Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Drug Resist Updat ; 74: 101081, 2024 May.
Article in English | MEDLINE | ID: mdl-38521003

ABSTRACT

Precision oncology has revolutionized the treatment of ALK-positive lung cancer with targeted therapies. However, an unmet clinical need still to address is the treatment of refractory tumors that contain drug-induced resistant mutations in the driver oncogene or exhibit resistance through the activation of diverse mechanisms. In this study, we established mouse tumor-derived cell models representing the two most prevalent EML4-ALK variants in human lung adenocarcinomas and characterized their proteomic profiles to gain insights into the underlying resistance mechanisms. We showed that Eml4-Alk variant 3 confers a worse response to ALK inhibitors, suggesting its role in promoting resistance to targeted therapy. In addition, proteomic analysis of brigatinib-treated cells revealed the upregulation of SRC kinase, a protein frequently activated in cancer. Co-targeting of ALK and SRC showed remarkable inhibitory effects in both ALK-driven murine and ALK-patient-derived lung tumor cells. This combination induced cell death through a multifaceted mechanism characterized by profound perturbation of the (phospho)proteomic landscape and a synergistic suppressive effect on the mTOR pathway. Our study demonstrates that the simultaneous inhibition of ALK and SRC can potentially overcome resistance mechanisms and enhance clinical outcomes in ALK-positive lung cancer patients. ONE SENTENCE SUMMARY: Co-targeting ALK and SRC enhances ALK inhibitor response in lung cancer by affecting the proteomic profile, offering hope for overcoming resistance and improving clinical outcomes.


Subject(s)
Anaplastic Lymphoma Kinase , Drug Resistance, Neoplasm , Lung Neoplasms , Organophosphorus Compounds , Protein Kinase Inhibitors , Proteome , src-Family Kinases , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Humans , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism , Mice , Proteome/metabolism , Protein Kinase Inhibitors/pharmacology , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Pyrimidines/pharmacology , Proteomics/methods , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Signal Transduction/drug effects
2.
Genome Biol ; 24(1): 267, 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-38001542

ABSTRACT

BACKGROUND: RNA editing has been described as promoting genetic heterogeneity, leading to the development of multiple disorders, including cancer. The cytosine deaminase APOBEC3B is implicated in tumor evolution through DNA mutation, but whether it also functions as an RNA editing enzyme has not been studied. RESULTS: Here, we engineer a novel doxycycline-inducible mouse model of human APOBEC3B-overexpression to understand the impact of this enzyme in tissue homeostasis and address a potential role in C-to-U RNA editing. Elevated and sustained levels of APOBEC3B lead to rapid alteration of cellular fitness, major organ dysfunction, and ultimately lethality in mice. Importantly, RNA-sequencing of mouse tissues expressing high levels of APOBEC3B identifies frequent UCC-to-UUC RNA editing events that are not evident in the corresponding genomic DNA. CONCLUSIONS: This work identifies, for the first time, a new deaminase-dependent function for APOBEC3B in RNA editing and presents a preclinical tool to help understand the emerging role of APOBEC3B as a driver of carcinogenesis.


Subject(s)
Neoplasms , RNA Editing , Humans , Animals , Mice , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Mutation , Neoplasms/pathology , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , DNA/metabolism
3.
Cell Death Dis ; 14(7): 430, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37452072

ABSTRACT

Tumor progression and evolution are frequently associated with chromosomal instability (CIN). Tumor cells often express high levels of the mitotic checkpoint protein MAD2, leading to mitotic arrest and cell death. However, some tumor cells are capable of exiting mitosis and consequently increasing CIN. How cells escape the mitotic arrest induced by MAD2 and proliferate with CIN is not well understood. Here, we explored loss-of-function screens and drug sensitivity tests associated with MAD2 levels in aneuploid cells and identified that aneuploid cells with high MAD2 levels are more sensitive to FOXM1 depletion. Inhibition of FOXM1 promotes MAD2-mediated mitotic arrest and exacerbates CIN. Conversely, elevating FOXM1 expression in MAD2-overexpressing human cell lines reverts prolonged mitosis and rescues mitotic errors, cell death and proliferative disadvantages. Mechanistically, we found that FOXM1 facilitates mitotic exit by inhibiting the spindle assembly checkpoint (SAC) and the expression of Cyclin B. Notably, we observed that FOXM1 is upregulated upon aneuploid induction in cells with dysfunctional SAC and error-prone mitosis, and these cells are sensitive to FOXM1 knockdown, indicating a novel vulnerability of aneuploid cells.


