scholarly journals Agricultural pesticide exposure and the molecular connection to lymphomagenesis

2009 ◽  
Vol 206 (7) ◽  
pp. 1473-1483 ◽  
Author(s):  
Julie Agopian ◽  
Jean-Marc Navarro ◽  
Anne-Claire Gac ◽  
Yannick Lecluse ◽  
Mélanie Briand ◽  
...  

The t(14;18) translocation constitutes the initiating event of a causative cascade leading to follicular lymphoma (FL). t(14;18) translocations are present in blood from healthy individuals, but there is a trend of increased prevalence in farmers exposed to pesticides, a group recently associated with higher risk of t(14;18)+ non-Hodgkin's lymphoma development. A direct connection between agricultural pesticide use, t(14;18) in blood, and malignant progression, however, has not yet been demonstrated. We followed t(14;18) clonal evolution over 9 yr in a cohort of farmers exposed to pesticides. We show that exposed individuals bear particularly high t(14;18) frequencies in blood because of a dramatic clonal expansion of activated t(14;18)+ B cells. We further demonstrate that such t(14;18)+ clones recapitulate the hallmark features of developmentally blocked FL cells, with some displaying aberrant activation-induced cytidine deaminase activity linked to malignant progression. Collectively, our data establish that expanded t(14;18)+ clones constitute bona fide precursors at various stages of FL development, and provide a molecular connection between agricultural pesticide exposure, t(14;18) frequency in blood, and clonal progression.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 375-375
Author(s):  
Sandrine Roulland ◽  
Julie Agopian ◽  
Yannick Lecluse ◽  
Jean-Marc Navarro ◽  
Anne-claire Gac ◽  
...  

Abstract Agricultural pesticide use has been repeatedly, but not consistently, associated with risk of non-Hodgkin lymphoma (NHL). Over the past years, it has become clear that NHL heterogeneity is setting many hurdles to this connection and that pertinent bio-markers must be identified according to malignant subtypes. Among those, t(14;18) translocation constitutes the early and promoting event of a cascade of causative effects leading to Follicular Lymphoma (FL): ectopic BCL2 expression leads to the rescue, accumulation, and developmental block of germinal center (GC) B-cells; there, protracted AID activity would favor the occurrence of complementary oncogenic hits and malignant progression, which arise from mistakes during somatic hypermutation (SHM) and class-switch recombination (CSR) processes. Although the significance of t(14;18) in blood from healthy individuals is yet unclear, evidence for a higher prevalence has been reported among farmers occupationally exposed to pesticides. Furthermore, recent findings have established that pesticide exposure is significantly associated with risk of t(14;18)+ NHL but not with t(14;18)-negative cases, suggesting that pesticides may act through a t(14;18)-dependant pathway. To get molecular insights into this connection, we followed and compared the status and clonal evolution of t(14;18)+ cells over a median of 9 years in blood samples issued from a cohort of farmers exposed to pesticides (n=112) with those of non-farmer controls (n=25). We report here that exposed individuals exhibit a much higher increase of t(14;18) frequency in blood, and that this increase result far less from a genotoxic effect of pesticides than from an immunogenic effect leading to an extensive clonal expansion of activated t(14;18)+ B cells. Strikingly, we demonstrate that such clones recapitulate the hallmark features of developmentally blocked FL cells as they 1) express high levels of BCL2/JH fusion transcripts; 2) retain the CD10 surface expression normally lost upon GC exit; and 3) sustain constitutive AID activity with the most advanced ones bearing, unexpectedly, early stigmata of AID-mediated genomic instability. Altogether these results show that expanded t(14;18)+ clones in farmers exposed to pesticides constitute bona fide FL precursors standing at various stages of tumor progression and support the notion of a direct connection between t(14;18) frequency in blood and malignant progression that could be could helpful in the identification of individuals at “high” risk for transformation to overt FL.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3444-3444
Author(s):  
Bay T Ho ◽  
Shyh-Jen Shih ◽  
Sheetal P Singh ◽  
Manuel O Diaz ◽  
Dawei Lin ◽  
...  

