Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
PLoS Pathog ; 19(11): e1011795, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38011215

ABSTRACT

Zika virus (ZIKV) serine protease, indispensable for viral polyprotein processing and replication, is composed of the membrane-anchored NS2B polypeptide and the N-terminal domain of the NS3 polypeptide (NS3pro). The C-terminal domain of the NS3 polypeptide (NS3hel) is necessary for helicase activity and contains an ATP-binding site. We discovered that ZIKV NS2B-NS3pro binds single-stranded RNA with a Kd of ~0.3 µM, suggesting a novel function. We tested various structural modifications of NS2B-NS3pro and observed that constructs stabilized in the recently discovered "super-open" conformation do not bind RNA. Likewise, stabilizing NS2B-NS3pro in the "closed" (proteolytically active) conformation using substrate inhibitors abolished RNA binding. We posit that RNA binding occurs when ZIKV NS2B-NS3pro adopts the "open" conformation, which we modeled using highly homologous dengue NS2B-NS3pro crystallized in the open conformation. We identified two positively charged fork-like structures present only in the open conformation of NS3pro. These forks are conserved across Flaviviridae family and could be aligned with the positively charged grove on NS3hel, providing a contiguous binding surface for the negative RNA strand exiting helicase. We propose a "reverse inchworm" model for a tightly intertwined NS2B-NS3 helicase-protease machinery, which suggests that NS2B-NS3pro cycles between open and super-open conformations to bind and release RNA enabling long-range NS3hel processivity. The transition to the closed conformation, likely induced by the substrate, enables the classical protease activity of NS2B-NS3pro.


Subject(s)
Zika Virus Infection , Zika Virus , Humans , Zika Virus/genetics , Viral Nonstructural Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Peptides , RNA , Protease Inhibitors
2.
Viruses ; 15(5)2023 04 30.
Article in English | MEDLINE | ID: mdl-37243192

ABSTRACT

The Zika virus (ZIKV), a member of the Flaviviridae family, is considered a major health threat causing multiple cases of microcephaly in newborns and Guillain-Barré syndrome in adults. In this study, we targeted a transient, deep, and hydrophobic pocket of the "super-open" conformation of ZIKV NS2B-NS3 protease to overcome the limitations of the active site pocket. After virtual docking screening of approximately seven million compounds against the novel allosteric site, we selected the top six candidates and assessed them in enzymatic assays. Six candidates inhibited ZIKV NS2B-NS3 protease proteolytic activity at low micromolar concentrations. These six compounds, targeting the selected protease pocket conserved in ZIKV, serve as unique drug candidates and open new opportunities for possible treatment against several flavivirus infections.


Subject(s)
Zika Virus Infection , Zika Virus , Infant, Newborn , Humans , Zika Virus/metabolism , Zika Virus Infection/drug therapy , Viral Nonstructural Proteins/chemistry , Serine Endopeptidases/metabolism , Peptide Hydrolases , Protein Conformation , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry
3.
Cell Rep Med ; 2(1): 100189, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33495758

ABSTRACT

The SARS-CoV-2 proteome shares regions of conservation with endemic human coronaviruses (CoVs), but it remains unknown to what extent these may be cross-recognized by the antibody response. Here, we study cross-reactivity using a highly multiplexed peptide assay (PepSeq) to generate an epitope-resolved view of IgG reactivity across all human CoVs in both COVID-19 convalescent and negative donors. PepSeq resolves epitopes across the SARS-CoV-2 Spike and Nucleocapsid proteins that are commonly targeted in convalescent donors, including several sites also recognized in some uninfected controls. By comparing patterns of homologous reactivity between CoVs and using targeted antibody-depletion experiments, we demonstrate that SARS-CoV-2 elicits antibodies that cross-recognize pandemic and endemic CoV antigens at two Spike S2 subunit epitopes. We further show that these cross-reactive antibodies preferentially bind endemic homologs. Our findings highlight sites at which the SARS-CoV-2 response appears to be shaped by previous CoV exposures and which have the potential to raise broadly neutralizing responses.