Subject(s)
Cell Cycle Proteins , Mitosis , Humans , Cell Cycle Proteins/metabolism , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Mitosis/genetics , Cell Line, Tumor , Spindle Apparatus/metabolism , Aneuploidy , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism
4.
Eur Respir J ; 60(1)2022 07.
Article in English | MEDLINE | ID: mdl-34887322

ABSTRACT

BACKGROUND: Survival after curative resection of early-stage lung adenocarcinoma (LUAD) varies and prognostic biomarkers are urgently needed. METHODS: Large-format tissue samples from a prospective cohort of 200 patients with resected LUAD were immunophenotyped for cancer hallmarks TP53, NF1, CD45, PD-1, PCNA, TUNEL and FVIII, and were followed for a median of 2.34 (95% CI 1.71-3.49) years. RESULTS: Unsupervised hierarchical clustering revealed two patient subgroups with similar clinicopathological features and genotype, but with markedly different survival: "proliferative" patients (60%) with elevated TP53, NF1, CD45 and PCNA expression had 50% 5-year overall survival, while "apoptotic" patients (40%) with high TUNEL had 70% 5-year survival (hazard ratio 2.23, 95% CI 1.33-3.80; p=0.0069). Cox regression and machine learning algorithms including random forests built clinically useful models: a score to predict overall survival and a formula and nomogram to predict tumour phenotype. The distinct LUAD phenotypes were validated in The Cancer Genome Atlas and KMplotter data, and showed prognostic power supplementary to International Association for the Study of Lung Cancer tumour-node-metastasis stage and World Health Organization histologic classification. CONCLUSIONS: Two molecular subtypes of LUAD exist and their identification provides important prognostic information.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Humans , Lung Neoplasms/pathology , Phenotype , Prognosis , Proliferating Cell Nuclear Antigen/genetics , Prospective Studies
5.
Cancers (Basel) ; 13(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34885137

ABSTRACT

Aneuploidy, an imbalance number of chromosomes, is frequently observed in lung cancer and inversely correlates with patient survival. Paradoxically, an aneuploid karyotype has detrimental consequences on cellular fitness, and it has been proposed that aneuploid cells, at least in vitro, generate signals for their own elimination by NK cells. However, how aneuploidy affects tumor progression as well as the interplay between aneuploid tumor cells and the tumor microenvironment is still unclear. We generated a new mouse model in which overexpression of Mad2 was almost entirely restricted to normal epithelial cells of the lung, and combined it with an oncogenic Eml4-Alk chromosome inversion. This combination resulted in a higher tumor burden and an increased number of tumor nodules compared to control Eml4-Alk mice alone. The FISH analysis detected significant differences in the aneuploidy levels in the non-tumor regions of Eml4-Alk+Mad2 compared to Eml4-Alk alone, although both tumor groups presented similar levels of aneuploidy. We further show that aneuploid cells in the non-tumor areas adjacent to lung tumors recruit immune cells, such as tumor-associated macrophages. In fact, these areas presented an increase in alveolar macrophages, neutrophils, decreased cytotoxic CD8+ T cells, and IFN-γ, suggesting that aneuploid cells in the surrounding tumor areas create an immunosuppressive signature that might contribute to lung tumor initiation and progression.

