Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Haemophilia ; 29(4): 1024-1031, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37228173

ABSTRACT

BACKGROUND: The overall burden of disease in persons with haemophilia continues to be high despite the latest advancements in therapeutics. Clinical trials testing prenatal treatments for several genetic disorders are underway or are recruiting subjects, attesting to the much-needed change in paradigm of how patients with monogenic disorders can be treated. Here we investigate the overall attitude towards prenatal diagnosis, preferences on types of prenatal therapies for haemophilia, the level of 'acceptable' risk tolerated, and which social and moral pressures or disease personal experiences may predict willingness of individuals to consider foetal therapy in a future pregnancy. RESULTS: A multidisciplinary team designed the survey, and the study was carried out using REDCap, and publicized through the National Haemophilia Foundation. Subjects ≥18 years of age were eligible to participate in the study. We assessed participants' attitudes towards prenatal therapy and their level of 'acceptable' risk towards the procedure and therapy. The survey was completed by 67 adults, the majority females. Respondents were willing to undergo prenatal diagnosis, and their main concerns related to the well-being of the pregnant woman and the foetus regarding lasting therapeutic efficacy, side effects of the therapy, and procedural risks, but they were likely to accept a wide range of prenatal therapeutic options, particularly if the foetal therapy proved to be long-lasting and safe. CONCLUSIONS: These data demonstrate the willingness of persons with haemophilia, and the haemophilia community, to explore new treatment options beyond the currently offered approaches.


Subject(s)
Hemophilia A , Pregnancy , Adult , Female , Humans , Hemophilia A/diagnosis , Hemophilia A/therapy , Hemophilia A/genetics , Prenatal Diagnosis , Surveys and Questionnaires
2.
Am J Health Syst Pharm ; 80(16): 1056-1062, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37061822

ABSTRACT

PURPOSE: To evaluate the impact of a collaborative drug therapy management (CDTM) agreement allowing a pharmacist to automatically prescribe refills of discharge medications to patients' preferred outpatient pharmacy on utilization of a hospital discharge prescription program and hospital readmission rates. METHODS: This was a single-center, quasi-experimental pre-post intervention study. Patients aged 18 years or older discharged from the cardiology services to home were eligible for inclusion in the study. The CDTM agreement was initiated on July 1, 2019. Patients discharged to home from July 1, 2018, to June 30, 2019, were assigned to the historical control group. The primary outcome was the difference in the proportion of patients who used the bedside medication delivery service at hospital discharge between the groups. Secondary outcomes included 30-day hospital readmissions and a descriptive analysis of medications prescribed by a pharmacist through the program. A χ2 test was used to assess the primary outcome, and multivariable logistic regression was used to assess hospital readmissions. RESULTS: In total, 1,704 and 2,200 patients were discharged in the control and CDTM groups, respectively. The CDTM group had a greater proportion of patients who participated in the discharge prescription program compared to the historical control group (77.8% vs 68.7%; P < 0.0001). There was no difference in 30-day hospital readmission rate between the groups (adjusted odds ratio, 1.01; 95% confidence interval, 0.83-1.23; P = 0.94). CONCLUSION: A CDTM protocol to improve the availability of medication refills at a patient's regular outpatient pharmacy improved utilization of a bedside medication delivery service but did not change 30-day readmission rates.


Subject(s)
Patient Discharge , Pharmacy Service, Hospital , Humans , Patient Readmission , Pharmacists , Medication Therapy Management , Prescriptions , Medication Reconciliation/methods
3.
Cancer Biol Ther ; 20(2): 169-182, 2019.
Article in English | MEDLINE | ID: mdl-30183475