4.
bioRxiv ; 2020 Jul 27.
Article in English | MEDLINE | ID: mdl-32743570

ABSTRACT

A high-resolution understanding of the antibody response to SARS-CoV-2 is important for the design of effective diagnostics, vaccines and therapeutics. However, SARS-CoV-2 antibody epitopes remain largely uncharacterized, and it is unknown whether and how the response may cross-react with related viruses. Here, we use a multiplexed peptide assay ('PepSeq') to generate an epitope-resolved view of reactivity across all human coronaviruses. PepSeq accurately detects SARS-CoV-2 exposure and resolves epitopes across the Spike and Nucleocapsid proteins. Two of these represent recurrent reactivities to conserved, functionally-important sites in the Spike S2 subunit, regions that we show are also targeted for the endemic coronaviruses in pre-pandemic controls. At one of these sites, we demonstrate that the SARS-CoV-2 response strongly and recurrently cross-reacts with the endemic virus hCoV-OC43. Our analyses reveal new diagnostic and therapeutic targets, including a site at which SARS-CoV-2 may recruit common pre-existing antibodies and with the potential for broadly-neutralizing responses.

5.
J Inherit Metab Dis ; 43(6): 1333-1348, 2020 11.
Article in English | MEDLINE | ID: mdl-32681751

ABSTRACT

Asparagine-linked glycosylation 13 homolog (ALG13) encodes a nonredundant, highly conserved, X-linked uridine diphosphate (UDP)-N-acetylglucosaminyltransferase required for the synthesis of lipid linked oligosaccharide precursor and proper N-linked glycosylation. De novo variants in ALG13 underlie a form of early infantile epileptic encephalopathy known as EIEE36, but given its essential role in glycosylation, it is also considered a congenital disorder of glycosylation (CDG), ALG13-CDG. Twenty-four previously reported ALG13-CDG cases had de novo variants, but surprisingly, unlike most forms of CDG, ALG13-CDG did not show the anticipated glycosylation defects, typically detected by altered transferrin glycosylation. Structural homology modeling of two recurrent de novo variants, p.A81T and p.N107S, suggests both are likely to impact the function of ALG13. Using a corresponding ALG13-deficient yeast strain, we show that expressing yeast ALG13 with either of the highly conserved hotspot variants rescues the observed growth defect, but not its glycosylation abnormality. We present molecular and clinical data on 29 previously unreported individuals with de novo variants in ALG13. This more than doubles the number of known cases. A key finding is that a vast majority of the individuals presents with West syndrome, a feature shared with other CDG types. Among these, the initial epileptic spasms best responded to adrenocorticotropic hormone or prednisolone, while clobazam and felbamate showed promise for continued epilepsy treatment. A ketogenic diet seems to play an important role in the treatment of these individuals.


Subject(s)
Congenital Disorders of Glycosylation/genetics , N-Acetylglucosaminyltransferases/deficiency , N-Acetylglucosaminyltransferases/genetics , Spasms, Infantile/genetics , Biomarkers , Child, Preschool , Congenital Disorders of Glycosylation/diagnosis , Diet, Ketogenic , Female , Glycosylation , Humans , Infant , Male , Mutation , N-Acetylglucosaminyltransferases/chemistry , Spasms, Infantile/diagnosis , Transferrin/metabolism
6.
Molecules ; 25(10)2020 05 22.
Article in English | MEDLINE | ID: mdl-32455942

