. Last, analysis of patients with cutaneous melanoma showed that GCN2-dependent transcriptional signatures correlated with macrophage polarization, T cell infiltrates, and overall survival. Thus, these data reveal a previously unknown dependence of tumors on myeloid GCN2 signals for protection from immune attack.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Macrophages/immunology , Melanoma/immunology , Myeloid-Derived Suppressor Cells/immunology , Protein Serine-Threonine Kinases/immunology , Tumor Microenvironment/immunology , Animals , Cells, Cultured , Humans , Mice
8.
Nat Immunol ; 19(6): 571-582, 2018 06.
Article in English | MEDLINE | ID: mdl-29760532

ABSTRACT

The transcription factor AhR modulates immunity at multiple levels. Here we report that phagocytes exposed to apoptotic cells exhibited rapid activation of AhR, which drove production of the cytokine IL-10. Activation of AhR was dependent on interactions between apoptotic-cell DNA and the pattern-recognition receptor TLR9 that was required for the prevention of immune responses to DNA and histones in vivo. Moreover, disease progression in mouse systemic lupus erythematosus (SLE) correlated with strength of the AhR signal, and the disease course could be altered by modulation of AhR activity. Deletion of AhR in the myeloid lineage caused systemic autoimmunity in mice, and an enhanced AhR transcriptional signature correlated with disease in patients with SLE. Thus, AhR activity induced by apoptotic cell phagocytes maintains peripheral tolerance.


Subject(s)
Apoptosis/immunology , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/immunology , Macrophages/immunology , Receptors, Aryl Hydrocarbon/immunology , Animals , Humans , Mice , Signal Transduction/immunology , Toll-Like Receptor 9/immunology
9.
J Immunol ; 197(7): 2618-26, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27559051

ABSTRACT

Autoimmune diseases are characterized by pathogenic immune responses to self-antigens. In systemic lupus erythematosus (SLE), many self-antigens are found in apoptotic cells (ACs), and defects in removal of ACs from the body are linked to a risk for developing SLE. This includes pathological memory that gives rise to disease flares. In this study, we investigated how memory to AC-derived self-antigens develops and the contribution of self-memory to the development of lupus-related pathology. Multiple injections of ACs without adjuvant into wild-type mice induce a transient primary autoimmune response without apparent anti-nuclear Ab reactivity or kidney pathology. Interestingly, as the transient Ab response reached baseline, a single boost injection fully recalled the immune response to ACs, and this memory response was furthermore transferable into naive mice. Additionally, the memory response contains elements of pathogenicity, accompanied by selective memory to selective Ags. Thus, we provide evidence for a selective self-memory that underlies progression of the response to self-antigens with implications for SLE development therapy.


Subject(s)
Apoptosis/immunology , Autoantibodies/immunology , Autoantigens/immunology , Immunologic Memory/immunology , Lupus Erythematosus, Systemic/immunology , Animals , Lupus Erythematosus, Systemic/therapy , Mice , Mice, Inbred C57BL
10.
Mol Cell Biol ; 35(23): 4006-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26391949

ABSTRACT

Synthesis of the p53 tumor suppressor and its subsequent activation following DNA damage are critical for its protection against tumorigenesis. We previously discovered an internal ribosome entry site (IRES) at the 5' untranslated region of the p53 mRNA. However, the connection between IRES-mediated p53 translation and p53's tumor suppressive function is unknown. In this study, we identified two p53 IRES trans-acting factors, translational control protein 80 (TCP80), and RNA helicase A (RHA), which positively regulate p53 IRES activity. Overexpression of TCP80 and RHA also leads to increased expression and synthesis of p53. Furthermore, we discovered two breast cancer cell lines that retain wild-type p53 but exhibit defective p53 induction and synthesis following DNA damage. The levels of TCP80 and RHA are extremely low in both cell lines, and expression of both proteins is required to significantly increase the p53 IRES activity in these cells. Moreover, we found cancer cells transfected with a shRNA against TCP80 not only exhibit decreased expression of TCP80 and RHA but also display defective p53 induction and diminished ability to induce senescence following DNA damage. Therefore, our findings reveal a novel mechanism of p53 inactivation that links deregulation of IRES-mediated p53 translation with tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , DNA Damage , Gene Expression Regulation, Neoplastic , Internal Ribosome Entry Sites , Protein Biosynthesis , Tumor Suppressor Protein p53/genetics , 5' Untranslated Regions , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Factor 90 Proteins/genetics , Nuclear Factor 90 Proteins/metabolism , Protein Binding , Proteolysis , Tumor Suppressor Protein p53/metabolism
11.
Biomed Res Int ; 2015: 708158, 2015.
Article in English | MEDLINE | ID: mdl-26273641

