100 results on '"Mock BA"'
Search Results
2. Regional Localization of the Human Glutaminase (gls) and Interleukin-9 (il9) Genes By Insitu Hybridization
- Author
-
UCL - Autre, Modi, WS., Pollock, DD., Mock, BA., Banner, C., Renauld, Jean-Christophe, Van Snick, Jacques, 2ND INTERNATIONAL WORKSHOP ON HUMAN CHROMOSOME 17, UCL - Autre, Modi, WS., Pollock, DD., Mock, BA., Banner, C., Renauld, Jean-Christophe, Van Snick, Jacques, and 2ND INTERNATIONAL WORKSHOP ON HUMAN CHROMOSOME 17
- Abstract
Phosphate-activated glutaminase is found in mammalian small intestine, brain, and kidney, but not in liver. The enzyme initiates the catabolism of glutamine as the principal respiratory fuel in the small intestine, may synthesize the neurotransmitter glutamate in the brain, and functions in the kidney to help maintain systemic pH homeostasis. Interleukin-9 (IL9) is a relatively new cytokine that supports the growth of helper T-cell clones, mast cells, and megakaryoblastic leukemia cells. cDNA clones have recently been obtained for each of these genes. The human loci for phosphate-activated glutaminase (GLS) and IL9 have previously been mapped to chromosomes 2 and 5, respectively, by analysis of somatic cell hybrid DNAs. By using chromosomal in situ hybridization, we have regionally mapped GLS to 2q32 --> q34 and IL9 to 5q31 --> q35.
- Published
- 1991
3. Multigenic control of skin tumor susceptibility in SENCARA/Pt mice.
- Author
-
Mock, BA, Lowry, DT, Rehman, I, Padlan, C, Yuspa, SH, and Hennings, H
- Abstract
Skin tumors induced in mice by initiation-promotion (2 μg DMBA-2 μg TPA) protocols were found to be under multigenic control. Eighty-one N2 mice from the cross (BALB/cAnPtxSENCARA/Pt)F1 x SENCARA/Pt that were either solidly resistant (no papillomas) or highly susceptible (7 papillomas/mouse) were subjected to a 'genome scan' using 89 microsatellite markers to check for associations with susceptible and resistant phenotypes. A locus on Chr 5 (Skts4) was found to control the susceptibility of SENCARA/Pt mice and the resistance of BALB/cAnPt mice to papilloma formation. In addition, higher than expected linkage scores were seen for the markers D9Mit271, D11Mit268 and D12Mit56. Further work is required to establish whether genes determining papilloma formation are located in these regions of the genome. In general, no evidence was seen for loss of heterozygosity in microsatellite markers on Chrs5, 9 and 11 in 17 microdissected papillomas from (BALB/c x SENCARA)F1 hybrid mice. [ABSTRACT FROM PUBLISHER]
- Published
- 1998
- Full Text
- View/download PDF
4. Transfer RNA acetylation regulates in vivo mammalian stress signaling.
- Author
-
Gamage ST, Khoogar R, Manage SH, Crawford MC, Georgeson J, Polevoda BV, Sanders C, Lee KA, Nance KD, Iyer V, Kustanovich A, Perez M, Thu CT, Nance SR, Amin R, Miller CN, Holewinski RJ, Meyer T, Koparde V, Yang A, Jailwala P, Nguyen JT, Andresson T, Hunter K, Gu S, Mock BA, Edmondson EF, Difilippantonio S, Chari R, Schwartz S, O'Connell MR, Chih-Chien Wu C, and Meier JL
- Abstract
Transfer RNA (tRNA) modifications are crucial for protein synthesis, but their position-specific physiological roles remain poorly understood. Here we investigate the impact of N4-acetylcytidine (ac
4 C), a highly conserved tRNA modification, using a Thumpd1 knockout mouse model. We find that loss of Thumpd1-dependent tRNA acetylation leads to reduced levels of tRNALeu , increased ribosome stalling, and activation of eIF2α phosphorylation. Thumpd1 knockout mice exhibit growth defects and sterility. Remarkably, concurrent knockout of Thumpd1 and the stress-sensing kinase Gcn2 causes penetrant postnatal lethality, indicating a critical genetic interaction. Our findings demonstrate that a modification restricted to a single position within type II cytosolic tRNAs can regulate ribosome-mediated stress signaling in mammalian organisms, with implications for our understanding of translation control as well as therapeutic interventions., Competing Interests: DECLARATION OF INTERESTS The authors have no positions or financial interests to declare.- Published
- 2024
- Full Text
- View/download PDF
5. A structure-based designed small molecule depletes hRpn13 Pru and a select group of KEN box proteins.
- Author
-
Lu X, Chandravanshi M, Sabbasani VR, Gaikwad S, Hughitt VK, Gyabaah-Kessie N, Scroggins BT, Das S, Myint W, Clapp ME, Schwieters CD, Dyba MA, Bolhuis DL, Koscielniak JW, Andresson T, Emanuele MJ, Brown NG, Matsuo H, Chari R, Citrin DE, Mock BA, Swenson RE, and Walters KJ
- Subjects
- Intracellular Signaling Peptides and Proteins, Ubiquitin metabolism, Cytoplasm metabolism, Transcription Factors, Proteasome Endopeptidase Complex metabolism, Membrane Glycoproteins metabolism
- Abstract
Proteasome subunit hRpn13 is partially proteolyzed in certain cancer cell types to generate hRpn13
Pru by degradation of its UCHL5/Uch37-binding DEUBAD domain and retention of an intact proteasome- and ubiquitin-binding Pru domain. By using structure-guided virtual screening, we identify an hRpn13 binder (XL44) and solve its structure ligated to hRpn13 Pru by integrated X-ray crystallography and NMR to reveal its targeting mechanism. Surprisingly, hRpn13Pru is depleted in myeloma cells following treatment with XL44. TMT-MS experiments reveal a select group of off-targets, including PCNA clamp-associated factor PCLAF and ribonucleoside-diphosphate reductase subunit M2 (RRM2), that are similarly depleted by XL44 treatment. XL44 induces hRpn13-dependent apoptosis and also restricts cell viability by a PCLAF-dependent mechanism. A KEN box, but not ubiquitination, is required for XL44-induced depletion of PCLAF. Here, we show that XL44 induces ubiquitin-dependent loss of hRpn13Pru and ubiquitin-independent loss of select KEN box containing proteins., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2024
- Full Text
- View/download PDF
6. Use of CRISPR-based screens to identify mechanisms of chemotherapy resistance.
- Author
-
Alyateem G, Wade HM, Bickert AA, Lipsey CC, Mondal P, Smith MD, Labib RM, Mock BA, Robey RW, and Gottesman MM
- Subjects
- Humans, RNA, Guide, CRISPR-Cas Systems, CRISPR-Cas Systems genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Despite the development of new classes of targeted anti-cancer drugs, the curative treatment of metastatic solid tumors remains out of reach owing to the development of resistance to current chemotherapeutics. Although many mechanisms of drug resistance have been described, there is still a general lack of understanding of the many means by which cancer cells elude otherwise effective chemotherapy. The traditional strategy of isolating resistant clones in vitro, defining their mechanism of resistance, and testing to see whether these mechanisms play a role in clinical drug resistance is time-consuming and in many cases falls short of providing clinically relevant information. In this review, we summarize the use of CRISPR technology, including the promise and pitfalls, to generate libraries of cancer cells carrying sgRNAs that define novel mechanisms of resistance. The existing strategies using CRISPR knockout, activation, and inhibition screens, and combinations of these approaches are described. In addition, specialized approaches to identify more than one gene that may be contributing to resistance, as occurs in synthetic lethality, are described. Although these CRISPR-based approaches to cataloguing drug resistance genes in cancer cells are just beginning to be utilized, appropriately used they promise to accelerate understanding of drug resistance in cancer., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2023
- Full Text
- View/download PDF
7. Drug combinations identified by high-throughput screening promote cell cycle transition and upregulate Smad pathways in myeloma.
- Author
-
Peat TJ, Gaikwad SM, Dubois W, Gyabaah-Kessie N, Zhang S, Gorjifard S, Phyo Z, Andres M, Hughitt VK, Simpson RM, Miller MA, Girvin AT, Taylor A, Williams D, D'Antonio N, Zhang Y, Rajagopalan A, Flietner E, Wilson K, Zhang X, Shinn P, Klumpp-Thomas C, McKnight C, Itkin Z, Chen L, Kazandijian D, Zhang J, Michalowski AM, Simmons JK, Keats J, Thomas CJ, and Mock BA
- Subjects
- Humans, High-Throughput Screening Assays, Drug Synergism, Cell Cycle, Drug Combinations, Cell Line, Tumor, Drug Resistance, Neoplasm, Multiple Myeloma drug therapy, Multiple Myeloma genetics
- Abstract
Drug resistance and disease progression are common in multiple myeloma (MM) patients, underscoring the need for new therapeutic combinations. A high-throughput drug screen in 47 MM cell lines and in silico Huber robust regression analysis of drug responses revealed 43 potentially synergistic combinations. We hypothesized that effective combinations would reduce MYC expression and enhance p16 activity. Six combinations cooperatively reduced MYC protein, frequently over-expressed in MM and also cooperatively increased p16 expression, frequently downregulated in MM. Synergistic reductions in viability were observed with top combinations in proteasome inhibitor-resistant and sensitive MM cell lines, while sparing fibroblasts. Three combinations significantly prolonged survival in a transplantable Ras-driven allograft model of advanced MM closely recapitulating high-risk/refractory myeloma in humans and reduced viability of ex vivo treated patient cells. Common genetic pathways similarly downregulated by these combinations promoted cell cycle transition, whereas pathways most upregulated were involved in TGFβ/SMAD signaling. These preclinical data identify potentially useful drug combinations for evaluation in drug-resistant MM and reveal potential mechanisms of combined drug sensitivity., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. mTOR inhibition overcomes RSK3-mediated resistance to BET inhibitors in small cell lung cancer.
- Author
-
Kumari A, Gesumaria L, Liu YJ, Hughitt VK, Zhang X, Ceribelli M, Wilson KM, Klumpp-Thomas C, Chen L, McKnight C, Itkin Z, Thomas CJ, Mock BA, Schrump DS, and Chen H
- Subjects
- Humans, Apoptosis drug effects, Apoptosis genetics, Apoptosis physiology, TOR Serine-Threonine Kinases, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms metabolism, MTOR Inhibitors pharmacology, MTOR Inhibitors therapeutic use, Small Cell Lung Carcinoma drug therapy, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma metabolism
- Abstract
Small cell lung cancer (SCLC) is a recalcitrant malignancy with limited treatment options. Bromodomain and extraterminal domain inhibitors (BETis) have shown promising preclinical activity in SCLC, but the broad sensitivity spectrum limits their clinical prospects. Here, we performed unbiased high-throughput drug combination screens to identify therapeutics that could augment the antitumor activities of BETis in SCLC. We found that multiple drugs targeting the PI-3K-AKT-mTOR pathway synergize with BETis, among which mTOR inhibitors (mTORis) show the highest synergy. Using various molecular subtypes of the xenograft models derived from patients with SCLC, we confirmed that mTOR inhibition potentiates the antitumor activities of BETis in vivo without substantially increasing toxicity. Furthermore, BETis induce apoptosis in both in vitro and in vivo SCLC models, and this antitumor effect is further amplified by combining mTOR inhibition. Mechanistically, BETis induce apoptosis in SCLC by activating the intrinsic apoptotic pathway. However, BET inhibition leads to RSK3 upregulation, which promotes survival by activating the TSC2-mTOR-p70S6K1-BAD cascade. mTORis block this protective signaling and augment the apoptosis induced by BET inhibition. Our findings reveal a critical role of RSK3 induction in tumor survival upon BET inhibition and warrant further evaluation of the combination of mTORis and BETis in patients with SCLC.