ABSTRACT

Processing of certain viral proteins and bacterial toxins by host serine proteases is a frequent and critical step in virulence. The coronavirus spike glycoprotein contains three (S1, S2, and S2') cleavage sites that are processed by human host proteases. The exact nature of these cleavage sites, and their respective processing proteases, can determine whether the virus can cross species and the level of pathogenicity. Recent comparisons of the genomes of the highly pathogenic SARS-CoV2 and MERS-CoV, with less pathogenic strains (e.g., Bat-RaTG13, the bat homologue of SARS-CoV2) identified possible mutations in the receptor binding domain and in the S1 and S2' cleavage sites of their spike glycoprotein. However, there remains some confusion on the relative roles of the possible serine proteases involved for priming. Using anthrax toxin as a model system, we show that in vivo inhibition of priming by pan-active serine protease inhibitors can be effective at suppressing toxicity. Hence, our studies should encourage further efforts in developing either pan-serine protease inhibitors or inhibitor cocktails to target SARS-CoV2 and potentially ward off future pandemics that could develop because of additional mutations in the S-protein priming sequence in coronaviruses.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Serine Proteases/metabolism , Serine Proteinase Inhibitors/pharmacology , Spike Glycoprotein, Coronavirus/metabolism , Animals , Antigens, Bacterial/toxicity , Antiviral Agents/therapeutic use , Bacterial Toxins/toxicity , Betacoronavirus/pathogenicity , Binding Sites , COVID-19 , Drug Delivery Systems , Female , Furin/pharmacology , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Pandemics , RAW 264.7 Cells , SARS-CoV-2 , Serine Proteinase Inhibitors/therapeutic use , Spike Glycoprotein, Coronavirus/chemistry
7.
Article in English | MEDLINE | ID: mdl-31160359

ABSTRACT

Jacobsen syndrome (OMIM #147791) is a rare contiguous gene disorder caused by deletions in distal 11q. The clinical phenotype is variable and can include dysmorphic features, varying degrees of intellectual disability, behavioral problems including autism and attention deficit hyperactivity disorder, congenital heart defects, structural kidney defects, genitourinary problems, immunodeficiency, and a bleeding disorder due to impaired platelet production and function. Previous studies combining both human and animal systems have implicated several disease-causing genes in distal 11q that contribute to the Jacobsen syndrome phenotype. One gene, ETS1, has been implicated in causing congenital heart defects, structural kidney defects, and immunodeficiency. We performed a comprehensive phenotypic analysis on a patient with congenital heart disease previously found to have a de novo frameshift mutation in ETS1, resulting in the loss of the DNA-binding domain of the protein. Our results suggest that loss of Ets1 causes a "partial Jacobsen syndrome phenotype" including congenital heart disease, facial dysmorphism, intellectual disability, and attention deficit hyperactivity disorder.


Subject(s)
Heart Defects, Congenital/genetics , Jacobsen Distal 11q Deletion Syndrome/genetics , Proto-Oncogene Protein c-ets-1/genetics , Frameshift Mutation , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/pathology , Humans , Infant, Newborn , Jacobsen Distal 11q Deletion Syndrome/diagnosis , Jacobsen Distal 11q Deletion Syndrome/pathology , Male , Phenotype , Sequence Deletion
8.
Neuron ; 101(6): 1089-1098.e4, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30713029

ABSTRACT

Zika virus (ZIKV) targets neural progenitor cells in the brain, attenuates cell proliferation, and leads to cell death. Here, we describe a role for the ZIKV protease NS2B-NS3 heterodimer in mediating neurotoxicity through cleavage of a host protein required for neurogenesis. Similar to ZIKV infection, NS2B-NS3 expression led to cytokinesis defects and cell death in a protease activity-dependent fashion. Among binding partners, NS2B-NS3 cleaved Septin-2, a cytoskeletal factor involved in cytokinesis. Cleavage of Septin-2 occurred at residue 306 and forced expression of a non-cleavable Septin-2 restored cytokinesis, suggesting a direct mechanism of ZIKV-induced neural toxicity. VIDEO ABSTRACT.


