Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Proc Natl Acad Sci U S A ; 116(8): 3100-3105, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718426

ABSTRACT

Denileukin diftitox (DAB-IL-2, Ontak) is a diphtheria-toxin-based fusion protein that depletes CD25-positive cells including regulatory T cells and has been approved for the treatment of persistent or recurrent cutaneous T cell lymphoma. However, the clinical use of denileukin diftitox was limited by vascular leak toxicity and production issues related to drug aggregation and purity. We found that a single amino acid substitution (V6A) in a motif associated with vascular leak induction yields a fully active, second-generation biologic, s-DAB-IL-2(V6A), which elicits 50-fold less human umbilical vein endothelial cell monolayer permeation and is 3.7-fold less lethal to mice by LD50 analysis than s-DAB-IL-2. Additionally, to overcome aggregation problems, we developed a production method for the fusion toxin using Corynebacterium diphtheriae that secretes fully folded, biologically active, monomeric s-DAB-IL-2 into the culture medium. Using the poorly immunogenic mouse B16F10 melanoma model, we initiated treatment 7 days after tumor challenge and observed that, while both s-DAB-IL-2(V6A) and s-DAB-IL-2 are inhibitors of tumor growth, the capacity to treat with higher doses of s-DAB-IL-2(V6A) could provide a superior activity window. In a sequential dual-therapy study in tumors that have progressed for 10 days, both s-DAB-IL-2(V6A) and s-DAB-IL-2 given before checkpoint inhibition with anti-programmed cell death-1 (anti-PD-1) antibodies inhibited tumor growth, while either drug given as monotherapy had less effect. s-DAB-IL-2(V6A), a fully monomeric protein with reduced vascular leak, is a second-generation diphtheria-toxin-based fusion protein with promise as a cancer immunotherapeutic both alone and in conjunction with PD-1 blockade.


Subject(s)
Diphtheria Toxin/administration & dosage , Interleukin-2/administration & dosage , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/genetics , Amino Acid Substitution/genetics , Antibodies/administration & dosage , Cell Proliferation/drug effects , Corynebacterium diphtheriae/chemistry , Corynebacterium diphtheriae/pathogenicity , Diphtheria Toxin/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Immunosuppressive Agents/administration & dosage , Immunotoxins/administration & dosage , Interleukin-2/chemistry , Interleukin-2 Receptor alpha Subunit/drug effects , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , T-Lymphocytes, Regulatory/drug effects
2.
J Infect Dis ; 224(11): 1962-1972, 2021 12 01.
Article in English | MEDLINE | ID: mdl-33955457

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are present in elevated numbers in tuberculosis patients and have been found to be permissive for Mycobacterium tuberculosis proliferation. To determine whether depletion of MDSCs may improve host control of tuberculosis, we used a novel diphtheria toxin-based fusion protein DABIL-4 that targets and depletes interleukin 4 (IL-4) receptor-positive cells. We show that DABIL-4 depletes both polymorphonuclear MDSCs and monocytic MDSCs, increases interferon-γ + T cells, and reduces the lung bacillary burden in a mouse tuberculosis model. These results indicate that MDSC-depleting therapies targeting the IL-4 receptor are beneficial in tuberculosis and offer an avenue towards host-directed tuberculosis therapy.


Subject(s)
Diphtheria Toxin/therapeutic use , Immunotherapy/methods , Mycobacterium tuberculosis/immunology , Myeloid-Derived Suppressor Cells/immunology , Tuberculosis/therapy , Animals , Disease Models, Animal , Mice , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes
3.
J Infect Dis ; 215(12): 1883-1887, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28863467

ABSTRACT

Host-directed therapies that augment host immune effector mechanisms may serve as important adjunctive therapies for tuberculosis treatment. We evaluated the activity of denileukin diftitox in an acute mouse model of tuberculosis (TB) infection and analyzed the cellular composition and bacterial burden in lungs and spleens. These in vivo studies show that denileukin diftitox potentiates standard TB treatment in the mouse model, an effect which may be due to depletion of T-regulatory and myeloid-derived suppressor cells during TB infection. Our results indicate that denileukin diftitox and other suppressor cell-depleting therapies may be useful adjunctive, host-directed therapies for TB.