6.
EMBO Mol Med ; 12(3): e10941, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32030896

ABSTRACT

Chromosome instability (CIN) has been associated with therapeutic resistance in many cancers. However, whether tumours become genomically unstable as an evolutionary mechanism to overcome the bottleneck exerted by therapy is not clear. Using a CIN model of Kras-driven breast cancer, we demonstrate that aneuploid tumours acquire genetic modifications that facilitate the development of resistance to targeted therapy faster than euploid tumours. We further show that the few initially chromosomally stable cancers that manage to persist during treatment do so concomitantly with the acquisition of CIN. Whole-genome sequencing analysis revealed that the most predominant genetic alteration in resistant tumours, originated from either euploid or aneuploid primary tumours, was an amplification on chromosome 6 containing the cMet oncogene. We further show that these tumours are dependent on cMet since its pharmacological inhibition leads to reduced growth and increased cell death. Our results highlight that irrespective of the initial CIN levels, cancer genomes are dynamic and the acquisition of a certain level of CIN, either induced or spontaneous, is a mechanism to circumvent oncogene addiction.


Subject(s)
Breast Neoplasms/genetics , Chromosomal Instability , Oncogene Addiction , Aneuploidy , Animals , Drug Resistance, Neoplasm/genetics , Female , Mice , Mice, Transgenic , Neoplasms, Experimental , Piperidines , Pyridazines , Pyrimidines
7.
Nat Commun ; 9(1): 3012, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30069007

ABSTRACT

Polo-like kinase 1 (Plk1) is overexpressed in a wide spectrum of human tumors, being frequently considered as an oncogene and an attractive cancer target. However, its contribution to tumor development is unclear. Using a new inducible knock-in mouse model we report here that Plk1 overexpression results in abnormal chromosome segregation and cytokinesis, generating polyploid cells with reduced proliferative potential. Mechanistically, these cytokinesis defects correlate with defective loading of Cep55 and ESCRT complexes to the abscission bridge, in a Plk1 kinase-dependent manner. In vivo, Plk1 overexpression prevents the development of Kras-induced and Her2-induced mammary gland tumors, in the presence of increased rates of chromosome instability. In patients, Plk1 overexpression correlates with improved survival in specific breast cancer subtypes. Therefore, despite the therapeutic benefits of inhibiting Plk1 due to its essential role in tumor cell cycles, Plk1 overexpression has tumor-suppressive properties by perturbing mitotic progression and cytokinesis.


Subject(s)
Carcinogenesis/genetics , Cell Cycle Proteins/metabolism , Chromosomal Instability/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Centrosome/metabolism , Chromosome Segregation , Cytokinesis , Disease Models, Animal , Embryo, Mammalian/cytology , Endosomal Sorting Complexes Required for Transport/metabolism , Female , Fibroblasts/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Oncogenes , Polo-Like Kinase 1
8.
Nat Commun ; 9(1): 200, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321627

ABSTRACT

A Supplementary Information file from a different paper was inadvertently published with the original version of this Article. This file was replaced with the correct Supplementary Information file on 24 October 2017.

9.
Nat Commun ; 8(1): 849, 2017 10 11.
Article in English | MEDLINE | ID: mdl-29021609

ABSTRACT

Surface contraction waves (SCWs) in oocytes and embryos lead to large-scale shape changes coupled to cell cycle transitions and are spatially coordinated with the cell axis. Here, we show that SCWs in the starfish oocyte are generated by a traveling band of myosin II-driven cortical contractility. At the front of the band, contractility is activated by removal of cdk1 inhibition of the RhoA/RhoA kinase/myosin II signaling module, while at the rear, contractility is switched off by negative feedback originating downstream of RhoA kinase. The SCW's directionality and speed are controlled by a spatiotemporal gradient of cdk1-cyclinB. This gradient is formed by the release of cdk1-cyclinB from the asymmetrically located nucleus, and progressive degradation of cyclinB. By combining quantitative imaging, biochemical and mechanical perturbations with mathematical modeling, we demonstrate that the SCWs result from the spatiotemporal integration of two conserved regulatory modules, cdk1-cyclinB for cell cycle regulation and RhoA/Rok/NMYII for actomyosin contractility.Surface contraction waves (SCWs) are prominent shape changes coupled to cell cycle transitions in oocytes. Here the authors show that SCWs are patterned by the spatiotemporal integration of two conserved modules, cdk1-cyclinB for cell cycle regulation and RhoA/Rok/NMYII for actomyosin contractility.


