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










Publication year range
1.
Immunity ; 56(12): 2803-2815.e6, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38035879

ABSTRACT

Somatic hypermutation (SHM) drives affinity maturation and continues over months in SARS-CoV-2-neutralizing antibodies (nAbs). However, several potent SARS-CoV-2 antibodies carry no or only a few mutations, leaving the question of how ongoing SHM affects neutralization unclear. Here, we reverted variable region mutations of 92 antibodies and tested their impact on SARS-CoV-2 binding and neutralization. Reverting higher numbers of mutations correlated with decreasing antibody functionality. However, for some antibodies, including antibodies of the public clonotype VH1-58, neutralization of Wu01 remained unaffected. Although mutations were dispensable for Wu01-induced VH1-58 antibodies to neutralize Alpha, Beta, and Delta variants, they were critical for Omicron BA.1/BA.2 neutralization. We exploited this knowledge to convert the clinical antibody tixagevimab into a BA.1/BA.2 neutralizer. These findings broaden our understanding of SHM as a mechanism that not only improves antibody responses during affinity maturation but also contributes to antibody diversification, thus increasing the chances of neutralizing viral escape variants.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/genetics , Antibodies, Viral , Mutation/genetics , Antibodies, Neutralizing
2.
iScience ; 25(10): 105193, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36188189

ABSTRACT

Blocking the interaction of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with its angiotensin-converting enzyme 2 (ACE2) receptor was proved to be an effective therapeutic option. Various protein binders as well as monoclonal antibodies that effectively target the receptor-binding domain (RBD) of SARS-CoV-2 to prevent interaction with ACE2 were developed. The emergence of SARS-CoV-2 variants that accumulate alterations in the RBD can severely affect the efficacy of such immunotherapeutic agents, as is indeed the case with Omicron that resists many of the previously isolated monoclonal antibodies. Here, we evaluate an ACE2-based immunoadhesin that we have developed early in the pandemic against some of the recent variants of concern (VoCs), including the Delta and the Omicron variants. We show that our ACE2-immunoadhesin remains effective in neutralizing these variants, suggesting that immunoadhesin-based immunotherapy is less prone to escape by the virus and has a potential to remain effective against future VoCs.

3.
Nature ; 603(7899): 174-179, 2022 03.
Article in English | MEDLINE | ID: mdl-35173332

ABSTRACT

Lassa virus (LASV) is a human pathogen, causing substantial morbidity and mortality1,2. Similar to other Arenaviridae, it presents a class-I spike complex on its surface that facilitates cell entry. The virus's cellular receptor is matriglycan, a linear carbohydrate that is present on α-dystroglycan3,4, but the molecular mechanism that LASV uses to recognize this glycan is unknown. In addition, LASV and other arenaviruses have a unique signal peptide that forms an integral and functionally important part of the mature spike5-8; yet the structure, function and topology of the signal peptide in the membrane remain uncertain9-11. Here we solve the structure of a complete native LASV spike complex, finding that the signal peptide crosses the membrane once and that its amino terminus is located in the extracellular region. Together with a double-sided domain-switching mechanism, the signal peptide helps to stabilize the spike complex in its native conformation. This structure reveals that the LASV spike complex is preloaded with matriglycan, suggesting the mechanism of binding and rationalizing receptor recognition by α-dystroglycan-tropic arenaviruses. This discovery further informs us about the mechanism of viral egress and may facilitate the rational design of novel therapeutics that exploit this binding site.


Subject(s)
Dystroglycans , Lassa virus , Receptors, Virus , Viral Envelope Proteins , Dystroglycans/chemistry , Dystroglycans/metabolism , Humans , Lassa Fever/virology , Lassa virus/chemistry , Lassa virus/metabolism , Protein Conformation , Protein Sorting Signals , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization
4.
J Mol Biol ; 433(15): 167099, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34119488