Subject(s)
Diphtheria Toxin/therapeutic use , Immunotherapy/methods , Interleukin-2/therapeutic use , Mycobacterium tuberculosis/immunology , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes, Regulatory/immunology , Tuberculosis/therapy , Animals , Disease Models, Animal , Humans , Lung/immunology , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/therapeutic use , Recombinant Proteins/therapeutic use , Spleen/immunology
4.
Nat Commun ; 14(1): 7427, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37973991

ABSTRACT

As one of the most successful human pathogens, Mycobacterium tuberculosis (Mtb) has evolved a diverse array of determinants to subvert host immunity and alter host metabolic patterns. However, the mechanisms of pathogen interference with host metabolism remain poorly understood. Here we show that a glutamine metabolism antagonist, JHU083, inhibits Mtb proliferation in vitro and in vivo. JHU083-treated mice exhibit weight gain, improved survival, a 2.5 log lower lung bacillary burden at 35 days post-infection, and reduced lung pathology. JHU083 treatment also initiates earlier T-cell recruitment, increased proinflammatory myeloid cell infiltration, and a reduced frequency of immunosuppressive myeloid cells when compared to uninfected and rifampin-treated controls. Metabolomic analysis of lungs from JHU083-treated Mtb-infected mice reveals citrulline accumulation, suggesting elevated nitric oxide (NO) synthesis, and lowered levels of quinolinic acid which is derived from the immunosuppressive metabolite kynurenine. JHU083-treated macrophages also produce more NO potentiating their antibacterial activity. When tested in an immunocompromised mouse model of Mtb infection, JHU083 loses its therapeutic efficacy suggesting the drug's host-directed effects are likely to be predominant. Collectively, these data reveal that JHU083-mediated glutamine metabolism inhibition results in dual antibacterial and host-directed activity against tuberculosis.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Mice , Humans , Animals , Glutamine/pharmacology , Tuberculosis/microbiology , Anti-Bacterial Agents/pharmacology
5.
bioRxiv ; 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36865287

ABSTRACT

As one of the most successful human pathogens, Mycobacterium tuberculosis (Mtb) has evolved a diverse array of determinants to subvert host immunity and alter host metabolic patterns. However, the mechanisms of pathogen interference with host metabolism remain poorly understood. Here we show that a novel glutamine metabolism antagonist, JHU083, inhibits Mtb proliferation in vitro and in vivo. JHU083-treated mice exhibit weight gain, improved survival, a 2.5 log lower lung bacillary burden at 35 days post-infection, and reduced lung pathology. JHU083 treatment also initiates earlier T-cell recruitment, increased proinflammatory myeloid cell infiltration, and a reduced frequency of immunosuppressive myeloid cells when compared to uninfected and rifampin-treated controls. Metabolomics analysis of lungs from JHU083-treated Mtb-infected mice revealed reduced glutamine levels, citrulline accumulation suggesting elevated NOS activity, and lowered levels of quinolinic acid which is derived from the immunosuppressive metabolite kynurenine. When tested in an immunocompromised mouse model of Mtb infection, JHU083 lost its therapeutic efficacy suggesting the drug's host-directed effects are likely to be predominant. Collectively, these data reveal that JHU083-mediated glutamine metabolism inhibition results in dual antibacterial and host-directed activity against tuberculosis.

6.
Mol Microbiol ; 81(5): 1390-401, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21797942

ABSTRACT

Anthrax toxin is an A/B bacterial protein toxin which is composed of the enzymatically active Lethal Factor (LF) and/or Oedema Factor (EF) bound to Protective Antigen 63 (PA63) which functions as both the receptor binding and transmembrane domains. Once the toxin binds to its cell surface receptors it is internalized into the cell and traffics through Rab5- and Rab7-associated endosomal vesicles. Following acidification of the vesicle lumen, PA63 undergoes a dynamic change forming a beta-barrel that inserts into and forms a pore through the endosomal membrane. It is widely recognized that LF, and the related fusion protein LFnDTA, must be completely denatured in order to transit through the PA63 formed pore and enter the eukaryotic cell cytosol. We demonstrate by protease protection assays that the molecular chaperone GRP78 mediates the unfolding of LFnDTA and LF at neutral pH and thereby converts these proteins from a trypsin resistant to sensitive conformation. We have used immunoelectron microscopy and gold-labelled antibodies to demonstrate that both GRP78 and GRP94 chaperones are present in the lumen of endosomal vesicles. Finally, we have used siRNA to demonstrate that knock-down of GRP78 results in the emergence of resistance to anthrax lethal toxin and oedema toxin action.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Bacillus anthracis/chemistry , Bacillus anthracis/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Heat-Shock Proteins/metabolism , Protein Transport/physiology , Animals , Anthrax/metabolism , Anthrax/microbiology , Bacterial Proteins/metabolism , Cell Line , Cytosol/enzymology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Membrane Glycoproteins/biosynthesis , Mice , Protein Unfolding , RNA Interference , RNA, Small Interfering , Trypsin/metabolism , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
7.
PLoS One ; 17(2): e0263370, 2022.
Article in English | MEDLINE | ID: mdl-35113934

