Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
N Engl J Med ; 374(17): 1635-46, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-25629663

ABSTRACT

BACKGROUND: The West African outbreak of Ebola virus disease that peaked in 2014 has caused more than 11,000 deaths. The development of an effective Ebola vaccine is a priority for control of a future outbreak. METHODS: In this phase 1 study, we administered a single dose of the chimpanzee adenovirus 3 (ChAd3) vaccine encoding the surface glycoprotein of Zaire ebolavirus (ZEBOV) to 60 healthy adult volunteers in Oxford, United Kingdom. The vaccine was administered in three dose levels--1×10(10) viral particles, 2.5×10(10) viral particles, and 5×10(10) viral particles--with 20 participants in each group. We then assessed the effect of adding a booster dose of a modified vaccinia Ankara (MVA) strain, encoding the same Ebola virus glycoprotein, in 30 of the 60 participants and evaluated a reduced prime-boost interval in another 16 participants. We also compared antibody responses to inactivated whole Ebola virus virions and neutralizing antibody activity with those observed in phase 1 studies of a recombinant vesicular stomatitis virus-based vaccine expressing a ZEBOV glycoprotein (rVSV-ZEBOV) to determine relative potency and assess durability. RESULTS: No safety concerns were identified at any of the dose levels studied. Four weeks after immunization with the ChAd3 vaccine, ZEBOV-specific antibody responses were similar to those induced by rVSV-ZEBOV vaccination, with a geometric mean titer of 752 and 921, respectively. ZEBOV neutralization activity was also similar with the two vaccines (geometric mean titer, 14.9 and 22.2, respectively). Boosting with the MVA vector increased virus-specific antibodies by a factor of 12 (geometric mean titer, 9007) and increased glycoprotein-specific CD8+ T cells by a factor of 5. Significant increases in neutralizing antibodies were seen after boosting in all 30 participants (geometric mean titer, 139; P<0.001). Virus-specific antibody responses in participants primed with ChAd3 remained positive 6 months after vaccination (geometric mean titer, 758) but were significantly higher in those who had received the MVA booster (geometric mean titer, 1750; P<0.001). CONCLUSIONS: The ChAd3 vaccine boosted with MVA elicited B-cell and T-cell immune responses to ZEBOV that were superior to those induced by the ChAd3 vaccine alone. (Funded by the Wellcome Trust and others; ClinicalTrials.gov number, NCT02240875.).


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Adenoviruses, Simian/immunology , Adult , Animals , Antibodies, Viral/blood , B-Lymphocytes/physiology , Cytokines/blood , Ebola Vaccines/administration & dosage , Female , Hemorrhagic Fever, Ebola/immunology , Humans , Immunity, Cellular , Immunization, Secondary , Male , Middle Aged , Pan troglodytes , T-Lymphocytes/physiology , Vaccinia , Young Adult
2.
Hepatology ; 63(5): 1455-70, 2016 May.
Article in English | MEDLINE | ID: mdl-26474390

ABSTRACT

UNLABELLED: Adenoviral vectors encoding hepatitis C virus (HCV) nonstructural (NS) proteins induce multispecific, high-magnitude, durable CD4(+) and CD8(+) T-cell responses in healthy volunteers. We assessed the capacity of these vaccines to induce functional HCV-specific immune responses and determine T-cell cross-reactivity to endogenous virus in patients with chronic HCV infection. HCV genotype 1-infected patients were vaccinated using heterologous adenoviral vectors (ChAd3-NSmut and Ad6-NSmut) encoding HCV NS proteins in a dose escalation, prime-boost regimen, with and without concomitant pegylated interferon-α/ribavirin therapy. Analysis of immune responses ex vivo used human leukocyte antigen class I pentamers, intracellular cytokine staining, and fine mapping in interferon-γ enzyme-linked immunospot assays. Cross-reactivity of T cells with population and endogenous viral variants was determined following viral sequence analysis. Compared to healthy volunteers, the magnitude of HCV-specific T-cell responses following vaccination was markedly reduced. CD8(+) HCV-specific T-cell responses were detected in 15/24 patients at the highest dose, whereas CD4(+) T-cell responses were rarely detectable. Analysis of the host circulating viral sequence showed that T-cell responses were rarely elicited when there was sequence homology between vaccine immunogen and endogenous virus. In contrast, T cells were induced in the context of genetic mismatch between vaccine immunogen and endogenous virus; however, these commonly failed to recognize circulating epitope variants and had a distinct partially functional phenotype. Vaccination was well tolerated but had no significant effect on HCV viral load. CONCLUSION: Vaccination with potent HCV adenoviral vectored vaccines fails to restore T-cell immunity except where there is genetic mismatch between vaccine immunogen and endogenous virus; this highlights the major challenge of overcoming T-cell exhaustion in the context of persistent antigen exposure with implications for cancer and other persistent infections.