Subject(s)
Actomyosin/physiology , CDC2 Protein Kinase/metabolism , Cell Shape/physiology , Meiosis , Oocytes/physiology , Animals , Cyclin B/metabolism , Myosin Type II/metabolism , Starfish , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
10.
Methods Mol Biol ; 1457: 145-66, 2016.
Article in English | MEDLINE | ID: mdl-27557579

ABSTRACT

High throughput DNA sequencing, the decreasing costs of DNA synthesis, and universal techniques for genetic manipulation have made it much easier and quicker to establish molecular tools for any organism than it has been 5 years ago. This opens a great opportunity for reviving "nonconventional" model organisms, which are particularly suited to study a specific biological process and many of which have already been established before the era of molecular biology. By taking advantage of transcriptomics, in particular, these systems can now be easily turned into full fetched models for molecular cell biology.As an example, here we describe how we established molecular tools in the starfish Patiria miniata, which has been a popular model for cell and developmental biology due to the synchronous and rapid development, transparency, and easy handling of oocytes, eggs, and embryos. Here, we detail how we used a de novo assembled transcriptome to produce molecular markers and established conditions for live imaging to investigate the molecular mechanisms underlying centriole elimination-a poorly understood process essential for sexual reproduction of animal species.


Subject(s)
Centrioles/metabolism , Meiosis , Molecular Imaging , Oocytes/cytology , Oocytes/metabolism , Starfish/cytology , Starfish/physiology , Animals , Databases, Protein , Female , Gene Expression , Genes, Reporter , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Molecular Imaging/methods , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Web Browser
11.
J Cell Biol ; 212(7): 815-27, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-27002173

ABSTRACT

Centriole elimination is an essential process that occurs in female meiosis of metazoa to reset centriole number in the zygote at fertilization. How centrioles are eliminated remains poorly understood. Here we visualize the entire elimination process live in starfish oocytes. Using specific fluorescent markers, we demonstrate that the two older, mother centrioles are selectively removed from the oocyte by extrusion into polar bodies. We show that this requires specific positioning of the second meiotic spindle, achieved by dynein-driven transport, and anchorage of the mother centriole to the plasma membrane via mother-specific appendages. In contrast, the single daughter centriole remaining in the egg is eliminated before the first embryonic cleavage. We demonstrate that these distinct elimination mechanisms are necessary because if mother centrioles are artificially retained, they cannot be inactivated, resulting in multipolar zygotic spindles. Thus, our findings reveal a dual mechanism to eliminate centrioles: mothers are physically removed, whereas daughters are eliminated in the cytoplasm, preparing the egg for fertilization.


Subject(s)
Centrioles/physiology , Meiosis , Oocytes/physiology , Starfish/physiology , Animals , Centrioles/metabolism , Cleavage Stage, Ovum/physiology , Dyneins/metabolism , Female , Fertilization , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Oocytes/metabolism , Polar Bodies/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Starfish/genetics , Starfish/metabolism , Time Factors
12.
Curr Biol ; 24(12): 1421-1428, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24909322

ABSTRACT

Animal cells disassemble and reassemble their nuclear envelopes (NEs) upon each division. Nuclear envelope breakdown (NEBD) serves as a major regulatory mechanism by which mixing of cytoplasmic and nuclear compartments drives the complete reorganization of cellular architecture, committing the cell for division. Breakdown is initiated by phosphorylation-driven partial disassembly of the nuclear pore complexes (NPCs), increasing their permeability but leaving the overall NE structure intact. Subsequently, the NE is rapidly broken into membrane fragments, defining the transition from prophase to prometaphase and resulting in complete mixing of cyto- and nucleoplasm. However, the mechanism underlying this rapid NE fragmentation remains largely unknown. Here, we show that NE fragmentation during NEBD in starfish oocytes is driven by an Arp2/3 complex-nucleated F-actin "shell" that transiently polymerizes on the inner surface of the NE. Blocking the formation of this F-actin shell prevents membrane fragmentation and delays entry of large cytoplasmic molecules into the nucleus. We observe spike-like protrusions extending from the F-actin shell that appear to "pierce" the NE during the fragmentation process. Finally, we show that NE fragmentation is essential for successful reproduction, because blocking this process in meiosis leads to formation of aneuploid eggs.


