1
|
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, Mock BA. Drug combinations identified by high-throughput screening promote cell cycle transition and upregulate Smad pathways in myeloma. Cancer Lett 2023; 568:216284. [PMID: 37356470 PMCID: PMC10408729 DOI: 10.1016/j.canlet.2023.216284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/27/2023]
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.
Collapse
Affiliation(s)
- Tyler J Peat
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.
| | - Snehal M Gaikwad
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wendy Dubois
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Nana Gyabaah-Kessie
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shuling Zhang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sayeh Gorjifard
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; University of Washington, Seattle, WA, USA
| | - Zaw Phyo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Johns Hopkins University, Baltimore, MD, USA
| | - Megan Andres
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Johns Hopkins University, Baltimore, MD, USA
| | - V Keith Hughitt
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - R Mark Simpson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Margaret A Miller
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | | | | | | | | | - Yong Zhang
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | | | - Evan Flietner
- McArdle Research Labs, University of Wisconsin, Madison, WI, USA
| | - Kelli Wilson
- Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Xiaohu Zhang
- Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Paul Shinn
- Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Carleen Klumpp-Thomas
- Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Crystal McKnight
- Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Zina Itkin
- Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Lu Chen
- Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Dickran Kazandijian
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Jing Zhang
- McArdle Research Labs, University of Wisconsin, Madison, WI, USA
| | - Aleksandra M Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Jonathan Keats
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Craig J Thomas
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Chemical Genomics Center, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, USA
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
2
|
Shinton SA, Brill-Dashoff J, Hayakawa K. Pla2g2a promotes innate Th2-type immunity lymphocytes to increase B1a cells. Sci Rep 2022; 12:14899. [PMID: 36050343 PMCID: PMC9437038 DOI: 10.1038/s41598-022-18876-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Newborns require early generation of effective innate immunity as a primary physiological mechanism for survival. The neonatal Lin28+Let7– developmental pathway allows increased generation of Th2-type cells and B1a (B-1 B) cells compared to adult cells and long-term maintenance of these initially generated innate cells. For initial B1a cell growth from the neonatal to adult stage, Th2-type IL-5 production from ILC2s and NKT2 cells is important to increase B1a cells. The Th17 increase is dependent on extracellular bacteria, and increased bacteria leads to lower Th2-type generation. Secreted group IIA-phospholipase A2 (sPLA2-IIA) from the Pla2g2a gene can bind to gram-positive bacteria and degrade bacterial membranes, controlling microbiota in the intestine. BALB/c mice are Pla2g2a+, and express high numbers of Th2-type cells and B1a cells. C57BL/6 mice are Pla2g2a-deficient and distinct from the SLAM family, and exhibit fewer NKT2 cells and fewer B1a cells from the neonatal to adult stage. We found that loss of Pla2g2a in the BALB/c background decreased IL-5 from Th2-type ILC2s and NKT2s but increased bacterial-reactive NKT17 cells and MAIT cells, and decreased the number of early-generated B1a cells and MZ B cells and the CD4/CD8 T cell ratio. Low IL-5 by decreased Th2-type cells in Pla2g2a loss led to low early-generated B1a cell growth from the neonatal to adult stage. In anti-thymocyte/Thy-1 autoreactive μκ transgenic (ATAμκ Tg) Pla2g2a+ BALB/c background C.B17 mice generated NKT2 cells that continuously control CD1d+ B1 B cells through old aging and lost CD1d in B1 B cells generating strong B1 ATA B cell leukemia/lymphoma. Pla2g2a-deficient ATAμκTg C57BL/6 mice suppressed the initial B1a cell increase, with low/negative spontaneous leukemia/lymphoma generation. These data confirmed that the presence of Pla2g2a to control bacteria is important to allow the neonatal to adult stage. Pla2g2a promotes innate Th2-type immunity lymphocytes to increase early generated B1a cells.
Collapse
Affiliation(s)
- Susan A Shinton
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
| | | | - Kyoko Hayakawa
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA.
| |
Collapse
|
3
|
Zhang S, Dubois W, Feng X, Nguyen JT, Young NS, Mock BA. Conditional deletion of mTOR discloses its essential role in early B-cell development. Mol Carcinog 2021; 61:408-416. [PMID: 34964999 DOI: 10.1002/mc.23386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 01/22/2023]
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.
Collapse
Affiliation(s)
- Shuling Zhang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Wendy Dubois
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute; National Institutes of Health, Bethesda, Maryland, USA
| | - Joe T Nguyen
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute; National Institutes of Health, Bethesda, Maryland, USA
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
4
|
Genetic modifiers regulating DNA replication and double-strand break repair are associated with differences in mammary tumors in mouse models of Li-Fraumeni syndrome. Oncogene 2021; 40:5026-5037. [PMID: 34183771 PMCID: PMC8349885 DOI: 10.1038/s41388-021-01892-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 05/16/2021] [Accepted: 06/04/2021] [Indexed: 01/04/2023]
Abstract
Breast cancer is the most common tumor among women with inherited variants in the TP53 tumor suppressor, but onset varies widely suggesting interactions with genetic or environmental factors. Rodent models haploinsufficent for Trp53 also develop a wide variety of malignancies associated with Li-Fraumeni Syndrome, but BALB/c mice are uniquely susceptible to mammary tumors and is genetically linked to the Suprmam1 locus on chromosome 7. To define mechanisms that interact with deficiencies in p53 to alter susceptibility to mammary tumors, we fine-mapped the Suprmam1 locus in females from an N2 backcross of BALB/cMed and C57BL/6J mice. A major modifier was localized within a 10 cM interval on chromosome 7. The effect of the locus on DNA damage responses was examined in the parental strains and mice that are congenic for C57BL/6J alleles on the BALB/cMed background (SM1-Trp53+/−). The mammary epithelium of C57BL/6J-Trp53+/− females exhibited little radiation-induced apoptosis compared to BALB/cMed-Trp53+/− and SM1-Trp53+/− females indicating that the Suprmam1B6/B6 alleles could not rescue repair of radiation-induced DNA double-strand breaks mostly relying on non-homologous end joining. In contrast, the Suprmam1B6/B6 alleles in SM1-Trp53+/− mice were sufficient to confer the C57BL/6J-Trp53+/− phenotypes in homology-directed repair and replication fork progression. The Suprmam1B6/B6 alleles in SM1-Trp53+/− mice appear to act in trans to regulate a panel of DNA repair and replication genes which lie outside the locus.
Collapse
|
5
|
Pisano M, Cheng Y, Sun F, Dhakal B, D’Souza A, Chhabra S, Knight JM, Rao S, Zhan F, Hari P, Janz S. Laboratory Mice - A Driving Force in Immunopathology and Immunotherapy Studies of Human Multiple Myeloma. Front Immunol 2021; 12:667054. [PMID: 34149703 PMCID: PMC8206561 DOI: 10.3389/fimmu.2021.667054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/28/2021] [Indexed: 11/13/2022] Open
Abstract
Mouse models of human cancer provide an important research tool for elucidating the natural history of neoplastic growth and developing new treatment and prevention approaches. This is particularly true for multiple myeloma (MM), a common and largely incurable neoplasm of post-germinal center, immunoglobulin-producing B lymphocytes, called plasma cells, that reside in the hematopoietic bone marrow (BM) and cause osteolytic lesions and kidney failure among other forms of end-organ damage. The most widely used mouse models used to aid drug and immunotherapy development rely on in vivo propagation of human myeloma cells in immunodeficient hosts (xenografting) or myeloma-like mouse plasma cells in immunocompetent hosts (autografting). Both strategies have made and continue to make valuable contributions to preclinical myeloma, including immune research, yet are ill-suited for studies on tumor development (oncogenesis). Genetically engineered mouse models (GEMMs), such as the widely known Vκ*MYC, may overcome this shortcoming because plasma cell tumors (PCTs) develop de novo (spontaneously) in a highly predictable fashion and accurately recapitulate many hallmarks of human myeloma. Moreover, PCTs arise in an intact organism able to mount a complete innate and adaptive immune response and tumor development reproduces the natural course of human myelomagenesis, beginning with monoclonal gammopathy of undetermined significance (MGUS), progressing to smoldering myeloma (SMM), and eventually transitioning to frank neoplasia. Here we review the utility of transplantation-based and transgenic mouse models of human MM for research on immunopathology and -therapy of plasma cell malignancies, discuss strengths and weaknesses of different experimental approaches, and outline opportunities for closing knowledge gaps, improving the outcome of patients with myeloma, and working towards a cure.
Collapse
Affiliation(s)
- Michael Pisano
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Yan Cheng
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Fumou Sun
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Binod Dhakal
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anita D’Souza
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Saurabh Chhabra
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jennifer M. Knight
- Departments of Psychiatry, Medicine, and Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sridhar Rao
- Division of Hematology, Oncology and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, United States
| | - Fenghuang Zhan
- Myeloma Center, Department of Internal Medicine and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Parameswaran Hari
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Siegfried Janz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
6
|
Zhang S, DuBois W, Zhang K, Simmons JK, Hughitt VK, Gorjifard S, Gaikwad S, Peat TJ, Mock BA. Mouse tumor susceptibility genes identify drug combinations for multiple myeloma. ACTA ACUST UNITED AC 2020; 6. [PMID: 32923678 PMCID: PMC7486007 DOI: 10.20517/2394-4722.2020.40] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
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.
Collapse
Affiliation(s)
- Shuling Zhang
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Wendy DuBois
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Ke Zhang
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - John K Simmons
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA.,Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | - V Keith Hughitt
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Sayeh Gorjifard
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA.,University of Washington School of Medicine, Department of Genome Sciences, Seattle, WA 98195, USA
| | - Snehal Gaikwad
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Tyler J Peat
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Godoy GJ, Paira DA, Olivera C, Breser ML, Sanchez LR, Motrich RD, Rivero VE. Differences in T regulatory cells between mouse strains frequently used in immunological research. Immunol Lett 2020; 223:17-25. [DOI: 10.1016/j.imlet.2020.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/25/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
|
8
|
Zhang S, Shi W, Ramsay ES, Bliskovsky V, Eiden AM, Connors D, Steinsaltz M, DuBois W, Mock BA. The transcription factor MZF1 differentially regulates murine Mtor promoter variants linked to tumor susceptibility. J Biol Chem 2019; 294:16756-16764. [PMID: 31548308 DOI: 10.1074/jbc.ra119.009779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/18/2019] [Indexed: 01/15/2023] Open
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.