ABSTRACT

Synthesis of the p53 tumor suppressor increases following DNA damage. This increase and subsequent activation of p53 are essential for the protection of normal cells against tumorigenesis. We previously discovered an internal ribosome entry site (IRES) that is located at the 5'-untranslated region (UTR) of p53 mRNA and found that the IRES activity increases following DNA damage. However, the mechanism underlying IRES-mediated p53 translation in response to DNA damage is still poorly understood. In this study, we discovered that translational control protein 80 (TCP80) has increased binding to the p53 mRNA in vivo following DNA damage. Overexpression of TCP80 also leads to increased p53 IRES activity in response to DNA damage. TCP80 has increased association with RNA helicase A (RHA) following DNA damage and overexpression of TCP80, along with RHA, leads to enhanced expression of p53. Moreover, we found that MCF-7 breast cancer cells with decreased expression of TCP80 and RHA exhibit defective p53 induction following DNA damage and diminished expression of its downstream target PUMA, a proapoptotic protein. Taken together, our discovery of the function of TCP80 and RHA in regulating p53 IRES and p53 induction following DNA damage provides a better understanding of the mechanisms that regulate IRES-mediated p53 translation in response to genotoxic stress.


Subject(s)
DNA Damage/physiology , Internal Ribosome Entry Sites/physiology , Nuclear Factor 90 Proteins/metabolism , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/metabolism , Humans , MCF-7 Cells
12.
Cell Cycle ; 12(17): 2733-7, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23966173

ABSTRACT

Ubiquitylation is currently recognized as a major posttranslational modification that regulates diverse cellular processes. Pirh2 is a ubiquitin E3 ligase that regulates the turnover and functionality of several proteins involved in cell proliferation and differentiation, cell cycle checkpoints, and cell death. Here we review the role of Pirh2 as a regulator of the DNA damage response through the ubiquitylation of p53, Chk2, p73, and PolH. By ubiquitylating these proteins, Pirh2 regulates cell cycle checkpoints and cell death in response to DNA double-strand breaks or the formation of bulky DNA lesions. We also discuss how Pirh2 affects cell proliferation and differentiation in unstressed conditions through ubiquitylation and degradation of c-Myc, p63, and p27(kip1). Finally, we link these different functions of Pirh2 to its role as a tumor suppressor in mice and as a prognosis marker in various human cancer subtypes.


Subject(s)
Cell Cycle , Cell Differentiation , DNA Damage , Ubiquitin-Protein Ligases/metabolism , Animals , Humans , Models, Biological , Ubiquitination
13.
Biochem Biophys Res Commun ; 435(4): 708-13, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23702482

ABSTRACT

Chloroquine is a pharmaceutical agent that has been widely used to treat patients with malaria. Chloroquine has also been reported to have hypoglycemic effects on humans and animal models of diabetes. Despite many previous studies, the mechanism responsible for its hypoglycemic effect is still unclear. Chloroquine was recently reported to be an activator of ATM, the protein deficient in the Ataxia-telagiectasia (A-T) disease. Since ATM is also known as an insulin responsive protein that mediates Akt activation, we tested the effect of chloroquine on the activity of Akt and its downstream targets. In L6 muscle cells treated with insulin and chloroquine, the phosphorylation of Akt and glucose uptake were dramatically increased compared to cells treated with insulin alone, suggesting that chloroquine is a potent activator of Akt and glucose uptake in these cells. We also found that the reduction of insulin-mediated Akt activity in muscle tissues of insulin resistant rats was partially reversed by chloroquine treatment. Moreover, insulin-mediated phosphorylation of glycogen synthase kinase-3ß in L6 cells was greatly enhanced by chloroquine. A substantial decrease in phosphorylation of glycogen synthase was also observed in chloroquine-treated L6 cells, indicating enhanced activity of glycogen synthase. Taken together, our results not only show that chloroquine is a novel activator of Akt that stimulates glucose uptake and glycogen synthase, but also validate chloroquine as a potential therapeutic agent for patients with type 2 diabetes mellitus.