ABSTRACT

Glycans decorate the cell surface, secreted glycoproteins and glycolipids, and altered glycans are often found in cancers. Despite their high diagnostic and therapeutic potential, however, glycans are polar and flexible molecules that are quite challenging for the development and design of high-affinity binding antibodies. To understand the mechanisms by which glycan neoantigens are specifically recognized by antibodies, we analyze the biomolecular recognition of the tumor-associated carbohydrate antigen CA19-9 by two distinct antibodies using X-ray crystallography. Despite the potential plasticity of glycans and the very different antigen-binding surfaces presented by the antibodies, both structures reveal an essentially identical extended CA19-9 conformer, suggesting that this conformer's stability selects the antibodies. Starting from the bound structure of one of the antibodies, we use the AbLIFT computational algorithm to design a variant with seven core mutations in the variable domain's light-heavy chain interface that exhibits tenfold improved affinity for CA19-9. The results reveal strategies used by antibodies to specifically recognize glycan antigens and show how automated antibody-optimization methods may be used to enhance the clinical potential of existing antibodies.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , CA-19-9 Antigen/immunology , Computational Biology/methods , Algorithms , Animals , Antibodies, Monoclonal/genetics , Antibody Affinity , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Mutation , Protein Conformation
6.
Cell ; 182(4): 843-854.e12, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32673567

ABSTRACT

The SARS-CoV-2 pandemic has unprecedented implications for public health, social life, and the world economy. Because approved drugs and vaccines are limited or not available, new options for COVID-19 treatment and prevention are in high demand. To identify SARS-CoV-2-neutralizing antibodies, we analyzed the antibody response of 12 COVID-19 patients from 8 to 69 days after diagnosis. By screening 4,313 SARS-CoV-2-reactive B cells, we isolated 255 antibodies from different time points as early as 8 days after diagnosis. Of these, 28 potently neutralized authentic SARS-CoV-2 with IC100 as low as 0.04 µg/mL, showing a broad spectrum of variable (V) genes and low levels of somatic mutations. Interestingly, potential precursor sequences were identified in naive B cell repertoires from 48 healthy individuals who were sampled before the COVID-19 pandemic. Our results demonstrate that SARS-CoV-2-neutralizing antibodies are readily generated from a diverse pool of precursors, fostering hope for rapid induction of a protective immune response upon vaccination.


Subject(s)
Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Betacoronavirus/immunology , COVID-19 , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunologic Memory , Longitudinal Studies , Pandemics , SARS-CoV-2 , Somatic Hypermutation, Immunoglobulin
7.
Cell Host Microbe ; 27(3): 418-427.e4, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32059794

ABSTRACT

Ebola virus disease is a severe health problem in Africa. Vaccines that display the Zaire ebolavirus glycoprotein spike complex are a prime component for the effort to combat it. The VH3-15/Vλ1-40-based class of antibodies was recently discovered to be a common response in individuals who received the Ebola virus vaccines. These antibodies display attractive properties, and thus likely contribute to the efficacy of the vaccines. Here, we use cryo-EM to elucidate how three VH3-15/Vλ1-40 antibodies from different individuals target the virus and found a convergent mechanism against a partially conserved site on the spike complex. Our study rationalizes the selection of the VH3-15/Vλ1-40 germline genes for specifically targeting this site and highlights Ebolavirus species-specific sequence divergences that may restrict breadth of VH3-15/Vλ1-40-based humoral response. The results from this study could help develop improved immunization schemes and further enable the design of immunogens that would be efficacious against a broader set of Ebolavirus species.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antibody Specificity , Cryoelectron Microscopy , Ebola Vaccines , Ebolavirus , Epitopes/immunology , HEK293 Cells , Humans , Protein Binding , Protein Structure, Tertiary
8.
Nat Commun ; 11(1): 67, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31900422

ABSTRACT

Certain arenaviruses that circulate in rodent populations can cause life-threatening hemorrhagic fevers when they infect humans. Due to their efficient transmission, arenaviruses pose a severe risk for outbreaks and might be exploited as biological weapons. Effective countermeasures against these viruses are highly desired. Ideally, a single remedy would be effective against many or even all the pathogenic viruses in this family. However, despite the fact that all pathogenic arenaviruses from South America utilize transferrin receptor 1 (TfR1) as a cellular receptor, their viral glycoproteins are highly diversified, impeding efforts to isolate cross-neutralizing antibodies. Here we address this problem using a rational design approach to target TfR1-tropic arenaviruses with high potency and breadth. The pan-reactive molecule is highly effective against all arenaviruses that were tested, offering a universal therapeutic approach. Our design scheme avoids the shortcomings of previous immunoadhesins and can be used to combat other zoonotic pathogens.