Abstract Abstract 3444 The MLL gene, encoding a histone methyl transferase, fuses with multiple different partner genes and is a common finding in patients with AML and ALL. This multiplicity of partners is in stark contrast to other fusion oncogenes such as BCR-ABL in CML or PML-RARA in APL, where fusions between the same genes predominate and which has aided the design of targeted therapy. In order to understand the fusion process in more detail, a hot spot for both MLL cleavage and gene fusion adjacent to exon 12 was examined in a series of breast cancer and lymphoma patients, using inverse PCR (IPCR). All patients received chemotherapy containing drugs targeting Topoisomerase II and samples of blood were examined both before and after therapy. Of the fifty patients enrolled in the study a subset was also examined by IPCR, parallel sequencing and custom bioinformatic analysis. The advantage of this approach is that all possible rearrangements may be examined within a single sample. In addition, the technique also records all sequences at the same location that are uninvolved in a translocation. The blood of three out of four patients examined in this way contained no evidence of MLL rearrangements. However in one patient, with a diagnosis of diffuse large B cell lymphoma, a total of thirteen MLL rearrangements were identified that were present prior to therapy. The majority of the fusions were detected for up to 6 months after the end of chemotherapy indicating they were likely of clonal origin. Of the thirteen fusions, five were predicted to provide functional fusion proteins and these included MLL-MLLT3 (AF9), the most common MLL fusion in myeloid leukemia. The remaining fusions predicted to generate functional proteins involved USP46, FER1L5, CCNJL and NKD1. None of these have been previously identified in clinical specimens of MLL linked disease. However, NKD1 is a negative regulator of the WNT pathway that has been linked to the maintenance of the stem cell phenotype in AML. In order to understand the fusion process in more detail, each fusion breakpoint was examined. All thirteen MLL fusions contained microhomology at the breakpoint, ranging from 1 to 6 bp, indicating NHEJ as the likely pathway generating the fusions. Though the presence of microhomology masked the precise breakpoint, using the 3' edge of microhomology as a reference, eight of the thirteen rearrangements were clustered within a 5 bp tract at the base of a putative stem-loop structure, as we have reported before. Such a restricted distribution of breakpoints found within a clonal population suggests a common mechanism is involved in either cleavage, and/or fusion, at this location. In order to address the mechanism driving these rearrangements, a screen of all sequenced material was undertaken. It was observed that within the residual sequenced material, a selective increase in C>T transitions were noted for two cytosines located within the 5bp breakpoint hotspot. The level of C>T transition was 50–100 fold higher than anywhere else within the sequence generated by IPCR. In addition, the region of C>T transition contained the WRC (A/T, A/G, C) motif characteristic of Activation-Induced Cytidine Deaminase (AID) attack. AID deaminates cytosine leading to a uracil which may be repaired by uracil-DNA glycosylase and the base excision repair (BER) pathway. AID has been implicated in both DNA breaks, via the BER pathway, and a linked increase in translocations. If the BER pathway fails to execute appropriately, a C>T transition may occur as the aberrantly located uracil undergoes replication at the next division. The presence of extensive C>T transitions therefore is indicative of both AID function and defective repair pathway(s). These factors may explain both the extensive number of rearrangements observed in this patient and the legacy C>T transitions from multiple AID attacks. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 181-181
Author(s):  
Tanja Gruber ◽  
Mi Sook Chang ◽  
Richard Sposto ◽  
Markus Müschen