Subject(s)
Hepacivirus/immunology , Hepatitis C, Chronic/immunology , T-Lymphocytes/immunology , Viral Hepatitis Vaccines/immunology , Adenoviridae/genetics , Adult , Aged , Amino Acid Sequence , Epitopes, T-Lymphocyte , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Humans , Interferon-alpha/administration & dosage , Male , Middle Aged , Molecular Sequence Data , Polyethylene Glycols/administration & dosage , Recombinant Proteins/administration & dosage , Riboflavin/administration & dosage , Vaccination
3.
Vaccines (Basel) ; 10(6)2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35746566

ABSTRACT

In this study, we invented and construct novel candidate HIV-1 vaccines. Through genetic and protein engineering, we unknowingly constructed an HIV-1-derived transgene with a homopolymeric run of 11 cytidines, which was inserted into an adenovirus vaccine vector. Here, we describe the virus rescue, three rounds of clonal purification and preparation of good manufacturing practise (GMP) starting material assessed for genetic stability in five additional virus passages. Throughout these steps, quality control assays indicated the presence of the transgene in the virus genome, expression of the correct transgene product and immunogenicity in mice. However, DNA sequencing of the transgene revealed additional cytidines inserted into the original 11-cytidine region, and the GMP manufacture had to be aborted. Subsequent analyses indicated that as little as 1/25th of the virus dose used for confirmation of protein expression (106 cells at a multiplicity of infection of 10) and murine immunogenicity (108 infectious units per animal) met the quality acceptance criteria. Similar frameshifts in the expressed proteins were reproduced in a one-reaction in vitro transcription/translation employing phage T7 polymerase and E. coli ribosomes. Thus, the most likely mechanism for addition of extra cytidines into the ChAdOx1.tHIVconsv6 genome is that the adenovirus DNA polymerase lost its fidelity on a stretch of 11 cytidines, which informs future adenovirus vaccine designs.

4.
J Lipid Res ; 52(1): 78-86, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20959675

ABSTRACT

Proprotein convertase subtilisin-like/kexin type 9 (PCSK9) regulates LDL cholesterol levels by inhibiting LDL receptor (LDLr)-mediated cellular LDL uptake. We have identified a fragment antigen-binding (Fab) 1D05 which binds PCSK9 with nanomolar affinity. The fully human antibody 1D05-IgG2 completely blocks the inhibitory effects of wild-type PCSK9 and two gain-of-function human PCSK9 mutants, S127R and D374Y. The crystal structure of 1D05-Fab bound to PCSK9 reveals that 1D05-Fab binds to an epitope on the PCSK9 catalytic domain which includes the entire LDLr EGF(A) binding site. Notably, the 1D05-Fab CDR-H3 and CDR-H2 loops structurally mimic the EGF(A) domain of LDLr. In a transgenic mouse model (CETP/LDLr-hemi), in which plasma lipid and PCSK9 profiles are comparable to those of humans, 1D05-IgG2 reduces plasma LDL cholesterol to 40% and raises hepatic LDLr protein levels approximately fivefold. Similarly, in healthy rhesus monkeys, 1D05-IgG2 effectively reduced LDL cholesterol 20%-50% for over 2 weeks, despite its relatively short terminal half-life (t(1/2) = 3.2 days). Importantly, the decrease in circulating LDL cholesterol corresponds closely to the reduction in free PCSK9 levels. Together these results clearly demonstrate that the LDL-lowering effect of the neutralizing anti-PCSK9 1D05-IgG2 antibody is mediated by reducing the amount of PCSK9 that can bind to the LDLr.