Subject(s)
Arenaviridae Infections/therapy , Arenavirus/immunology , Immunotherapy , Receptors, Transferrin/chemistry , Receptors, Transferrin/immunology , Receptors, Virus/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Arenavirus/chemistry , Arenavirus/genetics , Drug Design , Humans , Receptors, Transferrin/genetics , Receptors, Virus/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
9.
Nat Med ; 25(10): 1589-1600, 2019 10.
Article in English | MEDLINE | ID: mdl-31591605

ABSTRACT

Recombinant vesicular stomatitis virus-Zaire Ebola virus (rVSV-ZEBOV) is the most advanced Ebola virus vaccine candidate and is currently being used to combat the outbreak of Ebola virus disease (EVD) in the Democratic Republic of the Congo (DRC). Here we examine the humoral immune response in a subset of human volunteers enrolled in a phase 1 rVSV-ZEBOV vaccination trial by performing comprehensive single B cell and electron microscopy structure analyses. Four studied vaccinees show polyclonal, yet reproducible and convergent B cell responses with shared sequence characteristics. EBOV-targeting antibodies cross-react with other Ebolavirus species, and detailed epitope mapping revealed overlapping target epitopes with antibodies isolated from EVD survivors. Moreover, in all vaccinees, we detected highly potent EBOV-neutralizing antibodies with activities comparable or superior to the monoclonal antibodies currently used in clinical trials. These include antibodies combining the IGHV3-15/IGLV1-40 immunoglobulin gene segments that were identified in all investigated individuals. Our findings will help to evaluate and direct current and future vaccination strategies and offer opportunities for novel EVD therapies.


Subject(s)
Ebola Vaccines/administration & dosage , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Immunity, Humoral/immunology , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Formation/immunology , B-Lymphocytes/immunology , B-Lymphocytes/virology , Ebola Vaccines/adverse effects , Ebola Vaccines/immunology , Ebolavirus/pathogenicity , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Humans , Male , Middle Aged , Vaccination/adverse effects , Vesiculovirus/genetics , Volunteers
10.
J Mol Biol ; 431(11): 2095-2111, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31004664

ABSTRACT

Lassa virus (LASV) is a notorious human pathogen in West Africa. Its class I trimeric spike complex displays a distinct architecture, and its cell entry mechanism involves unique attributes not shared by other related viruses. We determined the crystal structure of the GP2 fusion glycoprotein from the spike complex of LASV (GP2LASV) in its post-fusion conformation. GP2LASV adopts a canonical helical bundle configuration similarly to other viruses in its family. The core packing of GP2LASV, however, is more organized compared to GP2 from other viruses reducing the formation of internal hydrophobic cavities. We demonstrate a link between the formation of such unfavorable hydrophobic cavities and the efficiencies of membrane fusion and cell entry. Our study suggests that LASV has evolved a more efficient membrane fusogen compared to other viruses from its family by optimizing the post-fusion configuration of its GP2 module.


Subject(s)
Lassa Fever/virology , Lassa virus/physiology , Virus Internalization , Animals , Cell Line , Crystallography, X-Ray , HEK293 Cells , Humans , Lassa Fever/metabolism , Lassa virus/chemistry , Membrane Fusion , Molecular Dynamics Simulation , Protein Conformation
11.
Commun Biol ; 1: 213, 2018.
Article in English | MEDLINE | ID: mdl-30534605

ABSTRACT

Characterization of overexpressed proteins is essential for assessing their quality, and providing input for iterative redesign and optimization. This process is typically carried out following purification procedures that require pronounced cost of time and labor. Therefore, quality assessment of recombinant proteins with no prior purification offers a major advantage. Here, we report a native mass spectrometry method that enables characterization of overproduced proteins directly from culture media. Properties such as solubility, molecular weight, folding, assembly state, overall structure, post-translational modifications and binding to relevant biomolecules are immediately revealed. We show the applicability of the method for in-depth characterization of secreted recombinant proteins from eukaryotic systems such as yeast, insect, and human cells. This method, which can be readily extended to high-throughput analysis, considerably shortens the time gap between protein production and characterization, and is particularly suitable for characterizing engineered and mutated proteins, and optimizing yield and quality of overexpressed proteins.