- Published
- 2023
- Full Text
- View/download PDF
9. Structural Optimization and Anticancer Activity of Polo-like Kinase 1 (Plk1) Polo-Box Domain (PBD) Inhibitors and Their Prodrugs.
- Author
-
Park JE, Lee H, Oliva P, Kirsch K, Kim B, Ahn JI, Alverez CN, Gaikwad S, Krausz KW, O'Connor R, Rai G, Simeonov A, Mock BA, Gonzalez FJ, Lee KS, and Jacobson KA
- Abstract
Polo-like kinase 1 (Plk1), a mitotic kinase whose activity is widely upregulated in various human cancers, is considered an attractive target for anticancer drug discovery. Aside from the kinase domain, the C-terminal noncatalytic polo-box domain (PBD), which mediates the interaction with the enzyme's binding targets or substrates, has emerged as an alternative target for developing a new class of inhibitors. Various reported small molecule PBD inhibitors exhibit poor cellular efficacy and/or selectivity. Here, we report structure-activity relationship (SAR) studies on triazoloquinazolinone-derived inhibitors, such as 43 (a 1-thioxo-2,4-dihydrothieno[2,3- e ][1,2,4]triazolo[4,3- a ]pyrimidin-5(1 H )-one) that effectively block Plk1, but not Plk2 and Plk3 PBDs, with improved affinity and drug-like properties. The range of prodrug moieties needed for thiol group masking of the active drugs has been expanded to increase cell permeability and mechanism-based cancer cell (L363 and HeLa) death. For example, a 5-thio-1-methyl-4-nitroimidazolyl prodrug 80 , derived from 43 , showed an improved cellular potency (GI
50 4.1 μM). As expected, 80 effectively blocked Plk1 from localizing to centrosomes and kinetochores and consequently induced potent mitotic block and apoptotic cell death. Another prodrug 78 containing 9-fluorophenyl in place of the thiophene-containing heterocycle in 80 also induced a comparable degree of anti-Plk1 PBD effect. However, orally administered 78 was rapidly converted in the bloodstream to parent drug 15, which was shown be relatively stable toward in vivo oxidation due to its 9-fluorophenyl group in comparison to unsubstituted phenyl. Further derivatization of these inhibitors, particularly to improve the systemic prodrug stability, could lead to a new class of therapeutics against Plk1-addicted cancers., Competing Interests: The authors declare no competing financial interest., (Not subject to U.S. Copyright. Published 2023 by American Chemical Society.)- Published
- 2023
- Full Text
- View/download PDF
10. Deep phenotyping and lifetime trajectories reveal limited effects of longevity regulators on the aging process in C57BL/6J mice.
- Author
-
Xie K, Fuchs H, Scifo E, Liu D, Aziz A, Aguilar-Pimentel JA, Amarie OV, Becker L, da Silva-Buttkus P, Calzada-Wack J, Cho YL, Deng Y, Edwards AC, Garrett L, Georgopoulou C, Gerlini R, Hölter SM, Klein-Rodewald T, Kramer M, Leuchtenberger S, Lountzi D, Mayer-Kuckuk P, Nover LL, Oestereicher MA, Overkott C, Pearson BL, Rathkolb B, Rozman J, Russ J, Schaaf K, Spielmann N, Sanz-Moreno A, Stoeger C, Treise I, Bano D, Busch DH, Graw J, Klingenspor M, Klopstock T, Mock BA, Salomoni P, Schmidt-Weber C, Weiergräber M, Wolf E, Wurst W, Gailus-Durner V, Breteler MMB, Hrabě de Angelis M, and Ehninger D
- Subjects
- Mice, Animals, Male, Mice, Inbred C57BL, Phenotype, Longevity genetics, Aging physiology
- Abstract
Current concepts regarding the biology of aging are primarily based on studies aimed at identifying factors regulating lifespan. However, lifespan as a sole proxy measure for aging can be of limited value because it may be restricted by specific pathologies. Here, we employ large-scale phenotyping to analyze hundreds of markers in aging male C57BL/6J mice. For each phenotype, we establish lifetime profiles to determine when age-dependent change is first detectable relative to the young adult baseline. We examine key lifespan regulators (putative anti-aging interventions; PAAIs) for a possible countering of aging. Importantly, unlike most previous studies, we include in our study design young treated groups of animals, subjected to PAAIs prior to the onset of detectable age-dependent phenotypic change. Many PAAI effects influence phenotypes long before the onset of detectable age-dependent change, but, importantly, do not alter the rate of phenotypic change. Hence, these PAAIs have limited effects on aging., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
11. Conditional deletion of mTOR discloses its essential role in early B-cell development.
- Author
-
Zhang S, Dubois W, Feng X, Nguyen JT, Young NS, and Mock BA
- Subjects
- Animals, B-Lymphocytes metabolism, Cell Differentiation, Lymphocyte Activation, Mice, Mice, Knockout, TOR Serine-Threonine Kinases metabolism, Signal Transduction, Sirolimus
- Abstract
Mechanistic target of rapamycin (mTOR) is a serine-threonine kinase and central regulator of cell growth, differentiation, and survival. mTOR is commonly hyperactivated in a diverse number of cancers and critical roles for mTOR in regulating immune cell differentiation and function have been demonstrated. However, there is little work investigating the roles of mTOR in early B-cell development. Here we demonstrate that conditional disruption of mTOR in developing mouse B cells results in reduced pre-B-cell proliferation and survival, as well as a developmental block at the pre-B-cell stage, with a corresponding lack of peripheral B cells. Upon immunization with NP-CGG antigen, mice with Mtor conditional disruption in early B cells lost their ability to form germinal centers and produce specific antibodies. In competitive BM repopulation assays, donor BM cells from conditional knock-out mice were completely impaired in their ability to reconstitute B cells. Our data reveal the essential role of mTOR in early pre-B-cell development and survival., (© 2021 Wiley Periodicals LLC. This article has been contributed to by US Government employees and their work is in the public domain in the USA.)
- Published
- 2022
- Full Text
- View/download PDF
12. Fatty acid oxidation is required for embryonic stem cell survival during metabolic stress.
- Author
-
Yan H, Malik N, Kim YI, He Y, Li M, Dubois W, Liu H, Peat TJ, Nguyen JT, Tseng YC, Ayaz G, Alzamzami W, Chan K, Andresson T, Tessarollo L, Mock BA, Lee MP, and Huang J
- Subjects
- Fatty Acids, Oxidation-Reduction, Stress, Physiological, Embryonic Stem Cells, Lipid Metabolism
- Abstract
Metabolic regulation is critical for the maintenance of pluripotency and the survival of embryonic stem cells (ESCs). The transcription factor Tfcp2l1 has emerged as a key factor for the naïve pluripotency of ESCs. Here, we report an unexpected role of Tfcp2l1 in metabolic regulation in ESCs-promoting the survival of ESCs through regulating fatty acid oxidation (FAO) under metabolic stress. Tfcp2l1 directly activates many metabolic genes in ESCs. Deletion of Tfcp2l1 leads to an FAO defect associated with upregulation of glucose uptake, the TCA cycle, and glutamine catabolism. Mechanistically, Tfcp2l1 activates FAO by inducing Cpt1a, a rate-limiting enzyme transporting free fatty acids into the mitochondria. ESCs with defective FAO are sensitive to cell death induced by glycolysis inhibition and glutamine deprivation. Moreover, the Tfcp2l1-Cpt1a-FAO axis promotes the survival of quiescent ESCs and diapause-like blastocysts induced by mTOR inhibition. Thus, our results reveal how ESCs orchestrate pluripotent and metabolic programs to ensure their survival in response to metabolic stress., (Published 2021. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2021
- Full Text
- View/download PDF
13. A Small Molecule Stabilizer of the MYC G4-Quadruplex Induces Endoplasmic Reticulum Stress, Senescence and Pyroptosis in Multiple Myeloma.
- Author
-
Gaikwad SM, Phyo Z, Arteaga AQ, Gorjifard S, Calabrese DR, Connors D, Huang J, Michalowski AM, Zhang S, Liu ZG, Schneekloth JS Jr, and Mock BA
- Abstract
New approaches to target MYC include the stabilization of a guanine-rich, G-quadruplex (G4) tertiary DNA structure in the NHE III region of its promoter. Recent screening of a small molecule microarray platform identified a benzofuran, D089, that can stabilize the MYC G4 and inhibit its transcription. D089 induced both dose- and time-dependent multiple myeloma cell death mediated by endoplasmic reticulum induced stress. Unexpectedly, we uncovered two mechanisms of cell death: cellular senescence, as evidenced by increased levels of p16, p21 and γ-H2AX proteins and a caspase 3-independent mechanism consistent with pyroptosis. Cells treated with D089 exhibited high levels of the cleaved form of initiator caspase 8; but failed to show cleavage of executioner caspase 3, a classical apoptotic marker. Cotreatment with the a pan-caspase inhibitor Q-VD-OPh did not affect the cytotoxic effect of D089. In contrast, cleaved caspase 1, an inflammatory caspase downstream of caspases 8/9, was increased by D089 treatment. Cells treated with D089 in addition to either a caspase 1 inhibitor or siRNA-caspase 1 showed increased IC
50 values, indicating a contribution of cleaved caspase 1 to cell death. Downstream effects of caspase 1 activation after drug treatment included increases in IL1B, gasdermin D cleavage, and HMGB1 translocation from the nucleus to the cytoplasm. Drug treated cells underwent a 'ballooning' morphology characteristic of pyroptosis, rather than 'blebbing' typically associated with apoptosis. ASC specks colocalized with NLRP3 in proximity ligation assays after drug treatment, indicating inflammasome activation and further confirming pyroptosis as a contributor to cell death. Thus, the small molecule MYC G4 stabilizer, D089, provides a new tool compound for studying pyroptosis. These studies suggest that inducing both tumor senescence and pyroptosis may have therapeutic potential for cancer treatment.- Published
- 2020
- Full Text
- View/download PDF
14. Hypomorphic mTOR Downregulates CDK6 and Delays Thymic Pre-T LBL Tumorigenesis.
- Author
-
Gary JM, Simmons JK, Xu J, Zhang S, Peat TJ, Watson N, Gamache BJ, Zhang K, Kovalchuk AL, Michalowski AM, Chen JQ, Thaiwong T, Kiupel M, Gaikwad S, Etienne M, Simpson RM, Dubois W, Testa JR, and Mock BA
- Subjects
- Animals, Carcinogenesis, Down-Regulation, Mice, Mice, Transgenic, Cyclin-Dependent Kinase 6 metabolism, Gene Expression Profiling methods, TOR Serine-Threonine Kinases metabolism
- Abstract
PI3K/AKT/mTOR pathway hyperactivation is frequent in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/LBL). To model inhibition of mTOR, pre-T-cell lymphoblastic leukemia/lymphoma (pre-T LBL) tumor development was monitored in mice with T lymphocyte-specific, constitutively active AKT (Lck-MyrAkt2) that were either crossed to mTOR knockdown (KD) mice or treated with the mTOR inhibitor everolimus. Lck-MyrAkt2;mTOR KD mice lived significantly longer than Lck-MyrAkt2;mTOR wild-type (WT) mice, although both groups ultimately developed thymic pre-T LBL. An increase in survival was also observed when Lck-MyrAkt2;mTOR WT mice were treated for 8 weeks with everolimus. The transcriptional profiles of WT and KD thymic lymphomas were compared, and Ingenuity Pathway Upstream Regulator Analysis of differentially expressed genes in tumors from mTOR WT versus KD mice identified let-7 and miR-21 as potential regulatory genes. mTOR KD mice had higher levels of let-7a and miR-21 than mTOR WT mice, and rapamycin induced their expression in mTOR WT cells. CDK6 was one of the most downregulated targets of both let-7 and miR21 in mTOR KD tumors. CDK6 overexpression and decreased expression of let-7 in mTOR KD cells rescued a G
1 arrest phenotype. Combined mTOR (rapamycin) and CDK4/6 (palbociclib) inhibition decreased tumor size and proliferation in tumor flank transplants, increased survival in an intravenous transplant model of disseminated leukemia compared with single agent treatment, and cooperatively decreased cell viability in human T-ALL/LBL cell lines. Thus, mTOR KD mice provide a model to explore drug combinations synergizing with mTOR inhibitors and can be used to identify downstream targets of inhibition., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
