Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Eng Biol ; 5(3): 60-63, 2021 Sep.
Article En | MEDLINE | ID: mdl-36968260

When we think about the potential that biology has to offer, the U.S. Bioindustrial Manufacturing and Design Ecosystem or BioMADE slogan could read, 'we don't make the products you buy, we make the products that you buy, with biology'. BioMADE is a non-profit public-private partnership between the U.S. government and the private sector to leverage the work already accomplished in industry, accelerate the bioindustrial revolution, and create a stronger, resilient, sustainable, and environmentally friendly manufacturing ecosystem. BioMADE endeavours to be a leader, an enabler, and a beacon for how contemporary manufacturing can be transformed with biology to mature the bioindustrial manufacturing ecosystem. The institute cannot go this path alone to solve all the problems and coalesce the existing ecosystem. It requires determination and commitment from the private sector, academia, non-profit research institutions and national laboratories; the entire community. Many technical challenges and adoption hurdles still loom high. Industry and consumers need to start accepting that engineering biology has a critical role to play in the manufacturing of many of the materials and products we use today.

2.
PLoS Pathog ; 10(2): e1003914, 2014 Feb.
Article En | MEDLINE | ID: mdl-24550726

Vector-borne viruses are an important class of emerging and re-emerging pathogens; thus, an improved understanding of the cellular factors that modulate infection in their respective vertebrate and insect hosts may aid control efforts. In particular, cell-intrinsic antiviral pathways restrict vector-borne viruses including the type I interferon response in vertebrates and the RNA interference (RNAi) pathway in insects. However, it is likely that additional cell-intrinsic mechanisms exist to limit these viruses. Since insects rely on innate immune mechanisms to inhibit virus infections, we used Drosophila as a model insect to identify cellular factors that restrict West Nile virus (WNV), a flavivirus with a broad and expanding geographical host range. Our genome-wide RNAi screen identified 50 genes that inhibited WNV infection. Further screening revealed that 17 of these genes were antiviral against additional flaviviruses, and seven of these were antiviral against other vector-borne viruses, expanding our knowledge of invertebrate cell-intrinsic immunity. Investigation of two newly identified factors that restrict diverse viruses, dXPO1 and dRUVBL1, in the Tip60 complex, demonstrated they contributed to antiviral defense at the organismal level in adult flies, in mosquito cells, and in mammalian cells. These data suggest the existence of broadly acting and functionally conserved antiviral genes and pathways that restrict virus infections in evolutionarily divergent hosts.


DNA Helicases/genetics , Drosophila Proteins/genetics , Flavivirus Infections/genetics , Host-Parasite Interactions/genetics , Karyopherins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Culicidae , Drosophila melanogaster , Flavivirus/genetics , Flavivirus Infections/immunology , Genome-Wide Association Study , Humans , RNA, Small Interfering/analysis , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , West Nile virus/genetics , West Nile virus/immunology , Exportin 1 Protein
3.
Cell Rep ; 5(6): 1737-48, 2013 Dec 26.
Article En | MEDLINE | ID: mdl-24332855

Alphaviruses are a large class of insect-borne human pathogens and little is known about the host-factor requirements for infection. To identify such factors, we performed a genome-wide RNAi screen using model Drosophila cells and validated 94 genes that impacted infection of Sindbis virus (SINV), the prototypical alphavirus. We identified a conserved role for SEC61A and valosin-containing protein (VCP) in facilitating SINV entry in insects and mammals. SEC61A and VCP selectively regulate trafficking of the entry receptor NRAMP2, and loss or pharmacological inhibition of these proteins leads to altered NRAMP2 trafficking to lysosomal compartments and proteolytic digestion within lysosomes. NRAMP2 is the major iron transporter in cells, and loss of NRAMP2 attenuates intracellular iron transport. Thus, this study reveals genes and pathways involved in both infection and iron homeostasis that may serve as targets for antiviral therapeutics or for iron-imbalance disorders.