12.
Nat Microbiol ; 3(10): 1153-1160, 2018 10.
Article in English | MEDLINE | ID: mdl-30150732

ABSTRACT

Lujo virus (LUJV) has emerged as a highly fatal human pathogen. Despite its membership among the Arenaviridae, LUJV does not classify with the known Old and New World groups of that viral family. Likewise, LUJV was recently found to use neuropilin-2 (NRP2) as a cellular receptor instead of the canonical receptors used by Old World and New World arenaviruses. The emergence of a deadly pathogen into human populations using an unprecedented entry route raises many questions regarding the mechanism of cell recognition. To provide the basis for combating LUJV in particular, and to increase our general understanding of the molecular changes that accompany an evolutionary switch to a new receptor for arenaviruses, we used X-ray crystallography to reveal how the GP1 receptor-binding domain of LUJV (LUJVGP1) recognizes NRP2. Structural data show that LUJVGP1 is more similar to Old World than to New World arenaviruses. Structural analysis supported by experimental validation further suggests that NRP2 recognition is metal-ion dependent and that the complete NRP2 binding site is formed in the context of the trimeric spike. Taken together, our data provide the mechanism for the cell attachment step of LUJV and present indispensable information for combating this phatogen.


Subject(s)
Lujo virus/chemistry , Neuropilin-2/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Dystroglycans/metabolism , HEK293 Cells , Humans , Lujo virus/metabolism , Lujo virus/physiology , Mutation , Protein Binding , Protein Domains , Viral Envelope Proteins/genetics , Virus Attachment
13.
PLoS Pathog ; 13(4): e1006337, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28448640

ABSTRACT

Cell entry of many enveloped viruses occurs by engagement with cellular receptors, followed by internalization into endocytic compartments and pH-induced membrane fusion. A previously unnoticed step of receptor switching was found to be critical during cell entry of two devastating human pathogens: Ebola and Lassa viruses. Our recent studies revealed the functional role of receptor switching to LAMP1 for triggering membrane fusion by Lassa virus and showed the involvement of conserved histidines in this switching, suggesting that other viruses from this family may also switch to LAMP1. However, when we investigated viruses that are genetically close to Lassa virus, we discovered that they cannot bind LAMP1. A crystal structure of the receptor-binding module from Morogoro virus revealed structural differences that allowed mapping of the LAMP1 binding site to a unique set of Lassa residues not shared by other viruses in its family, illustrating a key difference in the cell-entry mechanism of Lassa virus that may contribute to its pathogenicity.


Subject(s)
Arenaviridae Infections/virology , Arenaviruses, Old World/metabolism , Lassa Fever/virology , Lassa virus/metabolism , Lysosomal Membrane Proteins/chemistry , Amino Acid Sequence , Animals , Arenaviruses, Old World/chemistry , Arenaviruses, Old World/genetics , Binding Sites , Humans , Lassa virus/chemistry , Lassa virus/genetics , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Membrane Fusion , Models, Molecular , Models, Structural , Protein Binding , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Alignment , Species Specificity
14.
J Virol ; 90(22): 10329-10338, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27605678

ABSTRACT

To effectively infect cells, Lassa virus needs to switch in an endosomal compartment from its primary receptor, α-dystroglycan, to a protein termed LAMP1. A unique histidine triad on the surface of the receptor-binding domain from the glycoprotein spike complex of Lassa virus is important for LAMP1 binding. Here we investigate mutated spikes that have an impaired ability to interact with LAMP1 and show that although LAMP1 is important for efficient infectivity, it is not required for spike-mediated membrane fusion per se Our studies reveal important regulatory roles for histidines from the triad in sensing acidic pH and preventing premature spike triggering. We further show that LAMP1 requires a positively charged His230 residue to engage with the spike complex and that LAMP1 binding promotes membrane fusion. These results elucidate the molecular role of LAMP1 binding during Lassa virus cell entry and provide new insights into how pH is sensed by the spike. IMPORTANCE: Lassa virus is a devastating disease-causing agent in West Africa, with a significant yearly death toll and severe long-term complications associated with its infection in survivors. In recent years, we learned that Lassa virus needs to switch receptors in a pH-dependent manner to efficiently infect cells, but neither the molecular mechanisms that allow switching nor the actual effects of switching were known. Here we investigate the activity of the viral spike complex after abrogation of its ability to switch receptors. These studies inform us about the role of switching receptors and provide new insights into how the spike senses acidic pH.