Abstract Abstract 181 Activation-Induced Cytidine Deaminase (AID) is required for somatic hypermutation and immunoglobulin (Ig) class switch recombination in germinal center (GC) B cells. Occasionally, AID can target non-Ig genes and thereby promote GC B cell lymphomagenesis. We recently demonstrated that the oncogenic BCR-ABL1 kinase induces aberrant expression of AID in pre-B acute lymphoblastic leukemia (ALL). Compared to other ALL subtypes, BCR-ABL1 ALL is considered high risk and is characterized by a high degree of genetic instability. Because aberrant mutational activity of AID is associated with malignant transformation in B cell lymphoma, we sought to determine whether aberrant AID expression contributes to clonal evolution and genetic instability in Ph+ ALL. To investigate the function of AID expression in Ph+ ALL, we established a genetic loss-of-function model for Ph+ ALL: Bone marrow cells from AID−/− mice and AID+/+ controls were transformed by retroviral transduction with BCR-ABL1 under B lymphoid culture conditions and subsequently injected into lethally irradiated congenic recipients. Mice transplanted with AID−/−BCR-ABL1 ALL had prolonged median survival as compared to mice transplanted with leukemia cells generated from AID+/+ bone marrow (AID−/− 34 days (n=18) vs AID+/+ 13 days (n=21); p<0.0001). In secondary and tertiary transplant experiments, however, the difference between AID−/− and AID+/+BCR-ABL1 ALL narrowed as determined by a decreasing hazard ratio (from 25.5 in the primary transplant to 5.1 in the secondary and 2.9 in the tertiary transplantation). These findings suggest that aberrant AID expression accelerates clonal evolution of Ph+ ALL, but AID-independent factors exist that are sufficient for transformation. In support of enzymatic activity of AID in BCR-ABL1-transformed ALL cells, we observed that aberrant somatic hypermutation of non-immunoglobulin genes in these leukemias was largely dependent on AID: mutations in the known hypermutation target genes Pax5 and Rhoh were increased in AID+/+ but not AID−/−BCR-ABL1 ALL cells. Mutations in the first intron of Rhoh as observed here are relevant because they interfere with Rhoh transcription. Indeed, we found that Rhoh mRNA levels are significantly higher in AID−/− compared to AID+/+BCR-ABL1 ALL cells. Rhoh is a hematopoietic specific GTPase that negatively regulates Rac-mediated signaling downstream of the oncogenic BCR-ABL1 kinase. AID-dependent mutation and transcriptional inactivation of Rhoh in BCR-ABL1 ALL therefore likely augments oncogenic BCR-ABL1 signaling. Consistent with a causative role of AID in genetic instability, AID−/− leukemia had a lower frequency of amplifications (17+2 vs 45+7; p=0.002) and deletions (11+2 vs 40+7; p=0.003) as compared to AID+/+ leukemias. AID−/− and AID+/+ ALL cells showed a markedly distinct gene expression pattern with 2,365 differentially expressed genes (p=0.003; FDR 0.05). A detailed analysis of these differences in gene expression revealed that AID−/−BCR-ABL1 ALL cells failed to downregulate a number of tumor suppressor genes including p53, Rhoh, Cdkn1a (p21), and Blnk (SLP65). AID-dependent downregulation of p53 in BCR-ABL1 ALL cells is of particular importance, because previous work demonstrated that transcriptional repression of p53 in normal GC B cells is required to make these cells permissive to high levels of AID expression. AID-induced DNA damage would otherwise activate p53 and rapidly induce apoptosis. Compared to AID-deficient BCR-ABL1 ALL, AID+/+BCR-ABL1 ALL cells are more resistant to Imatinib-treatment. However, acquisition of BCR-ABL1 kinase domain mutations does not appear to be the main cause of drug-resistance in this experiment, since only one relevant mutation was amplified from AID+/+ ALL cells (no mutations in AID−/− ALL cells). We conclude that AID accelerates clonal evolution in BCR-ABL1 ALL by enhancing genetic instability, aberrant somatic hypermutation, and by negative regulation of tumor suppressor genes. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 214 (12) ◽  
pp. 3543-3552 ◽  
Author(s):  
Yunxiang Mu ◽  
Monika A. Zelazowska ◽  
Kevin M. McBride

Activation-induced cytidine deaminase (AID) is a mutator enzyme that targets immunoglobulin (Ig) genes to initiate antibody somatic hypermutation (SHM) and class switch recombination (CSR). Off-target AID association also occurs, which causes oncogenic mutations and chromosome rearrangements. However, AID occupancy does not directly correlate with DNA damage, suggesting that factors beyond AID association contribute to mutation targeting. CSR and SHM are regulated by phosphorylation on AID serine38 (pS38), but the role of pS38 in off-target activity has not been evaluated. We determined that lithium, a clinically used therapeutic, induced high AID pS38 levels. Using lithium and an AID-S38 phospho mutant, we compared the role of pS38 in AID activity at the Ig switch region and off-target Myc gene. We found that deficient pS38 abated AID chromatin association and CSR but not mutation at Myc. Enhanced pS38 elevated Myc translocation and mutation frequency but not CSR or Ig switch region mutation. Thus, AID activity can be differentially targeted by phosphorylation to induce oncogenic lesions.