Subject(s)
Cholesterol, LDL/blood , Immunoglobulin Fab Fragments/pharmacology , Receptors, LDL/chemistry , Serine Endopeptidases/immunology , Animals , Antibodies, Monoclonal/metabolism , Binding Sites , Cholesterol Ester Transfer Proteins/metabolism , Fluoroimmunoassay , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Macaca mulatta , Male , Mice , Mice, Transgenic , Proprotein Convertase 9 , Proprotein Convertases , Receptors, LDL/metabolism , Serine Endopeptidases/chemistry
5.
J Biol Chem ; 285(17): 12882-91, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20172854

ABSTRACT

PCSK9 binds to the low density lipoprotein receptor (LDLR) and leads to LDLR degradation and inhibition of plasma LDL cholesterol clearance. Consequently, the role of PCSK9 in modulating circulating LDL makes it a promising therapeutic target for treating hypercholesterolemia and coronary heart disease. Although the C-terminal domain of PCSK9 is not involved in LDLR binding, the location of several naturally occurring mutations within this region suggests that it has an important role for PCSK9 function. Using a phage display library, we identified an anti-PCSK9 Fab (fragment antigen binding), 1G08, with subnanomolar affinity for PCSK9. In an assay measuring LDL uptake in HEK293 and HepG2 cells, 1G08 Fab reduced 50% the PCSK9-dependent inhibitory effects on LDL uptake. Importantly, we found that 1G08 did not affect the PCSK9-LDLR interaction but inhibited the internalization of PCSK9 in these cells. Furthermore, proteolysis and site-directed mutagenesis studies demonstrated that 1G08 Fab binds a region of beta-strands encompassing Arg-549, Arg-580, Arg-582, Glu-607, Lys-609, and Glu-612 in the PCSK9 C-terminal domain. Consistent with these results, 1G08 fails to bind PCSK9DeltaC, a truncated form of PCSK9 lacking the C-terminal domain. Additional studies revealed that lack of the C-terminal domain compromised the ability of PCSK9 to internalize into cells, and to inhibit LDL uptake. Together, the present study demonstrate that the PCSK9 C-terminal domain contribute to its inhibition of LDLR function mainly through its role in the cellular uptake of PCSK9 and LDLR complex. 1G08 Fab represents a useful new tool for delineating the mechanism of PCSK9 uptake and LDLR degradation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunoglobulin Fab Fragments/pharmacology , Lipoproteins, LDL/metabolism , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Amino Acid Substitution , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Hep G2 Cells , Humans , Hypercholesterolemia/drug therapy , Hypercholesterolemia/genetics , Hypercholesterolemia/immunology , Hypercholesterolemia/metabolism , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Lipoproteins, LDL/genetics , Lipoproteins, LDL/immunology , Mutagenesis, Site-Directed , Proprotein Convertase 9 , Proprotein Convertases , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, LDL/genetics , Receptors, LDL/immunology , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology
6.
Biochemistry ; 48(2): 242-53, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19140693

ABSTRACT

RTN1-C protein is a membrane protein localized in the ER and expressed in the nervous system. Its biological role is still unclear, although interactions of the N-terminal region of RTN1-C with proteins involved in vesicle trafficking have been observed, but the role of the C-terminal region of this family protein remains to be investigated. By a homology analysis of the amino acid sequence, we identified in the C-terminal region of RTN1-C a unique consensus sequence characteristic of H4 histone protein. Thus, a 23-mer peptide (RTN1-C(CT)) corresponding to residues 186-208 of RTN1-C was synthesized, and its conformation and its interaction with nucleic acids were investigated. Here we demonstrate the strong ability of RTN1-C(CT) peptide to bind and condense the nucleic acids using electrophoretic and spectroscopic techniques. To determine if the binding of RTN1-C to nucleic acids could be regulated in vivo by an acetylation-deacetylation mechanism, as for the histone proteins, we studied the interaction of RTN1-C with one zinc-dependent histone deacetylase (HDAC) enzyme, HDAC8, with fluorescence and kinetic techniques using an acetylated form of RTN1-C(CT). The results reported here allow us to propose that the nucleic acid binding property of RTN1-C may have an important role in the biological function of this protein, the function of which could be regulated by an acetylation-deacetylation mechanism.


