scholarly journals Engineering species-like barriers to sexual reproduction

2016 ◽  
Author(s):  
Maciej Maselko ◽  
Stephen C. Heinsch ◽  
Jeremy Chacón ◽  
William Harcombe ◽  
Michael J. Smanski

We introduce a novel approach to engineer a genetic barrier to sexual reproduction between otherwise compatible populations. Programmable transcription factors drive lethal gene expression in hybrid offspring following undesired mating events. As a proof of concept, we target the ACT1 promoter of the model organism Saccharomyces cerevisiae using a dCas9-based transcriptional activator. Lethal over-expression of actin results from mating this engineered strain with a strain containing the wild-type ACT1 promoter.

2008 ◽  
Vol 68 (2) ◽  
pp. 257-263 ◽  
Author(s):  
M Corr ◽  
D L Boyle ◽  
L Ronacher ◽  
N Flores ◽  
G S Firestein

Objectives:The IκB kinase (IKK)-related kinase IKKϵ regulates type I interferon expression and responses as well as proinflammatory mediator production. We examined the role of IKKϵ in arthritis and its ability to enhance the therapeutic response to systemic interferon (IFN) β therapy in passive murine K/BxN arthritis.Methods:IKKϵ–/–, IFNα∼βR–/– and wild type mice were given K/BxN serum and treated with polyinosinic polycytidylic acid (poly(I:C)), IFNβ, or normal saline. Clinical response and histological scores were assessed. Gene expression in the paws was measured by quantitative PCR. Serum interleukin 1a receptor agonist (IL1Ra) and IL10 were measured by ELISA and multiplex bead array.Results:Arthritis was almost completely blocked in wild type mice if arthritogenic K/BxN serum and the Toll-like receptor (TLR)3 ligand, poly(I:C), were coadministered at the onset of the model, but not in established disease. Mice deficient in IFNα∼βR had an accelerated course of arthritis, and did not respond to poly(I:C). IKKϵ null mice had a modest decrease in clinical arthritis compared with heterozygous mice. Low doses of IFNβ that were ineffective in wild type mice significantly decreased clinical arthritis in IKKϵ null mice. Articular chemokine gene expression was reduced in the IKKϵ–/– mice with arthritis and secreted IL1Ra (sIL1Ra) mRNA was significantly increased. Serum levels of IL1Ra were increased in low dose IFNβ-treated IKKϵ–/– mice.Conclusions:Subtherapeutic doses of IFNβ enhance the anti-inflammatory effects of IKKϵ deficiency, possibly by increasing production of IL1Ra and unmasking the antichemokine effects. Combination therapy with low dose IFNβ and an IKKϵ inhibitor might improve efficacy of either agent alone and offers a novel approach to RA.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 743-743
Author(s):  
Jingya Wang ◽  
Andrew G. Muntean ◽  
Jay L. Hess