ABSTRACT

The cytokine Interferon-γ (IFN-γ) exerts powerful immunoregulatory effects on the adaptive immune system and also enhances functions of the neutrophil (PMN). The clinical use of IFN-γ has been driven by the finding that its administration to patients with chronic granulomatous disease (CGD) results in decreased incidence and severity of infections. However, IFN-γ has no effect on the characteristic defect of CGD, the inability to convert oxygen to microbicidal metabolites including superoxide anion (O2-) during the phagocytosis associated oxidative burst. We administered varying doses of IFN-γ to adult volunteers and studied the effects on plasma drug levels and response molecules and PMNs isolated from blood drawn at intervals over a 96- hour period. Plasma concentrations of IFN-γ, IP-10 and neopterin, and stimulated release of O2- from PMNs exhibited dose- and time-dependent increases after IFN-γ administration. Gene expression in PMNs was altered for 2775 genes; changes occurred rapidly after administration and returned to baseline in 24-36 hours. Several genes involved with neutrophil host defense were upregulated including those for components of the O2- generating NADPH oxidase; innate-immune and Fc receptors; proteins involved in MHCI and II; a regulator of circulating PMN number; guanylate binding proteins; and a key enzyme in synthesis of an essential NOS cofactor. Coordinate changes were detected in protein levels of representative products from several of these genes. Lysates from isolated neutrophils also demonstrated a spike in NO following IFN-γ administration. IFN-γ appears to increase non-oxygen dependent microbicidal functions of PMNs which could provide strategies to compensate for deficiencies, explain its clinical benefit for CGD patients and expand therapeutic applications of IFN-γ to other disorders. Trial registration: Protocol registered in ClinicalTrials.gov, NCT02609932, Effect of IFN-γ on Innate Immune Cells.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Granulomatous Disease, Chronic/drug therapy , Granulomatous Disease, Chronic/metabolism , Interferon-gamma/pharmacology , Neutrophils/drug effects , Adolescent , Adult , Chemokine CXCL10/biosynthesis , Granulomatous Disease, Chronic/genetics , Healthy Volunteers , Humans , Interferon-gamma/biosynthesis , Middle Aged , NADPH Oxidases/metabolism , Neopterin/biosynthesis , Neutrophils/metabolism , Phagocytosis , Phenotype , Respiratory Burst , Superoxides , Young Adult
8.
Mol Microbiol ; 76(4): 1010-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20398220

ABSTRACT

The translocation of the diphtheria toxin catalytic domain from the lumen of early endosomes into the cytosol of eukaryotic cells is an essential step in the intoxication process. We have previously shown that the in vitro translocation of the catalytic domain from the lumen of toxin pre-loaded endosomal vesicles to the external medium requires the addition of cytosolic proteins including coatomer protein complex I (COPI) to the reaction mixture. Further, we have shown that transmembrane helix 1 plays an essential, but as yet undefined role in the entry process. We have used both site-directed mutagenesis and a COPI complex precipitation assay to demonstrate that interaction(s) between at least three lysine residues in transmembrane helix 1 are essential for both COPI complex binding and the delivery of the catalytic domain into the target cell cytosol. Finally, a COPI binding domain swap was used to demonstrate that substitution of the lysine-rich transmembrane helix 1 with the COPI binding portion of the p23 adaptor cytoplasmic tail results in a mutant that displays full wild-type activity. Thus, irrespective of sequence, the ability of transmembrane helix 1 to bind to COPI complex appears to be the essential feature for catalytic domain delivery to the cytosol.