Subject(s)
Apoptosis , Cytokinesis , Mitosis , Neural Stem Cells/metabolism , Septins/metabolism , Viral Nonstructural Proteins/metabolism , Zika Virus/metabolism , Cytoskeleton/metabolism , HEK293 Cells , HeLa Cells , Humans , Neurogenesis , RNA Helicases/metabolism , Serine Endopeptidases/metabolism
10.
Cell Discov ; 4: 31, 2018.
Article in English | MEDLINE | ID: mdl-29872540

ABSTRACT

The re-emergence of Zika virus (ZIKV) and Ebola virus (EBOV) poses serious and continued threats to the global public health. Effective therapeutics for these maladies is an unmet need. Here, we show that emetine, an anti-protozoal agent, potently inhibits ZIKV and EBOV infection with a low nanomolar half maximal inhibitory concentration (IC50) in vitro and potent activity in vivo. Two mechanisms of action for emetine are identified: the inhibition of ZIKV NS5 polymerase activity and disruption of lysosomal function. Emetine also inhibits EBOV entry. Cephaeline, a desmethyl analog of emetine, which may be better tolerated in patients than emetine, exhibits a similar efficacy against both ZIKV and EBOV infections. Hence, emetine and cephaeline offer pharmaceutical therapies against both ZIKV and EBOV infection.

11.
Sci Rep ; 8(1): 1218, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352135

ABSTRACT

The outbreak of the Zika virus (ZIKV) has been associated with increased incidence of congenital malformations. Although recent efforts have focused on vaccine development, treatments for infected individuals are needed urgently. Sofosbuvir (SOF), an FDA-approved nucleotide analog inhibitor of the Hepatitis C (HCV) RNA-dependent RNA polymerase (RdRp) was recently shown to be protective against ZIKV both in vitro and in vivo. Here, we show that SOF protected human neural progenitor cells (NPC) and 3D neurospheres from ZIKV infection-mediated cell death and importantly restored the antiviral immune response in NPCs. In vivo, SOF treatment post-infection (p.i.) decreased viral burden in an immunodeficient mouse model. Finally, we show for the first time that acute SOF treatment of pregnant dams p.i. was well-tolerated and prevented vertical transmission of the virus to the fetus. Taken together, our data confirmed SOF-mediated sparing of human neural cell types from ZIKV-mediated cell death in vitro and reduced viral burden in vivo in animal models of chronic infection and vertical transmission, strengthening the growing body of evidence for SOF anti-ZIKV activity.

12.
Front Microbiol ; 9: 3252, 2018.
Article in English | MEDLINE | ID: mdl-30666246

ABSTRACT

The Zika virus (ZIKV) global epidemic prompted the World Health Organization to declare it a 2016 Public Health Emergency of International Concern. The overwhelming experience over the past several years teaches us that ZIKV and the associated neurological complications represent a long-term world-wide challenge to public health. Although the number of ZIKV cases in the Western Hemisphere has dropped since 2016, the need for basic research and anti-ZIKV drug development remains strong. Re-emerging viruses like ZIKV are an ever-present threat in the 21st century where fast transcontinental travel lends itself to viral epidemics. Here, we first present the origin story for ZIKV and review the rapid progress researchers have made toward understanding of the ZIKV pathology and in the design, re-purposing, and testing-particularly in vivo-drug candidates for ZIKV prophylaxis and therapy ZIKV. Quite remarkably, a short, but intensive, drug-repurposing effort has already resulted in several readily available FDA-approved drugs that are capable of effectively combating the virus in infected adult mouse models and, most importantly, in both preventing maternal-fetal transmission and severe microcephaly in newborns in pregnant mouse models.