ABSTRACT

Targeting the early steps of the glycolysis pathway in cancers is a well-established therapeutic strategy; however, the doses required to elicit a therapeutic effect on the cancer can be toxic to the patient. Consequently, numerous preclinical and clinical studies have combined glycolytic blockade with other therapies. However, most of these other therapies do not specifically target cancer cells, and thus adversely affect normal tissue. Here we first show that a diverse number of cancer models - spontaneous, patient-derived xenografted tumor samples, and xenografted human cancer cells - can be efficiently targeted by 2-deoxy-D-Glucose (2DG), a well-known glycolytic inhibitor. Next, we tested the cancer-cell specificity of a therapeutic compound using the MEC1 cell line, a chronic lymphocytic leukemia (CLL) cell line that expresses activation induced cytidine deaminase (AID). We show that MEC1 cells, are susceptible to 4,4'-Diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS), a specific RAD51 inhibitor. We then combine 2DG and DIDS, each at a lower dose and demonstrate that this combination is more efficacious than fludarabine, the current standard- of- care treatment for CLL. This suggests that the therapeutic blockade of glycolysis together with the therapeutic inhibition of RAD51-dependent homologous recombination can be a potentially beneficial combination for targeting AID positive cancer cells with minimal adverse effects on normal tissue. Implications: Combination therapy targeting glycolysis and specific RAD51 function shows increased efficacy as compared to standard of care treatments in leukemias.


Subject(s)
4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxyglucose/pharmacology , Neoplasms/drug therapy , Rad51 Recombinase/antagonists & inhibitors , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/administration & dosage , Animals , Cell Line, Tumor , Deoxyglucose/administration & dosage , Drug Synergism , Female , Glycolysis/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasms/metabolism , Rad51 Recombinase/metabolism , Xenograft Model Antitumor Assays
4.
J Immunol ; 198(11): 4255-4267, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28461573

ABSTRACT

B lymphocytes play a key role in type 1 diabetes (T1D) development by serving as a subset of APCs preferentially supporting the expansion of autoreactive pathogenic T cells. As a result of their pathogenic importance, B lymphocyte-targeted therapies have received considerable interest as potential T1D interventions. Unfortunately, the B lymphocyte-directed T1D interventions tested to date failed to halt ß cell demise. IgG autoantibodies marking humans at future risk for T1D indicate that B lymphocytes producing them have undergone the affinity-maturation processes of class switch recombination and, possibly, somatic hypermutation. This study found that CRISPR/Cas9-mediated ablation of the activation-induced cytidine deaminase gene required for class switch recombination/somatic hypermutation induction inhibits T1D development in the NOD mouse model. The activation-induced cytidine deaminase protein induces genome-wide DNA breaks that, if not repaired through RAD51-mediated homologous recombination, result in B lymphocyte death. Treatment with the RAD51 inhibitor 4,4'-diisothiocyanatostilbene-2, 2'-disulfonic acid also strongly inhibited T1D development in NOD mice. The genetic and small molecule-targeting approaches expanded CD73+ B lymphocytes that exert regulatory activity suppressing diabetogenic T cell responses. Hence, an initial CRISPR/Cas9-mediated genetic modification approach has identified the AID/RAD51 axis as a target for a potentially clinically translatable pharmacological approach that can block T1D development by converting B lymphocytes to a disease-inhibitory CD73+ regulatory state.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Carrier Proteins/antagonists & inhibitors , Cytidine Deaminase/antagonists & inhibitors , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Lymphocyte Activation , Nuclear Proteins/antagonists & inhibitors , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , 5'-Nucleotidase/immunology , Animals , Autoantibodies/immunology , CRISPR-Cas Systems , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA-Binding Proteins , Diabetes Mellitus, Experimental , Immunoglobulin Class Switching , Mice , Mice, Inbred NOD , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins , Somatic Hypermutation, Immunoglobulin
5.
Cancer Discov ; 6(5): 532-45, 2016 05.
Article in English | MEDLINE | ID: mdl-27001151