Adenosine Triphosphatases/metabolism , Drosophila Proteins/metabolism , Genome, Insect , Membrane Proteins/metabolism , Sindbis Virus/pathogenicity , Virus Internalization , Adenosine Triphosphatases/genetics , Aedes/genetics , Aedes/metabolism , Aedes/virology , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Drosophila/genetics , Drosophila/metabolism , Drosophila/virology , Drosophila Proteins/genetics , Humans , Iron/metabolism , Membrane Proteins/genetics , Protein Transport , RNA Interference , RNA, Small Interfering/genetics , SEC Translocation Channels , Valosin Containing Protein
4.
Cell Host Microbe ; 10(2): 97-104, 2011 Aug 18.
Article En | MEDLINE | ID: mdl-21843867

Alphaviruses, including several emerging human pathogens, are a large family of mosquito-borne viruses with Sindbis virus being a prototypical member of the genus. The host factor requirements and receptors for entry of this class of viruses remain obscure. Using a Drosophila system, we identified the divalent metal ion transporter natural resistance-associated macrophage protein (NRAMP) as a host cell surface molecule required for Sindbis virus binding and entry into Drosophila cells. Consequently, flies mutant for dNRAMP were protected from virus infection. NRAMP2, the ubiquitously expressed vertebrate homolog, mediated binding and infection of Sindbis virus into mammalian cells, and murine cells deficient for NRAMP2 were nonpermissive to infection. Alphavirus glycoprotein chimeras demonstrated that the requirement for NRAMP2 is at the level of Sindbis virus entry. Given the conserved structure of alphavirus glycoproteins, and the widespread use of transporters for viral entry, other alphaviruses may use conserved multipass membrane proteins for infection.


Cation Transport Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/virology , Receptors, Virus/metabolism , Sindbis Virus/pathogenicity , Alphavirus Infections/metabolism , Alphavirus Infections/virology , Animals , Biotinylation/methods , Cation Transport Proteins/genetics , Cell Line , Culicidae , Drosophila Proteins/genetics , Humans , Immunoprecipitation , Iron/pharmacology , Mammals , Mice , RNA Interference , RNA Stability , Sindbis Virus/drug effects , Sindbis Virus/genetics , Transfection , Vesiculovirus/genetics , Vesiculovirus/metabolism , Vesiculovirus/pathogenicity , Virus Attachment , Virus Internalization , West Nile virus/drug effects , West Nile virus/genetics , West Nile virus/pathogenicity
6.
J Interferon Cytokine Res ; 30(11): 843-52, 2010 Nov.
Article En | MEDLINE | ID: mdl-20586614

The cell autonomous response to viral infection is carefully regulated to induce type I interferons (IFNs), which in turn induce the establishment of an antiviral state. Leucine-rich repeat (in Flightless I) interacting protein-1 (LRRFIP1) and LRRFIP2 are 2 related proteins that have been identified as interacting with MyD88 and Flightless I homolog, a leucine-rich repeat protein. LRRFIP2 positively regulates NFκB and macrophage cytokine production after lipopolysaccharide, but less is known about LRRFIP1. We hypothesized that LRRFIP1 could be more important in antiviral responses, as overexpression led to type I IFN production in a pilot study. The induction of type I IFNs occurred even in the absence of virus, but was enhanced by the presence of virus. Conversely, knockdown of LRRFIP1 compromised IFN expression. We found that LRRFIP1 was rapidly recruited to influenza-containing early endosomes in a p38-dependent fashion. This was specific for virus-containing endosomes as there was almost no colocalization of LRRFIP1 with early endosomes in the absence of virus. Further, LRRFIP1 was recruited to RNA-containing vesicles. Taken together, these data suggest that LRRFIP1 participates in cell responses to virus at early time points and is important for type I IFN induction.


Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , RNA-Binding Proteins/metabolism , Animals , Endosomes/virology , Influenza A virus/pathogenicity , Interferon Type I/biosynthesis , Interferon Type I/genetics , Mice , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , NIH 3T3 Cells , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , Protein Binding , Protein Transport/immunology , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Transcriptional Activation/genetics , Transgenes/genetics
7.
J Virol ; 82(19): 9615-28, 2008 Oct.
Article En | MEDLINE | ID: mdl-18667499

Vascular endothelial cadherin (VE-cadherin) connects neighboring endothelial cells (ECs) via interendothelial junctions and regulates EC proliferation and adhesion during vasculogenesis and angiogenesis. The cytoplasmic domain of VE-cadherin recruits alpha- and beta-catenins and gamma-catenin, which interact with the actin cytoskeleton, thus modulating cell morphology. Dysregulation of the adherens junction/cytoskeletal axis is a hallmark of invasive tumors. We now demonstrate that the transmembrane ubiquitin ligase K5/MIR-2 of Kaposi's sarcoma-associated herpesvirus targets VE-cadherin for ubiquitin-mediated destruction, thus disturbing EC adhesion. In contrast, N-cadherin levels in K5-expressing cells were increased compared to those in control cells. Steady-state levels of alpha- and beta-catenins and gamma-catenin in K5-expressing ECs were drastically reduced due to proteasomal destruction. Moreover, the actin cytoskeleton was rearranged, resulting in the dysregulation of EC barrier function as measured by electric cell-substrate impedance sensing. Our data represent the first example of a viral protein targeting adherens junction proteins and suggest that K5 contributes to EC proliferation, vascular leakage, and the reprogramming of the EC proteome during Kaposi's sarcoma tumorigenesis.