Abstract Abstract 743 MLL gene rearrangements are among the most common chromosomal abnormalities associated with both acute lymphoid (ALL) and myeloid (AML) leukemia. Both MLL and leukemogenic MLL fusion proteins directly regulate expression of HoxA9 and the co-factor Meis1. Normally Hoxa9 and Meis1 are expressed at high levels in early hematopoietic progenitors, where they promote HSC self-renewal. MLL fusion proteins block this physiologic down regulation so that Hoxa9 and Meis1 are persistently expressed, resulting in leukemia. The mechanisms through which the activity of MLL is normally regulated are poorly understood. MLL fusion proteins include the first 1400 amino acids of MLL and invariably delete the plant homeodomain (PHD)/Bromodomain regions (including 4 PHD fingers, with bromodomain between the 3rd and 4th PHD finger) and C terminal SET domain. Previously we showed that inclusion of the MLL PHD domain in MLL fusion proteins prevents transformation. To further explore the function of this potential regulatory domain, we identified proteins that interact with the CxxC and PHD domains by immunoaffinity purification and mass spectroscopy. These studies identified subunits of the Elongin-Cullin-Socs box (ECS) complex including Elongin B, Elongin C, Cullin 5 and Ankyrin Repeat and SOCS box (ASB) E3 ligases. Biochemical experiments on a panel of ASB proteins revealed that ASB2, which functions as a substrate recognition subunit of the ECS complex specifically and potently degrades MLL. Over expression of ASB2 enhanced the degradation of MLL, while ASB2 knockdown results in MLL protein stabilization. Moreover, co-expressed ASB2 abolished MLL mediated transcriptional activation of a HoxA9 reporter. The interaction region on ASB2 maps to the five N terminal ankyrin repeats. Since the ASB2 interaction is mediated through a region of MLL invariably deleted from fusion proteins, we predicted that MLL fusions would be significantly more stable than full length MLL. Indeed, we measured the half-life of wild type MLL to be ~7hrs, and ~16hrs in the presence of MG132 proteasome inhibition. In contrast, MLL-AF9 displayed a half-life of ~60hrs, and MG132 treatment slightly increased the half-life to ~76hrs. Gene expression analysis of different hematopoietic cell populations indicates that Asb2 expression increases dramatically with differentiation. Also, All-trans retinoid acid (ATRA) induced differentiation of NB4 and K562 cells is associated with up regulation of Asb2 and decreased MLL protein level and Hox gene expression, while MLL transcription is largely unchanged. Furthermore, expression of ASB2 in primary mouse bone marrow cells results in accelerated differentiation and decreased expression of several MLL target genes compared to vector control and over expression of ASB2 in MLL-fusion transformed murine cell lines dramatically decreased colony formation, consistent with recently published data that wild type MLL is required for MLL fusion transformation. Together this work reveals a novel ubiquitination pathway that regulates MLL at the posttranslational level, which is likely to be important for both normal hematopoiesis and the dysregulated transcription that is seen in leukemias with MLL rearrangements. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1257-1257
Author(s):  
Meng Liu ◽  
Limengmeng Wang ◽  
Huafang Wang ◽  
Shan Fu ◽  
He Huang ◽  
...  

Introduction: CEBPA gene encodes CCAAT/enhancer-binding protein-alpha (C/EBPα), a crucial granulocytic differentiation factor and tumor suppressor in hematologic and many non-hematologic malignancies. We previously reported a donor-derived relapse of AML patient after allogeneic hematopoietic stem cell transplantation with multiple mutations in CEBPA gene: a N-terminal frameshift mutation (247dupC causing overproduction of truncated 30-KDa isoform, lacking the TAD1 domain, p30), a N-terminal germ-line mutation (584_589dup disrupting the TAD2 domain of protein, NM2), and a C-terminal mutation (914_916dup disrupting the bZIP domain, CM) (Blood 2011; 117: 5257-5260). Although studies from multiple laboratories have contributed immensely to our understanding that how different CEBPA mutations disturb C/EBPα functions including granulopoiesis and leukemic transformation in AML, whether C/EBPα might regulate immunosurveillance remains unknown. Methods: AML cell line cells infected with lentivirus to over-express of wild type C/EBPα as well as 3 types of C/EBPα mutants were co-cultured with NK92MI cells and detected cytotoxic lysis through FCM. NK92MI cells were stained with CD107a to detect degranulation.We performed gene expression microarray profiling analysis in AML cell line cells with over-expression of wild type C/EBPα and mutants . Flag tagged wide type C/EBPα was over-expressed in 293T cells and ChIP with anti-Flag antibody followed by sequencing assay was performed to explore candidate gene binding sites of C/EBPα. Finally, independent ChIP-qPCR of candidate sequences were performed to further verify the transcription factor binding sites of C/EBPα. Results: ULBPs expressed on the surface of tumor or infected cells are important ligands of NK cell receptor NKG2D. Our gene expression microarray profiling analysis showed that wild type C/EBPα could up-regulate the expression of ULBP2/5/6 in AML cell line cells. Consistent with the results of gene expression microarray profiling analysis, over-expression of wild type C/EBPα and a N-terminal germ-line mutant (NM2) can up-regulate ULBP2/5/6 expression in NB4 cells, whose endogenous expression of ULBPs was low. Meanwhile, the sensitivities of NB4 cells to the cytotoxicity of NK92MI cells were also increased by over-expression of wide type C/EBPα and NM2 mutant. In contrast, leukemia-associated somatic mutations, C/EBPα p30 and C-terminal mutant (CM), were disabled to up-regulate ULBPs expression. In dual-luciferase reporter assay, the ratio of level of firefly luciferase and renilla luciferase significantly increased when co-transduced report plasmid with wide type C/EBPα expressing plasmid compared with vector control, indicating that C/EBPα could up-regulate the transcription of ULBP2 as a transcription factor. Through ChIP-seq assay we identified 12 peaks nearby ULBP genes in chromosome 6. We further performed ChIP-qPCR to target the sequences acting as enhancers of ULBP genes, which located +7kb upstream of transcription start site of ULBP2 gene, +11kb upstream of ULBP5 gene, -9kb downstream of ULBP6 gene and -40kb downstream of ULBP1 gene. Wide type C/EBPα showed higher binding affinity to the ULBP2/5 enhancers with more than 50 folds' enrichment and to the ULBP6/1 enhancers 9 folds' enrichment compared with IgG control. The N-terminal germ-line mutant (NM2) conserved part of the binding affinity, but the enrich fold was lower than wide type. As expected, leukemia-associated C-terminal mutant (CM) totally lost its binding ability to both sequences due to the damage of DNA binding domain. Althoughleukemia-associated truncated 30-KDa isoform partly conserved its binding ability to these DNA sequences, the mutant lost the function of regulating ULBPs expression. Conclusions: C/EBPα played an important role in innate immunosurveillance of AML. C/EBPα could bind to the promoter and potential enhancers of ULBP genes as a transcription factor, up-regulate expression of ULBPs and eventually induce AML cells to be recognized and killed by NK cells. Mutations in TAD2 domain did not affect this regulation function, while mutations in TAD1 and bZIP domain lost the specific ability. Leukemia cells with N-terminal frameshift mutations (p30), or C-terminal mutations could escape from surveillance of NK cells and may play pivotal roles in leukemia relapse. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1919-1919
Author(s):  
Manujendra N. Saha ◽  
Yijun Yang ◽  
Hong Chang