ABSTRACT

UNLABELLED: Tumors are dynamic organs that evolve during disease progression with genetic, epigenetic, and environmental differences among tumor cells serving as the foundation for selection and evolution in tumors. Tumor-initiating cells (TIC) that are responsible for tumorigenesis are a source of functional cellular heterogeneity, whereas chromosomal instability (CIN) is a source of karyotypic genetic diversity. However, the extent that CIN contributes to TIC genetic diversity and its relationship to TIC function remains unclear. Here, we demonstrate that glioblastoma TICs display CIN with lagging chromosomes at anaphase and extensive nonclonal chromosome copy-number variations. Elevating the basal chromosome missegregation rate in TICs decreases both proliferation and the stem-like phenotype of TICs in vitro Consequently, tumor formation is abolished in an orthotopic mouse model. These results demonstrate that TICs generate genetic heterogeneity within tumors, but that TIC function is impaired if the rate of genetic change is elevated above a tolerable threshold. SIGNIFICANCE: Genetic heterogeneity among TICs may produce advantageous karyotypes that lead to therapy resistance and relapse; however, we found that TICs have an upper tolerable limit for CIN. Thus, increasing the chromosome missegregation rate offers a new therapeutic strategy to eliminate TICs from tumors. Cancer Discov; 6(5); 532-45. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 461.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomal Instability , Glioblastoma/genetics , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Biomarkers , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Chromosome Aberrations , Chromosome Segregation , DNA Fragmentation , Disease Models, Animal , Female , Genetic Heterogeneity , Genetic Predisposition to Disease , Glioblastoma/metabolism , Heterografts , Humans , In Situ Hybridization, Fluorescence , Mice , Mutation
7.
Nat Immunol ; 14(11): 1183-1189, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24097111

ABSTRACT

The ability of activation-induced cytidine deaminase (AID) to efficiently mediate class-switch recombination (CSR) is dependent on its phosphorylation at Ser38; however, the trigger that induces AID phosphorylation and the mechanism by which phosphorylated AID drives CSR have not been elucidated. Here we found that phosphorylation of AID at Ser38 was induced by DNA breaks. Conversely, in the absence of AID phosphorylation, DNA breaks were not efficiently generated at switch (S) regions in the immunoglobulin heavy-chain locus (Igh), consistent with a failure of AID to interact with the endonuclease APE1. Additionally, deficiency in the DNA-damage sensor ATM impaired the phosphorylation of AID at Ser38 and the interaction of AID with APE1. Our results identify a positive feedback loop for the amplification of DNA breaks at S regions through the phosphorylation- and ATM-dependent interaction of AID with APE1.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/immunology , DNA-(Apurinic or Apyrimidinic Site) Lyase/immunology , Feedback, Physiological , Immunoglobulin Class Switching , Immunoglobulin Heavy Chains/immunology , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/immunology , B-Lymphocytes/cytology , Cytidine Deaminase/genetics , DNA Breaks, Double-Stranded , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Gene Expression Regulation , Immunoglobulin Heavy Chains/genetics , Mice , Phosphorylation , Protein Binding , Serine/immunology , Serine/metabolism , Signal Transduction
8.
J Exp Med ; 210(5): 1021-33, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23589568

ABSTRACT

Activation-induced cytidine deaminase (AID) is critical in normal B cells to initiate somatic hypermutation and immunoglobulin class switch recombination. Accumulating evidence suggests that AID is also prooncogenic, inducing cancer-promoting mutations or chromosome rearrangements. In this context, we find that AID is expressed in >40% of primary human chronic lymphocytic leukemia (CLL) cases, consistent with other reports. Using a combination of human B lymphoid leukemia cells and mouse models, we now show that AID expression can be harnessed for antileukemic effect, after inhibition of the RAD51 homologous recombination (HR) factor with 4,4'-diisothiocyanatostilbene-2-2'-disulfonic acid (DIDS). As a proof of principle, we show that DIDS treatment inhibits repair of AID-initiated DNA breaks, induces apoptosis, and promotes cytotoxicity preferentially in AID-expressing human CLL. This reveals a novel antineoplastic role of AID that can be triggered by inhibition of HR, suggesting a potential new paradigm to treat AID-expressing tumors. Given the growing list of tumor types with aberrant AID expression, this novel therapeutic approach has potential to impact a significant patient population.