13.
Sci Rep ; 7(1): 15771, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29150641

ABSTRACT

One of the major challenges of the current Zika virus (ZIKV) epidemic is to prevent congenital foetal abnormalities, including microcephaly, following ZIKV infection of pregnant women. Given the urgent need for ZIKV prophylaxis and treatment, repurposing of approved drugs appears to be a viable and immediate solution. We demonstrate that the common anti-malaria drug chloroquine (CQ) extends the lifespan of ZIKV-infected interferon signalling-deficient AG129 mice. However, the severity of ZIKV infection in these mice precludes the study of foetal (vertical) viral transmission. Here, we show that interferon signalling-competent SJL mice support chronic ZIKV infection. Infected dams and sires are both able to transmit ZIKV to the offspring, making this an ideal model for in vivo validation of compounds shown to suppress ZIKV in cell culture. Administration of CQ to ZIKV-infected pregnant SJL mice during mid-late gestation significantly attenuated vertical transmission, reducing the ZIKV load in the foetal brain more than 20-fold. Given the limited side effects of CQ, its lack of contraindications in pregnant women, and its worldwide availability and low cost, we suggest that CQ could be considered for the treatment and prophylaxis of ZIKV.


Subject(s)
Antimalarials/therapeutic use , Chloroquine/therapeutic use , Drug Repositioning , Zika Virus Infection/drug therapy , Zika Virus Infection/prevention & control , Zika Virus/physiology , Animals , Antimalarials/pharmacology , Chloroquine/pharmacology , Disease Models, Animal , Humans , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Zika Virus/drug effects , Zika Virus Infection/transmission
14.
Antiviral Res ; 143: 218-229, 2017 07.
Article in English | MEDLINE | ID: mdl-28461069

ABSTRACT

The recent re-emergence of Zika virus (ZIKV)1, a member of the Flaviviridae family, has become a global emergency. Currently, there are no effective methods of preventing or treating ZIKV infection, which causes severe neuroimmunopathology and is particularly harmful to the developing fetuses of infected pregnant women. However, the pathology induced by ZIKV is unique among flaviviruses, and knowledge of the biology of other family members cannot easily be extrapolated to ZIKV. Thus, structure-function studies of ZIKV proteins are urgently needed to facilitate the development of effective preventative and therapeutic agents. Like other flaviviruses, ZIKV expresses an NS2B-NS3 protease, which consists of the NS2B cofactor and the NS3 protease domain and is essential for cleavage of the ZIKV polyprotein precursor and generation of fully functional viral proteins. Here, we report the enzymatic characterization of ZIKV protease, and we identify structural scaffolds for allosteric small-molecule inhibitors of this protease. Molecular modeling of the protease-inhibitor complexes suggests that these compounds bind to the druggable cavity in the NS2B-NS3 protease interface and affect productive interactions of the protease domain with its cofactor. The most potent compound demonstrated efficient inhibition of ZIKV propagation in vitro in human fetal neural progenitor cells and in vivo in SJL mice. The inhibitory scaffolds could be further developed into valuable research reagents and, ultimately, provide a roadmap for the selection of efficient inhibitors of ZIKV infection.


Subject(s)
Allosteric Site , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/chemistry , Zika Virus/enzymology , Amino Acid Sequence , Animals , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/chemistry , Base Sequence , Enzyme Activation , Female , Flavivirus/chemistry , Gene Expression , Humans , Inhibitory Concentration 50 , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , RNA Helicases/chemistry , RNA Helicases/drug effects , SOXB1 Transcription Factors/genetics , Sequence Alignment , Serine Endopeptidases/chemistry , Serine Endopeptidases/drug effects , Stem Cells , Viral Nonstructural Proteins/drug effects , Viral Proteins/chemistry , Viral Proteins/genetics , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus/growth & development , Zika Virus Infection/virology
15.
Oncotarget ; 8(2): 2781-2799, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27835863