Collapse
Affiliation(s)
- Shuling Zhang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Wei Shi
- Vaccine Research Center, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Edward S Ramsay
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Valery Bliskovsky
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Adrian Max Eiden
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Daniel Connors
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Matthew Steinsaltz
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Wendy DuBois
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
9
|
Scherger AK, Al-Maarri M, Maurer HC, Schick M, Maurer S, Öllinger R, Gonzalez-Menendez I, Martella M, Thaler M, Pechloff K, Steiger K, Sander S, Ruland J, Rad R, Quintanilla-Martinez L, Wunderlich FT, Rose-John S, Keller U. Activated gp130 signaling selectively targets B cell differentiation to induce mature lymphoma and plasmacytoma. JCI Insight 2019; 4:128435. [PMID: 31391340 DOI: 10.1172/jci.insight.128435] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/09/2019] [Indexed: 12/22/2022] Open
Abstract
Aberrant activity of the glycoprotein 130 130/JAK/STAT3 (gp130/JAK/STAT3) signaling axis is a recurrent event in inflammation and cancer. In particular, it is associated with a wide range of hematological malignancies, including multiple myeloma and leukemia. Novel targeted therapies have only been successful for some subtypes of these malignancies, underlining the need for developing robust mouse models to better dissect the role of this pathway in specific tumorigenic processes. Here, we investigated the role of selective gp130/JAK/STAT3 activation by generating a conditional mouse model. This model targeted constitutively active, cell-autonomous gp130 activity to B cells, as well as to the entire hematopoietic system. We found that regardless of the timing of activation in B cells, constitutively active gp130 signaling resulted in the formation specifically of mature B cell lymphomas and plasma cell disorders with full penetrance, only with different latencies, where infiltrating CD138+ cells were a dominant feature in every tumor. Furthermore, constitutively active gp130 signaling in all adult hematopoietic cells also led to the development specifically of largely mature, aggressive B cell cancers, again with a high penetrance of CD138+ tumors. Importantly, gp130 activity abrogated the differentiation block induced by a B cell-targeted Myc transgene and resulted in a complete penetrance of the gp130-associated, CD138+, mature B cell lymphoma phenotype. Thus, gp130 signaling selectively provides a strong growth and differentiation advantage for mature B cells and directs lymphomagenesis specifically toward terminally differentiated B cell cancers.
Collapse
Affiliation(s)
- Anna K Scherger
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Mona Al-Maarri
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Preventive Medicine and Diabetes, Cologne, Germany
| | | | - Markus Schick
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Sabine Maurer
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Rupert Öllinger
- Internal Medicine II.,Center for Translational Cancer Research, and.,Institute of Molecular Oncology and Functional Genomics, Technische Universität München, Munich, Germany
| | | | - Manuela Martella
- Institute of Pathology, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Markus Thaler
- Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany
| | - Konstanze Pechloff
- Center for Translational Cancer Research, and.,Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Katja Steiger
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany.,Institute of Pathology, Technische Universität München, Munich, Germany
| | - Sandrine Sander
- Adaptive Immunity and Lymphoma, German Cancer Research Center/National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Jürgen Ruland
- Center for Translational Cancer Research, and.,Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Roland Rad
- Internal Medicine II.,Center for Translational Cancer Research, and.,Institute of Molecular Oncology and Functional Genomics, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | | | - Frank T Wunderlich
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Preventive Medicine and Diabetes, Cologne, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Ulrich Keller
- Internal Medicine III, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany.,Department of Hematology, Oncology and Tumor Immunology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
10
|
Saito M, Okumura K, Isogai E, Araki K, Tanikawa C, Matsuda K, Kamijo T, Kominami R, Wakabayashi Y. A Polymorphic Variant in p19 Arf Confers Resistance to Chemically Induced Skin Tumors by Activating the p53 Pathway. J Invest Dermatol 2019; 139:1459-1469. [PMID: 30684556 DOI: 10.1016/j.jid.2018.12.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 12/14/2022]
Abstract
Identification of the specific genetic variants responsible for the increased susceptibility to familial or sporadic cancers is important. Using a forward genetics approach to map such loci in a mouse skin cancer model, we previously identified a strong genetic locus, Stmm3, conferring resistance to chemically induced skin papillomas on chromosome 4. Here, we report the cyclin-dependent kinase inhibitor gene Cdkn2a/p19Arf as a major responsible gene for the Stmm3 locus. We provide evidence that the function of Stmm3 is dependent on p53 and that p19ArfMSM confers stronger resistance to papillomas than p16Ink4aMSMin vivo. In addition, we found that genetic polymorphism in p19Arf between a resistant strain, MSM/Ms (Val), and a susceptible strain, FVB/N (Leu), alters the susceptibility to papilloma development, malignant conversion, and the epithelial-mesenchymal transition. Moreover, we demonstrated that the p19ArfMSM allele more efficiently activates the p53 pathway than the p19ArfFVB allele in vitro and in vivo. Furthermore, we found polymorphisms in CDKN2A in the vicinity of a polymorphism in mouse Cdkn2a associated with the risk of human cancers in the Japanese population. Genetic polymorphisms in Cdkn2a and CDKN2A may affect the cancer risk in both mice and humans.
Collapse
Affiliation(s)
- Megumi Saito
- Department of Carcinogenesis Research, Division of Experimental Animal Research, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Kazuhiro Okumura
- Department of Carcinogenesis Research, Division of Experimental Animal Research, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Eriko Isogai
- Department of Carcinogenesis Research, Division of Experimental Animal Research, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Kimi Araki
- Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto, Japan
| | - Chizu Tanikawa
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Koichi Matsuda
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan; Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Takehiko Kamijo
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Ryo Kominami
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
| | - Yuichi Wakabayashi
- Department of Carcinogenesis Research, Division of Experimental Animal Research, Chiba Cancer Center Research Institute, Chiba, Japan.
| |
Collapse
|
11
|
Hayakawa K, Formica AM, Nakao Y, Ichikawa D, Shinton SA, Brill-Dashoff J, Smith MR, Morse HC, Hardy RR. Early Generated B-1-Derived B Cells Have the Capacity To Progress To Become Mantle Cell Lymphoma-like Neoplasia in Aged Mice. THE JOURNAL OF IMMUNOLOGY 2018; 201:804-813. [PMID: 29898964 DOI: 10.4049/jimmunol.1800400] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/16/2018] [Indexed: 11/19/2022]
Abstract
In mice, fetal/neonatal B-1 cell development generates murine CD5+ B cells (B1a) with autoreactivity. We analyzed B1a cells at the neonatal stage in a VH11/D/JH knock-in mouse line (VH11t) that generates an autoreactive antiphosphatidylcholine BCR. Our study revealed that antiphosphatidylcholine B1a cells develop in liver, mature in spleen, and distribute in intestine/colon, mesenteric lymph node (mLN), and body cavity as the outcome of B-1 cell development before B-2 cell development. Throughout life, self-renewing B-1 B1a cells circulate through intestine, mesenteric vessel, and blood. The body cavity-deposited B1a cells also remigrate. In old age, some B1a cells proceed to monoclonal B cell lymphocytosis. When neonatal B-1 B1a cells express an antithymocyte/Thy-1 autoreactivity (ATA) BCR transgene in the C.B17 mouse background, ATA B cells increase in PBL and strongly develop lymphomas in aging mice that feature splenomegaly and mLN hyperplasia with heightened expression of CD11b, IL-10, and activated Stat3. At the adult stage, ATA B cells were normally present in the mantle zone area, including in intestine. Furthermore, frequent association with mLN hyperplasia suggests the influence by intestinal microenvironment on lymphoma development. When cyclin D1 was overexpressed by the Eμ-cyclin D1 transgene, ATA B cells progressed to further diffused lymphoma in aged mice, including in various lymph nodes with accumulation of IgMhiIgDloCD5+CD23-CD43+ cells, resembling aggressive human mantle cell lymphoma. Thus, our findings reveal that early generated B cells, as an outcome of B-1 cell development, can progress to become lymphocytosis, lymphoma, and mantle cell lymphoma-like neoplasia in aged mice.
Collapse
Affiliation(s)
| | | | - Yuka Nakao
- Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Daiju Ichikawa
- Fox Chase Cancer Center, Philadelphia, PA 19111.,Division of Clinical Physiology and Therapeutics, Keio University Faculty of Pharmacy, Tokyo 105-8512, Japan
| | | | | | - Mitchell R Smith
- Fox Chase Cancer Center, Philadelphia, PA 19111.,George Washington University Cancer Center, Washington, DC 20052; and
| | - Herbert C Morse
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | | |
Collapse
|
12
|
Lutful Kabir FM, Alvarez CE, Bird RC. Canine Mammary Carcinomas: A Comparative Analysis of Altered Gene Expression. Vet Sci 2015; 3:vetsci3010001. [PMID: 29056711 PMCID: PMC5644615 DOI: 10.3390/vetsci3010001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 11/19/2015] [Accepted: 12/21/2015] [Indexed: 12/19/2022] Open
Abstract
Breast cancer represents the second most frequent neoplasm in humans and sexually intact female dogs after lung and skin cancers, respectively. Many similar features in human and dog cancers including, spontaneous development, clinical presentation, tumor heterogeneity, disease progression and response to conventional therapies have supported development of this comparative model as an alternative to mice. The highly conserved similarities between canine and human genomes are also key to this comparative analysis, especially when compared to the murine genome. Studies with canine mammary tumor (CMT) models have shown a strong genetic correlation with their human counterparts, particularly in terms of altered expression profiles of cell cycle regulatory genes, tumor suppressor and oncogenes and also a large group of non-coding RNAs or microRNAs (miRNAs). Because CMTs are considered predictive intermediate models for human breast cancer, similarities in genetic alterations and cancer predisposition between humans and dogs have raised further interest. Many cancer-associated genetic defects critical to mammary tumor development and oncogenic determinants of metastasis have been reported and appear to be similar in both species. Comparative analysis of deregulated gene sets or cancer signaling pathways has shown that a significant proportion of orthologous genes are comparably up- or down-regulated in both human and dog breast tumors. Particularly, a group of cell cycle regulators called cyclin-dependent kinase inhibitors (CKIs) acting as potent tumor suppressors are frequently defective in CMTs. Interestingly, comparative analysis of coding sequences has also shown that these genes are highly conserved in mammals in terms of their evolutionary divergence from a common ancestor. Moreover, co-deletion and/or homozygous loss of the INK4A/ARF/INK4B (CDKN2A/B) locus, encoding three members of the CKI tumor suppressor gene families (p16/INK4A, p14ARF and p15/INK4B), in many human and dog cancers including mammary carcinomas, suggested their important conserved genetic order and localization in orthologous chromosomal regions. miRNAs, as powerful post-transcriptional regulators of most of the cancer-associated genes, have not been well evaluated to date in animal cancer models. Comprehensive expression profiles of miRNAs in CMTs have revealed their altered regulation showing a strong correlation with those found in human breast cancers. These genetic correlations between human and dog mammary cancers will greatly advance our understanding of regulatory mechanisms involving many critical cancer-associated genes that promote neoplasia and contribute to the promising development of future therapeutics.