Abstract PRIMA-1Met/APR246 (p53 reactivation and induction of massive apoptosis), is a small molecule with remarkable anti-tumor activities in various human tumor cells, and is currently under phase I/II clinical trial. We have previously demonstrated anti-tumor activity of PRIMA-1Met in multiple myeloma (MM) cells irrespective of p53 status. In addition, we have shown that PRIMA-1Met alone or in combination with dexamethasone triggers significant tumor growth inhibition in vivo in a murine xenograft model of human MM. However, the molecular mechanism underlying anti-myeloma activity of PRIMA-1Met has not been fully elucidated. MicroRNAs (miRNAs) are non-coding small RNA molecules that regulate post-transcriptional gene expression and play a critical role in tumor pathogenesis. Since the role of miRNAs and their regulation in response to PRIMA-1Met in MM is not known, here we investigated the relationship between PRIMA-1Met-induced apoptosis and miRNA expression in MM cells. Using a miRNA PCR array platform (Human Cancer Pathway Finder miScript miRNA PCR array, MIHS-102Z, Qiagen Inc), we analyzed the miRNA profiles in two MM cell lines of different p53 status (MM.1S with wild type p53 and 8226 with mutant p53) treated with either PRIMA-1Met or DMSO control. After normalization to a set of housekeeping genes, differential expressions of the miRNAs were analysed. miRNA-29a, miRNA-29b, and miRNA-34a were found significantly up-regulated (more than 2 fold, p<0.05) in cells treated with PRIMA-1Met compared to DMSO-treated cells. To evaluate the effect of over-expression of these miRNAs, we transfected two MM cell lines (MM.1S and 8226) with either miR-29a/b or miR-34a. Cells transfected with scramble miRNA were used as control. Over-expression of the miRNAs resulted in a dose-dependent inhibition of viability and increase in apoptosis of MM.1S or 8226 cells. Next, we examined the endogenous expression of these miRNAs in 5 primary MM samples by qPCR. Results showed a significant low expression of miR-29a/b and miR-34a in 3 of the 5 samples. Treatment of the two primary MM samples with low expression for miR-29a/b and miR-34a with PRIMA-1Met resulted in up-regulation of these miRNAs leading to inhibition of the viability and induction of apoptosis. To identify the possible targets of these miRNAs, we performed bioinformatics analysis. Results obtained from different searches by miRanda and TargetScan algorithm predicted c-Myc as a potential target for miRNA-29a/b and miRNA-34a. c-Myc is an oncogene whose over-expression has been associated with resistance to current chemotherapy in MM. Global gene expression profiling by microarray showed significant down-regulation of c-Myc in two MM cell lines with either wild type or mutant p53 treated with PRIMA-1Met compare to cells treated with DMSO. Importantly, down-regulation of c-Myc (∼2.6-fold) by PRIMA-1Met was also observed in a MM cell line (8226R5) lacking p53 expression suggesting an important role of c-Myc in p53-independent apoptosis of MM cells induced by PRIMA-1Met. By qPCR and Western blot analysis, we confirmed significant down-regulation of c-Myc in PRIMA-1Met-treated MM cells. These data provided the evidence for an inverse correlation between the expression of these miRNAs and c-Myc indicating that apoptosis of MM cells induced by PRIMA-1Met is regulated by miRNAs29a/b or miRNA34a targeting c-Myc. Our results suggest a novel mechanism for PRIMA-1Met-induced apoptotic signaling in MM cells mediated by up-regulation of miR-29a/b and miR-34a targeting c-Myc. Our findings also provide a preclinical framework for development of therapeutic strategies in combination of PRIMA-1Met and miRNA (miR-29a/b or miR-34a) mimics for the treatment of MM patients, especially for those with high c-Myc expressions. Disclosures: No relevant conflicts of interest to declare.