ABSTRACT

The invasion-promoting MT1-MMP is a cell surface-associated collagenase with a plethora of critical cellular functions. There is a consensus that MT1-MMP is a key protease in aberrant pericellular proteolysis in migrating cancer cells and, accordingly, a promising drug target. Because of high homology in the MMP family and a limited success in the design of selective small-molecule inhibitors, it became evident that the inhibitor specificity is required for selective and successful MT1-MMP therapies. Using the human Fab antibody library (over 1.25×109 individual variants) that exhibited the extended, 23-27 residue long, VH CDR-H3 segments, we isolated a panel of the inhibitory antibody fragments, from which the 3A2 Fab outperformed others as a specific and potent, low nanomolar range, inhibitor of MT1-MMP. Here, we report the in-depth characterization of the 3A2 antibody. Our multiple in vitro and cell-based tests and assays, and extensive structural modeling of the antibody/protease interactions suggest that the antibody epitope involves the residues proximal to the protease catalytic site and that, in contrast with tissue inhibitor-2 of MMPs (TIMP-2), the 3A2 Fab inactivates the protease functionality by binding to the catalytic domain outside the active site cavity. In agreement with the studies in metastasis by others, our animal studies in acute pulmonary melanoma metastasis support a key role of MT1-MMP in metastatic process. Conversely, the selective anti-MT1-MMP monotherapy significantly alleviated melanoma metastatic burden. It is likely that further affinity maturation of the 3A2 Fab will result in the lead inhibitor and a proof-of-concept for MT1-MMP targeting in metastatic cancers.


Subject(s)
Antibodies, Blocking/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Neoplasms/metabolism , Neoplasms/pathology , Animals , Antibodies, Blocking/chemistry , Antineoplastic Agents, Immunological/chemistry , Binding, Competitive , Catalytic Domain , Cell Line, Tumor , Cell Movement , Cell Survival , Collagen/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Female , Heterografts , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/pharmacology , Matrix Metalloproteinase Inhibitors/chemistry , Mice , Models, Molecular , Molecular Conformation , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/drug therapy , Protein Binding , Proteolysis , Recombinant Proteins/metabolism
16.
Hum Mutat ; 37(7): 653-60, 2016 07.
Article in English | MEDLINE | ID: mdl-26931382

ABSTRACT

Congenital disorders of glycosylation (CDG) arise from pathogenic mutations in over 100 genes leading to impaired protein or lipid glycosylation. ALG1 encodes a ß1,4 mannosyltransferase that catalyzes the addition of the first of nine mannose moieties to form a dolichol-lipid linked oligosaccharide intermediate required for proper N-linked glycosylation. ALG1 mutations cause a rare autosomal recessive disorder termed ALG1-CDG. To date 13 mutations in 18 patients from 14 families have been described with varying degrees of clinical severity. We identified and characterized 39 previously unreported cases of ALG1-CDG from 32 families and add 26 new mutations. Pathogenicity of each mutation was confirmed based on its inability to rescue impaired growth or hypoglycosylation of a standard biomarker in an alg1-deficient yeast strain. Using this approach we could not establish a rank order comparison of biomarker glycosylation and patient phenotype, but we identified mutations with a lethal outcome in the first two years of life. The recently identified protein-linked xeno-tetrasaccharide biomarker, NeuAc-Gal-GlcNAc2 , was seen in all 27 patients tested. Our study triples the number of known patients and expands the molecular and clinical correlates of this disorder.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Mannosyltransferases/genetics , Mutation , Polysaccharides/metabolism , Biomarkers/metabolism , Congenital Disorders of Glycosylation/metabolism , Female , Genes, Lethal , Glycosylation , Humans , Male , Sequence Analysis, DNA , Survival Analysis
17.
J Neuroinflammation ; 12: 158, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26337825