Adherens Junctions/metabolism , Gene Expression Regulation, Viral , Herpesvirus 8, Human/metabolism , Actins/metabolism , Biotinylation , Catenins/metabolism , Cell Adhesion , Cell Membrane/metabolism , Cell Proliferation , Cell Survival , Cytoskeleton/metabolism , Electric Impedance , Humans , Skin/metabolism , Ubiquitin/metabolism
8.
J Virol ; 81(15): 8050-62, 2007 Aug.
Article En | MEDLINE | ID: mdl-17507477

Kaposi's sarcoma-associated herpesvirus (KSHV) is the pathological agent of Kaposi's sarcoma (KS), a tumor characterized by aberrant proliferation of endothelial-cell-derived spindle cells. Since in many cancers tumorigenesis is associated with an increase in the activity of the cathepsin family, we studied the role of cathepsins in KS using an in vitro model of KSHV-mediated endothelial cell transformation. Small-molecule inhibitors and small interfering RNA (siRNA) targeting CTSB, but not other cathepsins, inhibited KSHV-induced postconfluent proliferation and the formation of spindle cells and foci of dermal microvascular endothelial cells. Interestingly, neither CTSB mRNA nor CTSB protein levels were induced in endothelial cells latently infected with KSHV. Secretion of CTSB was strongly diminished upon KSHV infection. Increased targeting of CTSB to endosomes was caused by the induction by KSHV of the expression of insulin-like growth factor-II receptor (IGF-IIR), a mannose-6-phosphate receptor (M6PR) that binds to cathepsins. Inhibition of IGF-IIR/M6PR expression by siRNA released CTSB for secretion. In contrast to the increased cathepsin secretion observed in most other tumors, viral inhibition of CTSB secretion via induction of an M6PR is crucial for the transformation of endothelial cells.


Cathepsin B/metabolism , Cell Transformation, Neoplastic , Herpesvirus 8, Human/physiology , Receptor, IGF Type 2/metabolism , Antigens, Viral/genetics , Antigens, Viral/metabolism , Apoptosis/physiology , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cells, Cultured , Contact Inhibition , Dipeptides/metabolism , Endothelial Cells/cytology , Endothelial Cells/physiology , Enzyme Precursors/antagonists & inhibitors , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, IGF Type 2/genetics , Sarcoma, Kaposi
9.
Blood ; 108(6): 1932-40, 2006 Sep 15.
Article En | MEDLINE | ID: mdl-16601245

The transmembrane ubiquitin ligase K5/MIR2 of Kaposi sarcoma herpesvirus (KSHV) mediates internalization and lysosomal degradation of glycoproteins involved in antigen presentation and co-stimulation. In endothelial cells (ECs), K5 additionally reduced expression of CD31/platelet-endothelial cell adhesion molecule (PECAM), an adhesion molecule regulating cell-cell interactions of ECs, platelets, monocytes, and T cells. K5 also reduced EC migration, a CD31-dependent process. Unlike other K5 substrates, both newly synthesized and pre-existing CD31 molecules were targeted by K5. K5 was transported to the cell surface and ubiquitinated pre-existing CD31, resulting in endocytosis and lysosomal degradation. In the endoplasmic reticulum, newly synthesized CD31 was degraded by proteasomes, which required binding of phosphofurin acidic cluster sorting protein-2 (PACS-2) to acidic residues in the carboxyterminal tail of K5. Thus, CD31, a novel target of K5, is efficiently removed from ECs by a dual degradation mechanism that is regulated by the subcellular sorting of the ubiquitin ligase. K5-mediated degradation of CD31 is likely to affect EC function in KS tumors.


Endothelial Cells/immunology , Endothelial Cells/virology , Herpesvirus 8, Human/enzymology , Herpesvirus 8, Human/pathogenicity , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Cells, Cultured , Down-Regulation , Endothelial Cells/cytology , Humans , Proteasome Endopeptidase Complex/metabolism , Sarcoma, Kaposi/enzymology , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/virology , Substrate Specificity , Vesicular Transport Proteins , Viral Proteins/metabolism
10.
J Virol ; 79(7): 4357-68, 2005 Apr.
Article En | MEDLINE | ID: mdl-15767436