2000 ◽  
Vol 72 (9) ◽  
pp. 1615-1622 ◽  
Author(s):  
Manfred T. Reetz

A novel approach to developing enantioselective enzymes for use in organic chemistry has been devised which is independent of structural or mechanistic aspects. The underlying idea is to combine appropriate methods of random mutagenesis, gene expression, and high-throughput screening for enantioselectivity. If these actions are performed in repetitive cycles, an evolutionary pressure is created that leads to sequential improvements of the enantioselectivity of a given enzyme-catalyzed reaction. The concept is illustrated by an example involving the lipase-catalyzed hydrolytic kinetic resolution of an α-chiral ester, the enantio-selectivity increasing from ee = 2% (E =1.1) for a wild-type enzyme to ee = 90-93% (E = 25) for the best mutants.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Simona K. Zahova ◽  
Trevor Humby ◽  
Jennifer R. Davies ◽  
Joanne E. Morgan ◽  
Anthony R. Isles

AbstractPrader-Willi Syndrome (PWS) is a neurodevelopmental disorder caused by mutations affecting paternal chromosome 15q11-q13, and characterized by hypotonia, hyperphagia, impaired cognition, and behavioural problems. Psychotic illness is a challenging problem for individuals with PWS and has different rates of prevalence in distinct PWS genotypes. Previously, we demonstrated behavioural and cognitive endophenotypes of relevance to psychiatric illness in a mouse model for one of the associated PWS genotypes, namely PWS-IC, in which deletion of the imprinting centre leads to loss of paternally imprinted gene expression and over-expression of Ube3a. Here we examine the broader gene expression changes that are specific to the psychiatric endophenotypes seen in this model. To do this we compared the brain transcriptomic profile of the PWS-IC mouse to the PWS-cr model that carries a deletion of the PWS minimal critical interval spanning the snoRNA Snord116 and Ipw. Firstly, we examined the same behavioural and cognitive endophenotypes of relevance to psychiatric illness in the PWS-cr mice. Unlike the PWS-IC mice, PWS-cr exhibit no differences in locomotor activity, sensory-motor gating, and attention. RNA-seq analysis of neonatal whole brain tissue revealed a greater number of transcriptional changes between PWS-IC and wild-type littermates than between PWS-cr and wild-type littermates. Moreover, the differentially expressed genes in the PWS-IC brain were enriched for GWAS variants of episodes of psychotic illness but, interestingly, not schizophrenia. These data illustrate the molecular pathways that may underpin psychotic illness in PWS and have implications for potential therapeutic interventions.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5111-5111
Author(s):  
Julia Abramowitz ◽  
Tzahi Neuman ◽  
Yoav Smith ◽  
Rania Samman ◽  
Alex Gural ◽  
...  