Subject(s)
Chloroquine/pharmacology , Glucose/pharmacokinetics , Glycogen Synthase/metabolism , Muscle Fibers, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Male , Muscle Fibers, Skeletal/drug effects , Rats , Rats, Wistar
14.
PLoS Genet ; 9(1): e1003259, 2013.
Article in English | MEDLINE | ID: mdl-23382699

ABSTRACT

Rnf8 is an E3 ubiquitin ligase that plays a key role in the DNA damage response as well as in the maintenance of telomeres and chromatin remodeling. Rnf8(-/-) mice exhibit developmental defects and increased susceptibility to tumorigenesis. We observed that levels of p53, a central regulator of the cellular response to DNA damage, increased in Rnf8(-/-) mice in a tissue- and cell type-specific manner. To investigate the role of the p53-pathway inactivation on the phenotype observed in Rnf8(-/-) mice, we have generated Rnf8(-/-)p53(-/-) mice. Double-knockout mice showed similar growth retardation defects and impaired class switch recombination compared to Rnf8(-/-) mice. In contrast, loss of p53 fully rescued the increased apoptosis and reduced number of thymocytes and splenocytes in Rnf8(-/-) mice. Similarly, the senescence phenotype of Rnf8(-/-) mouse embryonic fibroblasts was rescued in p53 null background. Rnf8(-/-)p53(-/-) cells displayed defective cell cycle checkpoints and DNA double-strand break repair. In addition, Rnf8(-/-)p53(-/-) mice had increased levels of genomic instability and a remarkably elevated tumor incidence compared to either Rnf8(-/-) or p53(-/-) mice. Altogether, the data in this study highlight the importance of p53-pathway activation upon loss of Rnf8, suggesting that Rnf8 and p53 functionally interact to protect against genomic instability and tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Neoplasms , Tumor Suppressor Protein p53 , Ubiquitin-Protein Ligases , Animals , Cell Transformation, Neoplastic/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair/genetics , Fibroblasts/cytology , Genomic Instability , Humans , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
15.
PLoS Genet ; 7(5): e1001385, 2011 May.
Article in English | MEDLINE | ID: mdl-21625617

ABSTRACT

Chk2 is an effector kinase important for the activation of cell cycle checkpoints, p53, and apoptosis in response to DNA damage. Mus81 is required for the restart of stalled replication forks and for genomic integrity. Mus81(Δex3-4/Δex3-4) mice have increased cancer susceptibility that is exacerbated by p53 inactivation. In this study, we demonstrate that Chk2 inactivation impairs the development of Mus81(Δex3-4/Δex3-4) lymphoid cells in a cell-autonomous manner. Importantly, in contrast to its predicted tumor suppressor function, loss of Chk2 promotes mitotic catastrophe and cell death, and it results in suppressed oncogenic transformation and tumor development in Mus81(Δex3-4/Δex3-4) background. Thus, our data indicate that an important role for Chk2 is maintaining lymphocyte development and that dual inactivation of Chk2 and Mus81 remarkably inhibits cancer.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Genomic Instability , Lymphocytes/cytology , Neoplasms/metabolism , Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Lineage , Cells, Cultured , Checkpoint Kinase 2 , DNA-Binding Proteins/genetics , Endonucleases/genetics , Enzyme Activation , Gene Expression Regulation, Developmental , Lymphocytes/immunology , Mice , Mice, Knockout , Mitosis , Neoplasms/genetics , Protein Serine-Threonine Kinases/deficiency , Thymus Gland/cytology , Thymus Gland/immunology , Tumor Suppressor Protein p53/metabolism
16.
Drug Discov Today ; 16(7-8): 332-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21315178