15. Conventional Co-Housing Modulates Murine Gut Microbiota and Hematopoietic Gene Expression.
- Author
-
Chen J, Zhang S, Feng X, Wu Z, Dubois W, Thovarai V, Ahluwalia S, Gao S, Chen J, Peat T, Sen SK, Trinchieri G, Young NS, and Mock BA
- Subjects
- Animals, Bacteria genetics, Bacteria isolation & purification, Bone Marrow Transplantation, Gastrointestinal Microbiome, Gene Expression Regulation, Housing, Animal, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Phylogeny, Single-Cell Analysis, Specific Pathogen-Free Organisms, Bacteria classification, Gene Expression Profiling methods, Hematopoiesis, Myeloid Cells metabolism, Sequence Analysis, RNA methods, T-Lymphocytes metabolism
- Abstract
Specific-pathogen-free (SPF) mice have improved hematopoietic characteristics relative to germ-free mice, however, it is not clear whether improvements in hematopoietic traits will continue when the level of microorganism exposure is further increased. We co-housed SPF C57BL/6 mice in a conventional facility (CVT) and found a significant increase in gut microbiota diversity along with increased levels of myeloid cells and T cells, especially effector memory T cells. Through single cell RNA sequencing of sorted KL (c-Kit
+ Lin- ) cells, we imputed a decline in long-term hematopoietic stem cells and an increase in granulocyte-monocyte progenitors in CVT mice with up-regulation of genes associated with cell survival. Bone marrow transplantation through competitive repopulation revealed a significant increase in KSL (c-Kit+ Sca-1+ Lin- ) cell reconstitution in recipients of CVT donor cells which occurred when donors were co-housed for both one and twelve months. However, there was minimal to no gain in mature blood cell engraftment in recipients of CVT donor cells relative to those receiving SPF donor cells. We conclude that co-housing SPF mice with mice born in a conventional facility increased gut microbiota diversity, augmented myeloid cell production and T cell activation, stimulated KSL cell reconstitution, and altered hematopoietic gene expression.- Published
- 2020
- Full Text
- View/download PDF
16. Attenuation of immune-mediated bone marrow damage in conventionally housed mice.
- Author
-
Li J, Dubois W, Thovarai V, Wu Z, Feng X, Peat T, Zhang S, Sen SK, Trinchieri G, Chen J, Mock BA, and Young NS
- Subjects
- Anemia, Aplastic immunology, Anemia, Aplastic pathology, Animals, Bone Marrow pathology, Feces microbiology, Hyaluronan Receptors immunology, Hyaluronan Receptors metabolism, Immunologic Memory immunology, Lymphocyte Activation immunology, Mice, Inbred BALB C, Mice, Inbred C57BL, Specific Pathogen-Free Organisms, T-Lymphocytes metabolism, T-Lymphocytes transplantation, Transplantation Immunology, Transplantation, Homologous, Anemia, Aplastic therapy, Bone Marrow immunology, Bone Marrow Transplantation methods, Gastrointestinal Microbiome immunology, T-Lymphocytes immunology
- Abstract
In humans, bone marrow (BM) failure syndromes, both constitutional and acquired, predispose to myeloid malignancies. We have modeled acquired immune aplastic anemia, the paradigmatic disease of these syndromes, in the mouse by infusing lymph node cells from specific pathogen-free (SPF) CD45.1 congenic C57BL/6 (B6) donors into hybrid CByB6F1 recipients housed either in conventional (CVB) or SPF facilities. The severity of BM damage was reduced in CVB recipients; they also had reduced levels of CD44
+ CD62L- effector memory T cells, reduced numbers of donor-type CD44+ T cells, and reduced expansion of donor-type CD8 T cells carrying T-cell receptor β-variable regions 07, 11, and 17. Analyses of fecal samples through 16S ribosomal RNA amplicon sequencing revealed greater gut microbial alpha diversity in CVB mice relative to that of SPF mice. Thus, the presence of a broader spectrum of gut microorganisms in CVB-housed CByB6F1 could have primed recipient animal's immune system leading to suppression of allogeneic donor T-cell activation and expansion and attenuation of host BM destruction. These results suggest the potential benefit of diverse gut microbiota in patients receiving BM transplants., (© 2020 Wiley Periodicals, Inc.)- Published
- 2020
- Full Text
- View/download PDF
17. Mouse tumor susceptibility genes identify drug combinations for multiple myeloma.
- Author
-
Zhang S, DuBois W, Zhang K, Simmons JK, Hughitt VK, Gorjifard S, Gaikwad S, Peat TJ, and Mock BA
- Abstract
Long-term genetic studies utilizing backcross and congenic strain analyses coupled with positional cloning strategies and functional studies identified Cdkn2a , Mtor , and Mndal as mouse plasmacytoma susceptibility/resistance genes. Tumor incidence data in congenic strains carrying the resistance alleles of Cdkn2a and Mtor led us to hypothesize that drug combinations affecting these pathways are likely to have an additive, if not synergistic effect in inhibiting tumor cell growth. Traditional and novel systems-level genomic approaches were used to assess combination activity, disease specificity, and clinical potential of a drug combination involving rapamycin/everolimus, an Mtor inhibitor, with entinostat, an histone deacetylase inhibitor. The combination synergistically repressed oncogenic MYC and activated the Cdkn2a tumor suppressor. The identification of MYC as a primary upstream regulator led to the identification of small molecule binders of the G-quadruplex structure that forms in the NHEIII region of the MYC promoter. These studies highlight the importance of identifying drug combinations which simultaneously upregulate tumor suppressors and downregulate oncogenes., Competing Interests: Conflicts of interest All authors declared that there are no conflicts of interest.
- Published
- 2020
- Full Text
- View/download PDF
18. The transcription factor MZF1 differentially regulates murine Mtor promoter variants linked to tumor susceptibility.
- Author
-
Zhang S, Shi W, Ramsay ES, Bliskovsky V, Eiden AM, Connors D, Steinsaltz M, DuBois W, and Mock BA
- Subjects
- Alleles, Animals, Base Sequence, Cell Line, Tumor, Down-Regulation, Mice, Plasmacytoma pathology, Genetic Predisposition to Disease genetics, Kruppel-Like Transcription Factors metabolism, Mutation, Plasmacytoma genetics, Promoter Regions, Genetic genetics, TOR Serine-Threonine Kinases genetics
- Abstract
Mechanistic target of rapamycin (MTOR) is a highly conserved serine/threonine kinase that critically regulates cell growth, proliferation, differentiation, and survival. Previously, we have implicated Mtor as a plasmacytoma-resistance locus, Pctr2 , in mice. Here, we report that administration of the tumor-inducing agent pristane decreases Mtor gene expression to a greater extent in mesenteric lymph nodes of BALB/cAnPt mice than of DBA/2N mice. We identified six allelic variants in the Mtor promoter region in BALB/cAnPt and DBA/2N mice. To determine the effects of these variants on Mtor transcription, we constructed a series of luciferase reporters containing these promoter variants and transfected them into mouse plasmacytoma cells. We could attribute the differences in Mtor promoter activity between the two mouse strains to a C → T change at the -6 position relative to the transcriptional start site Tssr 40273; a T at this position in the BALB promoter creates a consensus binding site for the transcription factor MZF1 (myeloid zinc finger 1). Results from electrophoretic mobility shift assays and DNA pulldown assays with ChIP-PCR confirmed that MZF1 binds to the cis -element TGGGGA located in the -6/-1 Mtor promoter region. Of note, MZF1 significantly and differentially down-regulated Mtor promoter activity, with MZF1 overexpression reducing Mtor expression more strongly in BALB mice than in DBA mice. Moreover, MZF1 overexpression reduced Mtor expression in both fibroblasts and mouse plasmacytoma cells, and Mzf1 knockdown increased Mtor expression in BALB3T3 and NIH3T3 fibroblast cells. Our results provide evidence that MZF1 down-regulates Mtor expression in pristane-induced plasmacytomas in mice.
- Published
- 2019
- Full Text
- View/download PDF
19. The transcription factor CBFB suppresses breast cancer through orchestrating translation and transcription.
- Author
-
Malik N, Yan H, Moshkovich N, Palangat M, Yang H, Sanchez V, Cai Z, Peat TJ, Jiang S, Liu C, Lee M, Mock BA, Yuspa SH, Larson D, Wakefield LM, and Huang J
- Subjects
- Animals, Breast pathology, Breast Neoplasms pathology, Cell Line, Tumor, Core Binding Factor Alpha 2 Subunit metabolism, Down-Regulation, Eukaryotic Initiation Factors metabolism, Female, HEK293 Cells, Humans, Mice, Mice, Nude, RNA, Messenger metabolism, Receptors, Notch metabolism, Signal Transduction genetics, Tissue Array Analysis, Xenograft Model Antitumor Assays, Breast Neoplasms genetics, Core Binding Factor Alpha 2 Subunit genetics, Core Binding Factor beta Subunit metabolism, Gene Expression Regulation, Neoplastic, Heterogeneous-Nuclear Ribonucleoprotein K metabolism
- Abstract
Translation and transcription are frequently dysregulated in cancer. These two processes are generally regulated by distinct sets of factors. The CBFB gene, which encodes a transcription factor, has recently emerged as a highly mutated driver in a variety of human cancers including breast cancer. Here we report a noncanonical role of CBFB in translation regulation. RNA immunoprecipitation followed by deep sequencing (RIP-seq) reveals that cytoplasmic CBFB binds to hundreds of transcripts and regulates their translation. CBFB binds to mRNAs via hnRNPK and enhances translation through eIF4B, a general translation initiation factor. Interestingly, the RUNX1 mRNA, which encodes the transcriptional partner of CBFB, is bound and translationally regulated by CBFB. Furthermore, nuclear CBFB/RUNX1 complex transcriptionally represses the oncogenic NOTCH signaling pathway in breast cancer. Thus, our data reveal an unexpected function of CBFB in translation regulation and propose that breast cancer cells evade translation and transcription surveillance simultaneously through downregulating CBFB.