Subject(s)
Lassa Fever/metabolism , Lassa Fever/virology , Lassa virus/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Protein Binding/physiology , Africa, Western , Animals , Cell Line , Chlorocebus aethiops , Dystroglycans/metabolism , Endosomes/metabolism , Endosomes/virology , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Membrane Fusion/physiology , Receptors, Virus/metabolism , Vero Cells , Viral Envelope Proteins/metabolism , Virus Internalization
15.
J Virol ; 89(15): 7584-92, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972533

ABSTRACT

UNLABELLED: Lassa virus is a notorious human pathogen that infects many thousands of people each year in West Africa, causing severe viral hemorrhagic fevers and significant mortality. The surface glycoprotein of Lassa virus mediates receptor recognition through its GP1 subunit. Here we report the crystal structure of GP1 from Lassa virus, which is the first representative GP1 structure for Old World arenaviruses. We identify a unique triad of histidines that forms a binding site for LAMP1, a known lysosomal protein recently discovered to be a critical receptor for internalized Lassa virus at acidic pH. We demonstrate that mutation of this histidine triad, which is highly conserved among Old World arenaviruses, impairs LAMP1 recognition. Our biochemical and structural data further suggest that GP1 from Lassa virus may undergo irreversible conformational changes that could serve as an immunological decoy mechanism. Together with a variable region that we identify on the surface of GP1, those could be two distinct mechanisms that Lassa virus utilizes to avoid antibody-based immune response. IMPORTANCE: Structural data at atomic resolution for viral proteins is key for understanding their function at the molecular level and can facilitate novel avenues for combating viral infections. Here we used X-ray protein crystallography to decipher the crystal structure of the receptor-binding domain (GP1) from Lassa virus. This is a pathogenic virus that causes significant illness and mortality in West Africa. This structure reveals the overall architecture of GP1 domains from the group of viruses known as the Old World arenaviruses. Using this structural information, we elucidated the mechanisms for pH switch and binding of Lassa virus to LAMP1, a recently identified host receptor that is critical for successful infection. Lastly, our structural analysis suggests two novel immune evasion mechanisms that Lassa virus may utilize to escape antibody-based immune response.


Subject(s)
Lassa Fever/metabolism , Lassa virus/metabolism , Lysosomal Membrane Proteins/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Cell Line , Humans , Lassa Fever/genetics , Lassa Fever/virology , Lassa virus/chemistry , Lassa virus/genetics , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Sequence Alignment , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
16.
Sci Signal ; 8(368): ra29, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25783158

ABSTRACT

Growth factors promote tumor growth and metastasis. We found that epidermal growth factor (EGF) induced a set of 22 microRNAs (miRNAs) before promoting the migration of mammary cells. These miRNAs were more abundant in human breast tumors relative to the surrounding tissue, and their abundance varied among breast cancer subtypes. One of these miRNAs, miR-15b, targeted the 3' untranslated region of MTSS1 (metastasis suppressor protein 1). Although xenografts in which MTSS1 was knocked down grew more slowly in mice initially, longer-term growth was unaffected. Knocking down MTSS1 increased migration and Matrigel invasion of nontransformed mammary epithelial cells. Overexpressing MTSS1 in an invasive cell line decreased cell migration and invasiveness, decreased the formation of invadopodia and actin stress fibers, and increased the formation of cellular junctions. In tissues from breast cancer patients with the aggressive basal subtype, an inverse correlation occurred with the high expression of miRNA-15b and the low expression of MTSS1. Furthermore, low abundance of MTSS1 correlated with poor patient prognosis. Thus, growth factor-inducible miRNAs mediate mechanisms underlying the progression of cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Epidermal Growth Factor/metabolism , MicroRNAs/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Epidermal Growth Factor/genetics , Female , Heterografts , Humans , Mice , Mice, SCID , MicroRNAs/genetics , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation
17.
EMBO Mol Med ; 7(3): 299-314, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25678558

ABSTRACT

Dissemination of primary tumor cells depends on migratory and invasive attributes. Here, we identify Navigator-3 (NAV3), a gene frequently mutated or deleted in human tumors, as a regulator of epithelial migration and invasion. Following induction by growth factors, NAV3 localizes to the plus ends of microtubules and enhances their polarized growth. Accordingly, NAV3 depletion trimmed microtubule growth, prolonged growth factor signaling, prevented apoptosis and enhanced random cell migration. Mathematical modeling suggested that NAV3-depleted cells acquire an advantage in terms of the way they explore their environment. In animal models, silencing NAV3 increased metastasis, whereas ectopic expression of the wild-type form, unlike expression of two, relatively unstable oncogenic mutants from human tumors, inhibited metastasis. Congruently, analyses of > 2,500 breast and lung cancer patients associated low NAV3 with shorter survival. We propose that NAV3 inhibits breast cancer progression by regulating microtubule dynamics, biasing directionally persistent rather than random migration, and inhibiting locomotion of initiated cells.