Subject(s)
Coat Protein Complex I/metabolism , Diphtheria Toxin/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Cattle , Cell Line , Diphtheria Toxin/chemistry , Diphtheria Toxin/genetics , Humans , Lysine/genetics , Lysine/metabolism , Molecular Sequence Data , Protein Structure, Secondary , Protein Transport
9.
Proc Natl Acad Sci U S A ; 105(13): 5254-9, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18356299

ABSTRACT

The delivery of the diphtheria toxin catalytic domain (DTA) from acidified endosomes into the cytoplasm of eukaryotic cells requires protein-protein interactions between the toxin and a cytosolic translocation factor (CTF) complex. A conserved peptide motif, T1, within the DT transmembrane helix 1 mediates these interactions. Because the T1 motif is also present in the N-terminal segments of lethal factor (LF) and edema factor (EF) in anthrax toxin, we asked whether LF entry into the cell might also be facilitated by target cell cytosolic proteins. In this study, we have used LFnDTA and its associated ADP-ribosyltransferase activity (DTA) to determine the requirements for LF translocation from the lumen of endosomal vesicles to the external medium in vitro. Although low-level release of LFnDTA from enriched endosomal vesicles occurs in the absence of added factors, translocation was enhanced by the addition of cytosolic proteins and ATP to the reaction mixture. We show by GST-LFn pull-down assays that LFn specifically interacts with at least zeta-COP and beta-COP of the COPI coatomer complex. Immunodepletion of COPI coatomer complex and associated proteins from cytosolic extracts blocks in vitro LFnDTA translocation. Translocation may be reconstituted by the addition of partially purified bovine COPI to the translocation assay mixture. Taken together, these data suggest that the delivery of LF to the cytosol requires either COPI coatomer complex or a COPI subcomplex for translocation from the endosomal lumen. This facilitated delivery appears to use a mechanism that is analogous to that of DT entry.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Coat Protein Complex I/metabolism , Intracellular Membranes/metabolism , Bacillus anthracis/metabolism , Cell Line, Tumor , Coat Protein Complex I/genetics , Cytosol/metabolism , Humans , Microscopy, Electron , Protein Binding , Protein Subunits/metabolism , Protein Transport , Sensitivity and Specificity
10.
Mol Oncol ; 15(5): 1330-1344, 2021 05.
Article in English | MEDLINE | ID: mdl-33682324

ABSTRACT

In many solid tumors including triple-negative breast cancer (TNBC), upregulation of the interleukin-4 receptor (IL-4R) has been shown to promote cancer cell proliferation, apoptotic resistance, metastatic potential, and a Th2 response in the tumor microenvironment (TME). Since immunosuppressive cells in the TME and spleen including myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) also express the IL-4R, we hypothesized that selective depletion of IL-4R-bearing cells in TNBC would result in the direct killing of tumor cells and the depletion of immunosuppressive cells and lead to an enhanced antitumor response. To selectively target IL-4R+ cells, we employed DABIL-4, a fusion protein toxin consisting of the catalytic and translocation domains of diphtheria toxin fused to murine IL-4. As anticipated, DABIL-4 has potent cytotoxic activity against TNBC cells both in vitro and in vivo. We demonstrate in the murine 4T1 TNBC model that DABIL-4 significantly reduces tumor growth, splenomegaly, and lung metastases. Importantly, we also show that the administration of DABIL-4 results in the selective depletion of MDSCs, TAMs, and regulatory T cells in treated mice, with a concomitant increase in IFN-γ+ CD8 effector T cells in the TME. Since the 4T1 antitumor activity of DABIL-4 was largely diminished in IL-4R knockout mice, we postulate that DABIL-4 functions primarily as an immunotherapeutic by the depletion of MDSCs, TAMs, and regulatory T cells. NanoString analysis of control and treated tumors confirmed and extended these observations by showing a marked decline of mRNA transcripts that are associated with tumorigenesis and metastasis. In conclusion, we demonstrate that DABIL-4 targeting of both tumor and immunosuppressive host cells likely represents a novel and effective treatment strategy for 4T1 TNBC and warrants further study.