- Published
- 2019
- Full Text
- View/download PDF
20. Chemical and structural studies provide a mechanistic basis for recognition of the MYC G-quadruplex.
- Author
-
Calabrese DR, Chen X, Leon EC, Gaikwad SM, Phyo Z, Hewitt WM, Alden S, Hilimire TA, He F, Michalowski AM, Simmons JK, Saunders LB, Zhang S, Connors D, Walters KJ, Mock BA, and Schneekloth JS Jr
- Subjects
- Binding Sites genetics, Binding Sites physiology, Blotting, Western, Cell Line, Cell Line, Tumor, Cell Survival genetics, Humans, Hydrogen Bonding, Magnetic Resonance Spectroscopy, Molecular Structure, Promoter Regions, Genetic genetics, Promoter Regions, Genetic physiology, Protein Binding genetics, Protein Binding physiology, Protein Structure, Secondary, Proto-Oncogene Proteins c-myc genetics, Structure-Activity Relationship, Surface Plasmon Resonance, Cell Survival physiology, G-Quadruplexes, Proto-Oncogene Proteins c-myc metabolism
- Abstract
G-quadruplexes (G4s) are noncanonical DNA structures that frequently occur in the promoter regions of oncogenes, such as MYC, and regulate gene expression. Although G4s are attractive therapeutic targets, ligands capable of discriminating between different G4 structures are rare. Here, we describe DC-34, a small molecule that potently downregulates MYC transcription in cancer cells by a G4-dependent mechanism. Inhibition by DC-34 is significantly greater for MYC than other G4-driven genes. We use chemical, biophysical, biological, and structural studies to demonstrate a molecular rationale for the recognition of the MYC G4. We solve the structure of the MYC G4 in complex with DC-34 by NMR spectroscopy and illustrate specific contacts responsible for affinity and selectivity. Modification of DC-34 reveals features required for G4 affinity, biological activity, and validates the derived NMR structure. This work advances the design of quadruplex-interacting small molecules to control gene expression in therapeutic areas such as cancer.
- Published
- 2018
- Full Text
- View/download PDF
21. Characterization of clinically used oral antiseptics as quadruplex-binding ligands.
- Author
-
Calabrese DR, Zlotkowski K, Alden S, Hewitt WM, Connelly CM, Wilson RM, Gaikwad S, Chen L, Guha R, Thomas CJ, Mock BA, and Schneekloth JS Jr
- Subjects
- Anti-Infective Agents, Local pharmacology, Binding Sites, Cell Line, Tumor, Cell Survival drug effects, Chlorhexidine pharmacology, DNA genetics, DNA metabolism, Gene Expression drug effects, Humans, Ligands, Magnetic Resonance Spectroscopy, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Small Molecule Libraries pharmacology, Surface Plasmon Resonance, Anti-Infective Agents, Local metabolism, Chlorhexidine metabolism, G-Quadruplexes, Small Molecule Libraries metabolism
- Abstract
Approaches to characterize the nucleic acid-binding properties of drugs and druglike small molecules are crucial to understanding the behavior of these compounds in cellular systems. Here, we use a Small Molecule Microarray (SMM) profiling approach to identify the preferential interaction between chlorhexidine, a widely used oral antiseptic, and the G-quadruplex (G4) structure in the KRAS oncogene promoter. The interaction of chlorhexidine and related drugs to the KRAS G4 is evaluated using multiple biophysical methods, including thermal melt, fluorescence titration and surface plasmon resonance (SPR) assays. Chlorhexidine has a specific low micromolar binding interaction with the G4, while related drugs have weaker and/or less specific interactions. Through NMR experiments and docking studies, we propose a plausible binding mode driven by both aromatic stacking and groove binding interactions. Additionally, cancer cell lines harbouring oncogenic mutations in the KRAS gene exhibit increased sensitivity to chlorhexidine. Treatment of breast cancer cells with chlorhexidine decreases KRAS protein levels, while a KRAS gene transiently expressed by a promoter lacking a G4 is not affected. This work confirms that known ligands bind broadly to G4 structures, while other drugs and druglike compounds can have more selective interactions that may be biologically relevant.
- Published
- 2018
- Full Text
- View/download PDF
22. The Reign of Antibodies: A Celebration of and Tribute to Michael Potter and His Homogeneous Immunoglobulin Workshops.
- Author
-
Gearhart PJ, Mock BA, Casellas R, and Cancro MP
- Subjects
- Antibodies, Immunoglobulins
- Published
- 2018
- Full Text
- View/download PDF
23. mTOR intersects antibody-inducing signals from TACI in marginal zone B cells.
- Author
-
Sintes J, Gentile M, Zhang S, Garcia-Carmona Y, Magri G, Cassis L, Segura-Garzón D, Ciociola A, Grasset EK, Bascones S, Comerma L, Pybus M, Lligé D, Puga I, Gutzeit C, He B, DuBois W, Crespo M, Pascual J, Mensa A, Aróstegui JI, Juan M, Yagüe J, Serrano S, Lloreta J, Meffre E, Hahne M, Cunningham-Rundles C, Mock BA, and Cerutti A
- Subjects
- Animals, Cell Line, Cell Proliferation, Enzyme Activation, Gene Expression Profiling, HEK293 Cells, Humans, Immunoglobulin Class Switching genetics, Immunoglobulin Class Switching immunology, Immunoglobulin G biosynthesis, Lymphocyte Activation immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, NF-kappa B metabolism, Signal Transduction immunology, Sirolimus pharmacology, B-Lymphocytes immunology, Immunoglobulin G immunology, Mechanistic Target of Rapamycin Complex 1 immunology, Myeloid Differentiation Factor 88 metabolism, TOR Serine-Threonine Kinases immunology, Transmembrane Activator and CAML Interactor Protein immunology
- Abstract
Mechanistic target of rapamycin (mTOR) enhances immunity in addition to orchestrating metabolism. Here we show that mTOR coordinates immunometabolic reconfiguration of marginal zone (MZ) B cells, a pre-activated lymphocyte subset that mounts antibody responses to T-cell-independent antigens through a Toll-like receptor (TLR)-amplified pathway involving transmembrane activator and CAML interactor (TACI). This receptor interacts with mTOR via the TLR adapter MyD88. The resulting mTOR activation instigates MZ B-cell proliferation, immunoglobulin G (IgG) class switching, and plasmablast differentiation through a rapamycin-sensitive pathway that integrates metabolic and antibody-inducing transcription programs, including NF-κB. Disruption of TACI-mTOR interaction by rapamycin, truncation of the MyD88-binding domain of TACI, or B-cell-conditional mTOR deficiency interrupts TACI signaling via NF-κB and cooperation with TLRs, thereby hampering IgG production to T-cell-independent antigens but not B-cell survival. Thus, mTOR drives innate-like antibody responses by linking proximal TACI signaling events with distal immunometabolic transcription programs.
- Published
- 2017
- Full Text
- View/download PDF
24. Cooperative Targets of Combined mTOR/HDAC Inhibition Promote MYC Degradation.
- Author
-
Simmons JK, Michalowski AM, Gamache BJ, DuBois W, Patel J, Zhang K, Gary J, Zhang S, Gaikwad S, Connors D, Watson N, Leon E, Chen JQ, Kuehl WM, Lee MP, Zingone A, Landgren O, Ordentlich P, Huang J, and Mock BA
- Subjects
- Animals, Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Tumor, DNA Repair, DNA Replication drug effects, Disease Models, Animal, Drug Synergism, Female, Gene Expression Profiling, Humans, Mice, Pharmacogenetics, Pharmacogenomic Variants, Protein Stability, Proteolysis, Transcriptome, Xenograft Model Antitumor Assays, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-myc metabolism, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Cancer treatments often require combinations of molecularly targeted agents to be effective. mTORi (rapamycin) and HDACi (MS-275/entinostat) inhibitors have been shown to be effective in limiting tumor growth, and here we define part of the cooperative action of this drug combination. More than 60 human cancer cell lines responded synergistically (CI<1) when treated with this drug combination compared with single agents. In addition, a breast cancer patient-derived xenograft, and a BCL-XL plasmacytoma mouse model both showed enhanced responses to the combination compared with single agents. Mice bearing plasma cell tumors lived an average of 70 days longer on combination treatment compared with single agents. A set of 37 genes cooperatively affected (34 downregulated; 3 upregulated) by the combination responded pharmacodynamically in human myeloma cell lines, xenografts, and a P493 model, and were both enriched in tumors, and correlated with prognostic markers in myeloma patient datasets. Genes downregulated by the combination were overexpressed in several untreated cancers (breast, lung, colon, sarcoma, head and neck, myeloma) compared with normal tissues. The MYC/E2F axis, identified by upstream regulator analyses and validated by immunoblots, was significantly inhibited by the drug combination in several myeloma cell lines. Furthermore, 88% of the 34 genes downregulated have MYC-binding sites in their promoters, and the drug combination cooperatively reduced MYC half-life by 55% and increased degradation. Cells with MYC mutations were refractory to the combination. Thus, integrative approaches to understand drug synergy identified a clinically actionable strategy to inhibit MYC/E2F activity and tumor cell growth in vivo Mol Cancer Ther; 16(9); 2008-21. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
25. Molecular Pathways: Increased Susceptibility to Infection Is a Complication of mTOR Inhibitor Use in Cancer Therapy.
- Author
-
Eiden AM, Zhang S, Gary JM, Simmons JK, and Mock BA
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Humans, Immunosuppressive Agents adverse effects, Neoplasms drug therapy, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Antineoplastic Agents adverse effects, Communicable Diseases etiology, Disease Susceptibility etiology, Protein Kinase Inhibitors adverse effects, Signal Transduction drug effects, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
As one of the earliest examples of "chemical biology," the M: echanistic T: arget of R: apamycin (mTOR) protein and its chemical inhibitors have been extensively studied across a spectrum of physiologic and pathologic processes at the molecular, organismal, and patient population levels. There are several FDA-approved mTOR inhibitors (sirolimus, everolimus, and temsirolimus) with indications for cancer treatment and for prevention of solid organ rejection. Dozens of mTOR inhibitors are currently being evaluated in hundreds of ongoing clinical trials across a spectrum of diseases, including numerous cancer indications, autoimmune diseases, and a number of congenital disorders. As many of the approved and investigational indications for mTOR inhibitors require long-term treatment, the magnitude and incidence of particular side effects differ from those observed in shorter-term treatments. Here, we focus on the increased risk of infections in patients being treated with mTOR inhibitors. While increased infection rates might be expected from a class of drugs approved as posttransplant immunosuppressants, we review reports from clinical, mechanistic, and genetically engineered mouse model studies detailing a much more nuanced view of mTOR inhibitor drug action and target biology., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
26. Small Molecule Microarrays Enable the Identification of a Selective, Quadruplex-Binding Inhibitor of MYC Expression.
- Author
-
Felsenstein KM, Saunders LB, Simmons JK, Leon E, Calabrese DR, Zhang S, Michalowski A, Gareiss P, Mock BA, and Schneekloth JS Jr
- Subjects
- Blotting, Western, Cell Line, Tumor, Humans, Inhibitory Concentration 50, Molecular Structure, Small Molecule Libraries chemistry, G-Quadruplexes, Gene Expression Regulation drug effects, Oligonucleotide Array Sequence Analysis, Proto-Oncogene Proteins c-myc antagonists & inhibitors, Proto-Oncogene Proteins c-myc genetics, Small Molecule Libraries pharmacology
- Abstract
The transcription factor MYC plays a pivotal role in cancer initiation, progression, and maintenance. However, it has proven difficult to develop small molecule inhibitors of MYC. One attractive route to pharmacological inhibition of MYC has been the prevention of its expression through small molecule-mediated stabilization of the G-quadruplex (G4) present in its promoter. Although molecules that bind globally to quadruplex DNA and influence gene expression are well-known, the identification of new chemical scaffolds that selectively modulate G4-driven genes remains a challenge. Here, we report an approach for the identification of G4-binding small molecules using small molecule microarrays (SMMs). We use the SMM screening platform to identify a novel G4-binding small molecule that inhibits MYC expression in cell models, with minimal impact on the expression of other G4-associated genes. Surface plasmon resonance (SPR) and thermal melt assays demonstrated that this molecule binds reversibly to the MYC G4 with single digit micromolar affinity, and with weaker or no measurable binding to other G4s. Biochemical and cell-based assays demonstrated that the compound effectively silenced MYC transcription and translation via a G4-dependent mechanism of action. The compound induced G1 arrest and was selectively toxic to MYC-driven cancer cell lines containing the G4 in the promoter but had minimal effects in peripheral blood mononucleocytes or a cell line lacking the G4 in its MYC promoter. As a measure of selectivity, gene expression analysis and qPCR experiments demonstrated that MYC and several MYC target genes were downregulated upon treatment with this compound, while the expression of several other G4-driven genes was not affected. In addition to providing a novel chemical scaffold that modulates MYC expression through G4 binding, this work suggests that the SMM screening approach may be broadly useful as an approach for the identification of new G4-binding small molecules.