2014 ◽  
Vol 192 (10) ◽  
pp. 4887-4896 ◽  
Author(s):  
Lyne Khair ◽  
Jeroen E. J. Guikema ◽  
Erin K. Linehan ◽  
Anna J. Ucher ◽  
Niek G. J. Leus ◽  
...  

2016 ◽  
Vol 214 (1) ◽  
pp. 49-58 ◽  
Author(s):  
Qiao Wang ◽  
Kyong-Rim Kieffer-Kwon ◽  
Thiago Y. Oliveira ◽  
Christian T. Mayer ◽  
Kaihui Yao ◽  
...  

Activation-induced cytidine deaminase (AID) converts cytosine into uracil to initiate somatic hypermutation (SHM) and class switch recombination (CSR) of antibody genes. In addition, this enzyme produces DNA lesions at off-target sites that lead to mutations and chromosome translocations. However, AID is mostly cytoplasmic, and how and exactly when it accesses nuclear DNA remains enigmatic. Here, we show that AID is transiently in spatial contact with genomic DNA from the time the nuclear membrane breaks down in prometaphase until early G1, when it is actively exported into the cytoplasm. Consistent with this observation, the immunoglobulin (Igh) gene deamination as measured by uracil accumulation occurs primarily in early G1 after chromosomes decondense. Altering the timing of cell cycle–regulated AID nuclear residence increases DNA damage at off-target sites. Thus, the cell cycle–controlled breakdown and reassembly of the nuclear membrane and the restoration of transcription after mitosis constitute an essential time window for AID-induced deamination, and provide a novel DNA damage mechanism restricted to early G1.


2007 ◽  
Vol 67 (14) ◽  
pp. 6555-6564 ◽  
Author(s):  
Sergey W. Popov ◽  
Gerhard Moldenhauer ◽  
Beate Wotschke ◽  
Silke Brüderlein ◽  
Thomas F. Barth ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4816-4816
Author(s):  
Rebecca J Leeman-Neill ◽  
Daniel E Johnson ◽  
Steven H Swerdlow

Abstract Several epidemiologic studies have demonstrated an increased risk of non-Hodgkin B cell lymphoma with exposure to pesticides, including herbicides, fungicides and insecticides. Few studies have specifically demonstrated an increased risk for follicular lymphoma in individuals exposed to pesticides. The molecular mechanisms of these associations have not been fully investigated. Activation-induced cytidine deaminase (AID), which physiologically mediates somatic hypermutation in germinal center B cells, has been found to have off-target effects that play an important role in oncogenic mutagenesis driving germinal center type B cell lymphomas. Elevated levels of AID among peripheral blood mononuclear cells has been found to be associated with higher numbers of circulating follicular lymphoma-like cells in individuals exposed to pesticides. Increased AID expression and/or activity represents a possible mechanistic link between pesticide exposure and lymphomagenesis. In order to test the hypothesis that pesticides might lead to an increased risk of germinal center neoplasia by increasing AID levels, four pesticides including isoproturon (a phenylurea herbicide), chlorpyrifos (an organophosphate insecticide), DDT (an organochlorine insecticide), and lindane (an organochlorine insecticide), were screened for their ability to increase AID expression in germinal center type B cells (Ramos and Raji, Burkitt lymphoma cell lines). After 48 hours of treatment with concentrations ranging from 50-200 μM, RNA was extracted, reverse transcribed and cDNA subject to SYBR green Realtime PCR amplification of AID. AID was found to be expressed, at low levels, in untreated Ramos and Raji cells, compared to acute myeloid leukemia cell lines HL60 and U937, which served as negative controls showing no AID expression. Isoproturon treatment resulted in a dose dependent, up to 6-fold, increase in AID in Ramos cells. A smaller, up to 1.9 fold increase was seen in Raji cells. None of the other pesticide treatments resulted in increased expression of AID (Fig. 1). The findings demonstrate that AID expression is induced by isoproturon, a member of the class of phenylurea herbicides which is among the most commonly used herbicides worldwide and generally considered to be relatively safe for humans. These results support a possible role for off-target AID activity in pesticide-exposure related lymphomagenesis. Future studies will investigate the mechanism by which AID is upregulated andwhether or not there is induction of oncogenic mutagenesis in germinal center B cells exposed to phenylurea herbicides. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document