Subject(s)
DNA/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Proteins/metabolism , RNA/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites/genetics , Biophysical Phenomena , Consensus Sequence , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , DNA, Bacterial/metabolism , Escherichia coli/chemistry , Humans , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Protein Binding/genetics , Protein Conformation , Protein Processing, Post-Translational , Protein Structure, Secondary , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , RNA, Bacterial/metabolism
7.
PLoS Negl Trop Dis ; 11(5): e0005527, 2017 May.
Article in English | MEDLINE | ID: mdl-28498840

ABSTRACT

BACKGROUND: Visceral leishmaniasis (VL or kala azar) is the most serious form of human leishmaniasis, responsible for over 20,000 deaths annually, and post kala azar dermal leishmaniasis (PKDL) is a stigmatizing skin condition that often occurs in patients after successful treatment for VL. Lack of effective or appropriately targeted cell mediated immunity, including CD8+ T cell responses, underlies the progression of VL and progression to PKDL, and can limit the therapeutic efficacy of anti-leishmanial drugs. Hence, in addition to the need for prophylactic vaccines against leishmaniasis, the development of therapeutic vaccines for use alone or in combined immuno-chemotherapy has been identified as an unmet clinical need. Here, we report the first clinical trial of a third-generation leishmaniasis vaccine, developed intentionally to induce Leishmania-specific CD8+ T cells. METHODS: We conducted a first-in-human dose escalation Phase I trial in 20 healthy volunteers to assess the safety, tolerability and immunogenicity of a prime-only adenoviral vaccine for human VL and PKDL. ChAd63-KH is a replication defective simian adenovirus expressing a novel synthetic gene (KH) encoding two Leishmania proteins KMP-11 and HASPB. Uniquely, the latter was engineered to reflect repeat domain polymorphisms and arrangements identified from clinical isolates. We monitored innate immune responses by whole blood RNA-Seq and antigen specific CD8+ T cell responses by IFNγ ELISPOT and intracellular flow cytometry. FINDINGS: ChAd63-KH was safe at intramuscular doses of 1x1010 and 7.5x1010 vp. Whole blood transcriptomic profiling indicated that ChAd63-KH induced innate immune responses characterized by an interferon signature and the presence of activated dendritic cells. Broad and quantitatively robust CD8+ T cell responses were induced by vaccination in 100% (20/20) of vaccinated subjects. CONCLUSION: The results of this study support the further development of ChAd63-KH as a novel third generation vaccine for VL and PKDL. TRIAL REGISTRATION: This clinical trial (LEISH1) was registered at EudraCT (2012-005596-14) and ISRCTN (07766359).


Subject(s)
Leishmaniasis Vaccines/immunology , Leishmaniasis Vaccines/isolation & purification , Leishmaniasis, Cutaneous/prevention & control , Leishmaniasis, Cutaneous/therapy , Leishmaniasis, Visceral/prevention & control , Leishmaniasis, Visceral/therapy , Adenoviruses, Simian/genetics , Adolescent , Adult , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Drug Carriers , Enzyme-Linked Immunospot Assay , Female , Flow Cytometry , Healthy Volunteers , Humans , Injections, Intramuscular , Interferon-gamma/metabolism , Leishmania/genetics , Leishmania/immunology , Leishmaniasis Vaccines/administration & dosage , Leishmaniasis Vaccines/adverse effects , Male , Middle Aged , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification , Young Adult
8.
JCI Insight ; 2(21)2017 11 02.
Article in English | MEDLINE | ID: mdl-29093263