Subject(s)
Adenocarcinoma/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Recombinant Fusion Proteins/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Count , Cell Line, Tumor , Female , Humans , Interleukin-4/chemistry , Interleukin-4/therapeutic use , Interleukin-4 Receptor alpha Subunit/chemistry , Interleukin-4 Receptor alpha Subunit/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Targeted Therapy , Myeloid-Derived Suppressor Cells/pathology , Recombinant Fusion Proteins/pharmacology , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
11.
J Cell Biol ; 160(7): 1139-50, 2003 Mar 31.
Article in English | MEDLINE | ID: mdl-12668662

ABSTRACT

In vitro delivery of the diphtheria toxin catalytic (C) domain from the lumen of purified early endosomes to the external milieu requires the addition of both ATP and a cytosolic translocation factor (CTF) complex. Using the translocation of C-domain ADP-ribosyltransferase activity across the endosomal membrane as an assay, the CTF complex activity was 650-800-fold purified from human T cell and yeast extracts, respectively. The chaperonin heat shock protein (Hsp) 90 and thioredoxin reductase were identified by mass spectrometry sequencing in CTF complexes purified from both human T cell and yeast. Further analysis of the role played by these two proteins with specific inhibitors, both in the in vitro translocation assay and in intact cell toxicity assays, has demonstrated their essential role in the productive delivery of the C-domain from the lumen of early endosomes to the external milieu. These results confirm and extend earlier observations of diphtheria toxin C-domain unfolding and refolding that must occur before and after vesicle membrane translocation. In addition, results presented here demonstrate that thioredoxin reductase activity plays an essential role in the cytosolic release of the C-domain. Because analogous CTF complexes have been partially purified from mammalian and yeast cell extracts, results presented here suggest a common and fundamental mechanism for C-domain translocation across early endosomal membranes.


Subject(s)
Catalytic Domain , Cytosol/metabolism , Diphtheria Toxin/metabolism , T-Lymphocytes/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Benzoquinones , Cell Line , Endocytosis , Endosomes/metabolism , HSP90 Heat-Shock Proteins/analysis , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic , Lactones/pharmacology , Macrolides , Mass Spectrometry , Peptide Elongation Factor 2/metabolism , Quinones/pharmacology , Thioredoxin-Disulfide Reductase/analysis , Thioredoxin-Disulfide Reductase/drug effects , Yeasts
12.
Microbiol Spectr ; 7(4)2019 07.
Article in English | MEDLINE | ID: mdl-31267892

ABSTRACT

Diphtheria is one of the most well studied of all the bacterial infectious diseases. These milestone studies of toxigenic Corynebacterium diphtheriae along with its primary virulence determinant, diphtheria toxin, have established the paradigm for the study of other related bacterial protein toxins. This review highlights those studies that have contributed to our current understanding of the structure-function relationships of diphtheria toxin, the molecular mechanism of its entry into the eukaryotic cell cytosol, the regulation of diphtheria tox expression by holo-DtxR, and the molecular basis of transition metal ion activation of apo-DtxR itself. These seminal studies have laid the foundation for the protein engineering of diphtheria toxin and the development of highly potent eukaryotic cell-surface receptor-targeted fusion protein toxins for the treatment of human diseases that range from T cell malignancies to steroid-resistant graft-versus-host disease to metastatic melanoma. This deeper scientific understanding of diphtheria toxin and the regulation of its expression have metamorphosed the third-most-potent bacterial toxin known into a life-saving targeted protein therapeutic, thereby at least partially fulfilling Paul Erlich's concept of a magic bullet-"a chemical that binds to and specifically kills microbes or tumor cells."


Subject(s)
Bacterial Proteins/metabolism , Corynebacterium diphtheriae/metabolism , DNA-Binding Proteins/metabolism , Diphtheria Toxin/metabolism , Diphtheria/microbiology , Gene Expression Regulation, Bacterial , Iron/metabolism , Animals , Bacterial Proteins/genetics , Corynebacterium diphtheriae/genetics , DNA-Binding Proteins/genetics , Humans , Operon
13.
Immunotherapy ; 11(13): 1117-1128, 2019 09.
Article in English | MEDLINE | ID: mdl-31361167