- Published
- 2016
- Full Text
- View/download PDF
27. Loss-of-function RNAi screens in breast cancer cells identify AURKB, PLK1, PIK3R1, MAPK12, PRKD2, and PTK6 as sensitizing targets of rapamycin activity.
- Author
-
Ou O, Huppi K, Chakka S, Gehlhaus K, Dubois W, Patel J, Chen J, Mackiewicz M, Jones TL, Pitt JJ, Martin SE, Goldsmith P, Simmons JK, Mock BA, and Caplen NJ
- Subjects
- Animals, Aurora Kinase B antagonists & inhibitors, Benzamides administration & dosage, Benzamides pharmacology, Breast Neoplasms enzymology, Cell Cycle Proteins antagonists & inhibitors, Cell Line, Tumor, Class Ia Phosphatidylinositol 3-Kinase, Drug Synergism, Female, Humans, MCF-7 Cells, Mice, Mice, SCID, Mitogen-Activated Protein Kinase 12 antagonists & inhibitors, Neoplasm Proteins antagonists & inhibitors, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase D2, Protein Kinase Inhibitors administration & dosage, Protein Kinases metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors, Pyridines administration & dosage, Pyridines pharmacology, RNA Interference, Random Allocation, Sirolimus administration & dosage, Xenograft Model Antitumor Assays, Polo-Like Kinase 1, Antineoplastic Combined Chemotherapy Protocols pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Drug Screening Assays, Antitumor methods, Protein Kinase Inhibitors pharmacology, Sirolimus pharmacology
- Abstract
The use of molecularly targeted drugs as single agents has shown limited utility in many tumor types, largely due to the complex and redundant nature of oncogenic signaling networks. Targeting of the PI3K/AKT/mTOR pathway through inhibition of mTOR in combination with aromatase inhibitors has seen success in particular sub-types of breast cancer and there is a need to identify additional synergistic combinations to maximize the clinical potential of mTOR inhibitors. We have used loss-of-function RNAi screens of the mTOR inhibitor rapamycin to identify sensitizers of mTOR inhibition. RNAi screens conducted in combination with rapamycin in multiple breast cancer cell lines identified six genes, AURKB, PLK1, PIK3R1, MAPK12, PRKD2, and PTK6 that when silenced, each enhanced the sensitivity of multiple breast cancer lines to rapamycin. Using selective pharmacological agents we confirmed that inhibition of AURKB or PLK1 synergizes with rapamycin. Compound-associated gene expression data suggested histone deacetylation (HDAC) inhibition as a strategy for reducing the expression of several of the rapamycin-sensitizing genes, and we tested and validated this using the HDAC inhibitor entinostat in vitro and in vivo. Our findings indicate new approaches for enhancing the efficacy of rapamycin including the use of combining its application with HDAC inhibition., (Published by Elsevier Ireland Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
28. TORC1 and class I HDAC inhibitors synergize to suppress mature B cell neoplasms.
- Author
-
Simmons JK, Patel J, Michalowski A, Zhang S, Wei BR, Sullivan P, Gamache B, Felsenstein K, Kuehl WM, Simpson RM, Zingone A, Landgren O, and Mock BA
- Subjects
- Animals, Cell Line, Tumor, Humans, Lymphoma, B-Cell genetics, Lymphoma, B-Cell metabolism, Lymphoma, B-Cell mortality, Mechanistic Target of Rapamycin Complex 1, Mice, Mice, Nude, Multiple Myeloma genetics, Multiple Myeloma metabolism, Multiple Myeloma pathology, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, Plasmacytoma genetics, Plasmacytoma metabolism, Plasmacytoma pathology, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Benzamides pharmacology, Histone Deacetylase Inhibitors pharmacology, Lymphoma, B-Cell drug therapy, Multiple Myeloma drug therapy, Multiprotein Complexes antagonists & inhibitors, Plasmacytoma drug therapy, Pyridines pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Enhanced proliferative signaling and loss of cell cycle regulation are essential for cancer progression. Increased mitogenic signaling through activation of the mTOR pathway, coupled with deregulation of the Cyclin D/retinoblastoma (Rb) pathway is a common feature of lymphoid malignancies, including plasmacytoma (PCT), multiple myeloma (MM), Burkitt's lymphoma (BL), and mantle cell lymphoma (MCL). Here we evaluate the synergy of pharmacologically affecting both of these critical pathways using the mTOR inhibitor sirolimus and the histone deacetylase inhibitor entinostat. A dose-matrix screening approach found this combination to be highly active and synergistic in a panel of genetically diverse human MM cell lines. Synergy and activity was observed in mouse PCT and human BL and MCL cell lines tested in vitro, as well as in freshly isolated primary MM patient samples tested ex vivo. This combination had minimal effects on healthy donor cells and retained activity when tested in a co-culture system simulating the protective interaction of cancer cells with the tumor microenvironment. Combining sirolimus with entinostat enhanced cell cycle arrest and apoptosis. At the molecular level, entinostat increased the expression of cell cycle negative regulators including CDKN1A (p21) and CDKN2A (p16), while the combination decreased critical growth and survival effectors including Cyclin D, BCL-XL, BIRC5, and activated MAPK., (Published by Elsevier B.V.)
- Published
- 2014
- Full Text
- View/download PDF
29. B cell-specific deficiencies in mTOR limit humoral immune responses.
- Author
-
Zhang S, Pruitt M, Tran D, Du Bois W, Zhang K, Patel R, Hoover S, Simpson RM, Simmons J, Gary J, Snapper CM, Casellas R, and Mock BA
- Subjects
- Animals, Antibodies, Bacterial biosynthesis, Antibodies, Bacterial immunology, Antibody Affinity, Bacterial Capsules immunology, Cell Lineage, Enzyme Activation, Gene Knock-In Techniques, Germinal Center immunology, Germinal Center pathology, Haptens immunology, Housing, Animal, Immunization, Immunoglobulin Class Switching, Immunoglobulin G immunology, Mice, Mice, Knockout, Nitrophenols immunology, Phenylacetates immunology, Signal Transduction immunology, Somatic Hypermutation, Immunoglobulin, Spleen immunology, Spleen pathology, Streptococcal Infections immunology, Streptococcus pneumoniae immunology, Antibody Diversity, Antibody Formation, B-Lymphocytes immunology, Cytidine Deaminase immunology, TOR Serine-Threonine Kinases immunology
- Abstract
Generation of high-affinity Abs in response to Ags/infectious agents is essential for developing long-lasting immune responses. B cell maturation and Ab responses to Ag stimulation require Ig somatic hypermutation (SHM) and class-switch recombination (CSR) for high-affinity responses. Upon immunization with either the model Ag 4-hydroxy-3-nitrophenylacetyl hapten (NP) conjugated to chicken γ globulin lysine (NP-CGG) or heat-killed Streptococcus pneumoniae capsular type 14 protein (Pn14), knock-in (KI) mice hypomorphic for mTOR function had a decreased ability to form germinal centers, develop high-affinity anti-NP-specific or anti-Pn14-specific Abs, and perform SHM/CSR. Hypomorphic mTOR mice also had a high mortality (40%) compared with wild-type (WT) (0%) littermates and had lower pneumococcal surface protein A-specific Ab titers when immunized and challenged with live S. pneumoniae infection. Mice with mTOR deleted in their B cell lineage (knockout [KO]) also produced fewer splenic germinal centers and decreased high-affinity Ab responses to NP-CGG than did their WT littermates. CSR rates were lower in mTOR KI and KO mice, and pharmacologic inhibition of mTOR in WT B cells resulted in decreased rates of ex vivo CSR. RNA and protein levels of activation-induced cytidine deaminase (AID), a protein essential for SHM and CSR, were lower in B cells from both KI and B cell-specific KO mice, concomitant with increases in phosphorylated AKT and FOXO1. Rescue experiments increasing AID expression in KI B cells restored CSR levels to those in WT B cells. Thus, mTOR plays an important immunoregulatory role in the germinal center, at least partially through AID signaling, in generating high-affinity Abs.
- Published
- 2013
- Full Text
- View/download PDF
30. A novel KIT-deficient mouse mast cell model for the examination of human KIT-mediated activation responses.
- Author
-
Smrž D, Bandara G, Zhang S, Mock BA, Beaven MA, Metcalfe DD, and Gilfillan AM
- Subjects
- Animals, Benzamides immunology, Benzamides pharmacology, Bone Marrow Cells immunology, Bone Marrow Cells metabolism, Calcium immunology, Calcium metabolism, Cell Degranulation drug effects, Cell Survival drug effects, Cell Survival immunology, Cells, Cultured, Chemotaxis drug effects, Chemotaxis immunology, Cytokines immunology, Cytokines metabolism, Flow Cytometry, Humans, Imatinib Mesylate, Immunoblotting, Mast Cells metabolism, Mast Cells physiology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microscopy, Confocal, Mutation immunology, Phosphorylation drug effects, Phosphorylation immunology, Piperazines immunology, Piperazines pharmacology, Protein Kinase Inhibitors immunology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-kit genetics, Proto-Oncogene Proteins c-kit metabolism, Pyrimidines immunology, Pyrimidines pharmacology, Receptors, IgE genetics, Receptors, IgE immunology, Receptors, IgE metabolism, Stem Cell Factor immunology, Stem Cell Factor pharmacology, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases immunology, TOR Serine-Threonine Kinases metabolism, Antigens immunology, Cell Degranulation immunology, Mast Cells immunology, Proto-Oncogene Proteins c-kit immunology
- Abstract
Activation of KIT, by its ligand, stem cell factor (SCF), results in the initiation of signal transduction pathways that influence mast cell survival and proliferation. Activating mutations in KIT have thus been linked to clonal MC proliferation associated with systemic mastocytosis. SCF also modulates MC function by inducing MC chemotaxis and by potentiating antigen (Ag)/IgE-mediated MC degranulation. Thus, mutations in KIT also have the potential to affect these processes in allergic and other mast cell-related diseases. Studies to determine how native and mutated KIT may modulate MC chemotaxis and activation have, however, been limited due to the lack of availability of a suitable functional MC line lacking native KIT which would allow transduction of KIT constructs. Here we describe a novel mouse MC line which allows the study of normal and mutated KIT constructs. These cells originated from a bone marrow-derived mouse MC culture out of which a rapidly dividing mast cell sub-population spontaneously arose. Over time, these cells lost KIT expression while continuing to express functional high affinity receptors for IgE (FcεRI). As a consequence, these cells degranulated in response to Ag/IgE but did not migrate nor show any evidence of potentiation of Ag/IgE degranulation in response to SCF. Retroviral transduction of the cells with a human (hu)KIT construct resulted in surface expression of huKIT which responded to huSCF by potentiation of Ag/IgE-induced degranulation and chemotaxis. This cell line thus presents a novel system to delineate how MC function is modulated by native and mutated KIT and for the identification of novel inhibitors of these processes., (Published by Elsevier B.V.)