Subject(s)
Cytidine Deaminase/metabolism , Homologous Recombination/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/radiation effects , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , B-Lymphocytes/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Transformed , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytidine Deaminase/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/radiation effects , Histones/metabolism , Homologous Recombination/drug effects , Homologous Recombination/radiation effects , Humans , Mice , Rad51 Recombinase/metabolism , Radiation, Ionizing
9.
J Immunol ; 189(5): 2374-82, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22826323

ABSTRACT

Activation-induced cytidine deaminase (AID) initiates DNA double-strand breaks (DSBs) in the IgH gene (Igh) to stimulate isotype class switch recombination (CSR), and widespread breaks in non-Igh (off-target) loci throughout the genome. Because the DSBs that initiate class switching occur during the G1 phase of the cell cycle, and are repaired via end joining, CSR is considered a predominantly G1 reaction. By contrast, AID-induced non-Igh DSBs are repaired by homologous recombination. Although little is known about the connection between the cell cycle and either induction or resolution of AID-mediated non-Igh DSBs, their repair by homologous recombination implicates post-G1 phases. Coordination of DNA breakage and repair during the cell cycle is critical to promote normal class switching and prevent genomic instability. To understand how AID-mediated events are regulated through the cell cycle, we have investigated G1-to-S control in AID-dependent genome-wide DSBs. We find that AID-mediated off-target DSBs, like those induced in the Igh locus, are generated during G1. These data suggest that AID-mediated DSBs can evade G1/S checkpoint activation and persist beyond G1, becoming resolved during S phase. Interestingly, DSB resolution during S phase can promote not only non-Igh break repair, but also Ig CSR. Our results reveal novel cell cycle dynamics in response to AID-initiated DSBs, and suggest that the regulation of the repair of these DSBs through the cell cycle may ensure proper class switching while preventing AID-induced genomic instability.


Subject(s)
Cytidine Deaminase/physiology , DNA Breaks, Double-Stranded , Immunoglobulin Class Switching/genetics , Immunoglobulin Isotypes/genetics , S Phase/genetics , S Phase/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , DNA Repair/genetics , DNA Repair/immunology , G1 Phase/genetics , G1 Phase/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
PLoS One ; 6(8): e23498, 2011.
Article in English | MEDLINE | ID: mdl-21853140

ABSTRACT

Rhabdomyosarcomas (RSCs) are skeletal muscle neoplasms found in humans and domestic mammals. The A/J inbred strain developed a high frequency (between 70-80%) of adult pleomorphic type (APT) RSC at >20 months of age while BALB/cByJ also develop RSC but less frequently. These neoplasms invaded skeletal muscle surrounding either the axial or proximal appendicular skeleton and were characterized by pleomorphic cells with abundant eosinophilic cytoplasm, multiple nuclei, and cross striations. The diagnosis was confirmed by detection of alpha-sarcomeric actin and myogenin in the neoplastic cells using immunocytochemistry. The A/J strain, but not the related BALB/c substrains, is also characterised by a progressive muscular dystrophy homologous to limb-girdle muscular dystrophy type 2B. The association between the development of RSC in similar muscle groups to those most severely affected by the progressive muscular dystrophy suggested that these neoplasms developed from abnormal regeneration of the skeletal muscle exacerbated by the dysferlin mutation. Transcriptome analyses of RSCs revealed marked downregulation of genes in muscular development and function signaling networks. Non-synonymous coding SNPs were found in Myl1, Abra, Sgca, Ttn, and Kcnj12 suggesting these may be important in the pathogenesis of RSC. These studies suggest that A strains of mice can be useful models for dissecting the molecular genetic basis for development, progression, and ultimately for testing novel anticancer therapeutic agents dealing with rhabdomyosarcoma.


Subject(s)
Aging/pathology , Rhabdomyosarcoma/pathology , Animals , Female , Gene Expression Regulation, Neoplastic , Immunohistochemistry , Male , Mice , Mice, Inbred Strains , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Rhabdomyosarcoma/genetics , Signal Transduction/genetics
11.
Chromosoma ; 120(1): 61-71, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20703494