By selecting the R5 human immunodeficiency virus type 1 (HIV-1) strain JR-CSF for efficient use of a CCR5 coreceptor with a badly damaged amino terminus [i.e., CCR5(Y14N)], we previously isolated variants that weakly utilize CCR5(Delta18), a low-affinity mutant lacking the normal tyrosine sulfate-containing amino-terminal region of the coreceptor. These previously isolated HIV-1(JR-CSF) variants contained adaptive mutations situated exclusively in the V3 loop of their gp120 envelope glycoproteins. We now have weaned the virus from all dependency on the CCR5 amino terminus by performing additional selections with HeLa-CD4 cells that express only a low concentration of CCR5(Delta18). The adapted variants had additional mutations in their V3 loops, as well as one in the V2 stem (S193N) and four alternative mutations in the V4 loop that eliminated the same N-linked oligosaccharide from position N403. Assays using pseudotyped viruses suggested that these new gp120 mutations all made strong contributions to use of CCR5(Delta18) by accelerating a rate-limiting CCR5-dependent conformational change in gp41 rather than by increasing viral affinity for this damaged coreceptor. Consistent with this interpretation, loss of the V4 N-glycan at position N403 also enhanced HIV-1 use of a different low-affinity CCR5 coreceptor with a mutation in extracellular loop 2 (ECL2) [i.e., CCR5(G163R)], whereas the double mutant CCR5(Delta18,G163R) was inactive. We conclude that loss of the N-glycan at position N403 helps to convert the HIV-1 envelope into a hair-trigger form that no longer requires strong interactions with both the CCR5 amino terminus and ECL2 but efficiently uses either site alone. These results demonstrate a novel functional role for a gp120 N-linked oligosaccharide and a high degree of adaptability in coreceptor usage by HIV-1.


HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/physiology , HIV-1/genetics , HIV-1/physiology , Receptors, CCR5/physiology , Adaptation, Biological , Amides/pharmacology , Amino Acid Substitution , Cell Line , Cloning, Molecular , DNA Mutational Analysis , Enfuvirtide , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/pharmacology , HIV Envelope Protein gp41/physiology , HIV Fusion Inhibitors/pharmacology , HeLa Cells , Humans , Models, Molecular , Mutation , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Polysaccharides/chemistry , Polysaccharides/physiology , Quaternary Ammonium Compounds/pharmacology , Receptors, CCR5/chemistry , Receptors, CCR5/genetics , Virus Replication
11.
J Virol ; 77(22): 12336-45, 2003 Nov.
Article En | MEDLINE | ID: mdl-14581570

The genetic evolution of human immunodeficiency virus type 1 (HIV-1) in the brain is distinct from that in lymphoid tissues, indicating tissue-specific compartmentalization of the virus. Few primary HIV-1 envelope glycoproteins (Envs) from uncultured brain tissues have been biologically well characterized. In this study, we analyzed 37 full-length env genes from uncultured brain biopsy and blood samples from four patients with AIDS. Phylogenetic analysis of intrapatient sequence sets showed distinct clustering of brain relative to blood env sequences. However, no brain-specific signature sequence was identified. Furthermore, there was no significant difference in the number or positions of N-linked glycosylation sites between brain and blood env sequences. The patterns of coreceptor usage were heterogeneous, with no clear distinction between brain and blood env clones. Nine Envs used CCR5 as a coreceptor, one used CXCR4, and two used both CCR5 and CXCR4 in cell-to-cell fusion assays. Eight Envs could also use CCR3, CCR8, GPR15, STRL33, Apj, and/or GPR1, but these coreceptors did not play a major role in virus entry into microglia. Recognition of epitopes by the 2F5, T30, AG10H9, F105, 17b, and C11 monoclonal antibodies varied among env clones, reflecting genetic and conformational heterogeneity. Envs from two patients contained 28 to 32 N-glycosylation sites in gp120, compared to around 25 in lab strains and well-characterized primary isolates. These results suggest that HIV-1 Envs in brain cannot be distinguished from those in blood on the basis of coreceptor usage or the number or positions of N-glycosylation sites, indicating that other properties underlie neurotropism. The study also demonstrates characteristics of primary HIV-1 Envs from uncultured tissues and implies that Env variants that are glycosylated more extensively than lab strains and well-characterized primary isolates should be considered during development of vaccines and neutralizing antibodies.


Acquired Immunodeficiency Syndrome/virology , Brain/virology , Genes, env , HIV-1/genetics , Viremia/virology , Amino Acid Sequence , CCR5 Receptor Antagonists , HIV-1/classification , Humans , Molecular Sequence Data , Phylogeny , Receptors, CCR5/physiology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/physiology , Structure-Activity Relationship , Virus Replication
...