- Published
- 2013
- Full Text
- View/download PDF
31. Mouse genetics 2011: meeting report.
- Author
-
Simmons JK, Amlin-Van Schaick JC, Geiger TR, Reilly K, Hunter K, and Mock BA
- Subjects
- Animals, Disease Models, Animal, Humans, Mice growth & development, Mice physiology, Disease genetics, Mice genetics
- Published
- 2012
- Full Text
- View/download PDF
32. mTORC1 and mTORC2 differentially regulate homeostasis of neoplastic and non-neoplastic human mast cells.
- Author
-
Smrz D, Kim MS, Zhang S, Mock BA, Smrzová S, DuBois W, Simakova O, Maric I, Wilson TM, Metcalfe DD, and Gilfillan AM
- Subjects
- Adaptor Proteins, Signal Transducing antagonists & inhibitors, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Animals, Antibiotics, Antineoplastic pharmacology, Blotting, Western, Bone Marrow Cells drug effects, Bone Marrow Cells metabolism, Carrier Proteins antagonists & inhibitors, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Cycle drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Cells, Cultured, Gene Expression Profiling, Humans, Mast Cells drug effects, Mastocytosis, Systemic genetics, Mastocytosis, Systemic pathology, Mechanistic Target of Rapamycin Complex 1, Mice, Mice, Transgenic, Multiprotein Complexes, Naphthyridines pharmacology, Proteins antagonists & inhibitors, Proteins genetics, RNA Interference, Rapamycin-Insensitive Companion of mTOR Protein, Regulatory-Associated Protein of mTOR, Reverse Transcriptase Polymerase Chain Reaction, Sirolimus pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Tumor Cells, Cultured, Homeostasis, Mast Cells metabolism, Mastocytosis, Systemic metabolism, Proteins metabolism
- Abstract
Increased mast cell burden is observed in the inflamed tissues and affected organs and tissues of patients with mast cell proliferative disorders. However, normal mast cells participate in host defense, so approaches to preferentially target clonally expanding mast cells are needed. We found that mammalian target of rapamycin complex 1 (mTORC1) and 2 (mTORC2) are up-regulated in neoplastic and developing immature mast cells compared with their terminally differentiated counterparts. Elevated mTOR mRNA was also observed in bone marrow mononuclear cells of patients exhibiting mast-cell hyperplasia. Selective inhibition of mTORC1 and mTORC2 through genetic and pharmacologic manipulation revealed that, whereas mTORC1 may contribute to mast-cell survival, mTORC2 was only critical for homeostasis of neoplastic and dividing immature mast cells. The cytostatic effect of mTORC2 down-regulation in proliferating mast cells was determined to be via inhibition of cell-cycle progression. Because mTORC2 was observed to play little role in the homeostasis of differentiated, nonproliferating, mature mast cells, these data provide a rationale for adopting a targeted approaching selectively inhibiting mTORC2 to effectively reduce the proliferation of mast cells associated with inflammation and disorders of mast cell proliferation while leaving normal differentiated mast cells largely unaffected.
- Published
- 2011
- Full Text
- View/download PDF
33. Constitutive reductions in mTOR alter cell size, immune cell development, and antibody production.
- Author
-
Zhang S, Readinger JA, DuBois W, Janka-Junttila M, Robinson R, Pruitt M, Bliskovsky V, Wu JZ, Sakakibara K, Patel J, Parent CA, Tessarollo L, Schwartzberg PL, and Mock BA
- Subjects
- Animals, B-Lymphocytes metabolism, Body Size genetics, Cell Differentiation genetics, Cell Differentiation immunology, Cell Lineage genetics, Cell Lineage immunology, Cell Size, Down-Regulation immunology, Down-Regulation physiology, Gene Knockdown Techniques, Mice, Mice, Inbred BALB C, Mice, Transgenic, Organ Size genetics, Spleen anatomy & histology, Spleen metabolism, Antibody Formation genetics, B-Lymphocytes cytology, B-Lymphocytes physiology, TOR Serine-Threonine Kinases genetics
- Abstract
Mammalian TOR (mTOR) regulates cell growth, proliferation, and migration. Because mTOR knock-outs are embryonic lethal, we generated a viable hypomorphic mouse by neo-insertion that partially disrupts mTOR transcription and creates a potential physiologic model of mTORC1/TORC2 inhibition. Homozygous knock-in mice exhibited reductions in body, organ, and cell size. Although reductions in most organ sizes were proportional to decreased body weight, spleens were disproportionately smaller. Decreases in the total number of T cells, particularly memory cells, and reduced responses to chemokines suggested alterations in T-cell homing/homeostasis. T-cell receptor-stimulated T cells proliferated less, produced lower cytokine levels, and expressed FoxP3. Decreased neutrophil numbers were also observed in the spleen, despite normal development and migration in the bone marrow. However, B-cell effects were most pronounced, with a partial block in B-cell development in the bone marrow, altered splenic populations, and decreases in proliferation, antibody production, and migration to chemokines. Moreover, increased AKT(Ser473) phosphorylation was observed in activated B cells, reminiscent of cancers treated with rapamycin, and was reduced by a DNA-pk inhibitor. Thus, mTOR is required for the maturation and differentiation of multiple immune cell lineages. These mice provide a novel platform for studying the consequences of constitutively reduced mTORC1/TORC2 activity.
- Published
- 2011
- Full Text
- View/download PDF
34. Mndal, a new interferon-inducible family member, is highly polymorphic, suppresses cell growth, and may modify plasmacytoma susceptibility.
- Author
-
Zhang K, Kagan D, DuBois W, Robinson R, Bliskovsky V, Vass WC, Zhang S, and Mock BA
- Subjects
- Animals, Antigens, Differentiation, Myelomonocytic genetics, Antigens, Differentiation, Myelomonocytic metabolism, B-Lymphocytes metabolism, B-Lymphocytes pathology, Cloning, Molecular, Colony-Forming Units Assay, DNA, Complementary genetics, Gene Expression Profiling, Humans, Mice, Mice, Inbred BALB C, Mice, Inbred DBA, NIH 3T3 Cells, Oligonucleotide Array Sequence Analysis, Plasmacytoma genetics, Plasmacytoma metabolism, Transcription Factors genetics, Transcription Factors metabolism, Antineoplastic Agents pharmacology, Interferons pharmacology, Nuclear Proteins genetics, Nuclear Proteins metabolism, Plasmacytoma pathology, Polymorphism, Genetic, Proteins genetics, Proteins metabolism
- Abstract
The human HIN-200 gene cluster and its mouse counterpart, the interferon inducible-200 (Ifi200) family, both on Chr 1, are associated with several diseases, including solid tumors and lupus. Our study was initiated to identify the modifier gene(s) encoded by the Pctm locus, in which mouse B-cell plasmacytomas induced by pristane are associated with heterozygosity of Chr 1 genes near the Ifi200 cluster. A screen for differentially expressed genes in granulomatous tissues induced by pristane in resistant and susceptible strains identified a new Ifi200 member whose expression was 1000-fold higher in the strain carrying the resistant allele of Pctm and was the most highly expressed Ifi200 gene. The gene, designated Mndal (for MNDA-like, myeloid nuclear differentiation antigen-like), was absent in the susceptible genome, as were genomic sequences upstream of Ifi203, the gene adjacent to Mndal. Ectopic expression of MNDAL suppressed cell growth, which, together with the disease susceptibility of heterozygotes at the Pctm locus, suggests that Mndal, perhaps with Ifi203, acts as a tumor suppressor and display(s) haploinsufficiency. Mndal is highly polymorphic among inbred mouse strains, because it is absent in 10 of 24 strains. This polymorphism may have implications for other disease modifiers mapping to the same region.
- Published
- 2009
- Full Text
- View/download PDF
35. The Collaborative Cross, a community resource for the genetic analysis of complex traits.
- Author
-
Churchill GA, Airey DC, Allayee H, Angel JM, Attie AD, Beatty J, Beavis WD, Belknap JK, Bennett B, Berrettini W, Bleich A, Bogue M, Broman KW, Buck KJ, Buckler E, Burmeister M, Chesler EJ, Cheverud JM, Clapcote S, Cook MN, Cox RD, Crabbe JC, Crusio WE, Darvasi A, Deschepper CF, Doerge RW, Farber CR, Forejt J, Gaile D, Garlow SJ, Geiger H, Gershenfeld H, Gordon T, Gu J, Gu W, de Haan G, Hayes NL, Heller C, Himmelbauer H, Hitzemann R, Hunter K, Hsu HC, Iraqi FA, Ivandic B, Jacob HJ, Jansen RC, Jepsen KJ, Johnson DK, Johnson TE, Kempermann G, Kendziorski C, Kotb M, Kooy RF, Llamas B, Lammert F, Lassalle JM, Lowenstein PR, Lu L, Lusis A, Manly KF, Marcucio R, Matthews D, Medrano JF, Miller DR, Mittleman G, Mock BA, Mogil JS, Montagutelli X, Morahan G, Morris DG, Mott R, Nadeau JH, Nagase H, Nowakowski RS, O'Hara BF, Osadchuk AV, Page GP, Paigen B, Paigen K, Palmer AA, Pan HJ, Peltonen-Palotie L, Peirce J, Pomp D, Pravenec M, Prows DR, Qi Z, Reeves RH, Roder J, Rosen GD, Schadt EE, Schalkwyk LC, Seltzer Z, Shimomura K, Shou S, Sillanpää MJ, Siracusa LD, Snoeck HW, Spearow JL, Svenson K, Tarantino LM, Threadgill D, Toth LA, Valdar W, de Villena FP, Warden C, Whatley S, Williams RW, Wiltshire T, Yi N, Zhang D, Zhang M, and Zou F
- Subjects
- Animals, Community Networks, Crosses, Genetic, Databases, Genetic, Health Services Research, Humans, Mice, Recombination, Genetic, Breeding, Health Resources, Mice, Inbred Strains
- Abstract
The goal of the Complex Trait Consortium is to promote the development of resources that can be used to understand, treat and ultimately prevent pervasive human diseases. Existing and proposed mouse resources that are optimized to study the actions of isolated genetic loci on a fixed background are less effective for studying intact polygenic networks and interactions among genes, environments, pathogens and other factors. The Collaborative Cross will provide a common reference panel specifically designed for the integrative analysis of complex systems and will change the way we approach human health and disease.
- Published
- 2004
- Full Text
- View/download PDF
36. Frap, FKBP12 rapamycin-associated protein, is a candidate gene for the plasmacytoma resistance locus Pctr2 and can act as a tumor suppressor gene.