ABSTRACT

Nuclear localization influences the expression of certain genes. Chromosomal rearrangements can reposition genes in the nucleus and thus could impact the expression of genes far from chromosomal breakpoints. However, the extent to which chromosomal rearrangements influence nuclear organization and gene expression is poorly understood. We examined mouse progenitor B cell lymphomas with a common translocation, der(12)t(12;15), which fuses a gene-rich region of mouse chromosome 12 (Mmu 12) with a gene-poor region of mouse chromosome 15 (Mmu 15). We found that sequences 2.3 Mb proximal and 2.7 Mb distal to the der(12)t(12;15) breakpoint had different nuclear positions measured relative to the nuclear radius. However, their positions were similar on unrearranged chromosomes in the same tumor cells and normal progenitor B cells. In addition, higher-order chromatin folding marked by three-dimensional gene clustering was not significantly altered for the 7 Mb of Mmu 15 sequence distal to this translocation breakpoint. Translocation also did not correspond to significant changes in gene expression in this region. Thus, any changes to Mmu 15 structure and function imposed by the der(12)t(12;15) translocation are constrained to sequences near (<2.5 Mb) the translocation junction. These data contrast with those of certain other chromosomal rearrangements and suggest that significant changes to Mmu 15 sequence are structurally and functionally tolerated in the tumor cells examined.


Subject(s)
Chromatin/metabolism , Chromosomes, Mammalian/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/metabolism , Translocation, Genetic , Animals , Cell Line, Tumor , Chromatin/genetics , Chromosomes, Mammalian/genetics , Lymphoma, B-Cell/genetics , Mice
12.
BMC Biol ; 8: 132, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20979627

ABSTRACT

BACKGROUND: Unrepaired DNA double-stranded breaks (DSBs) cause chromosomal rearrangements, loss of genetic information, neoplastic transformation or cell death. The nonhomologous end joining (NHEJ) pathway, catalyzing sequence-independent direct rejoining of DSBs, is a crucial mechanism for repairing both stochastically occurring and developmentally programmed DSBs. In lymphocytes, NHEJ is critical for both development and genome stability. NHEJ defects lead to severe combined immunodeficiency (SCID) and lymphoid cancer predisposition in both mice and humans. While NHEJ has been thoroughly investigated in lymphocytes, the importance of NHEJ in other cell types, especially with regard to tumor suppression, is less well documented. We previously reported evidence that the NHEJ pathway functions to suppress a range of nonlymphoid tumor types, including various classes of sarcomas, by unknown mechanisms. RESULTS: Here we investigate roles for the NHEJ factor ARTEMIS in multipotent mesenchymal stem/progenitor cells (MSCs), as putative sarcomagenic cells of origin. We demonstrate a key role for ARTEMIS in sarcoma suppression in a sensitized mouse tumor model. In this context, we found that ARTEMIS deficiency led to chromosomal damage but, paradoxically, enhanced resistance and proliferative potential in primary MSCs subjected to various stresses. Gene expression analysis revealed abnormally regulated stress response, cell proliferation, and signal transduction pathways in ARTEMIS-defective MSCs. Finally, we identified candidate regulatory genes that may, in part, mediate a stress-resistant, hyperproliferative phenotype in preneoplastic ARTEMIS-deficient MSCs. CONCLUSIONS: Our discoveries suggest that Art prevents genome damage and restrains proliferation in MSCs exposed to various stress stimuli. We propose that deficiency leads to a preneoplastic state in primary MSCs and is associated with aberrant proliferative control and cellular stress resistance. Thus, our data reveal surprising new roles for ARTEMIS and the NHEJ pathway in normal MSC function and fitness relevant to tumor suppression in mesenchymal tissues.


Subject(s)
DNA Repair/genetics , Genomic Instability/physiology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Nuclear Proteins/metabolism , Sarcoma/genetics , Signal Transduction/physiology , Animals , Cell Proliferation , DNA-Binding Proteins , Endonucleases , Gene Expression Profiling , Genes, Tumor Suppressor/physiology , Genomic Instability/genetics , Humans , Mesenchymal Stem Cells/metabolism , Mice , Multipotent Stem Cells/metabolism , Nuclear Proteins/genetics , Signal Transduction/genetics
13.
Nat Immunol ; 11(9): 820-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20657597

ABSTRACT

Activation-induced cytidine deaminase (AID) is required for somatic hypermutation and immunoglobulin class switching in activated B cells. Because AID has no known target-site specificity, there have been efforts to identify non-immunoglobulin AID targets. We show here that AID acts promiscuously, generating widespread DNA double-strand breaks (DSBs), genomic instability and cytotoxicity in B cells with less homologous recombination ability. We demonstrate that the homologous-recombination factor XRCC2 suppressed AID-induced off-target DSBs, promoting B cell survival. Finally, we suggest that aberrations that affect human chromosome 7q36, including XRCC2, correlate with genomic instability in B cell cancers. Our findings demonstrate that AID has promiscuous genomic DSB-inducing activity, identify homologous recombination as a safeguard against off-target AID action, and have implications for genomic instability in B cell cancers.