Subject(s)
Actin-Related Protein 2-3 Complex/genetics , Actinin/genetics , Actins/genetics , Nuclear Envelope/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actinin/metabolism , Actins/metabolism , Animals , Meiosis , Molecular Sequence Data , Oocytes/metabolism , Polymerization , Sequence Analysis, DNA , Starfish
13.
Genetics ; 195(2): 469-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23852386

ABSTRACT

The first microRNAs were discovered some 20 years ago, but only a small fraction of the microRNA-encoding genes have been described in detail yet. Here we report the molecular analysis of a computationally predicted Drosophila melanogaster microRNA gene, mir-282. We show that the mir-282 gene is the source of a 4.9-kb-long primary transcript with a 5' cap and a 3'-poly(A) sequence and a mature microRNA of ∼25 bp. Our data strongly suggest the existence of an independent mir-282 gene conserved in holometabolic insects. We give evidence that the mir-282 locus encodes a functional transcript that influences viability, longevity, and egg production in Drosophila. We identify the nervous system-specific adenylate cyclase (rutabaga) as a target of miR-282 and assume that one of the main functions of mir-282 is the regulation of adenylate cyclase activity in the nervous system during metamorphosis.


Subject(s)
Adenylyl Cyclases/genetics , Drosophila melanogaster/genetics , Longevity/genetics , MicroRNAs/genetics , Adenylyl Cyclases/metabolism , Animals , Cell Survival , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Metamorphosis, Biological/genetics , Nervous System/growth & development , Nervous System/metabolism , Ovum/growth & development
14.
BMC Dev Biol ; 12: 20, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22827336

ABSTRACT

BACKGROUND: Calpains are calcium regulated intracellular cysteine proteases implicated in a variety of physiological functions and pathological conditions. The Drosophila melanogaster genome contains only two genes, CalpA and CalpB coding for canonical, active calpain enzymes. The movement of the border cells in Drosophila egg chambers is a well characterized model of the eukaryotic cell migration. Using this genetically pliable model we can investigate the physiological role of calpains in cell motility. RESULTS: We demonstrate at the whole organism level that CalpB is implicated in cell migration, while the structurally related CalpA paralog can not fulfill the same function. The downregulation of the CalpB gene by mutations or RNA interference results in a delayed migration of the border cells in Drosophila egg chambers. This phenotype is significantly enhanced when the focal adhesion complex genes encoding for α-PS2 integrin ( if), ß-PS integrin (mys) and talin (rhea) are silenced. The reduction of CalpB activity diminishes the release of integrins from the rear end of the border cells. The delayed migration and the reduced integrin release phenotypes can be suppressed by expressing wild-type talin-head in the border cells but not talin-head(R367A), a mutant form which is not able to bind ß-PS integrin. CalpB can cleave talin in vitro, and the two proteins coimmunoprecipitate from Drosophila extracts. CONCLUSIONS: The physiological function of CalpB in border cell motility has been demonstrated in vivo. The genetic interaction between the CalpB and the if, mys, as well as rhea genes, the involvement of active talin head-domains in the process, and the fact that CalpB and talin interact with each other collectively suggest that the limited proteolytic cleavage of talin is one of the possible mechanisms through which CalpB regulates cell migration.


Subject(s)
Calpain/metabolism , Cell Movement , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Animals , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Focal Adhesions/metabolism , Talin/metabolism
15.
FEBS Lett ; 584(21): 4375-8, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20951134

ABSTRACT

The Nimrod gene superfamily is an important component of the innate immune response. The majority of its member genes are located in close proximity within the Drosophila melanogaster genome and they lie in a larger conserved cluster ("Nimrod cluster"), made up of non-related groups (families, superfamilies) of genes. This cluster has been a part of the Arthropod genomes for about 300-350 million years. The available data suggest that the Nimrod cluster is a functional module of the insect innate immune response.