- Author
-
Bliskovsky V, Ramsay ES, Scott J, DuBois W, Shi W, Zhang S, Qian X, Lowy DR, and Mock BA
- Subjects
- Alleles, Amino Acid Sequence, Animals, Blotting, Western, Cell Division, Cell Line, Tumor, Cell Transformation, Neoplastic, DNA chemistry, Genotype, Humans, Mice, Mice, Inbred BALB C, Mice, Inbred DBA, Models, Genetic, Molecular Sequence Data, NIH 3T3 Cells, Phenotype, Phosphorylation, Plasmacytoma metabolism, Plasmids metabolism, Polymerase Chain Reaction, Sequence Homology, Amino Acid, Sirolimus pharmacology, TOR Serine-Threonine Kinases, Time Factors, Carrier Proteins, Genes, Tumor Suppressor, Phosphotransferases (Alcohol Group Acceptor) genetics, Plasmacytoma genetics
- Abstract
Susceptibility to mouse plasmacytomagenesis is a complex genetic trait controlled by several Pctr loci (Pctr1, Pctr2, etc). Congenic strain analysis narrowed the genetic interval surrounding the Pctr2 locus, and genes identified in the interval were sequenced from susceptible BALB/c and resistant DBA/2 mice. Frap (FKBP12 rapamycin-associated protein, mTOR, RAFT) was the only gene differing in amino acid sequence between alleles that correlated with strain sensitivity to tumor development. The in vitro kinase activity of the BALB/c FRAP allele was lower than the DBA/2 allele; phosphorylation of p53 and PHAS1/4EBP1 (properties of heat and acid stability/eukaryotic initiation factor 4E-binding protein) and autophosphorylation of FRAP were less efficient with the BALB/c allele. FRAP also suppressed transformation of NIH 3T3 cells by ras, with DBA/2 FRAP being more efficient than BALB/c FRAP. Rapamycin, a specific inhibitor of FRAP, did not inhibit growth of plasmacytoma cell lines. These studies identify Frap as a candidate tumor suppressor gene, in contrast to many reports that have focused on its prooncogenic properties. Frap may be similar to Tgfb and E2f in exerting both positive and negative growth-regulatory signals, depending on the timing, pathway, or tumor system involved. The failure of rapamycin to inhibit plasma cell tumor growth suggests that FRAP antagonists may not be appropriate for the treatment of plasma cell tumors. Pctr2 joins Pctr1 in possessing alleles that modify susceptibility to plasmacytomagenesis by encoding differences in efficiency of function (efficiency alleles), rather than all-or-none, gain-of-function, or loss-of-function alleles. By analogy, human cancer may also result from the combined effects of several inefficient alleles.
- Published
- 2003
- Full Text
- View/download PDF
37. The nature and identification of quantitative trait loci: a community's view.
- Author
-
Abiola O, Angel JM, Avner P, Bachmanov AA, Belknap JK, Bennett B, Blankenhorn EP, Blizard DA, Bolivar V, Brockmann GA, Buck KJ, Bureau JF, Casley WL, Chesler EJ, Cheverud JM, Churchill GA, Cook M, Crabbe JC, Crusio WE, Darvasi A, de Haan G, Dermant P, Doerge RW, Elliot RW, Farber CR, Flaherty L, Flint J, Gershenfeld H, Gibson JP, Gu J, Gu W, Himmelbauer H, Hitzemann R, Hsu HC, Hunter K, Iraqi FF, Jansen RC, Johnson TE, Jones BC, Kempermann G, Lammert F, Lu L, Manly KF, Matthews DB, Medrano JF, Mehrabian M, Mittlemann G, Mock BA, Mogil JS, Montagutelli X, Morahan G, Mountz JD, Nagase H, Nowakowski RS, O'Hara BF, Osadchuk AV, Paigen B, Palmer AA, Peirce JL, Pomp D, Rosemann M, Rosen GD, Schalkwyk LC, Seltzer Z, Settle S, Shimomura K, Shou S, Sikela JM, Siracusa LD, Spearow JL, Teuscher C, Threadgill DW, Toth LA, Toye AA, Vadasz C, Van Zant G, Wakeland E, Williams RW, Zhang HG, and Zou F
- Subjects
- Animals, Animals, Genetically Modified, Humans, Chromosome Mapping standards, Quantitative Trait Loci
- Abstract
This white paper by eighty members of the Complex Trait Consortium presents a community's view on the approaches and statistical analyses that are needed for the identification of genetic loci that determine quantitative traits. Quantitative trait loci (QTLs) can be identified in several ways, but is there a definitive test of whether a candidate locus actually corresponds to a specific QTL?
- Published
- 2003
- Full Text
- View/download PDF
38. p16 INK4a gene promoter variation and differential binding of a repressor, the ras-responsive zinc-finger transcription factor, RREB.
- Author
-
Zhang S, Qian X, Redman C, Bliskovski V, Ramsay ES, Lowy DR, and Mock BA
- Subjects
- 3T3 Cells, Alleles, Animals, Animals, Congenic, B-Lymphocytes metabolism, Binding Sites, Consensus Sequence, DNA Mutational Analysis, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Genes, Reporter, Genetic Predisposition to Disease, Genetic Variation, Luciferases biosynthesis, Luciferases genetics, MAP Kinase Kinase 1, Mice, Mitogen-Activated Protein Kinase Kinases physiology, Plasmacytoma genetics, Protein Serine-Threonine Kinases physiology, RNA, Messenger biosynthesis, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins genetics, Species Specificity, Spleen metabolism, Terpenes pharmacology, Transfection, ras Proteins physiology, DNA-Binding Proteins physiology, Genes, p16, Mice, Inbred BALB C genetics, Mice, Inbred DBA genetics, Promoter Regions, Genetic genetics, Transcription Factors physiology
- Abstract
BALB/c mice are susceptible to the development of pristane-induced plasma cell tumors, and have a rare allelic variant in the coding region of the p16(INK4a) (p16) tumor suppressor gene that produces a protein with impaired activity. We have now found that the BALB/c p16 promoter has an allelic variant that may also compromise p16 activity. Following pristane treatment, BALB/c p16 mRNA levels in B cells were lower than that in DBA/2 or C.D2-Pctr1, a resistant BALB/c congenic strain that harbors DBA/2 chromatin surrounding the p16 locus. Four sequence variants were found between BALB/c and DBA/2 in the p16 promoter region. In reporter assays, the DBA promoter was at least four times more active in driving luciferase expression than the BALB/c promoter. Most of the difference in activity was localized to a single nucleotide deletion in BALB/c. This deletion created a consensus binding site for RREB, a ras-responsive transcriptional element with zinc-finger binding motifs. Transient transfections with RREB confirmed that the p16 promoter can be downregulated by RREB, in a Ras- or Mek-dependent manner, and that the BALB/c promoter is more sensitive than DBA/2 to regulation by RREB. BALB/c mice have both regulatory and coding region defects that may contribute to the impairment of p16 gene function.
- Published
- 2003
- Full Text
- View/download PDF
39. Might wasp venom desensitization induced Th2 to Th1 shift cause pregnancy failure?
- Author
-
Markert UR, Arck PC, Peiker G, and Mock BA
- Subjects
- Adult, Animals, Contraindications, Decidua immunology, Female, Humans, Killer Cells, Natural immunology, Models, Immunological, Placenta immunology, Pregnancy, Th1 Cells immunology, Th2 Cells immunology, Wasp Venoms administration & dosage, Abortion, Spontaneous etiology, Abortion, Spontaneous immunology, Desensitization, Immunologic adverse effects, Wasp Venoms adverse effects
- Abstract
The case of a 28-year-old woman under wasp venom desensitization having a premature birth in her 24th week of pregnancy 16 days after the last injection is described. To test the hypothesis that a special profile of immune cells in the decidua may trigger abortions, placental and decidual tissue sections were stained with antibodies against T cells (CD3), cytotoxic cells (CD8), natural killer cells (CD56), and mast cells, and an in-situ-hybridization was performed for tumor necrosis factor-alpha (TNF-alpha). CD56+ Natural killer cells were the dominating population. In earlier analyses of healthy first trimester decidua the percentage of NK cells and T cells was in a similar range, but the CD8:CD3 ratio was only 2.2% in contrast to 27% in the present case. Mast cells, which are known to be able to secrete abortogenic TNF-alpha, were only detectable in the decidua (10 cells/mm2) and decidua sections were TNF-alpha positive. Since SIT induces a shift of the interleukin and functional profile from a Th2 type towards a Th1 type, and pregnancy is dependent on a Th2 pronounced profile, SIT may trigger abortions or immature births. This is supported by the present results and might have happened in this case.
- Published
- 2002
- Full Text
- View/download PDF
40. Efficiency alleles of the Pctr1 modifier locus for plasmacytoma susceptibility.
- Author
-
Zhang SL, DuBois W, Ramsay ES, Bliskovski V, Morse HC 3rd, Taddesse-Heath L, Vass WC, DePinho RA, and Mock BA
- Subjects
- 3T3 Cells, Alleles, Animals, Carrier Proteins genetics, Cell Division, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Chromosome Mapping, Cyclin-Dependent Kinase Inhibitor p16, Flow Cytometry, G1 Phase, Genes, ras genetics, Genetic Variation genetics, Histocytochemistry, Mice, Mice, Congenic, Mice, Inbred BALB C, Mice, Inbred DBA, Mice, Knockout, Plasmacytoma pathology, Proteins genetics, Tumor Stem Cell Assay, Tumor Suppressor Protein p14ARF, Cell Transformation, Neoplastic chemically induced, Genes, p16 genetics, Genetic Predisposition to Disease genetics, Plasmacytoma chemically induced, Plasmacytoma genetics, Terpenes pharmacology
- Abstract
The susceptibility of BALB/c mice to pristane-induced plasmacytomas is a complex genetic trait involving multiple loci, while DBA/2 and C57BL/6 strains are genetically resistant to the plasmacytomagenic effects of pristane. In this model system for human B-cell neoplasia, one of the BALB/c susceptibility and modifier loci, Pctr1, was mapped to a 5.7-centimorgan (cM) chromosomal region that included Cdkn2a, which encodes p16(INK4a) and p19(ARF), and the coding sequences for the BALB/c p16(INK4a) and p19(ARF) alleles were found to be polymorphic with respect to their resistant Pctr1 counterparts in DBA/2 and C57BL/6 mice (45). In the present study, alleles of Pctr1, Cdkn2a, and D4Mit15 from a resistant strain (BALB/cDAG) carrying DBA/2 chromatin were introgressively backcrossed to the susceptible BALB/c strain. The resultant C.DAG-Pctr1 Cdkn2a D4Mit15 congenic was more resistant to plasmacytomagenesis than BALB/c, thus narrowing Pctr1 to a 1.5-cM interval. Concomitantly, resistant C57BL/6 mice, from which both gene products of the Cdkn2a gene have been eliminated, developed pristane-induced plasma cell tumors over a shorter latency period than the traditionally susceptible BALB/cAn strain. Biological assays of the p16(INK4a) and p19(ARF) alleles from BALB/c and DBA/2 indicated that the BALB/c p16(INK4a) allele was less active than its DBA/2 counterpart in inducing growth arrest of mouse plasmacytoma cell lines and preventing ras-induced transformation of NIH 3T3 cells, while the two p19(ARF) alleles displayed similar potencies in both assays. We propose that the BALB/c susceptibility/modifier locus, Pctr1, is an "efficiency" allele of the p16(INK4a) gene.
- Published
- 2001
- Full Text
- View/download PDF
41. [Specific immunotherapy (hyposensitization) with insect venom in pregnancy].
- Author
-
Mock BA, Peiker G, Markert UR, and Jäger L
- Subjects
- Adult, Animals, Contraindications, Dose-Response Relationship, Drug, Female, Humans, Infant, Newborn, Male, Pregnancy, Risk Factors, Wasp Venoms administration & dosage, Wasp Venoms immunology, Desensitization, Immunologic, Obstetric Labor, Premature chemically induced, Wasp Venoms adverse effects, Wasps immunology
- Abstract
Background: Pregnancy typically prohibits the specific immunotherapy (SIT) of various allergic conditions, with the exception of pre-existing Hymenoptera venom allergies. International consensus currently recommends the continuation of a well-tolerated SIT with insect venom during pregnancy, since there is a significant risk of anaphylaxis after insect stings with potentially dismal outcomes for mother and fetus., Case Report: We report on a 28-year old woman, becoming pregnant during specific immunotherapy with Hymenoptera venom. SIT was continued during pregnancy and a premature birth occurred at the 24th week., Discussion and Conclusion: Unfortunately, there are still conflicting opinions in Germany regarding SIT during pregnancy, and the decision to perform such therapy is entirely based on knowledge and/or level of comfort of the primary physician. Thus, obstetricians should closely work together with an allergologist in cases of pregnant women with insect sting allergies.