Collapse
Affiliation(s)
- Farruk M Lutful Kabir
- Auburn University Research Initiative in Cancer (AURIC), Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL 36849, USA.
- Current address: Department of Pediatrics, Division of Pulmonology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Carlos E Alvarez
- Center for Molecular and Human Genetics, The Research Institute at Nationwide Children's Hospital Departments of Pediatrics and Veterinary Clinical Sciences, The Ohio State University Colleges of Medicine and Veterinary Medicine, Columbus, OH 43205, USA.
| | - R Curtis Bird
- Auburn University Research Initiative in Cancer (AURIC), Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL 36849, USA.
| |
Collapse
|
13
|
Rosean TR, Tompkins VS, Tricot G, Holman CJ, Olivier AK, Zhan F, Janz S. Preclinical validation of interleukin 6 as a therapeutic target in multiple myeloma. Immunol Res 2014; 59:188-202. [PMID: 24845460 PMCID: PMC4209159 DOI: 10.1007/s12026-014-8528-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Studies on the biologic and molecular genetic underpinnings of multiple myeloma (MM) have identified the pleiotropic, pro-inflammatory cytokine, interleukin-6 (IL-6), as a factor crucial to the growth, proliferation and survival of myeloma cells. IL-6 is also a potent stimulator of osteoclastogenesis and a sculptor of the tumor microenvironment in the bone marrow of patients with myeloma. This knowledge has engendered considerable interest in targeting IL-6 for therapeutic purposes, using a variety of antibody- and small-molecule-based therapies. However, despite the early recognition of the importance of IL-6 for myeloma and the steady progress in our knowledge of IL-6 in normal and malignant development of plasma cells, additional efforts will be required to translate the promise of IL-6 as a target for new myeloma therapies into significant clinical benefits for patients with myeloma. This review summarizes published research on the role of IL-6 in myeloma development and describes ongoing efforts by the University of Iowa Myeloma Multidisciplinary Oncology Group to develop new approaches to the design and testing of IL-6-targeted therapies and preventions of MM.
Collapse
Affiliation(s)
- Timothy R Rosean
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Simmons JK, Patel J, Michalowski A, Zhang S, Wei BR, Sullivan P, Gamache B, Felsenstein K, Kuehl WM, Simpson RM, Zingone A, Landgren O, Mock BA. TORC1 and class I HDAC inhibitors synergize to suppress mature B cell neoplasms. Mol Oncol 2013; 8:261-72. [PMID: 24429254 DOI: 10.1016/j.molonc.2013.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/20/2013] [Accepted: 11/21/2013] [Indexed: 12/12/2022] Open
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.
Collapse
Affiliation(s)
- John K Simmons
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Jyoti Patel
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Aleksandra Michalowski
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Shuling Zhang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Patrick Sullivan
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Ben Gamache
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Kenneth Felsenstein
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - W Michael Kuehl
- Genetics Branch, National Cancer Institute, National Institutes of Health, USA
| | - R Mark Simpson
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Adriana Zingone
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, USA
| | - Ola Landgren
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, USA
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA.
| |
Collapse
|
15
|
Hu Y, Zheng M, Gali R, Tian Z, Topal Görgün G, Munshi NC, Mitsiades CS, Anderson KC. A novel rapid-onset high-penetrance plasmacytoma mouse model driven by deregulation of cMYC cooperating with KRAS12V in BALB/c mice. Blood Cancer J 2013; 3:e156. [PMID: 24185503 PMCID: PMC3880436 DOI: 10.1038/bcj.2013.53] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 09/24/2013] [Indexed: 12/28/2022] Open
Abstract
Our goal is to develop a rapid and scalable system for functionally evaluating deregulated genes in multiple myeloma (MM). Here, we forcibly expressed human cMYC and KRAS12V in mouse T2 B cells (IgM(+)B220(+)CD38(+)IgD(+)) using retroviral transduction and transplanted these cells into lethally irradiated recipient mice. Recipients developed plasmacytomas with short onset (70 days) and high penetrance, whereas neither cMYC nor KRAS12V alone induced disease in recipient mice. Tumor cell morphology and cell surface biomarkers (CD138(+)B220(-)IgM(-)GFP(+)) indicate a plasma cell neoplasm. Gene set enrichment analysis further confirms that the tumor cells have a plasma cell gene expression signature. Plasmacytoma cells infiltrated multiple loci in the bone marrow, spleen and liver; secreted immunoglobulins; and caused glomerular damage. Our findings therefore demonstrate that deregulated expression of cMYC with KRAS12V in T2 B cells rapidly generates a plasma cell disease in mice, suggesting utility of this model both to elucidate molecular pathogenesis and to validate novel targeted therapies.
Collapse
Affiliation(s)
- Y Hu
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Waldenström macroglobulinemia: clinical and immunological aspects, natural history, cell of origin, and emerging mouse models. ISRN HEMATOLOGY 2013; 2013:815325. [PMID: 24106612 PMCID: PMC3782845 DOI: 10.1155/2013/815325] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 07/26/2013] [Indexed: 12/22/2022]
Abstract
Waldenström macroglobulinemia (WM) is a rare and currently incurable neoplasm of IgM-expressing B-lymphocytes that is characterized by the occurrence of a monoclonal IgM (mIgM) paraprotein in blood serum and the infiltration of the hematopoietic bone marrow with malignant lymphoplasmacytic cells. The symptoms of patients with WM can be attributed to the extent and tissue sites of tumor cell infiltration and the magnitude and immunological specificity of the paraprotein. WM presents fascinating clues on neoplastic B-cell development, including the recent discovery of a specific gain-of-function mutation in the MYD88 adapter protein. This not only provides an intriguing link to new findings that natural effector IgM+IgD+ memory B-cells are dependent on MYD88 signaling, but also supports the hypothesis that WM derives from primitive, innate-like B-cells, such as marginal zone and B1 B-cells. Following a brief review of the clinical aspects and natural history of WM, this review discusses the thorny issue of WM's cell of origin in greater depth. Also included are emerging, genetically engineered mouse models of human WM that may enhance our understanding of the biologic and genetic underpinnings of the disease and facilitate the design and testing of new approaches to treat and prevent WM more effectively.
Collapse
|
17
|
Jorgenson TC, Williams BR, Wendland A, Bilger A, Sandgren EP, Drinkwater NR. Identification of susceptibility loci in a mouse model of KRASG12D-driven pancreatic cancer. Cancer Res 2010; 70:8398-406. [PMID: 20959479 DOI: 10.1158/0008-5472.can-09-3980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Genetic background affects susceptibility to pancreatic ductal adenocarcinoma in the Ela-KRAS(G12D) mouse model. In this model, KRAS oncogene expression is driven by an elastase promoter in acinar cells of the pancreas on an FVB/NTac (FVB) background [FVB-Tg(Ela-KRAS(G12D))] with the transgene carried on the Y chromosome. Through linkage analysis of crosses between the C57BL/6J (B6), BALB/cJ (BALB), and DBA/2J (D2) inbred strains of mice and resistant FVB-Tg(Ela-KRAS(G12D)), we have identified six susceptibility loci that affect mean preinvasive lesion multiplicity. Markers on chromosome 2 segregated with high tumor multiplicity in all three strains; these loci were designated Prsq1-3 (pancreatic ras susceptibility quantitative trait loci 1-3; combined F2 and N2 LOD(W), 6.0, 4.1, and 2.7, respectively). Susceptibility loci on chromosome 4, designated Prsq4 and Prsq5, were identified in crosses between FVB transgenic mice and B6 or BALB mice (combined F2 and N2 LOD(W), 3.6 and 2.9, respectively). A marker on chromosome 12 segregated with tumor multiplicity in a BALB × FVB-Tg(Ela-KRAS(G12D)) cross and was designated Prsq6 (LOD(W), ∼2.5). B6-Chr Y(FVB-Tg(Ela-KRASG12D)) and BALB-Chr Y(FVB-Tg(Ela-KRASG12D)) consomics, which carry the KRAS transgene on the FVB Y chromosome on an otherwise inbred B6 or BALB background, developed ∼4-fold (B6) and ∼10-fold (BALB) more lesions than FVB-Tg(Ela-KRAS(G12D)) mice. By 12 months of age, 10% of BALB-Chr Y(FVB-Tg(Ela-KRASG12D)) mice developed invasive carcinomas. Our findings provide evidence that regions of chromosomes 2, 4, and 12 influence the development and progression of pancreatic neoplasms initiated by an oncogenic allele of KRAS in mice.