Subject(s)
Conserved Sequence , Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , Immunity, Innate/genetics , Multigene Family , Animals , Genome, Insect/genetics
16.
Proc Natl Acad Sci U S A ; 106(12): 4805-9, 2009 Mar 24.
Article in English | MEDLINE | ID: mdl-19261847

ABSTRACT

The blood cells, or hemocytes, in Drosophila participate in the immune response through the production of antimicrobial peptides, the phagocytosis of bacteria, and the encapsulation of larger foreign particles such as parasitic eggs; these immune reactions are mediated by phylogenetically conserved mechanisms. The encapsulation reaction is analogous to the formation of granuloma in vertebrates, and is mediated by large specialized cells, the lamellocytes. The origin of the lamellocytes has not been formally established, although it has been suggested that they are derived from the lymph gland, which is generally considered to be the main hematopoietic organ in the Drosophila larva. However, it was recently observed that a subepidermal population of sessile blood cells is released into the circulation in response to a parasitoid wasp infection. We set out to analyze this phenomenon systematically. As a result, we define the sessile hemocytes as a novel hematopoietic compartment, and the main source of lamellocytes.


Subject(s)
Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/immunology , Hematopoiesis , Hemocytes/cytology , Animals , Cell Count , Cell Differentiation , Cell Separation , Drosophila melanogaster/cytology , Green Fluorescent Proteins/metabolism , Hemocytes/transplantation , Immunity , Larva/cytology , Larva/immunology , Larva/parasitology , Phenotype , Time Factors
17.
Mol Biol Evol ; 25(11): 2337-47, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18703524

ABSTRACT

The recently identified Nimrod superfamily is characterized by the presence of a special type of EGF repeat, the NIM repeat, located right after a typical CCXGY/W amino acid motif. On the basis of structural features, nimrod genes can be divided into three types. The proteins encoded by Draper-type genes have an EMI domain at the N-terminal part and only one copy of the NIM motif, followed by a variable number of EGF-like repeats. The products of Nimrod B-type and Nimrod C-type genes (including the eater gene) have different kinds of N-terminal domains, and lack EGF-like repeats but contain a variable number of NIM repeats. Draper and Nimrod C-type (but not Nimrod B-type) proteins carry a transmembrane domain. Several members of the superfamily were claimed to function as receptors in phagocytosis and/or binding of bacteria, which indicates an important role in the cellular immunity and the elimination of apoptotic cells. In this paper, the evolution of the Nimrod superfamily is studied with various methods on the level of genes and repeats. A hypothesis is presented in which the NIM repeat, along with the EMI domain, emerged by structural reorganizations at the end of an EGF-like repeat chain, suggesting a mechanism for the formation of novel types of repeats. The analyses revealed diverse evolutionary patterns in the sequences containing multiple NIM repeats. Although in the Nimrod B and Nimrod C proteins show characteristics of independent evolution, many internal NIM repeats in Eater sequences seem to have undergone concerted evolution. An analysis of the nimrod genes has been performed using phylogenetic and other methods and an evolutionary scenario of the origin and diversification of the Nimrod superfamily is proposed. Our study presents an intriguing example how the evolution of multigene families may contribute to the complexity of the innate immune response.


Subject(s)
Evolution, Molecular , Genes, Insect , Multigene Family , Repetitive Sequences, Amino Acid , Amino Acid Motifs , Animals , Anopheles/genetics , Bees/genetics , Drosophila/genetics , Phylogeny , Sequence Alignment , Tribolium/genetics
18.
BMC Bioinformatics ; 9: 27, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18205906