ABSTRACT

BACKGROUND: Mechanical pain hypersensitivity associated with physical trauma to peripheral nerve depends on T-helper (Th) cells expressing the algesic cytokine, interleukin (IL)-17A. Fibronectin (FN) isoform alternatively spliced within the IIICS region encoding the 25-residue-long connecting segment 1 (CS1) regulates T cell recruitment to the sites of inflammation. Herein, we analyzed the role of CS1-containing FN (FN-CS1) in IL-17A expression and pain after peripheral nerve damage. METHODS: Mass spectrometry, immunoblotting, and FN-CS1-specific immunofluorescence analyses were employed to examine FN expression after chronic constriction injury (CCI) in rat sciatic nerves. The acute intra-sciatic nerve injection of the synthetic CS1 peptide (a competitive inhibitor of the FN-CS1/α4 integrin binding) was used to elucidate the functional significance of FN-CS1 in mechanical and thermal pain hypersensitivity and IL-17A expression (by quantitative Taqman RT-PCR) after CCI. The CS1 peptide effects were analyzed in cultured primary Schwann cells, the major source of FN-CS1 in CCI nerves. RESULTS: Following CCI, FN expression in sciatic nerve increased with the dominant FN-CS1 deposition in endothelial cells, Schwann cells, and macrophages. Acute CS1 therapy attenuated mechanical allodynia (pain from innocuous stimulation) but not thermal hyperalgesia and reduced the levels of IL-17A expression in the injured nerve. CS1 peptide inhibited the LPS- or starvation-stimulated activation of the stress ERK/MAPK pathway in cultured Schwann cells. CONCLUSIONS: After physical trauma to the peripheral nerve, FN-CS1 contributes to mechanical pain hypersensitivity by increasing the number of IL-17A-expressing (presumably, Th17) cells. CS1 peptide therapy can be developed for pharmacological control of neuropathic pain.


Subject(s)
Hyperalgesia/etiology , Hyperalgesia/metabolism , Interleukin-17/metabolism , Peptides/metabolism , Sciatic Neuropathy/complications , Animals , Animals, Newborn , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cells, Cultured , Disease Models, Animal , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hyperalgesia/drug therapy , Intercellular Signaling Peptides and Proteins , Interleukin-17/genetics , Pain Measurement , Peptides/therapeutic use , Rats , Rats, Sprague-Dawley , Schwann Cells/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Neuropathy/pathology , Time Factors
18.
Chem Biol ; 22(8): 1122-33, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26256476

ABSTRACT

Matrix metalloproteinases (MMPs) play incompletely understood roles in health and disease. Knowing the MMP cleavage preferences is essential for a better understanding of the MMP functions and design of selective inhibitors. To elucidate the cleavage preferences of MMPs, we employed a high-throughput multiplexed peptide-centric profiling technology involving the cleavage of 18,583 peptides by 18 proteinases from the main sub-groups of the MMP family. Our results enabled comparison of the MMP substrates on a global scale, leading to the most efficient and selective substrates. The data validated the accuracy of our cleavage prediction software. This software allows us and others to locate, with nearly 100% accuracy, the MMP cleavage sites in the peptide sequences. In addition to increasing our understanding of both the selectivity and the redundancy of the MMP family, our study generated a roadmap for the subsequent MMP structural-functional studies and efficient substrate and inhibitor design.


Subject(s)
High-Throughput Screening Assays/methods , Metalloproteases/chemistry , Metalloproteases/metabolism , Amino Acid Sequence , Catalysis , Humans , Hydrolysis , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Peptides , Protein Structure, Tertiary , Structure-Activity Relationship , Substrate Specificity
19.
J Biol Chem ; 290(6): 3693-707, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25488667