ABSTRACT

The development of a highly effective vaccine remains a key strategic goal to aid the control and eventual eradication of Plasmodium falciparum malaria. In recent years, the reticulocyte-binding protein homolog 5 (RH5) has emerged as the most promising blood-stage P. falciparum candidate antigen to date, capable of conferring protection against stringent challenge in Aotus monkeys. We report on the first clinical trial to our knowledge to assess the RH5 antigen - a dose-escalation phase Ia study in 24 healthy, malaria-naive adult volunteers. We utilized established viral vectors, the replication-deficient chimpanzee adenovirus serotype 63 (ChAd63), and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA), encoding RH5 from the 3D7 clone of P. falciparum. Vaccines were administered i.m. in a heterologous prime-boost regimen using an 8-week interval and were well tolerated. Vaccine-induced anti-RH5 serum antibodies exhibited cross-strain functional growth inhibition activity (GIA) in vitro, targeted linear and conformational epitopes within RH5, and inhibited key interactions within the RH5 invasion complex. This is the first time to our knowledge that substantial RH5-specific responses have been induced by immunization in humans, with levels greatly exceeding the serum antibody responses observed in African adults following years of natural malaria exposure. These data support the progression of RH5-based vaccines to human efficacy testing.


Subject(s)
Antibodies, Neutralizing , Carrier Proteins/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Vaccination , Adaptive Immunity , Adult , Antibodies, Protozoan/blood , Carrier Proteins/genetics , Epitopes/immunology , Female , Genetic Vectors , Humans , Immunization , Male , Middle Aged , Plasmodium falciparum/genetics , Vaccinia virus , Young Adult
9.
J Med Chem ; 48(14): 4547-57, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-15999993

ABSTRACT

Infections caused by hepatitis C virus (HCV) are a significant world health problem for which novel therapies are in urgent demand. Compounds that block replication of subgenomic HCV RNA in liver cells are of interest because of their demonstrated antiviral effect in the clinic. In followup to our recent report that indole-N-acetamides (e.g., 1) are potent allosteric inhibitors of the HCV NS5B polymerase enzyme, we describe here their optimization as cell-based inhibitors. The crystal structure of 1 bound to NS5B was a guide in the design of a two-dimensional compound array that highlighted that formally zwitterionic inhibitors have strong intracellular potency and that pregnane X receptor (PXR) activation (an undesired off-target activity) is linked to a structural feature of the inhibitor. Optimized analogues devoid of PXR activation (e.g., 55, EC(50) = 127 nM) retain strong cell-based efficacy under high serum conditions and show acceptable pharmacokinetics parameters in rat and dog.


Subject(s)
Acetamides/chemical synthesis , Antiviral Agents/chemical synthesis , Hepacivirus/enzymology , Indoles/chemical synthesis , RNA, Viral/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Acetamides/chemistry , Acetamides/pharmacology , Allosteric Regulation , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Biological Availability , Cell Line, Tumor , Dogs , Genome, Viral , Half-Life , Hepacivirus/genetics , Humans , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Pregnane X Receptor , RNA-Dependent RNA Polymerase/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Steroid/agonists , Structure-Activity Relationship , Tissue Distribution , Viral Nonstructural Proteins/chemistry
10.
Sci Transl Med ; 7(300): 300ra126, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26268313