ABSTRACT

Ataxia-telangiectasia (A-T) is an autosomal recessive disorder characterized by cerebellar ataxia and oculocutaneous telangiectasias. The gene mutated in this disease, Atm (A-T mutated), encodes a serine/threonine protein kinase that has been traditionally considered to be a nuclear protein controlling cell-cycle progression. However, many of the growth abnormalities observed in patients with A-T, including neuronal degeneration and insulin resistance, remain difficult to explain with nuclear localization of ATM. Here, recent advances in elucidating the cytoplasmic localization and function of ATM are reviewed. Particular attention is given to the role of ATM in insulin signaling and Akt activation. The potential for cytoplasmic ATM protein kinase to be an emerging therapeutic target for treating diabetes, cancer and neuronal degeneration is discussed.


Subject(s)
Ataxia Telangiectasia/physiopathology , Cell Cycle Proteins/metabolism , Cytoplasm/enzymology , DNA-Binding Proteins/metabolism , Diabetes Mellitus/drug therapy , Neoplasms/drug therapy , Nerve Degeneration/drug therapy , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics
17.
J Exp Med ; 207(5): 983-97, 2010 May 10.
Article in English | MEDLINE | ID: mdl-20385750

ABSTRACT

Signaling and repair of DNA double-strand breaks (DSBs) are critical for preventing immunodeficiency and cancer. These DNA breaks result from exogenous and endogenous DNA insults but are also programmed to occur during physiological processes such as meiosis and immunoglobulin heavy chain (IgH) class switch recombination (CSR). Recent studies reported that the E3 ligase RNF8 plays important roles in propagating DNA DSB signals and thereby facilitating the recruitment of various DNA damage response proteins, such as 53BP1 and BRCA1, to sites of damage. Using mouse models for Rnf8 mutation, we report that Rnf8 deficiency leads to impaired spermatogenesis and increased sensitivity to ionizing radiation both in vitro and in vivo. We also demonstrate the existence of alternative Rnf8-independent mechanisms that respond to irradiation and accounts for the partial recruitment of 53bp1 to sites of DNA damage in activated Rnf8(-/-) B cells. Remarkably, IgH CSR is impaired in a gene dose-dependent manner in Rnf8 mutant mice, revealing that these mice are immunodeficient. In addition, Rnf8(-/-) mice exhibit increased genomic instability and elevated risks for tumorigenesis indicating that Rnf8 is a novel tumor suppressor. These data unravel the in vivo pleiotropic effects of Rnf8.


Subject(s)
Genetic Predisposition to Disease , Immunoglobulin Class Switching/physiology , Neoplasms/genetics , Spermatogenesis/physiology , Ubiquitin-Protein Ligases/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Division/genetics , Cell Division/immunology , DNA Damage/genetics , Genomic Instability/genetics , Growth/genetics , Growth/immunology , Immunoglobulin Class Switching/genetics , Mice , Mice, Mutant Strains , Neoplasms/immunology , Spermatogenesis/genetics , Spermatogenesis/immunology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
18.
Cell Signal ; 20(8): 1555-63, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18534819