ABSTRACT

BACKGROUND: The models developed to characterize the evolution of multigene families (such as the birth-and-death and the concerted models) have also been applied on the level of sequence repeats inside a gene/protein. Phylogenetic reconstruction is the method of choice to study the evolution of gene families and also sequence repeats in the light of these models. The characterization of the gene family evolution in view of the evolutionary models is done by the evaluation of the clustering of the sequences with the originating loci in mind. As the locus represents positional information, it is straightforward that in the case of the repeats the exact position in the sequence should be used, as the simple numbering according to repeat order can be misleading. RESULTS: We have developed a novel rapid visual approach to study repeat evolution, that takes into account the exact repeat position in a sequence. The "pairwise repeat homology diagram" visualizes sequence repeats detected by a profile HMM in a pair of sequences and highlights their homology relations inferred by a phylogenetic tree. The method is implemented in a Perl script (t2prhd) available for downloading at http://t2prhd.sourceforge.net and is also accessible as an online tool at http://t2prhd.brc.hu. The power of the method is demonstrated on the EGF-like and fibronectin-III-like (Fn-III) domain repeats of three selected mammalian Tenascin sequences. CONCLUSION: Although pairwise repeat homology diagrams do not carry all the information provided by the phylogenetic tree, they allow a rapid and intuitive assessment of repeat evolution. We believe, that t2prhd is a helpful tool with which to study the pattern of repeat evolution. This method can be particularly useful in cases of large datasets (such as large gene families), as the command line interface makes it possible to automate the generation of pairwise repeat homology diagrams with the aid of scripts.


Subject(s)
Biological Evolution , Chromosome Mapping/methods , Evolution, Molecular , Multigene Family/genetics , Repetitive Sequences, Nucleic Acid/genetics , Sequence Analysis, DNA/methods , Software , Algorithms , Base Sequence , DNA Mutational Analysis , Molecular Sequence Data , Pattern Recognition, Automated , Phylogeny
19.
PLoS Genet ; 1(4): e55, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16254604

ABSTRACT

This article reports the production of an EP-element insertion library with more than 3,700 unique target sites within the Drosophila melanogaster genome and its use to systematically identify genes that affect embryonic muscle pattern formation. We designed a UAS/GAL4 system to drive GAL4-responsive expression of the EP-targeted genes in developing apodeme cells to which migrating myotubes finally attach and in an intrasegmental pattern of cells that serve myotubes as a migration substrate on their way towards the apodemes. The results suggest that misexpression of more than 1.5% of the Drosophila genes can interfere with proper myotube guidance and/or muscle attachment. In addition to factors already known to participate in these processes, we identified a number of enzymes that participate in the synthesis or modification of protein carbohydrate side chains and in Ubiquitin modifications and/or the Ubiquitin-dependent degradation of proteins, suggesting that these processes are relevant for muscle pattern formation.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Genetic Techniques , Muscles/metabolism , Animals , Body Patterning , Cell Cycle , Cell Movement , Cytoskeleton/metabolism , Genes, Insect , Muscle Fibers, Skeletal/metabolism , Muscles/cytology , Muscles/pathology , Ubiquitin/metabolism
20.
Dev Cell ; 7(1): 85-93, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15239956

ABSTRACT

Cells migrating through a tissue exert force via their cytoskeleton and are themselves subject to tension, but the effects of physical forces on cell behavior in vivo are poorly understood. Border cell migration during Drosophila oogenesis is a useful model for invasive cell movement. We report that this migration requires the activity of the transcriptional factor serum response factor (SRF) and its cofactor MAL-D and present evidence that nuclear accumulation of MAL-D is induced by cell stretching. Border cells that cannot migrate lack nuclear MAL-D but can accumulate it if they are pulled by other migrating cells. Like mammalian MAL, MAL-D also responds to activated Diaphanous, which affects actin dynamics. MAL-D/SRF activity is required to build a robust actin cytoskeleton in the migrating cells; mutant cells break apart when initiating migration. Thus, tension-induced MAL-D activity may provide a feedback mechanism for enhancing cytoskeletal strength during invasive migration.


Subject(s)
Cell Movement/genetics , Drosophila Proteins/metabolism , Drosophila/metabolism , Nuclear Proteins/metabolism , Oocytes/metabolism , Oogenesis/genetics , Serum Response Factor/metabolism , Actin Cytoskeleton/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Carrier Proteins/metabolism , Cell Size/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , DNA, Complementary/analysis , DNA, Complementary/genetics , Drosophila/cytology , Drosophila/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/isolation & purification , Feedback/physiology , Feedback, Physiological/physiology , Formins , Molecular Sequence Data , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/isolation & purification , Oocytes/cytology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Serum Response Factor/genetics , Stress, Mechanical , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...