ABSTRACT

T regulatory cells (Tregs) are an important T cell population for immune tolerance, prevention of autoimmune diseases and inhibition of antitumor immunity. The tumor-promoting role played by Tregs in cancer has prompted numerous approaches to develop immunotherapeutics targeting Tregs. One approach to depletion of Treg cells is retargeting the highly potent cytotoxic activity of bacterial toxins. These agents capitalize on the well-characterized bacterial toxins, diphtheria toxin and Pseudomonas aeruginosa exotoxin A-both of which harbor membrane translocation domains and enzymatic domains that catalytically halt protein synthesis within intoxicated eukaryotic cells and act at picomolar or subpicomolar concentrations. In this review, we summarize the preclinical and clinical development of several Treg-depleting cancer immunotherapies based on these two bacterial toxins.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , Diphtheria Toxin/therapeutic use , Exotoxins/therapeutic use , Immunotherapy/methods , Lymphocyte Depletion/methods , Neoplasms/therapy , T-Lymphocytes, Regulatory/physiology , Virulence Factors/therapeutic use , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Immunity, Cellular/drug effects , Neoplasms/immunology , Tumor Microenvironment/drug effects , Pseudomonas aeruginosa Exotoxin A
14.
Infect Immun ; 76(2): 486-96, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18025095

ABSTRACT

Francisella tularensis can cause severe disseminated disease after respiratory infection. The identification of factors involved in mortality or recovery following induction of tularemia in the mouse will improve our understanding of the natural history of this disease and facilitate future evaluation of vaccine candidate preparations. BALB/c mice were infected intranasally with the live vaccine strain (LVS) of F. tularensis subsp. holarctica and euthanized at different stages of disease to analyze the induction of immune molecules, gross anatomical features of organs, bacterial burdens, and progression of the histopathological changes in lung and spleen. Tissue-specific interleukin-6 (IL-6), macrophage inflammatory protein 2, and monocyte chemotactic protein 1 were immune markers of mortality, while anti-LVS immunoglobulin M and IL-1beta were associated with survival. Moribund mice had enlarged spleens and lungs, while surviving mice had even more prominent splenomegaly and normal-appearing lungs. Histopathology of the spleens of severely ill mice was characterized by disrupted lymphoid follicles and fragmented nuclei, while the spleens of survivors appeared healthy but with increased numbers of megakaryocytes and erythrocytes. Histopathology of the lungs of severely ill mice indicated severe pneumonia. Lungs of survivors at early time points showed increased inflammation, while at late times they appeared healthy with peribronchial lymphoid aggregates. Our results suggest that host immune factors are able to affect bacterial dissemination after respiratory tularemia, provide new insights regarding the pathological characteristics of pulmonary tularemia leading to systemic disease, and potentially identify immune markers associated with recovery from the disease.


Subject(s)
Francisella tularensis/immunology , Pneumonia/immunology , Pneumonia/pathology , Tularemia/immunology , Tularemia/pathology , Animals , Antibodies, Bacterial/analysis , Body Weight , Chemokine CCL2/analysis , Chemokine CXCL2/analysis , Colony Count, Microbial , Female , Immunoglobulin M/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , Lung/chemistry , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Organ Size , Pneumonia/microbiology , Spleen/chemistry , Spleen/microbiology , Spleen/pathology
15.
Biochem Biophys Res Commun ; 376(1): 200-5, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18774771

ABSTRACT

Peptide nucleic acid (PNA) is highly stable and binds to complementary RNA and DNA with high affinity, but it resists cellular uptake, thereby limiting its bioavailability. We investigated whether protectiveantigen (PA, a non-toxic component of anthrax toxin) could transport antisense PNA oligomers into reporter cells that contain luciferase transgenes with mutant beta-globin IVS2 intronic inserts, which permit aberrant pre-mRNA splicing and impair luciferase expression. PNA oligomers antisense to mutant splice sites in these IVS2 inserts induced luciferase expression when effectively delivered into the cells. PNA 18-mers with C-terminal poly-lysine tails [PNA(Lys)(8)] demonstrated modest sequence-specific antisense activity by themselves at micromolar concentrations in luc-IVS2 reporter cell cultures. However, this activity was greatly amplified by PA. Antisense PNA(Lys)(8) with but not without PA also corrected the IVS2-654 beta-globin splice defect in cultured erythroid precursor cells from a patient with beta-thalassemia [genotype, IVS2-654(beta(0)/beta(E))], providing further evidence that anthrax PA can effectively transport antisense PNA oligomers into cells.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Oligodeoxyribonucleotides, Antisense/metabolism , Peptide Nucleic Acids/metabolism , Transfection/methods , Biological Transport , Globins/genetics , Humans , Introns , Luciferases/genetics , Oligodeoxyribonucleotides, Antisense/genetics , Peptide Nucleic Acids/genetics , RNA Splice Sites , RNA Splicing , beta-Thalassemia/genetics
16.
J Med Chem ; 60(2): 767-786, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27983835