Subject(s)
Breast Neoplasms/complications , Breast Neoplasms/pathology , Cell Movement , Membrane Proteins/metabolism , Neoplasm Metastasis/pathology , Nerve Tissue Proteins/metabolism , Animals , Disease Models, Animal , Humans , Mice
18.
Sci Signal ; 8(360): ra7, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25605973

ABSTRACT

Amplified HER2, which encodes a member of the epidermal growth factor receptor (EGFR) family, is a target of effective therapies against breast cancer. In search for similarly targetable genomic aberrations, we identified copy number gains in SYNJ2, which encodes the 5'-inositol lipid phosphatase synaptojanin 2, as well as overexpression in a small fraction of human breast tumors. Copy gain and overexpression correlated with shorter patient survival and a low abundance of the tumor suppressor microRNA miR-31. SYNJ2 promoted cell migration and invasion in culture and lung metastasis of breast tumor xenografts in mice. Knocking down SYNJ2 impaired the endocytic recycling of EGFR and the formation of cellular lamellipodia and invadopodia. Screening compound libraries identified SYNJ2-specific inhibitors that prevented cell migration but did not affect the related neural protein SYNJ1, suggesting that SYNJ2 is a potentially druggable target to block cancer cell migration.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Metastasis/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Movement/drug effects , Drug Discovery , ErbB Receptors/metabolism , Female , Fluorescent Antibody Technique , Gene Dosage , Humans , Image Processing, Computer-Assisted , Immunoblotting , Immunohistochemistry , Mice , Mice, SCID , Microscopy, Electron, Scanning , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Podosomes/genetics , Podosomes/physiology , Pseudopodia/genetics , Pseudopodia/physiology , RNA, Small Interfering/genetics , Statistics, Nonparametric
19.
Nat Commun ; 5: 5073, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25278152

ABSTRACT

Signal transduction by receptor tyrosine kinases (RTKs) and nuclear receptors for steroid hormones is essential for body homeostasis, but the cross-talk between these receptor families is poorly understood. We observed that glucocorticoids inhibit signalling downstream of EGFR, an RTK. The underlying mechanism entails suppression of EGFR's positive feedback loops and simultaneous triggering of negative feedback loops that normally restrain EGFR. Our studies in mice reveal that the regulation of EGFR's feedback loops by glucocorticoids translates to circadian control of EGFR signalling: EGFR signals are suppressed by high glucocorticoids during the active phase (night-time in rodents), while EGFR signals are enhanced during the resting phase. Consistent with this pattern, treatment of animals bearing EGFR-driven tumours with a specific kinase inhibitor was more effective if administered during the resting phase of the day, when glucocorticoids are low. These findings support a circadian clock-based paradigm in cancer therapy.


Subject(s)
ErbB Receptors/metabolism , Glucocorticoids/metabolism , Neoplasms/pathology , Signal Transduction , Animals , Cell Line, Tumor , Cell Movement , Circadian Rhythm , Disease Progression , Female , Humans , Ligands , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Oscillometry , Receptors, Glucocorticoid/metabolism , Treatment Outcome
20.
PLoS One ; 8(12): e80566, 2013.
Article in English | MEDLINE | ID: mdl-24324612

ABSTRACT

Signal-induced transcript isoform variation (TIV) includes alternative promoter usage as well as alternative splicing and alternative polyadenylation of mRNA. To assess the phenotypic relevance of signal-induced TIV, we employed exon arrays and breast epithelial cells, which migrate in response to the epidermal growth factor (EGF). We show that EGF rapidly--within one hour--induces widespread TIV in a significant fraction of the transcriptome. Importantly, TIV characterizes many genes that display no differential expression upon stimulus. In addition, similar EGF-dependent changes are shared by a panel of mammary cell lines. A functional screen, which utilized isoform-specific siRNA oligonucleotides, indicated that several isoforms play essential, non-redundant roles in EGF-induced mammary cell migration. Taken together, our findings highlight the importance of TIV in the rapid evolvement of a phenotypic response to extracellular signals.


Subject(s)
Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Exons , Genetic Variation , RNA, Messenger/genetics , Transcriptome , Alternative Splicing , Cell Line , Cell Movement/drug effects , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Oligonucleotide Array Sequence Analysis , Polyadenylation , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...