Subject(s)
Cytidine Deaminase/metabolism , DNA Breaks , Recombination, Genetic/genetics , B-Lymphocytes/immunology , Cell Cycle , Cell Survival , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Flow Cytometry , Genomic Instability , Humans , Reverse Transcriptase Polymerase Chain Reaction
14.
Cancer Res ; 69(24): 9422-30, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19934316

ABSTRACT

Molecular cancer diagnostics are an important clinical advance in cancer management, but new methods are still needed. In this context, gene expression signatures obtained by microarray represent a useful molecular diagnostic. Here, we describe novel probe-level microarray analyses that reveal connections between mRNA processing and neoplasia in multiple tumor types, with diagnostic potential. We now show that characteristic differences in mRNA processing, primarily in the 3'-untranslated region, define molecular signatures that can distinguish similar tumor subtypes with different survival characteristics, with at least 74% accuracy. Using a mouse model of B-cell leukemia/lymphoma, we find that differences in transcript isoform abundance are likely due to both alternative polyadenylation (APA) and differential degradation. While truncation of the 3'-UTR is the most common observed pattern, genes with elongated transcripts were also observed, and distinct groups of affected genes are found in related but distinct tumor types. Genes with elongated transcripts are overrepresented in ontology categories related to cell-cell adhesion and morphology. Analysis of microarray data from human primary tumor samples revealed similar phenomena. Western blot analysis of selected proteins confirms that changes in the 3'-UTR can correlate with changes in protein expression. Our work suggests that alternative mRNA processing, particularly APA, can be a powerful molecular biomarker with prognostic potential. Finally, these findings provide insights into the molecular mechanisms of gene deregulation in tumorigenesis.


Subject(s)
Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , RNA, Messenger/genetics , 3' Untranslated Regions , Animals , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Protein Isoforms , Tumor Suppressor Protein p53/deficiency
15.
Genes Chromosomes Cancer ; 48(10): 925-30, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19603522

ABSTRACT

Both somatic and meiotic recombinations involve the repair of DNA double strand breaks (DSBs) that occur at preferred locations in the genome. Improper repair of DSBs during either mitosis or meiosis can lead to mutations, chromosomal aberration such as translocations, cancer, and/or cell death. Currently, no model exists that explains the locations of either spontaneous somatic DSBs or programmed meiotic DSBs or relates them to each other. One common class of tumorigenic translocations arising from DSBs is chromosomal rearrangements near the Myc oncogene. Myc translocations have been associated with Burkitt lymphoma in humans, plasmacytoma in mice, and immunocytoma in rats. Comparing the locations of somatic and meiotic DSBs near the mouse Myc oncogene, we demonstrated that the placement of these DSBs is not random and that both events clustered in the same short discrete region of the genome. Our work shows that both somatic and meiotic DSBs tend to occur in proximity to each other within the Myc region, suggesting that they share common originating features. It is likely that some regions of the genome are more susceptible to both somatic and meiotic DSBs, and the locations of meiotic hotspots may be an indicator of genomic regions more susceptible to DNA damage.