Collapse
Affiliation(s)
- Tonia C Jorgenson
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706, USA
| | | | | | | | | | | |
Collapse
|
18
|
Mori N. Two loci controlling susceptibility to radiation-induced lymphomagenesis on mouse chromosome 4: cdkn2a, a candidate for one locus, and a novel locus distinct from cdkn2a. Radiat Res 2010; 173:158-64. [PMID: 20095847 DOI: 10.1667/rr1855.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BALB/c mice are sensitive to radiation-induced lymphomagenesis, while STS mice are resistant. Using 219 [(BALB/c x STS)F(1) x BALB/c] (N2C) and 197 [(BALB/c x STS)F(1) x STS] (N2S) animals, we performed a genome-wide search for loci controlling susceptibility to lymphomagenesis induced by radiation. Association of markers with the survival of animals was analyzed by the log rank test. For N2C mice, a significant correlation was detected, with four markers on the proximal to mid portion of chromosome 4: D4Mit302 and D4Mit255, P = 0.0075; D4Mit17, P = 0.034; and D4Mit86, P = 0.048. On the other hand, no significant linkage was detected in N2S mice. We analyzed BALB/c mice congenic for the STS allele in different regions of chromosome 4 and identified a locus with a conspicuous effect on survival located within a 7-Mb region between D4Mit302 and D4Mit144, where BALB/c mice harbor hypomorphic variant alleles of the tumor suppressor gene Cdkn2a, which encodes the cyclin-dependent kinase inhibitor protein p16INK4a. Using pooled F(2) intercrosses between the BALB/c and congenic lines carrying the STS allele near D4Mit17, but not in the range from D4Mit302 to D4Mit144, we assigned the second locus to an 11.4-Mb region in the vicinity of D4Mit17. Although Cdkn2a is a likely candidate for the locus controlling susceptibility to lymphomagenesis on chromosome 4, a novel tumor susceptibility gene different from Cdkn2a exists near the primary locus.
Collapse
Affiliation(s)
- Nobuko Mori
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai-shi, Osaka 599-8570, Japan.
| |
Collapse
|
19
|
Mndal, a new interferon-inducible family member, is highly polymorphic, suppresses cell growth, and may modify plasmacytoma susceptibility. Blood 2009; 114:2952-60. [PMID: 19654412 DOI: 10.1182/blood-2009-01-198812] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
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.
Collapse
|
20
|
Abstract
Multiple myeloma (MM) remains incurable despite high-dose chemotherapy with stem cell support. There is need, therefore, for continuous efforts directed toward the development of novel rational-based therapeutics for MM, which requires a detailed knowledge of the mutations driving this malignancy. In improving the success rate of effective drug development, it is equally imperative that biologic systems be developed to better validate these target genes. Here we review the recent developments in the generation of mouse models of MM and their impact as preclinical models for designing and assessing target-based therapeutic approaches.
Collapse
|
21
|
Jawad M, Giotopoulos G, Cole C, Plumb M. Target cell frequency is a genetically determined risk factor in radiation leukaemogenesis. Br J Radiol 2007; 80 Spec No 1:S56-62. [PMID: 17704327 DOI: 10.1259/bjr/99530445] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Whole body exposure to ionizing radiation increases the risk of radiation-induced acute myeloid leukaemia (r-AML). r-AML is the result of the accumulation of mutations in a single haemopoietic stem cell, so risk is therefore a function of the number of mutations required to transform the stem cell and the mutation rate. There is a genetic component to the risk of AML within the general population, and low penetrance variant alleles encoding DNA repair enzymes have been genetically implicated in therapy-related AML susceptibility. However, what is largely ignored is that target cell number, which defines the number of genomes at risk from DNA damaging agents, is also part of the equation that defines risk. We will review the evidence from genetic studies of inbred mouse models that target cell frequency is a risk factor in radiation leukaemogenesis. Inbred mouse strains that differ in their susceptibility to radiation-induced r-AML and thymic lymphoma (r-TL), spontaneous TL and pristane-induced plasmacytoma (PCT) have been exploited to identify susceptibility loci. The target cell in AML is the haemopoietic stem cell, whereas TLs and PCT arise from more mature lymphoid progenitor cells. Inbred mice also differ significantly in all aspects of haemopoiesis, and these differences have been used to identify quantitative trait loci (QTL) that determine the frequency of specific haemopoietic stem, progenitor or mature blood cells. The co-localization of QTL that determine risk and target cell frequency in all three haemopoietic malignancies is strong evidence that target cell frequency is a risk factor in radiation leukaemogenesis.
Collapse
Affiliation(s)
- M Jawad
- Department of Genetics, University of Leicester, University Road, Leicester LE1 7RH, UK
| | | | | | | |
Collapse
|
22
|
Dib A, Barlogie B, Shaughnessy JD, Kuehl WM. Methylation and expression of the p16INK4A tumor suppressor gene in multiple myeloma. Blood 2007; 109:1337-8. [PMID: 17244692 DOI: 10.1182/blood-2006-09-049510] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
23
|
Dib A, Peterson TR, Raducha-Grace L, Zingone A, Zhan F, Hanamura I, Barlogie B, Shaughnessy J, Kuehl WM. Paradoxical expression of INK4c in proliferative multiple myeloma tumors: bi-allelic deletion vs increased expression. Cell Div 2006; 1:23. [PMID: 17049078 PMCID: PMC1634742 DOI: 10.1186/1747-1028-1-23] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Accepted: 10/18/2006] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND A high proliferative capacity of tumor cells usually is associated with shortened patient survival. Disruption of the RB pathway, which is critically involved in regulating the G1 to S cell cycle transition, is a frequent target of oncogenic events that are thought to contribute to increased proliferation during tumor progression. Previously, we determined that p18INK4c, an essential gene for normal plasma cell differentiation, was bi-allelically deleted in five of sixteen multiple myeloma (MM) cell lines. The present study was undertaken to investigate a possible role of p18INK4c in increased proliferation of myeloma tumors as they progress. RESULTS Thirteen of 40 (33%) human myeloma cell lines do not express normal p18INK4c, with bi-allelic deletion of p18 in twelve, and expression of a mutated p18 fragment in one. Bi-allelic deletion of p18, which appears to be a late progression event, has a prevalence of about 2% in 261 multiple myeloma (MM) tumors, but the prevalence is 6 to 10% in the 50 tumors with a high expression-based proliferation index. Paradoxically, 24 of 40 (60%) MM cell lines, and 30 of 50 (60%) MM tumors with a high proliferation index express an increased level of p18 RNA compared to normal bone marrow plasma cells, whereas this occurs in only five of the 151 (3%) MM tumors with a low proliferation index. Tumor progression is often accompanied by increased p18 expression and an increased proliferation index. Retroviral-mediated expression of exogenous p18 results in marked growth inhibition in three MM cell lines that express little or no endogenous p18, but has no effect in another MM cell line that already expresses a high level of p18. CONCLUSION Paradoxically, although loss of p18 appears to contribute to increased proliferation of nearly 10% of MM tumors, most MM cell lines and proliferative MM tumors have increased expression of p18. Apart from a small fraction of cell lines and tumors that have inactivated the RB1 protein, it is not yet clear how other MM cell lines and tumors have become insensitive to the anti-proliferative effects of increased p18 expression.
Collapse
Affiliation(s)
- Amel Dib
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Naval Hospital, Bldg 8, Rm 5101, Bethesda, MD20889-5105, USA
| | - Timothy R Peterson
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Naval Hospital, Bldg 8, Rm 5101, Bethesda, MD20889-5105, USA
- Department of Biology, Masssachusetts Institutes of Technology, Whitehead Institute, 9 Cambridge Center, Rm 359, MA02142, USA
| | - Laura Raducha-Grace
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Naval Hospital, Bldg 8, Rm 5101, Bethesda, MD20889-5105, USA
- University of Pittsburgh School of Medicine, M218 Scaife Hall, Pittsburgh, PA15261, USA
| | - Adriana Zingone
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Naval Hospital, Bldg 8, Rm 5101, Bethesda, MD20889-5105, USA
| | - Fenghuang Zhan
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, 4301 Markham St., #816, Little Rock, AR72205-7199, USA
| | - Ichiro Hanamura
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, 4301 Markham St., #816, Little Rock, AR72205-7199, USA
- Department of Internal Medicine, Division of Hematology, Aichi Medical University, 21 Karimata, Yazako, Nagakute, Aichi-gun, Aichi 480-1195, Japan
| | - Bart Barlogie
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, 4301 Markham St., #816, Little Rock, AR72205-7199, USA
| | - John Shaughnessy
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, 4301 Markham St., #816, Little Rock, AR72205-7199, USA
| | - W Michael Kuehl
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Naval Hospital, Bldg 8, Rm 5101, Bethesda, MD20889-5105, USA
| |
Collapse
|
24
|
Abstract
Chromosomal translocations that join the cellular oncogene Myc (c-myc) with immunoglobulin (Ig) heavy-chain (Igh) or light-chain (Igk, Igl) loci are widely believed to be the crucial initiating oncogenic events in the development of B cell and plasma cell neoplasms in three mammalian species: Burkitt lymphoma (BL) in human beings, plasmacytoma (PCT) in mice, and immunocytoma in rats. Among the Myc-Ig translocations found in these neoplasms, mouse PCT T(12;15)(Igh-Myc) is of special interest because it affords a uniquely useful model system to study the fundamental outstanding questions on the mechanisms, genetics, and biological consequences of Myc translocations. Mouse T(12;15) is the direct counterpart of the human BL t(8;14)(q24;q32) translocation and thus of great relevance for human cancer. Mouse T(12;15) is the only cancer-associated translocation in mice that occurs with high incidence, spontaneity, and cell-type specificity. Due to the development of PCR methods for the detection of the underlying reciprocal Myc-Igh junction fragments, it is now known that mouse T(12;15) can be a dynamic process that begins with the genetic exchange of Myc and the Igh switch mu region (Smu), progresses by class switch recombination (CSR) just 3' of the translocation break site, and then undergoes further clonal diversification by micro-deletions in the junction flanks. The molecular pathway that subverts CSR to mediate trans-chromosomal joining of Myc and Smu (translocation origin) and secondary modification of Myc-Igh junctions (translocation "remodeling") has not been elucidated, but recent evidence indicates that it includes CSR factors, such as the activation-induced cytidine deaminase (AID), that may also be involved in the ongoing neoplastic progression of the translocation-bearing tumor precursor. Transgenic mouse models of T(12;15)/t(8;14), including newly developed "iMyc" gene-insertion mice, will be useful in elucidating the role of these CSR factors in the progression of Myc-induced B cell tumors.