Abstract Abstract 5111 The pathway controlled by the p53 tumor-suppressor protein is altered in most, if not all, human cancers and the TP53 gene itself is mutated in half of all human tumors. However, mutations in the TP53 gene are rare in human hematological malignancies. This implies that p53 is inactivated by alternative mechanisms such as over expression of its negative regulators. p53 is negatively regulated by Mdm2 through ubiquitin-dependent degradation and by Mdmx through inhibition of transcriptional function. To date, there is no information on the role of Mdm2 and Mdmx in human Acute Promyeloytic Leukemia (APL). We investigated the involvement of these negative regulators of p53 in APL at the gene and protein expression levels. First, we directly sequenced TP53 in 21 APL samples. In all cases TP53 was found to be wild type as expected. We studied Mdm2 and Mdmx gene expression in bone marrow samples of 30 APL patients at diagnosis in comparison to 35 normal bone marrow samples. Quantitative Real-Time PCR analysis showed no statistically significant differences in expression of Mdm2, Mdmx full length or its splicing variant (Mdmx-S) between APL and control samples. Bioinformatics analysis of gene expression of 39 APL patients at diagnosis, using publicly available arrays, showed homogeneous gene expression pattern between patients. We compared the APL arrays to 5 normal promyelocytes arrays. In agreement with our results, no significant difference was detected between APL and normal promyelocytes in the levels of Mdm2 or Mdmx gene expression. Mdm2 and Mdmx are subjected to complex regulation at the protein level. We therefore investigated the level of these proteins by immunohistochemical staining. Bone marrow biopsies from 23 APL patients at diagnosis were compared to 30 normal biopsies and protein levels were evaluated by semi-quantitative score. We found that Mdmx protein was low in APL samples and not significantly different from normal bone marrow. Thus, in APL, Mdmx does not appear to play a major role in p53 inhibition. Remarkably, APL samples showed a bi-modal expression of Mdm2 protein: 11/23 (48%) APL samples had significantly down-regulated Mdm2 protein (p<0.001). This might imply that Mdm2 is not involved in p53 inactivation in these patients. The other 12/23 (52%) patients showed significant over-expression of the Mdm2 protein (p<0.001). This result suggests that in half of APL patients, the p53 pathway may be inactivated by Mdm2. Interestingly, the subset of patients with high Mdm2 protein expression, was also characterized by high levels of p53 (5/12, p=0.058) and the pro-apoptotic p53 target, Puma (5/12). In summary, TP53 is wild type in APL. We found that the Mdm2 protein, a negative regulator of p53, may be accountable for p53 inactivation in about half of APL patients. In the other cases, a different, yet to be determined, mechanism is involved in inhibiting p53. Thus, APL is not a homogeneous disease regarding the inhibition of p53 pathway. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1807-1807
Author(s):  
Yuan Xiao Zhu ◽  
Chang-Xin Shi ◽  
Laura Bruins ◽  
Klaus Martin Kortuem ◽  
Jessica Schmidt ◽  
...  

Abstract Abstract 1807 We have recently demonstrated that cereblon (CRBN) mediates the direct anti-myeloma activity of immunomodulatory drugs (IMiDs). However, the genes/pathways downstream of CRBN associated with anti-myeloma activity remain unclear. We, and others, identified interferon regulatory factor 4 (IRF4) as one of the downstream targets of CRBN-associated signaling. Both lenalidomide treatment and CRBN knockdown downregulate IRF4. IRF4 levels return to baseline in IMiD resistant cells surviving CRBN silencing. To determine whether IMiD-induced IRF4 downregulation is critical to anti-MM activity, we overexpressed IRF4 in two IMiD-sensitive human MM cell lines (HMCLs), KMS11 and MM1.S, followed by lenalidomide treatment. Lenalidomide-induced cytotoxicity was greatly impaired in both HMCLs overexpressing IRF4 compared with the control virus infected cells. Further analysis indicated that IRF4 over-expression does not completely prevent lenalidomide-induced growth arrest, but reduces cell death by 70% after lenalidomide treatment. Immunoblotting analysis of KMS11 cells indicated that IRF4 over-expression blocks lenalidomide-induced activation of caspase 8, reduces up-regulation of p21waf and increases CDK6 expression but does not significantly affect lenalidomide-induced MYC down-regulation. Although cereblon and IRF4 are broadly expressed in MM, baseline levels of expression are only weakly correlated (r=0.22) in primary MM patient gene expression analysis. Gene expression studies revealed statistical changes in 1,368 genes when comparing high versus low CRBN expression in primary myeloma samples. Interestingly genes associated with high CRBN expression included cyclin D2, SOCS3 and IL4 while genes associated with low cereblon expression included cyclin D1, FRZB and CD200. In order to understand how CRBN is connected with downstream anti-myeloma signaling, a structure-function study was performed to determine which CRBN domain is required for lenalidomide-induced IRF4 down-regulation and cytotoxicity. Lentiviral constructs expressing wild-type CRBN and a series of mutated CRBN were generated, including mutations at thalidomide binding site (Y384A/W386A), deletion of DDB1 binding region (ΔMid) and truncations at N-terminal and C-terminal. Lentiviruses from these constructs were used to infect IMiDs resistant HMCLs, OCI-MY5 and MM1.S res. Both of these cell lines have very low endogenous CRBN expression and they became sensitive to lenalidomide after introduction of wild-type CRBN. Conversely, introduction of CRBN with mutated thalidomide-binding site or with DDB1 binding region depletion failed to mediate lenalidomide toxicity and down-regulation of IRF4. OCI-MY5 cells expressing either N-terminal or C-terminal truncated CRBN showed substantial reduced responses (more than 50%) to lenalidomide compared with wild-type CRBN expressing cells. Deletion of only 20–30 amino acids at either ends of CRBN greatly impaired the protein function, suggesting that protein folding might be important for CRBN-mediated IMiD response. Our data indicate that IMiD induced myeloma cytotoxicity is largely mediated by modifying CRBN associated E3 ubiqutin ligase and subsequent IRF4 downregulation, suggesting the CRBN-IRF4 axis is a potential target for development of new anti-myeloma drugs. Disclosures: Schmidt: Karyopharm: Research Funding. Stewart:Millenium: Consultancy, Honoraria, Research Funding; Onyx: Consultancy; Celgene: Consultancy.