Subject(s)
Chromosomes, Mammalian , DNA Breaks, Double-Stranded , Genes, myc , Proto-Oncogene Proteins c-myc/genetics , Animals , Female , Lymphoma, B-Cell/genetics , Male , Meiosis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitosis , Recombination, Genetic
16.
Aging Cell ; 8(3): 277-87, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19627267

ABSTRACT

To better characterize aging in mice, the Jackson Aging Center carried out a lifespan study of 31 genetically-diverse inbred mouse strains housed in a specific pathogen-free facility. Clinical assessments were carried out every 6 months, measuring multiple age-related phenotypes including neuromuscular, kidney and heart function, body composition, bone density, hematology, hormonal levels, and immune system parameters. In a concurrent cross-sectional study of the same 31 strains at 6, 12, and 20 months, more invasive measurements were carried out followed by necropsy to assess apoptosis, DNA repair, chromosome fragility, and histopathology. In this report, which is the initial paper of a series, the study design, median lifespans, and circulating insulin-like growth factor 1 (IGF1) levels at 6, 12, and 18 months are described for the first cohort of 32 females and 32 males of each strain. Survival curves varied dramatically among strains with the median lifespans ranging from 251 to 964 days. Plasma IGF1 levels, which also varied considerably at each time point, showed an inverse correlation with a median lifespan at 6 months (R = -0.33, P = 0.01). This correlation became stronger if the short-lived strains with a median lifespan < 600 days were removed from the analysis (R = -0.53, P < 0.01). These results support the hypothesis that the IGF1 pathway plays a key role in regulating longevity in mice and indicates that common genetic mechanisms may exist for regulating IGF1 levels and lifespan.


Subject(s)
Insulin-Like Growth Factor I/analysis , Longevity , Animals , Female , Male , Mice , Mice, Inbred Strains , Research Design , Survival Analysis
17.
Cancer Res ; 69(10): 4454-60, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19435904

ABSTRACT

Chromosomal instability is a hallmark of many tumor types. Complex chromosomal rearrangements with associated gene amplification, known as complicons, characterize many hematologic and solid cancers. Whereas chromosomal aberrations, including complicons, are useful diagnostic and prognostic cancer markers, their molecular origins are not known. Although accumulating evidence has implicated DNA double-strand break repair in suppression of oncogenic genome instability, the genomic elements required for chromosome rearrangements, especially complex lesions, have not been elucidated. Using a mouse model of B-lineage lymphoma, characterized by complicon formation involving the immunoglobulin heavy chain (Igh) locus and the c-myc oncogene, we have now investigated the requirement for specific genomic segments as donors for complex rearrangements. We now show that specific DNA double-strand breaks, occurring within a narrow segment of Igh, are necessary to initiate complicon formation. By contrast, neither specific DNA breaks nor the powerful intronic enhancer Emu are required for complicon-independent oncogenesis. This study is the first to delineate mechanisms of complex versus simple instability and the first to identify specific chromosomal elements required for complex chromosomal aberrations. These findings will illuminate genomic cancer susceptibility and risk factors.


Subject(s)
Chromosome Aberrations , DNA Damage , DNA Repair , Gene Amplification , Gene Rearrangement , Genes, myc , Immunoglobulin Heavy Chains/genetics , Lymphocytes/physiology , Lymphoma, B-Cell/genetics , Translocation, Genetic , Animals , Disease Models, Animal , Genetic Predisposition to Disease , Immunoglobulin Joining Region/genetics , Lymphoma, B-Cell/epidemiology , Lymphoma, B-Cell/immunology , Mice , Risk Factors
18.
Cell ; 135(5): 907-18, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-19041753

ABSTRACT

Genomic instability and alterations in gene expression are hallmarks of eukaryotic aging. The yeast histone deacetylase Sir2 silences transcription and stabilizes repetitive DNA, but during aging or in response to a DNA break, the Sir complex relocalizes to sites of genomic instability, resulting in the desilencing of genes that cause sterility, a characteristic of yeast aging. Using embryonic stem cells, we show that mammalian Sir2, SIRT1, represses repetitive DNA and a functionally diverse set of genes across the mouse genome. In response to DNA damage, SIRT1 dissociates from these loci and relocalizes to DNA breaks to promote repair, resulting in transcriptional changes that parallel those in the aging mouse brain. Increased SIRT1 expression promotes survival in a mouse model of genomic instability and suppresses age-dependent transcriptional changes. Thus, DNA damage-induced redistribution of SIRT1 and other chromatin-modifying proteins may be a conserved mechanism of aging in eukaryotes.