Collapse
Affiliation(s)
- Siegfried Janz
- Laboratory of Genetics, Center for Cancer Research, National Cancer Institute, NIH, Building 37, Room 3140A, Bethesda, MD 20892-4256, USA.
| |
Collapse
|
25
|
Rosemann M, Kuosaite V, Kremer M, Favor J, Quintanilla-Martinez L, Atkinson MJ. Multilocus inheritance determines predisposition to α-radiation induced bone tumourigenesis in mice. Int J Cancer 2006; 118:2132-8. [PMID: 16331598 DOI: 10.1002/ijc.21612] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In a recent study, we presented evidence for genetic predisposition governing radiation osteosarcomagenesis in mice. Following the incorporation of the bone-seeking alpha emitter 227Th, approximately 25% of the variance in osteosarcoma incidence was determined by inherited genetic factors. We have now mapped 5 susceptibility loci in crosses between the more susceptible BALB/c and the more resistant CBA/Ca strains. The major QTL on chromosome 14 overlaps with a locus that was already found in our previous study, using different strains of mice. Here, we investigate the effect by which the major susceptibility locus and 4 minor modifier loci interact to influence osteosarcoma predisposition. Following incorporation of the bone-seeking isotope, 100% of mice that harbour high-risk genotypes at all 5 susceptibility loci develop osteosarcoma with an average of 472 days latency times. In 10 mice inheriting exclusively low-risk genotypes only 1 osteosarcoma was found, arising after 733 days latency time. Inheritance of distinct combinations of BALB/c and CBA/Ca alleles at the susceptibility loci confer more extreme phenotypes in terms of susceptibility or resistance than observed in either of the two parental inbred strains. From the present study, we demonstrate that additive effects of multiple alleles, each making only a minor phenotypic contribution, can combine and significantly alter tumour risk. This mechanism can be of particular importance in genetically heterogeneous populations such as man.
Collapse
Affiliation(s)
- Michael Rosemann
- Institute of Pathology, Clinical Cooperation Group Osteosarcoma, GSF National Research Center for Environment and Health, Neuherberg, Germany.
| | | | | | | | | | | |
Collapse
|
26
|
Fischer M, Schwieger M, Horn S, Niebuhr B, Ford A, Roscher S, Bergholz U, Greaves M, Löhler J, Stocking C. Defining the oncogenic function of the TEL/AML1 (ETV6/RUNX1) fusion protein in a mouse model. Oncogene 2005; 24:7579-91. [PMID: 16044150 DOI: 10.1038/sj.onc.1208931] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The t(12;21) translocation, generating the TEL/AML1 fusion protein, is the most common genetic lesion in childhood cancer. Using a bone marrow transplantation model, we demonstrate that TEL/AML1 expression impinges on normal hematopoietic differentiation, leading to the in vivo accumulation and persistence of an early progenitor compartment with a Sca1(+)/Kit(hi)/CD11b(+) phenotype and an increased self-renewal capacity, as documented by replating assays in vitro. Differentiation of these cells is not blocked, but the frequency of mature blood cells arising from TEL/AML1-transduced progenitors is low. Impaired differentiation is prominently observed in the pro-B-cell compartment, resulting in an proportional increase in early progenitors in vivo, consistent with the t(12;21) ALL phenotype. Despite the accumulation of both multipotent and B-cell progenitors in vivo, no leukemia induction was observed during an observation period of over 1 year. These results are consistent with findings in twins with concordant ALL, showing that TEL/AML1 generates a preleukemic clone in utero that persists for several years in a clinically covert fashion. Furthermore, our studies showed that the pointed domain of TEL/AML1, which recruits transcriptional repressors and directs oligomerization with either TEL/AML1 or wild-type TEL, was essential for the observed differentiation impairment and could not be replaced with another oligomerization domain.
Collapse
MESH Headings
- Animals
- B-Lymphocytes
- Bone Marrow Transplantation
- Cell Differentiation
- Cell Transformation, Neoplastic/genetics
- Chromosomes, Human, Pair 12
- Chromosomes, Human, Pair 21
- Core Binding Factor Alpha 2 Subunit/biosynthesis
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/physiology
- Hematopoietic Stem Cells
- Humans
- Mice
- Mice, Inbred C57BL
- Oncogene Proteins, Fusion/biosynthesis
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/physiology
- Phenotype
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Preleukemia/genetics
- Preleukemia/physiopathology
- Translocation, Genetic
Collapse
Affiliation(s)
- Meike Fischer
- Molecular Pathology Group, Heinrich-Pette-Institut für Experimentelle Immunologie und Virologie, D-20251 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Multiple myeloma, the second most common haematopoietic cancer, represents a collection of plasma-cell neoplasms that invariably become fatal when self-renewing myeloma cells begin unrestrained proliferation. Myeloma cells are arrested as intermediates in plasma-cell differentiation as a consequence of transformation. Unlike normal plasma cells, myeloma cells retain the self-renewing potential. Although impaired apoptosis accounts for the accumulation of myeloma cells in the bone marrow during the plateau phase of the disease, cell-cycle deregulation underlies unrestrained proliferation of self-renewing myeloma cells in aggressive myelomas and during relapse. The mechanism that governs deregulated cell-cycle re-entry and progression in multiple myeloma is unknown, and the relationship between myeloma cells and their normal counterparts is undefined. Plasma-cell differentiation is a complex multi-step process. This chapter will address recent advances in the mechanism of normal plasma-cell differentiation and our current understanding of the relationship between plasma-cell differentiation and myeloma pathogenesis.
Collapse
Affiliation(s)
- S Chen-Kiang
- Department of Pathology and Laboratory Medicine and Graduate Program in Immunology and Microbial Pathogenesis, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY 10021, USA.
| |
Collapse
|
28
|
Hill R, Song Y, Cardiff RD, Van Dyke T. Heterogeneous Tumor Evolution Initiated by Loss of pRb Function in a Preclinical Prostate Cancer Model. Cancer Res 2005; 65:10243-54. [PMID: 16288012 DOI: 10.1158/0008-5472.can-05-1579] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Because each change in the evolution of a cancer is predicated on the effects of previous events, a full understanding of selective changes and their effect on tumor progression can only be understood in the context of appropriate initiating events. Here, we define the effect of pRb function inactivation in prostate epithelium on both the initiation of prostate cancer and the establishment of selective pressures that lead to diminished Pten function and tumor evolution. Using genetically engineered mice, we show that inactivation of the pRb family proteins (Rb/p107/p130) induces epithelial proliferation and apoptosis and is sufficient to produce prostatic intraepithelial neoplasia (PIN) lesions. Over time, adenocarcinomas develop in all mice with no evidence of neuroendocrine tumors. Apoptosis is dependent on Pten function and not p53, unlike other epithelial cell types tested previously. Consequently, Pten hemizygosity reduces apoptosis by 50%, accelerating progression to adenocarcinomas with heterogeneous composition. Heterogeneity is associated with concurrent Pten haploinsufficiency and focal selective progression to complete Pten loss, which yields distinct tumor properties. Given that this analysis models the apparent timing of highly penetrant events in human prostate cancer, observed effects may recapitulate the natural evolution of prostate cancer development.
Collapse
Affiliation(s)
- Reginald Hill
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | | |
Collapse
|
29
|
Park SS, Shaffer AL, Kim JS, duBois W, Potter M, Staudt LM, Janz S. Insertion of Myc into Igh accelerates peritoneal plasmacytomas in mice. Cancer Res 2005; 65:7644-52. [PMID: 16140930 DOI: 10.1158/0008-5472.can-05-1222] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gene-targeted mice that contain a His6-tagged mouse c-Myc cDNA, Myc(His), inserted head to head into different sites of the mouse immunoglobulin heavy-chain locus, Igh, mimic the chromosomal T(12;15)(Igh-Myc) translocation that results in the activation of Myc in the great majority of mouse plasmacytomas. Mice carrying Myc(His) just 5' of the intronic heavy-chain enhancer Emu (strain iMyc(Emu)) provide a specific model of the type of T(12;15) found in a subset (approximately 20%) of plasmacytomas that develop "spontaneously" in the gut-associated lymphoid tissue (GALT) of interleukin-6 transgenic BALB/c (C) mice. Here we show that the transfer of the iMyc(Emu) transgene from a mixed genetic background of segregating C57BL/6 x 129/SvJ alleles to the background of C increased the incidence of GALT plasmacytomas by a factor of 2.5 in first-generation backcross mice (C.iMyc(Emu) N1). Third-generation backcross mice (C.iMyc(Emu) N3, approximately 94% C alleles) were hypersusceptible to inflammation-induced peritoneal plasmacytomas (tumor incidence, 100%; mean tumor onset, 86 +/- 28 days) compared with inbred C mice (tumor incidence, 5% on day 150 after tumor induction). Peritoneal plasmacytomas of C.iMyc(Emu) N3 mice overexpressed Myc(His), produced monoclonal immunoglobulin, and exhibited a unique plasma cell signature upon gene expression profiling on mouse Lymphochip cDNA microarrays. These findings indicated that the iMyc(Emu) transgene accelerates plasmacytoma development by collaborating with tumor susceptibility alleles of strain C and circumventing the requirement for tumor precursors to acquire deregulated Myc by chromosomal translocation.