ABSTRACT

Neuronal glial antigen 2 (NG2) is an integral membrane chondroitin sulfate proteoglycan expressed by vascular pericytes, macrophages (NG2-Mφ), and progenitor glia of the nervous system. Herein, we revealed that NG2 shedding and axonal growth, either independently or jointly, depended on the pericellular remodeling events executed by membrane-type 1 matrix metalloproteinase (MT1-MMP/MMP-14). Using purified NG2 ectodomain constructs, individual MMPs, and primary NG2-Mφ cultures, we demonstrated for the first time that MMP-14 performed as an efficient and unconventional NG2 sheddase and that NG2-Mφ infiltrated into the damaged peripheral nervous system. We then characterized the spatiotemporal relationships among MMP-14, MMP-2, and tissue inhibitor of metalloproteinases-2 in sciatic nerve. Tissue inhibitor of metalloproteinases-2-free MMP-14 was observed in the primary Schwann cell cultures using the inhibitory hydroxamate warhead-based MP-3653 fluorescent reporter. In teased nerve fibers, MMP-14 translocated postinjury toward the nodes of Ranvier and its substrates, laminin and NG2. Inhibition of MMP-14 activity using the selective, function-blocking DX2400 human monoclonal antibody increased the levels of regeneration-associated factors, including laminin, growth-associated protein 43, and cAMP-dependent transcription factor 3, thereby promoting sensory axon regeneration after nerve crush. Concomitantly, DX2400 therapy attenuated mechanical hypersensitivity associated with nerve crush in rats. Together, our findings describe a new model in which MMP-14 proteolysis regulates the extracellular milieu and presents a novel therapeutic target in the damaged peripheral nervous system and neuropathic pain.


Subject(s)
Antigens/metabolism , Macrophages/metabolism , Matrix Metalloproteinase 14/metabolism , Peripheral Nerve Injuries/metabolism , Proteoglycans/metabolism , Animals , Axons/physiology , Cell Membrane/metabolism , Cells, Cultured , Extracellular Space/metabolism , Female , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , HEK293 Cells , Humans , Laminin/genetics , Laminin/metabolism , MCF-7 Cells , Male , Matrix Metalloproteinase 2/metabolism , Nerve Regeneration , Peripheral Nerve Injuries/physiopathology , Proteolysis , Rats , Rats, Sprague-Dawley , Schwann Cells/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/physiology
20.
PLoS One ; 9(11): e113896, 2014.
Article in English | MEDLINE | ID: mdl-25411788

ABSTRACT

Bacteroides fragilis causes the majority of Gram-negative anaerobic infections in the humans. The presence of a short, 6-kb, pathogenicity island in the genome is linked to enterotoxigenic B. fragilis (ETBF). The role of the enterotoxin in B. fragilis virulence, however, remains to be determined, as the majority of clinical isolates lack ETBF genes and healthy individuals carry enterotoxin-positive B. fragilis. The island encodes secretory metalloproteinase II (MPII) and one of three homologous enterotoxigenic fragilysin isoenzymes (FRA; also termed B. fragilis toxin or BFT). The secretory metalloproteinases expressed from the genes on the B. fragilis pathogenicity island may have pathological importance within the gut, not linked to diarrhea. MPII and FRA are counter-transcribed in the bacterial genome, implying that regardless of their structural similarity and overlapping cleavage preferences these proteases perform distinct and highly specialized functions in the course of B. fragilis infection. The earlier data by us and others have demonstrated that FRA cleaves cellular E-cadherin, an important adherens junction protein, and weakens cell-to-cell contacts. Using E-cadherin-positive and E-cadherin-deficient cancer cells, and the immunostaining, direct cell binding and pull-down approaches, we, however, demonstrated that MPII via its catalytic domain efficiently binds, rather than cleaves, E-cadherin. According to our results, E-cadherin is an adherens junction cellular receptor, rather than a proteolytic target, of the B. fragilis secretory MPII enzyme. As a result of the combined FRA and MPII proteolysis, cell-to-cell contacts and adherens junctions are likely to weaken further.


Subject(s)
Bacterial Proteins/metabolism , Bacteroides fragilis/metabolism , Cadherins/metabolism , Metalloproteases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacteroides fragilis/genetics , Bacteroides fragilis/pathogenicity , Catalytic Domain , Cell Line, Tumor , Cell Membrane/metabolism , Genome, Bacterial , Genomic Islands , HCT116 Cells , HT29 Cells , Humans , MCF-7 Cells , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Metalloproteases/chemistry , Metalloproteases/genetics , Microscopy, Fluorescence , Protein Binding , Proteolysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...