Diseases ◽  
2020 ◽  
Vol 8 (1) ◽  
pp. 2
Author(s):  
Chalongchai Chalermwat ◽  
Thitipa Thosapornvichai ◽  
Laran T. Jensen ◽  
Duangrurdee Wattanasirichaigoon

Citrin is a liver-specific mitochondrial aspartate–glutamate carrier encoded by SLC25A13. Citrin deficiency caused by SLC25A13 mutation results in carbohydrate toxicity, citrullinemia type II, and fatty liver diseases, the mechanisms of some of which remain unknown. Citrin shows a functional homolog in yeast aspartate-glutamate carrier (Agc1p) and agc1Δ yeasts are used as a model organism of citrin deficiency. Here, we found that agc1Δ yeasts decreased fat utilization, impaired NADH balance in peroxisomes, and decreased chronological lifespan. The activation of GPD1-mediated NAD+ regeneration in peroxisomes by GPD1 over-expression or activation of the malate–oxaloacetate NADH peroxisomal shuttle, by increasing flux in this NADH shuttle and over-expression of MDH3, resulted in lifespan extension of agc1Δ yeasts. In addition, over-expression of PEX34 restored longevity of agc1Δ yeasts as well as wild-type cells. The effect of PEX34-mediated longevity required the presence of the GPD1-mediated NADH peroxisomal shuttle, which was independent of the presence of the peroxisomal malate–oxaloacetate NADH shuttle and PEX34-induced peroxisome proliferation. These data confirm that impaired NAD+ regeneration in peroxisomes is a key defect in the yeast model of citrin deficiency, and enhancing peroxisome function or inducing NAD+ regeneration in peroxisomes is suggested for further study in patients’ hepatocytes.


2021 ◽  
Author(s):  
Ambuj Upadhyay ◽  
Nathan Feltman ◽  
Adam Sychla ◽  
Siba Das ◽  
Maciej Maselko ◽  
...  

Engineered Genetic Incompatibility (EGI) is a method to create species-like barriers to sexual reproduction. It has applications in pest control that mimic Sterile Insect Technique when only EGI males are released. This can be facilitated by introducing conditional female-lethality to EGI strains to generate a sex-sorting incompatible male system (SSIMS). Here we demonstrate a proof of concept by combining tetracycline-controlled female lethality constructs with a pyramus-targeting EGI line in the model insect Drosophila melanogaster. We show that both functions (incompatibility and sex-sorting) are robustly maintained in the SSIMS line and that this approach is effective for population suppression in cage experiments. Further we show that SSIMS males remain competitive with wild-type males for reproduction with wild-type females, including at the level of sperm competition.


Sign in / Sign up

Export Citation Format

Share Document