Collapse
Affiliation(s)
- Sung Sup Park
- Laboratory of Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892-4256, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Feo F, De Miglio MR, Simile MM, Muroni MR, Calvisi DF, Frau M, Pascale RM. Hepatocellular carcinoma as a complex polygenic disease. Interpretive analysis of recent developments on genetic predisposition. Biochim Biophys Acta Rev Cancer 2005; 1765:126-47. [PMID: 16216419 DOI: 10.1016/j.bbcan.2005.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 08/25/2005] [Accepted: 08/26/2005] [Indexed: 01/11/2023]
Abstract
The different frequency of hepatocellular carcinoma (HCC) in humans at risk suggests a polygenic predisposition. However, detection of genetic variants is difficult in genetically heterogeneous human population. Studies on mouse and rat models identified 7 hepatocarcinogenesis susceptibility (Hcs) and 2 resistance (Hcr) loci in mice, and 7 Hcs and 9 Hcr loci in rats, controlling multiplicity and size of neoplastic liver lesions. Six liver neoplastic nodule remodeling (Lnnr) loci control number and volume of re-differentiating lesions in rat. A Hcs locus, with high phenotypic effects, and various epistatic gene-gene interactions were identified in rats, suggesting a genetic model of predisposition to hepatocarcinogenesis with different subset of low-penetrance genes, at play in different subsets of population, and a major locus. This model is in keeping with human HCC epidemiology. Several putative modifier genes in rodents, deregulated in HCC, are located in chromosomal segments syntenic to sites of chromosomal aberrations in humans, suggesting possible location of predisposing loci. Resistance to HCC is associated with lower genomic instability and downregulation of cell cycle key genes in preneoplastic and neoplastic lesions. p16(INK4A) upregulation occurs in susceptible and resistant rat lesions. p16(INK4A)-induced growth restraint was circumvented by Hsp90/Cdc37 chaperons and E2f4 nuclear export by Crm1 in susceptible, but not in resistant rats and human HCCs with better prognosis. Thus, protective mechanisms seem to be modulated by HCC modifiers, and differences in their efficiency influence the susceptibility to hepatocarcinogenesis and probably the prognosis of human HCC.
Collapse
Affiliation(s)
- F Feo
- Department of Biomedical Sciences, Division of Experimental Pathology and Oncology, University of Sassari, Via P. Manzella 4, 07100 Sasssari, Italy.
| | | | | | | | | | | | | |
Collapse
|
31
|
Sharpless NE. INK4a/ARF: a multifunctional tumor suppressor locus. Mutat Res 2005; 576:22-38. [PMID: 15878778 DOI: 10.1016/j.mrfmmm.2004.08.021] [Citation(s) in RCA: 290] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2004] [Revised: 07/12/2004] [Accepted: 08/12/2004] [Indexed: 02/02/2023]
Abstract
The INK4a/ARF locus encodes two physically linked tumor suppressor proteins, p16(INK4a) and ARF, which regulate the RB and p53 pathways, respectively. The unusual genomic relationship of the open reading frames of these proteins initially fueled speculation that only one of the two was the true tumor suppressor, and loss of the other merely coincidental in cancer. Recent human and mouse genetic data, however, have firmly established that both proteins possess significant in vivo tumor suppressor activity, although there appear to be species- and cell-type specific differences between the two. For example, ARF plays a clear role in preventing Myc-induced lymphomagenesis in mice, whereas the role for p16(INK4a) is human carcinomas is more firmly established. In this review, I discuss the evolutionary history of the locus, the relative importance of these tumor suppressor genes in human cancer, and recent information suggesting novel biochemical and physiologic functions of these proteins in vivo.
Collapse
Affiliation(s)
- Norman E Sharpless
- Department of Medicine, The Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, 27599-7295, USA.
| |
Collapse
|
32
|
Luo H, Li Q, O'Neal J, Kreisel F, Le Beau MM, Tomasson MH. c-Myc rapidly induces acute myeloid leukemia in mice without evidence of lymphoma-associated antiapoptotic mutations. Blood 2005; 106:2452-61. [PMID: 15972450 DOI: 10.1182/blood-2005-02-0734] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Ectopic expression of c-Myc (Myc) in most primary cell types results in programmed cell death, and malignant transformation cannot occur without additional mutations that block apoptosis. The development of Myc-induced lymphoid tumors has been well studied and supports this model. Myc can be upregulated in acute myeloid leukemia (AML), but its exact role in myeloid leukemogenesis is unclear. To study its role in AML, we used a murine stem cell virus (MSCV) retroviral gene transfer/transplantation system to broadly express Myc in the bone marrow of mice either alone or in combination with antiapoptotic mutations. Myc expression in the context either of Arf/Ink4a loss or Bcl-2 coexpression induced a mixture of acute myeloid and acute lymphoid leukemias (AML+ALL). In the absence of antiapoptotic mutations however, all mice transplanted with MSCV-Myc (100%, n = 110) developed AML exclusively. MSCV-Myc-induced AML was polyclonal, readily transplantable, possessed an intact Arf-p53 pathway, and did not display cytogenetic abnormalities by spectral karyotyping (SKY) analysis. Lastly, we found that Myc preferentially stimulated the growth of myeloid progenitor cells in methylcellulose. These data provide the first direct evidence that Myc is a critical downstream effector of myeloid leukemogenesis and suggest that myeloid progenitors are intrinsically resistant to Myc-induced apoptosis.
Collapse
Affiliation(s)
- Hui Luo
- Department of Medicine and Genetics, Division of Oncology, Saint Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Mice likely represent the most-studied mammalian organism, except for humans. Genetic engineering in embryonic stem cells has allowed derivation of mouse strains lacking particular cell cycle proteins. Analyses of these mutant mice, and cells derived from them, facilitated the studies of the functions of cell cycle apparatus at the organismal and cellular levels. In this review, we give some background about the cell cycle progression during mouse development. We next discuss some insights about in vivo functions of the cell cycle proteins, gleaned from mouse knockout experiments. Our text is meant to provide examples of the recent experiments, rather than to supply an extensive and complete list.
Collapse
Affiliation(s)
- Maria A Ciemerych
- Department of Embryology, Institute of Zoology, Faculty of Biology, Warsaw University, Miecznikowa 1, 02-096 Warsaw, Poland
| | | |
Collapse
|
34
|
Flint J, Valdar W, Shifman S, Mott R. Strategies for mapping and cloning quantitative trait genes in rodents. Nat Rev Genet 2005; 6:271-86. [PMID: 15803197 DOI: 10.1038/nrg1576] [Citation(s) in RCA: 382] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Over the past 15 years, more than 2,000 quantitative trait loci (QTLs) have been identified in crosses between inbred strains of mice and rats, but less than 1% have been characterized at a molecular level. However, new resources, such as chromosome substitution strains and the proposed Collaborative Cross, together with new analytical tools, including probabilistic ancestral haplotype reconstruction in outbred mice, Yin-Yang crosses and in silico analysis of sequence variants in many inbred strains, could make QTL cloning tractable. We review the potential of these strategies to identify genes that underlie QTLs in rodents.
Collapse
Affiliation(s)
- Jonathan Flint
- Wellcome Trust Centre for Human Genetics, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom.
| | | | | | | |
Collapse
|
35
|
Hahn H, Nitzki F, Schorban T, Hemmerlein B, Threadgill D, Rosemann M. Genetic mapping of a Ptch1-associated rhabdomyosarcoma susceptibility locus on mouse chromosome 2. Genomics 2005; 84:853-8. [PMID: 15475264 DOI: 10.1016/j.ygeno.2004.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Revised: 06/03/2004] [Accepted: 07/08/2004] [Indexed: 01/05/2023]
Abstract
Mutations in the Patched (Ptch1) gene are responsible for various familial and sporadic cancers. Ptch1(neo67/+) mice, in which exons 6 and 7 are deleted, show genetic background-dependent susceptibility to the development of muscle tumors resembling human rhabdomyosarcoma (RMS); BALB/c (BALB) is a susceptible strain whereas C57BL/6 (B6) shows resistance. A genome-wide linkage analysis was carried out using Ptch1(neo67/+)mice produced from B6 x (BALB x B6) backcrosses to identify loci involved in the control of RMS susceptibility. Quantitative trait locus mapping with the censored tumor latency time as the quantitative parameter was used to detect a significant RMS susceptibility modifier locus, Parms1 (Patched-Associated RMS 1), on chromosome 2 between D2Mit37 and D2Mit102 (LRS = 10). A Kaplan-Meier survival curve revealed that mice with the B6/BALB genotype develop tumors more frequently and much faster as compared to mice homozygous for the B6 allele (P = 0.02). Additional loci not reaching linkage significance were also detected for medulloblastoma resistance.
Collapse
Affiliation(s)
- Heidi Hahn
- Institute of Human Genetics, University of Göttingen, Heinrich-Düker-Weg 12, 37073 Göttingen, Federal Republic of Germany.
| | | | | | | | | | | |
Collapse
|
36
|
Bauer AK, Malkinson AM, Kleeberger SR. Susceptibility to neoplastic and non-neoplastic pulmonary diseases in mice: genetic similarities. Am J Physiol Lung Cell Mol Physiol 2004; 287:L685-703. [PMID: 15355860 DOI: 10.1152/ajplung.00223.2003] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chronic inflammation predisposes toward many types of cancer. Chronic bronchitis and asthma, for example, heighten the risk of lung cancer. Exactly which inflammatory mediators (e.g., oxidant species and growth factors) and lung wound repair processes (e.g., proangiogenic factors) enhance pulmonary neoplastic development is not clear. One approach to uncover the most relevant biochemical and physiological pathways is to identify genes underlying susceptibilities to inflammation and to cancer development at the same anatomic site. Mice develop lung adenocarcinomas similar in histology, molecular characteristics, and histogenesis to this most common human lung cancer subtype. Over two dozen loci, called Pas or pulmonary adenoma susceptibility, Par or pulmonary adenoma resistance, and Sluc or susceptibility to lung cancer genes, regulate differential lung tumor susceptibility among inbred mouse strains as assigned by QTL (quantitative trait locus) mapping. Chromosomal sites that determine responsiveness to proinflammatory pneumotoxicants such as ozone (O3), particulates, and hyperoxia have also been mapped in mice. For example, susceptibility QTLs have been identified on chromosomes 17 and 11 for O3-induced inflammation (Inf1, Inf2), O3-induced acute lung injury (Aliq3, Aliq1), and sulfate-associated particulates. Sites within the human and mouse genomes for asthma and COPD phenotypes have also been delineated. It is of great interest that several susceptibility loci for mouse lung neoplasia also contain susceptibility genes for toxicant-induced lung injury and inflammation and are homologous to several human asthma loci. These QTLs are described herein, candidate genes are suggested within these sites, and experimental evidence that inflammation enhances lung tumor development is provided.