ABSTRACT

Ataxia-telangiectasia (A-T) is an autosomal recessive disorder characterized by cerebellar ataxia and oculocutaneous telangiectasias. Patients with A-T also have high incidences of type 2 diabetes mellitus. The gene mutated in this disease, ATM (A-T, mutated), encodes a protein kinase. Previous studies have demonstrated that cytoplasmic ATM is an insulin-responsive protein and a major upstream activator of Akt following insulin treatment. To further investigate the function of ATM in insulin signal transduction, insulin resistance was induced in rats by feeding them a high-fat diet. Muscle tissue of rats with insulin resistance had both dramatically reduced ATM levels and substantially decreased Akt phosphorylation at Ser473 in comparison to that of regular chow-fed controls. The decreased ATM expression suggests that ATM is involved in the development of insulin resistance through down-regulation of Akt activity. The role of ATM in activation of Akt was further confirmed in mouse embryonic fibroblast (MEF) A29 (ATM+/+) and A38 (ATM-/-) cells. In addition, insulin-mediated Akt phosphorylation in mouse L6 muscle cells was greatly reduced by KU-55933, a specific inhibitor of ATM. A 2-deoxyglucose incorporation assay showed that this inhibitor also caused a significant reduction in insulin-mediated glucose uptake in L6 cells. An immunofluorescence experiment demonstrated that in L6 cells transfected with wild-type (WT) ATM, insulin caused a dramatic increase of the cell surface glucose transporter 4 (GLUT4), while in cells transfected with kinase-dead (KD) ATM, translocation of GLUT4 to the cell surface in response to insulin was markedly inhibited.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Glucose Transporter Type 4/metabolism , Insulin Resistance , Insulin/pharmacology , Muscle, Skeletal/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line , DNA-Binding Proteins/genetics , Enzyme Activation , Male , Mice , Protein Serine-Threonine Kinases/genetics , Protein Transport , Rats , Rats, Wistar , Tumor Suppressor Proteins/genetics
19.
Anat Rec (Hoboken) ; 290(3): 243-50, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17525940

ABSTRACT

Ataxia-telangiectasia (A-T) is a human autosomal recessive disorder characterized by neuronal degeneration as well as many other physiological and somatic defects. ATM (A-T, mutated), the gene mutated in A-T, encodes a 370 kDa protein kinase. ATM knockout mouse models (ATM(-/-)) show growth retardation, infertility, neurological dysfunction, defects in T-lymphocytes, and extreme sensitivity to ionizing radiation. We have recently established multiple ATM(+/-) breeding pairs and discovered that all ATM(-/-) offspring exhibit a nonpigmented section of tail, usually at or near the tip. To our knowledge, this is the first time that a phenotype of nonpigmented tail has been reported in ATM(-/-) knockout mice. We believe that the sections of nonpigmented tail of 129S6/SvEvTac-ATM(tm1Awb)/J mice provide a novel phenotypic marker for research using this ATM knockout mouse model. Results from histochemistry and immunoblotting analysis further demonstrate that while melanocyte precursors or melanoblasts are present in the nonpigmented tail tissue of ATM(-/-) mice, they fail to differentiate fully into mature melanocytes. The potential connection between this phenotype and other clinical symptoms caused by ATM deficiency, such as progressive neuronal degeneration, is discussed in this article.


Subject(s)
Ataxia Telangiectasia/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Hypopigmentation/metabolism , Melanins/metabolism , Melanocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Tail/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Biomarkers/metabolism , Cell Cycle Proteins/genetics , Cell Differentiation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Genotype , Hypopigmentation/genetics , Hypopigmentation/pathology , Intramolecular Oxidoreductases/metabolism , Melanins/deficiency , Melanocytes/pathology , Mice , Mice, Knockout , Mice, Transgenic , Monophenol Monooxygenase/metabolism , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Tail/pathology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
20.
Gene ; 395(1-2): 1-7, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17395405

ABSTRACT

While posttranslational regulation of p53 levels by its interaction with the ubiquitin ligase MDM2 is widely accepted, it has recently become clear that regulation of p53 translation also contributes to p53 induction following DNA damage. However, the mechanisms underlying the translational control of p53 are still poorly understood. In this review, we will focus on the translational regulation of p53 through the 5'- and 3'-untranslated regions of its mRNA. We will also discuss in detail the recent discovery of the p53 internal ribosome entry site (IRES), its role in p53 translation in response to DNA damage, and how it might lead to a better understanding of the process of oncogenesis and provide new avenues for cancer therapeutics.


Subject(s)
Genes, p53 , Neoplasms/genetics , Protein Biosynthesis , 3' Untranslated Regions , 5' Untranslated Regions , Animals , DNA Damage , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/metabolism , Neoplasms/therapy , Ribosomes/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...