ABSTRACT

By use of a structure-based computational method for identification of structurally novel Janus kinase (JAK) inhibitors predicted to bind beyond the ATP binding site, a potent series of indazoles was identified as selective pan-JAK inhibitors with a type 1.5 binding mode. Optimization of the series for potency and increased duration of action commensurate with inhaled or topical delivery resulted in potent pan-JAK inhibitor 2 (PF-06263276), which was advanced into clinical studies.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Heterocyclic Compounds, 2-Ring/pharmacology , Indazoles/pharmacology , Janus Kinases/antagonists & inhibitors , Lung Diseases/drug therapy , Protein Kinase Inhibitors/pharmacology , Skin Diseases/drug therapy , Administration, Cutaneous , Administration, Inhalation , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/toxicity , Binding Sites , Crystallography, X-Ray , Dogs , Drug Design , Hepatocytes/metabolism , Heterocyclic Compounds, 2-Ring/administration & dosage , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/toxicity , Humans , Indazoles/administration & dosage , Indazoles/chemical synthesis , Indazoles/toxicity , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/toxicity , Rats , Solubility
17.
J Microbiol Methods ; 51(1): 63-72, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12069891

ABSTRACT

While the native diphtheria tox promoter/operator (toxPO)-lacZ transcriptional fusion has allowed initial isolation and characterization of the diphtheria toxin repressor (DtxR), the low level of reporter gene expression has limited the detection and analysis of mutations affecting subtle changes in repressor-operator binding. In order to overcome this difficulty, we have constructed a novel hybrid promoter/operator-lacZ transcriptional fusion in which the "-35" and spacing of the tac promoter was fused to the "-10" and interrupted palindromic sequence of toxO. We show that the hybrid tacPtoxO is regulated by the transition metal ion-dependent DtxR and that lacZ expression is increased approximately 70-fold in the reporter strain Escherichia coli DH5alpha/lambdaRS45-tacPtoxO-lacZ relative to DH5alpha/lambdaRS45-toxPO-lacZ. In addition, we have constructed a transcriptional fusion between tacPtoxO and luc, pJL1. We have used pJL1 to program S30 extracts of E. coli in order to direct in vitro the coupled transcription and translation of luciferase. We demonstrate the utility of this in vitro system in providing a direct functional link between in vivo and in vitro observations with DtxR and mutants of DtxR, which display subtle changes in activity in a manner not previously possible.


Subject(s)
Bacterial Proteins/genetics , DNA Probes/genetics , DNA-Binding Proteins/genetics , Diphtheria Toxin/genetics , Operator Regions, Genetic/genetics , Promoter Regions, Genetic/genetics , Bacterial Proteins/metabolism , Base Sequence , DNA Probes/metabolism , DNA-Binding Proteins/metabolism , Diphtheria Toxin/metabolism , Electrophoretic Mobility Shift Assay , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription, Genetic , beta-Galactosidase/analysis
18.
Virology ; 441(2): 107-13, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23601786

ABSTRACT

Epstein-Barr virus (EBV) is associated with multiple malignancies including nasopharyngeal carcinoma (NPC). In nasopharynx cancer, CD8+ T cells specific for EBV Nuclear Antigen-1 (EBNA-1) and Latent Membrane Protein 2 (LMP2) are important components of anti-tumor immunity since both are consistently expressed in NPC. We have previously shown that EBNA-1-specific CD8+ T cell responses were suppressed in NPC patients compared to healthy controls. We now find that CD8+ T cell responses specific for LMP2 are also abnormal in NPC patients, and both EBNA-1- and LMP2-specific responses are suppressed by regulatory T cells (Treg). EBNA-1 and LMP2-specific CD8+ T cell responses, as well as immune control of EBV-infected cells in vitro, could be restored by the depletion of Tregs and by use of a clinically approved drug targeting Tregs. Thus, in vivo modulation of Tregs may be an effective means of enhancing these anti-tumor immune responses in NPC patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Nasopharyngeal Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Carcinoma , Epstein-Barr Virus Nuclear Antigens/immunology , Humans , Immune Tolerance , Nasopharyngeal Carcinoma , Viral Matrix Proteins/immunology
19.
Toxins (Basel) ; 3(3): 294-308, 2011 03.
Article in English | MEDLINE | ID: mdl-22069710