Collapse
Affiliation(s)
- Alison K Bauer
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA.
| | | | | |
Collapse
|
37
|
Matheu A, Pantoja C, Efeyan A, Criado LM, Martín-Caballero J, Flores JM, Klatt P, Serrano M. Increased gene dosage of Ink4a/Arf results in cancer resistance and normal aging. Genes Dev 2004; 18:2736-46. [PMID: 15520276 PMCID: PMC528894 DOI: 10.1101/gad.310304] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mammalian genes frequently present allelic variants that differ in their expression levels and that, in the case of tumor suppressor genes, can be of relevance for cancer susceptibility and aging. We report here the characterization of a novel mouse model with increased activity for the Ink4a and Arf tumor suppressors. We have generated a "super Ink4a/Arf" mouse strain carrying a transgenic copy of the entire Ink4a/Arf locus. Cells derived from super Ink4a/Arf mice have increased resistance to in vitro immortalization and oncogenic transformation. Importantly, super Ink4a/Arf mice manifest higher resistance to cancer compared to normal, nontransgenic, mice. Finally, super Ink4a/Arf mice have normal aging and lifespan. Together, these results indicate that modest increases in the activity of the Ink4a/Arf tumor suppressor result in a beneficial cancer-resistant phenotype without affecting normal viability or aging.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene/toxicity
- Animals
- Carcinogens/toxicity
- Cell Survival
- Cell Transformation, Neoplastic/genetics
- Cells, Cultured
- Cellular Senescence
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Cyclin-Dependent Kinase Inhibitor p16/physiology
- Embryo, Mammalian/cytology
- Embryo, Mammalian/physiology
- Female
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Gene Dosage
- Genes, Tumor Suppressor
- Heterozygote
- Homozygote
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms, Experimental/chemically induced
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/prevention & control
Collapse
Affiliation(s)
- Ander Matheu
- Spanish National Cancer Center (CNIO), Madrid E-28029, Spain
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Cheung WC, Kim JS, Linden M, Peng L, Van Ness B, Polakiewicz RD, Janz S. Novel targeted deregulation of c-Myc cooperates with Bcl-X(L) to cause plasma cell neoplasms in mice. J Clin Invest 2004; 113:1763-73. [PMID: 15199411 PMCID: PMC420503 DOI: 10.1172/jci20369] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2003] [Accepted: 04/14/2004] [Indexed: 01/10/2023] Open
Abstract
Deregulated expression of both Myc and Bcl-X(L) are consistent features of human plasma cell neoplasms (PCNs). To investigate whether targeted expression of Myc and Bcl-X(L) in mouse plasma cells might lead to an improved model of human PCN, we generated Myc transgenics by inserting a single-copy histidine-tagged mouse Myc gene, Myc(His), into the mouse Ig heavy-chain Calpha locus. We also generated Bcl-X(L) transgenic mice that contain a multicopy Flag-tagged mouse Bcl-x(Flag) transgene driven by the mouse Ig kappa light-chain 3' enhancer. Single-transgenic Bcl-X(L) mice remained tumor free by 380 days of age, whereas single-transgenic Myc mice developed B cell tumors infrequently (4 of 43, 9.3%). In contrast, double-transgenic Myc/Bcl-X(L) mice developed plasma cell tumors with short onset (135 days on average) and full penetrance (100% tumor incidence). These tumors produced monoclonal Ig, infiltrated the bone marrow, and contained elevated amounts of Myc(His) and Bcl-X(L)(Flag) proteins compared with the plasma cells that accumulated in large numbers in young tumor-free Myc/Bcl-X(L) mice. Our findings demonstrate that the enforced expression of Myc and Bcl-X(L) by Ig enhancers with peak activity in plasma cells generates a mouse model of human PCN that recapitulates some features of human multiple myeloma.
Collapse
|
39
|
Cheung WC, Kim JS, Linden M, Peng L, Van Ness B, Polakiewicz RD, Janz S. Novel targeted deregulation of c-Myc cooperates with Bcl-XL to cause plasma cell neoplasms in mice. J Clin Invest 2004. [DOI: 10.1172/jci200420369] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
40
|
Thangaraju M, Sharan S, Sterneck E. Comparison of mammary gland involution between 129S1 and C57BL/6 inbred mouse strains: differential regulation of Bcl2a1, Trp53, Cebpb, and Cebpd expression. Oncogene 2004; 23:2548-53. [PMID: 14981542 DOI: 10.1038/sj.onc.1207363] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genetic engineering has made the mouse an invaluable tool to address the function of individual genes in a targeted manner. Over the last decade it has become apparent that the genetic mouse strain background can significantly influence the phenotype of an engineered mouse. Therefore, it is essential to characterize the biology of the different wild-type background strains. In this study, we have compared mouse mammary gland involution in the 129S1 and C57BL/6 inbred strains and report significant differences at the molecular level with differential expression of Bcl2a1 (Bfl1), Trp53 (p53), Cebpb (C/EBP beta), and Cebpd (C/EBP delta). The C57BL/6 strain exhibits dynamic responses with induction of Trp53 and Cebpd and concomitant downregulation of Bcl2a1 during the first phase of involution. In contrast, expression of these genes does not change significantly in 129S1 mice. During the second phase, C57BL/6 glands contain more Cebpb than 129S1 glands. Nevertheless, involution proceeds morphologically with similar kinetics in both strains. The data demonstrate that the genetic response of mammary tissue varies significantly between 129S1 and C57BL/6. These results may provide a basis for the interpretation of strain-specific phenotypes in engineered mice and underline the importance of pure strains for large-scale expression studies with mutant mice.
Collapse
Affiliation(s)
- Muthusamy Thangaraju
- Regulation of Cell Growth Laboratory, Center for Cancer Research, National Cancer Institute, Natinal Institutes of Health, PO Box B, Frederick, MD 21702, USA
| | | | | |
Collapse
|
41
|
Felix K, Gerstmeier S, Kyriakopoulos A, Howard OMZ, Dong HF, Eckhaus M, Behne D, Bornkamm GW, Janz S. Selenium Deficiency Abrogates Inflammation-Dependent Plasma Cell Tumors in Mice. Cancer Res 2004; 64:2910-7. [PMID: 15087411 DOI: 10.1158/0008-5472.can-03-2672] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The role of the micronutrient, selenium, in human cancers associated with chronic inflammations and persistent infections is poorly understood. Peritoneal plasmacytomas (PCTs) in strain BALB/c (C), the premier experimental model of inflammation-dependent plasma cell transformation in mice, may afford an opportunity to gain additional insights into the significance of selenium in neoplastic development. Here, we report that selenium-depleted C mice (n = 32) maintained on a torula-based low-selenium diet (5-8 micro g of selenium/kg) were totally refractory to pristane induction of PCT. In contrast, 11 of 26 (42.3%) control mice maintained on a selenium adequate torula diet (300 micro g of selenium/kg) and 15 of 40 (37.5%) control mice fed standard Purina chow (440 micro g of selenium/kg) developed PCT by 275 days postpristane. Abrogation of PCT was caused in part by the striking inhibition of the formation of the inflammatory tissue in which PCT develop (pristane granuloma). This was associated with the reduced responsiveness of selenium-deficient inflammatory cells (monocytes and neutrophils) to chemoattractants, such as thioredoxin and chemokines. Selenium-deficient C mice exhibited little evidence of disturbed redox homeostasis and increased mutant frequency of a transgenic lacZ reporter gene in vivo. These findings implicate selenium, via the selenoproteins, in the promotion of inflammation-induced PCT and suggest that small drug inhibitors of selenoproteins might be useful for preventing human cancers linked with chronic inflammations and persistent infections.
Collapse
Affiliation(s)
- Klaus Felix
- Laboratory of Genetics, Center for Cancer Research, National Cancer Institute and Veterinary Resources Program, NIH, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sachs Z, Sharpless NE, DePinho RA, Rosenberg N. p16(Ink4a) interferes with Abelson virus transformation by enhancing apoptosis. J Virol 2004; 78:3304-11. [PMID: 15016851 PMCID: PMC371071 DOI: 10.1128/jvi.78.7.3304-3311.2004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pre-B-cell transformation by Abelson virus (Ab-MLV) is a multistep process in which primary transformants are stimulated to proliferate but subsequently undergo crisis, a period of erratic growth marked by high levels of apoptosis. Inactivation of the p53 tumor suppressor pathway is an important step in this process and can be accomplished by mutation of p53 or down-modulation of p19(Arf), a p53 regulatory protein. Consistent with these data, pre-B cells from either p53 or Ink4a/Arf null mice bypass crisis. However, the Ink4a/Arf locus encodes both p19(Arf) and a second tumor suppressor, p16(Ink4a), that blocks cell cycle progression by inhibiting Cdk4/6. To determine if p16(Ink4a) plays a role in Ab-MLV transformation, primary transformants derived from Arf(-/-) and p16(Ink4a(-/-)) mice were compared. A fraction of those derived from Arf(-/-) animals underwent crisis, and even though all p16(Ink4a(-/-)) primary transformants experienced crisis, these cells became established more readily than cells derived from +/+ mice. Analyses of Ink4a/Arf(-/-) cells infected with a virus that expresses both v-Abl and p16(Ink4a) revealed that p16(Ink4a) expression does not alter cell cycle profiles but does increase the level of apoptosis in primary transformants. These results indicate that both products of the Ink4a/Arf locus influence Ab-MLV transformation and reveal that in addition to its well-recognized effects on the cell cycle, p16(Ink4a) can suppress transformation by inducing apoptosis.