Subject(s)
Aging/genetics , Chromatin/metabolism , Genomic Instability , Sirtuins/genetics , Animals , Brain/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair , Embryonic Stem Cells , Gene Knockout Techniques , Humans , Lymphoma/metabolism , Mice , Molecular Sequence Data , Oxidative Stress , Sirtuin 1 , Specific Pathogen-Free Organisms , Thymus Neoplasms/metabolism , Yeasts/cytology , Yeasts/metabolism
19.
Mol Cell Biol ; 28(7): 2295-303, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18212067

ABSTRACT

Primary immunodeficiencies are rare but serious diseases with diverse genetic causes. Accumulating evidence suggests that defects in DNA double-strand break (DSB) repair can underlie many of these syndromes. In this context, the nonhomologous end joining pathway of DSB repair is absolutely required for lymphoid development, but possible roles for the homologous recombination (HR) pathway have remained more controversial. While recent evidence suggests that HR may indeed be important to suppress lymphoid transformation, the specific relationship of HR to normal lymphocyte development remains unclear. We have investigated roles of the X-ray cross-complementing 2 (Xrcc2) HR gene in lymphocyte development. We show that HR is critical for normal B-cell development, with Xrcc2 nullizygosity leading to p53-dependent early S-phase arrest. In the absence of p53 (encoded by Trp53), Xrcc2-null B cells can fully develop but show high rates of chromosome and chromatid fragmentation. We present a molecular model wherein Xrcc2 is important to preserve or restore replication forks during rapid clonal expansion of developing lymphocytes. Our findings demonstrate a key role for HR in lymphoid development and suggest that Xrcc2 defects could underlie some human primary immunodeficiencies.


Subject(s)
B-Lymphocytes/cytology , DNA Repair , DNA-Binding Proteins/physiology , Lymphopoiesis/physiology , Recombination, Genetic , Animals , Cells, Cultured/cytology , Chromosome Aberrations , Chromosome Breakage , Coculture Techniques , Gene Deletion , Genes, p53 , Immunoglobulin M/biosynthesis , Interleukin-7/metabolism , Leukocyte Common Antigens/biosynthesis , Liver/cytology , Liver/embryology , Lymphopoiesis/genetics , Mice , Mice, Knockout , NIH 3T3 Cells/metabolism , S Phase , Sequence Homology, Nucleic Acid , Tumor Suppressor Protein p53/physiology
20.
Chromosome Res ; 15(8): 1061-73, 2007.
Article in English | MEDLINE | ID: mdl-18060570

ABSTRACT

Radiation exposure is an occupational hazard for military personnel, some health care professionals, airport security screeners, and medical patients, with some individuals at risk for acute, high-dose exposures. Therefore, the biological effects of radiation, especially the potential for chromosome damage, are major occupational and health concerns. However, the biophysical mechanisms of chromosome instability subsequent to radiation-induced DNA damage are poorly understood. It is clear that interphase chromosomes occupy discrete structural and functional subnuclear domains, termed chromosome territories (CT), which may be organized into 'neighborhoods' comprising groups of specific CTs. We directly evaluated the relationship between chromosome positioning, neighborhood composition, and translocation partner choice in primary lymphocytes, using a cell-based system in which we could induce multiple, concentrated DNA breaks via high-dose irradiation. We critically evaluated mis-rejoining profiles and tested whether breaks occurring nearby were more likely to fuse than breaks occurring at a distance. We show that CT neighborhoods comprise heterologous chromosomes, within which inter-CT distances directly relate to translocation partner choice. These findings demonstrate that interphase chromosome arrangement is a principal factor in genomic instability outcomes in primary lymphocytes, providing a structural context for understanding the biological effects of radiation exposure, and the molecular etiology of tumor-specific translocation patterns.


Subject(s)
Chromosome Aberrations/radiation effects , Chromosomes/genetics , Precursor Cells, B-Lymphoid/physiology , Radiation Dosage , Translocation, Genetic/genetics , Tumor Suppressor Protein p53/physiology , Animals , DNA Damage , Gamma Rays , Genomic Instability , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C57BL , Precursor Cells, B-Lymphoid/radiation effects , Spectral Karyotyping , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...