ABSTRACT

Research on diphtheria and anthrax toxins over the past three decades has culminated in a detailed understanding of their structure function relationships (e.g., catalytic (C), transmembrane (T), and receptor binding (R) domains), as well as the identification of their eukaryotic cell surface receptor, an understanding of the molecular events leading to the receptor-mediated internalization of the toxin into an endosomal compartment, and the pH triggered conformational changes required for pore formation in the vesicle membrane. Recently, a major research effort has been focused on the development of a detailed understanding of the molecular interactions between each of these toxins and eukaryotic cell factors that play an essential role in the efficient translocation of their respective catalytic domains through the trans-endosomal vesicle membrane pore and delivery into the cell cytosol. In this review, I shall focus on recent findings that have led to a more detailed understanding of the mechanism by which the diphtheria toxin catalytic domain is delivered to the eukaryotic cell cytosol. While much work remains, it is becoming increasingly clear that the entry process is facilitated by specific interactions with a number of cellular factors in an ordered sequential fashion. In addition, since diphtheria, anthrax lethal factor and anthrax edema factor all carry multiple coatomer I complex binding motifs and COPI complex has been shown to play an essential role in entry process, it is likely that the initial steps in catalytic domain entry of these divergent toxins follow a common mechanism.


Subject(s)
Coatomer Protein/metabolism , Cytosol/drug effects , Diphtheria Toxin/pharmacokinetics , Eukaryotic Cells/drug effects , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Cell Line , Cytosol/metabolism , Diphtheria Toxin/chemistry , Diphtheria Toxin/genetics , Eukaryotic Cells/metabolism , Humans , Molecular Sequence Data , Protein Binding , Protein Transport
20.
Clin Vaccine Immunol ; 15(9): 1322-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18614668

ABSTRACT

Francisella tularensis causes severe pneumonia that can be fatal if it is left untreated. Due to its potential use as a biological weapon, research is being conducted to develop an effective vaccine and to select and study adjuvant molecules able to generate a better and long-lasting protective effect. PorB, a porin from Neisseria meningitidis, is a well-established Toll-like receptor 2 ligand and has been shown to be a promising vaccine adjuvant candidate due to its ability to enhance the T-cell costimulatory activity of antigen-presenting cells both in vitro and in vivo. BALB/c mice were immunized with lipopolysaccharide (LPS) isolated from the F. tularensis subsp. holarctica live vaccine strain (LVS), with or without PorB from N. meningitidis, and the antibody levels induced during the vaccination regimen and the level of protection against intranasal challenge with LVS were determined. Antigen administered alone induced a specific F. tularensis LPS immunoglobulin M (IgM) response that was not maintained over the weeks and that conferred protection to only 25% of the mice. In contrast, F. tularensis LPS given in combination with neisserial PorB induced consistent levels of specific IgM throughout the immunization and increased the proportion of surviving mice to 70%. Postchallenge cytokine analysis showed that interleukin-6 (IL-6), monocyte chemoattractant protein 1, and gamma interferon were markers of mortality and that IL-1beta was a correlate of survival, independent of the presence of PorB as an adjuvant. These data indicate that neisserial PorB might be an optimal candidate adjuvant for improving the protective effect of F. tularensis LPS and other subunit vaccines against tularemia, but there is still a need to test its efficacy against virulent type A and type B F. tularensis strains.


Subject(s)
Adjuvants, Immunologic/pharmacology , Francisella tularensis/immunology , Lipopolysaccharides/immunology , Porins/pharmacology , Tularemia/prevention & control , Animals , Antibodies, Bacterial/blood , Cytokines/blood , Female , Humans , Immunoglobulin M/blood , Mice , Mice, Inbred BALB C , Survival Analysis , Tularemia/immunology
SELECTION OF CITATIONS
SEARCH DETAIL