Collapse
Affiliation(s)
- Zohar Sachs
- Department of Pathology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | | | |
Collapse
|
43
|
Fonseca R, Barlogie B, Bataille R, Bastard C, Bergsagel PL, Chesi M, Davies FE, Drach J, Greipp PR, Kirsch IR, Kuehl WM, Hernandez JM, Minvielle S, Pilarski LM, Shaughnessy JD, Stewart AK, Avet-Loiseau H. Genetics and cytogenetics of multiple myeloma: a workshop report. Cancer Res 2004; 64:1546-58. [PMID: 14989251 DOI: 10.1158/0008-5472.can-03-2876] [Citation(s) in RCA: 479] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Much has been learned regarding the biology and clinical implications of genetic abnormalities in multiple myeloma. Because of recent advances in the field, an International Workshop was held in Paris in february of 2003. This summary describes the consensus recommendations arising from that meeting with special emphasis on novel genetic observations. For instance, it is increasingly clear that translocations involving the immunoglobin heavy-chain locus are important for the pathogenesis of one-half of patients. As a corollary, it also clear that the remaining patients, lacking IgH translocations, have hyperdiploidy as the hallmark of their disease. Several important genetic markers are associated with a shortened survival such as chromosome 13 monosomy, hypodiploidy, and others. The events leading the transformation of the monoclonal gammopathy of undetermined significance (MGUS) to myeloma are still unclear. One of the few differential genetic lesions between myeloma and MGUS is the presence of ras mutations in the latter. Gene expression platforms are capable of detecting many of the genetic aberrations found in the clonal cells of myeloma. Areas in need of further study were identified. The study of the genetic aberrations will likely form the platform for targeted therapy for the disease.
Collapse
Affiliation(s)
- Rafael Fonseca
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Arbiser JL. Molecular regulation of angiogenesis and tumorigenesis by signal transduction pathways: evidence of predictable and reproducible patterns of synergy in diverse neoplasms. Semin Cancer Biol 2004; 14:81-91. [PMID: 15018892 DOI: 10.1016/j.semcancer.2003.09.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A large number of oncogenes, tumor suppressor genes, and signal transduction pathways have been described. Currently, a framework that allows prediction of tumor behavior based upon oncogenes, tumor suppressors, and signal transduction pathways is lacking. In 1869, Mendeleev published a periodic table of elements which allowed prediction of properties of elements based upon atomic weights that allowed prediction of chemical and physical properties of elements yet to be discovered. In this paper, I will discuss recurrent patterns of synergy found in the literature and our laboratory between tumor suppressor genes, oncogenes, and signaling pathways that allows one to predict the signaling pathway in a given tumor based upon the inactivation of a tumor suppressor gene. These patterns can be found in multiple different human neoplasms. Conversely, one can predict the inactivation of a tumor suppressor based upon the activation status of a signaling pathway. This knowledge can be used by a clinician or pathologist with access to immunohistochemistry to make predictions based upon simple technologies and determine the signaling pathways involved in a patient's tumor. These strategies may be useful in the design of prevention and treatment strategies for cancer.
Collapse
Affiliation(s)
- Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, WMB 5309, 1639 Pierce Drive, Atlanta, GA 30322, USA.
| |
Collapse
|
45
|
Huang AM, Montagna C, Sharan S, Ni Y, Ried T, Sterneck E. Loss of CCAAT/enhancer binding protein delta promotes chromosomal instability. Oncogene 2004; 23:1549-57. [PMID: 14716301 DOI: 10.1038/sj.onc.1207285] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The transcription factor CCAAT/enhancer binding protein delta (Cebpd, also known as C/EBPdelta, CRP3, CELF, NF-IL6beta) is implicated in diverse cellular functions such as the acute phase response, adipocyte differentiation, learning and memory, and mammary epithelial cell growth control. Here, we report that lack of Cebpd causes genomic instability and centrosome amplifications in primary embryonic fibroblasts derived from 129S1 mice. Upon spontaneous immortalization, Cebpd-deficient fibroblasts acquire transformed features such as impaired contact inhibition and reduced serum dependence. These data identify a novel role for Cebpd in the maintenance of chromosomal stability and suggest a potential tumor suppressor function in vivo.
Collapse
Affiliation(s)
- A-Mei Huang
- Regulation of Cell Growth Laboratory, PO Box B, Frederick, MD 21702, USA
| | | | | | | | | | | |
Collapse
|
46
|
Felix K, Polack A, Pretsch W, Jackson SH, Feigenbaum L, Bornkamm GW, Janz S. Moderate Hypermutability of a TransgeniclacZReporter Gene inMyc-Dependent Inflammation-Induced Plasma Cell Tumors in Mice. Cancer Res 2004; 64:530-7. [PMID: 14744766 DOI: 10.1158/0008-5472.can-03-2602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mutator phenotypes, a common and largely unexplained attribute of human cancer, might be better understood in mouse tumors containing reporter genes for accurate mutation enumeration and analysis. Previous work on peritoneal plasmacytomas (PCTs) in mice suggested that PCTs have a mutator phenotype caused by Myc-deregulating chromosomal translocations and/or phagocyte-induced mutagenesis due to chronic inflammation. To investigate this hypothesis, we generated PCTs that harbored the transgenic shuttle vector, pUR288, with a lacZ reporter gene for the assessment of mutations in vivo. PCTs exhibited a 5.5 times higher mutant frequency in lacZ (40.3 +/- 5.1 x 10(-5)) than in normal B cells (7.36 +/- 0.77 x 10(-5)), demonstrating that the tumors exhibit the phenotype of increased mutability. Studies on lacZ mutant frequency in serially transplanted PCTs and phagocyte-induced lacZ mutations in B cells in vitro indicated that mutant levels in tumors are not determined by exogenous damage inflicted by inflammatory cells. In vitro studies with a newly developed transgenic model of inducible Myc expression (Tet-off/MYC) showed that deregulated Myc sensitizes B cells to chemically induced mutations, but does not cause, on its own, mutations in lacZ. These findings suggested that the hypermutability of PCT is governed mainly by intrinsic features of tumor cells, not by deregulated Myc or chronic inflammation.
Collapse
Affiliation(s)
- Klaus Felix
- Laboratory of Genetics, Center for Cancer Research, National Cancer Institute, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Bliskovsky V, Ramsay ES, Scott J, DuBois W, Shi W, Zhang S, Qian X, Lowy DR, Mock BA. Frap, FKBP12 rapamycin-associated protein, is a candidate gene for the plasmacytoma resistance locus Pctr2 and can act as a tumor suppressor gene. Proc Natl Acad Sci U S A 2003; 100:14982-7. [PMID: 14634209 PMCID: PMC299869 DOI: 10.1073/pnas.2431627100] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2003] [Accepted: 10/06/2003] [Indexed: 11/18/2022] Open
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.
Collapse
Affiliation(s)
- Valery Bliskovsky
- Laboratory of Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4258, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Mice have become the mammalian model of choice for the application of genetics in biomedical research due to the evolutionary conservation of physiological systems and their attendant pathologies among all mammals as well as the exceptional power of genetic research technologies in the species. Beginning from aberrant phenotypes, a large number of mouse mutants and natural polymorphisms have been cloned, providing much information about the molecular basis of physiological processes. Additionally, the variable expression of these mutations in different inbred strain backgrounds has demonstrated the importance of modifier genes, which are also susceptible to cloning. Research efforts are keeping pace with these developments. In the area of gene discovery, large, government-funded mutagenesis programs now exist, and as a matter of great practical importance, recent evidence suggests that the same genes may be involved in the natural polymorphisms affecting disease in mice and humans. In parallel, dramatic advances are also being made in our ability to measure physiological processes in mice, and the advent of expression profiling promises revolutionary advances in understanding phenotype at the molecular level. Gene-driven approaches have relied on engineering the mouse genome, including adding, subtracting, and replacing genes and, most recently, the ability to control gene activity reversibly. Together, these multiple advances in our technical abilities have created extraordinary opportunities for future discovery.
Collapse
|
49
|
Abstract
Cell-cycle control is a major determinant of homeostasis during B-cell development, differentiation, and tumorigenesis. The generation of an antibody response requires activation and expansion of antigen-specific B cells and terminal differentiation of these cells into plasma cells. Plasma cells arrest in the G1 phase of the cell cycle, but the mechanism that underlies timely cell-cycle entry and exit in the humoral immune response is not known. The mammalian cell-cycle is regulated primarily at the G1 to S transition by the balance between positive regulators, the cyclin-dependent kinases (CDK) together with cyclins, and negative regulators, the CDK inhibitors. One such inhibitor, p18INK4c, has been shown to be required for cell-cycle termination and final differentiation of non-secreting plasmacytoid cells to antibody-secreting plasma cells. This finding provides the first direct evidence for cell-cycle control of B-cell immunity. It also raises important questions regarding cell-cycle control of cellular differentiation, apoptosis, and earlier steps of B-cell terminal differentiation. This article discusses the biochemical mechanism of cell-cycle control in the context of antibody response and plasma cell differentiation along with the role of cell-cycle dysregulation in the pathogenesis of multiple myeloma, the plasma cell cancer.
Collapse
Affiliation(s)
- Selina Chen-Kiang
- Department of Pathology, Weill Medical College of Cornell University, New York, NY 10021, USA.
| |
Collapse
|
50
|
Abstract
An increasing number of model systems of plasma cell tumor (PCT) formation have been and are being developed. Discussed here are six models in mice and multiple myeloma (MM) in humans. Each model illustrates a unique set of biological factors. There are two general types of model systems: those that depend upon naturally arising mutagenic changes (pristane-induced PCTs, 5TMM, and MM) and those that are associated with oncogenes (Emu-v-abl), growth factors [interleukin-6 (IL-6)], and anti-apoptotic factors (Bcl-xL/Bcl-2). PCTs develop in several special tissue microenvironments that provide essential cytokines (IL-6) and cell-cell interactions. In mice, the activation and deregulation of c-myc by chromosomal translocations is a major feature in many of the models. This mechanism is much less a factor in MM and the 5T model in mice. Genetically determined susceptibility is involved in many of the mouse models, but only a few genes have been implicated thus far.
Collapse
Affiliation(s)
- Michael Potter
- Laboratory of Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|