- Published
- 2000
- Full Text
- View/download PDF
42. Structure and localization of mouse Pmscl1 and Pmscl2 genes.
- Author
-
Bliskovski V, Liddell R, Ramsay ES, Miller MJ, and Mock BA
- Subjects
- 5' Untranslated Regions, Amino Acid Sequence, Amino Acid Substitution, Animals, Autoantigens chemistry, Base Sequence, DNA, Complementary, Exoribonucleases, Exosome Multienzyme Ribonuclease Complex, Humans, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Nuclear Proteins chemistry, Sequence Homology, Amino Acid, Autoantigens genetics, Nuclear Proteins genetics
- Abstract
Sera from some patients with polymyositis-scleoderma overlap syndrome (PM-SCL) recognize two antigenically unrelated proteins, PMSCL1 and PMSCL2. Complete mouse Pmscl1 and Pmscl2 cDNA sequences, chromosomal localizations, exon/intron structure, and promoter region sequences of the mouse Pmscl2 gene are presented. The PMSCL1 gene was found to overlap significantly with cyclin A2 in both human and mouse. As such, it may be deduced that PMSCL1 sequences map to human chromosome 4q27 and the proximal portion of mouse chromosome (Chr) 3 where human and mouse cyclin A2 genes reside. Analysis of human and mouse PMSCL1 cDNA sequences provides evidence that the PMSCL1 protein is 68 amino acids longer than previously thought. A BAC containing mouse Pmscl2 was localized to distal mouse Chr 4 by FISH. This BAC contains the microsatellite D4Mit310. D4Mit310 colocalizes with a number of genes that map to human 1p36. In fact, a STS (G25404) located 54.6 cR from the top of human chromosome 1 was found to contain PMSCL2 sequence upon BLAST search., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
43. Mapping of the SWAP70 gene to mouse chromosome 7 and human chromosome 11p15.
- Author
-
Masat L, Liddell RA, Mock BA, Kuo WL, Jessberger R, Wabl M, and Morse HC 3rd
- Subjects
- Animals, Haplotypes, Humans, In Situ Hybridization, Fluorescence, Mice, Mice, Inbred BALB C, Microsatellite Repeats, Minor Histocompatibility Antigens, Polymorphism, Restriction Fragment Length, Chromosomes, Human, Pair 11 genetics, DNA-Binding Proteins genetics, Guanine Nucleotide Exchange Factors, Nuclear Proteins genetics, Physical Chromosome Mapping
- Abstract
The protein SWAP-70 was isolated as part of a DNA recombination complex in B lymphocytes, where it is predominantly expressed. In resting B cells, SWAP-70 is found in the cytoplasm; upon B-cell activation, it is transported both into the nucleus and to the cell membrane, where it is associated with the B-cell receptor complex and may play a role in signal transduction. In the nucleus, its involvement in heavy-chain class switch recombination has been suggested. In this report, using restriction fragment length polymorphism, simple sequence length polymorphism, and fluorescence in situ hybridization, we map the chromosomal localization of the mouse and the human genes to syntenic regions of mouse mid Chromosome (Chr) 7 and human Chr 11p15.
- Published
- 2000
- Full Text
- View/download PDF
44. Mouse chromosome 4.
- Author
-
Liddell RA, Hirano MC, and Mock BA
- Subjects
- Animals, Chromosome Mapping, Genetic Linkage, Genetic Markers, Mice, Physical Chromosome Mapping, Chromosomes genetics
- Published
- 1999
- Full Text
- View/download PDF
45. The role of p16INK4a (Cdkn2a) in mouse plasma cell tumors.
- Author
-
Zhang S and Mock BA
- Subjects
- Alleles, Animals, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Genetic Variation, Methylation, Mice, Mice, Inbred BALB C, Mice, Inbred DBA, Mutation, Plasmacytoma metabolism, Genes, p16, Plasmacytoma genetics
- Published
- 1999
- Full Text
- View/download PDF
46. Cdkn2a, the cyclin-dependent kinase inhibitor encoding p16INK4a and p19ARF, is a candidate for the plasmacytoma susceptibility locus, Pctr1.
- Author
-
Zhang S, Ramsay ES, and Mock BA
- Subjects
- Amino Acid Sequence, Animals, Ankyrins chemistry, Ankyrins genetics, Carrier Proteins biosynthesis, Carrier Proteins chemistry, Chromosome Mapping, Crosses, Genetic, Cyclin-Dependent Kinase Inhibitor p15, Cyclin-Dependent Kinase Inhibitor p16 chemistry, Female, Genes, Tumor Suppressor, Genetic Predisposition to Disease, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Inbred DBA, Molecular Sequence Data, Muridae, Polymerase Chain Reaction, Protein Biosynthesis, Proteins chemistry, Rats, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins chemistry, Repetitive Sequences, Nucleic Acid, Species Specificity, Tumor Suppressor Protein p14ARF, Carrier Proteins genetics, Cell Cycle Proteins, Cyclin-Dependent Kinase Inhibitor p16 biosynthesis, Genes, p16, Plasmacytoma genetics, Point Mutation, Proteins genetics, Tumor Suppressor Proteins
- Abstract
Plasma cell tumor induction in mice by pristane is under multigenic control. BALB/c mice are susceptible to tumor development; whereas DBA/2 mice are resistant. Restriction fragment length polymorphisms between BALB/c and DBA/2 for Cdkn2a(p16) and Cdkn2b(p15), and between BALB/c and Mus spretus for Cdkn2c(p18(INK4c)) were used to position these loci with respect to the Pctr1 locus. These cyclin-dependent kinase (CDK) inhibitors mapped to a 6 cM interval of chromosome 4 between Ifna and Tal1. C.D2-Chr 4 congenic strains harboring DBA/2 alleles associated with the Pctr1 locus contained DBA/2 "resistant" alleles of the CDK4/CDK6 inhibitors p16 and p15. On sequencing p16 and p18 cDNAs, two different allelic variants within ankyrin repeat regions of p16 were found between BALB/c and DBA/2 mice. By using an assay involving PCR amplification and restriction enzyme digestion, allelic variants were typed among several inbred strains of mice. One of the variants, G232A, was specific to two inbred strains, BALB/cAn and ABP/Le, of mice and occurred in a highly conserved amino acid in both human and rat p16. When tested with wild-type (DBA/2) p16, both A134C and G232A BALB/c-specific variants of p16 were inefficient in their ability to inhibit the activity of cyclin D2/CDK4 in kinase assays with retinoblastoma protein, suggesting this defective, inherited allele plays an important role in the genetic susceptibility of BALB/c mice for plasmacytoma induction and that p16(INK4a) is a strong candidate for the Pctr1 locus.
- Published
- 1998
- Full Text
- View/download PDF
47. Gene organization and chromosome location of the neural-specific RNA binding protein Elavl4.
- Author
-
Inman MV, Levy S, Mock BA, and Owens GC
- Subjects
- Aging, Alternative Splicing, Animals, Base Sequence, Brain growth & development, Drosophila genetics, ELAV Proteins, ELAV-Like Protein 4, Exons, Gene Expression Regulation, Developmental, Genetic Markers, Genetic Variation, Introns, Mice, Neurologic Mutants, Molecular Sequence Data, Oligodeoxyribonucleotides, Protein Structure, Secondary, RNA, Messenger biosynthesis, RNA-Binding Proteins biosynthesis, Restriction Mapping, Transcription, Genetic, Brain metabolism, Chromosome Mapping, Drosophila Proteins, Mice genetics, Nerve Tissue Proteins, RNA-Binding Proteins genetics
- Abstract
We have isolated the gene that encodes the neural-specific RNA binding protein HuD in the mouse (Elavl4), and have mapped its location to the mid-distal region of chromosome 4, close to the neurological mutant clasper. The coding region of the Elavl4 gene covers approximately 44 kb; the first two RNA binding domains (RBDs) that are homologous to the two RBDs found in the Drosophila sex-lethal gene are each encoded in two exons, whereas the third RBD is encoded in a single exon. Elavl4 mRNAs are alternatively spliced in the region between RBDs 2 and 3 due to the variable use of two micro-exons, and RNase protection analysis indicates that two of four possible splice variants are the predominant isoforms expressed in the central nervous system. The high degree of sequence conservation between the Hu proteins suggests that the exon organization of all the Hu protein genes will be similar, if not identical, to the Elavl4 gene.
- Published
- 1998
- Full Text
- View/download PDF
48. Encyclopedia of the mouse genome VII. Mouse chromosome 4.
- Author
-
Mock BA and Hirano MC
- Subjects
- Animals, Chromosomes genetics, Genome, Chromosome Mapping, Mice genetics
- Published
- 1998
- Full Text
- View/download PDF
49. The plasmacytoma resistance gene, Pctr2, delays the onset of tumorigenesis and resides in the telomeric region of chromosome 4.
- Author
-
Mock BA, Hartley J, Le Tissier P, Wax JS, and Potter M
- Subjects
- Animals, Chromosomes, Human, Pair 1, Humans, Mice, Mice, Inbred BALB C, Mice, Inbred DBA, Alleles, Chromosome Mapping, Genetic Predisposition to Disease, Plasmacytoma genetics
- Abstract
Mouse plasmacytomas share pathogenetic features in common with both multiple myeloma and Burkitt's lymphoma in humans. Susceptibility to plasmacytoma induction by intraperitoneal pristane in mice is controlled by multiple genes. At least two of these genes reside on mouse chromosome 4 in regions of the genome sharing linkage homology with human chromosomes 9p21, 1p32, and 1p36. A series of congenic strains recombinant for regions of mouse chromosome 4 in the vicinity of the Pctr2 predisposition locus were created and typed for their tumor susceptibility/resistance phenotypes. These strains were derived by introgressively backcrossing alleles from resistant DBA/2 mice onto the susceptible BALB/cAnPt background. Six resistant and two susceptible strains were allelotyped for 10 genes and 49 random DNA markers to identify the smallest region of overlap in the resistant strains. These studies have determined that the Pctr2 locus resides in either a 500-kb interval proximal to Nppa, or in a 1- to 2-centiMorgan (cM) interval distal to Nppa. In these congenic strain analyses, the Nppa and Fv1 loci, in addition to genes within about 1 cM of these loci, have been excluded as candidates for the Pctr2 locus. A relevant locus that may reside in this interval is Rep2; it is associated with the efficiency of repairing X-ray induced DNA damage sustained during the G2 phase of the mitotic cycle. The Pctr2 locus acts in a codominant fashion. F1 hybrids between resistant and susceptible congenic strains exhibit a reduced tumor incidence and a significant delay in the onset of tumorigenesis. Identification and eventual cloning of the Pctr2 locus may assist in the identification of genes involved in many types of cancer showing aberrations in human chromosome 1p36.
- Published
- 1997
50. Mouse chromosome 4.
- Author
-
Mock BA, Neumann PE, and Fiedorek FT Jr
- Subjects
- Animals, Chromosomes genetics, Genetic Markers genetics, Chromosome Mapping, Mice genetics
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.