ABSTRACT

Respiratory syncytial virus (RSV) causes respiratory infection in annual epidemics, with infants and the elderly at particular risk of developing severe disease and death. However, despite its importance, no vaccine exists. The chimpanzee adenovirus, PanAd3-RSV, and modified vaccinia virus Ankara, MVA-RSV, are replication-defective viral vectors encoding the RSV fusion (F), nucleocapsid (N), and matrix (M2-1) proteins for the induction of humoral and cellular responses. We performed an open-label, dose escalation, phase 1 clinical trial in 42 healthy adults in which four different combinations of prime/boost vaccinations were investigated for safety and immunogenicity, including both intramuscular (IM) and intranasal (IN) administration of the adenovirus-vectored vaccine. The vaccines were safe and well tolerated, with the most common reported adverse events being mild injection site reactions. No vaccine-related serious adverse events occurred. RSV neutralizing antibody titers rose in response to IM prime with PanAd3-RSV and after IM boost for individuals primed by the IN route. Circulating anti-F immunoglobulin G (IgG) and IgA antibody-secreting cells (ASCs) were observed after the IM prime and IM boost. RSV-specific T cell responses were increased after the IM PanAd3-RSV prime and were most efficiently boosted by IM MVA-RSV. Interferon-γ (IFN-γ) secretion after boost was from both CD4(+) and CD8(+) T cells, without detectable T helper cell 2 (TH2) cytokines that have been previously associated with immune pathogenesis following exposure to RSV after the formalin-inactivated RSV vaccine. In conclusion, PanAd3-RSV and MVA-RSV are safe and immunogenic in healthy adults. These vaccine candidates warrant further clinical evaluation of efficacy to assess their potential to reduce the burden of RSV disease.


Subject(s)
Adenoviruses, Simian/genetics , Genetic Vectors/genetics , Pan troglodytes/virology , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Vaccinia virus/genetics , Adult , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Body Temperature , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Immunologic , Genetic Vectors/adverse effects , HEK293 Cells , Healthy Volunteers , Humans , Immunization, Secondary , Interferon-gamma/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/genetics , Vaccination
11.
J Med Chem ; 47(26): 6443-6, 2004 Dec 16.
Article in English | MEDLINE | ID: mdl-15588076

ABSTRACT

The design of a series of peptidomimetic inhibitors of the hepatitis C virus NS3 protease is described. These inhibitors feature an indoline-2-carboxamide as a novel heterocyclic replacement for the P3 amino acid residue and N-terminal capping group of tripeptide based inhibitors. The crystal structure of the ternary NS3/NS4A/inhibitor complex for the most active molecule in this series highlights its suitability as an N-terminal capping group of a dipeptide inhibitor of the NS3 protease.


Subject(s)
Antiviral Agents/chemical synthesis , Hepacivirus/enzymology , Indoles/chemical synthesis , Oligopeptides/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Antiviral Agents/chemistry , Crystallography, X-Ray , Indoles/chemistry , Models, Molecular , Molecular Mimicry , Molecular Structure , Protein Binding , Stereoisomerism
12.
Article in English | MEDLINE | ID: mdl-22921442

ABSTRACT

OBJECTIVE: The aim of this work was to evaluate both the level of endogenous cyclic nucleotides cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cAMP) and phosphodiesterase activity in mesenchymal stem cells (MSCs) before and during the osteogenic induction. STUDY DESIGN: Samples were organized into control (nondifferentiated) and test groups which were analyzed at 3 different time points: 1, 2, and 4 weeks. Periodontal ligament MSCs were isolated and then expanded in an MSCM medium while cyclic nucleotide levels and phosphodiesterase activity were assessed. RESULTS: cAMP and cGMP levels were markedly higher in the first week than in the following stages. Similarly, PDE activity increased during the first week and reached the peak in the second week. CONCLUSIONS: This work validates that cAMP, cGMP, and PDE activities are important factors in the first phase of the osteogenic induction of a human stem cell.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Periodontal Ligament/metabolism , Phosphoric Diester Hydrolases/metabolism , Adult , Alkaline Phosphatase/metabolism , Cell Culture Techniques , Cell Differentiation/physiology , Humans , Microscopy, Electron , Periodontal Ligament/cytology
13.
J Med Chem ; 54(1): 289-301, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21141896

ABSTRACT

Infections caused by hepatitis C virus (HCV) are a significant world health problem for which novel therapies are in urgent demand. The polymerase of HCV is responsible for the replication of viral genome and has been a prime target for drug discovery efforts. Here, we report on the further development of tetracyclic indole inhibitors, binding to an allosteric site on the thumb domain. Structure-activity relationship (SAR) studies around an indolo-benzoxazocine scaffold led to the identification of compound 33 (MK-3281), an inhibitor with good potency in the HCV subgenomic replication assay and attractive molecular properties suitable for a clinical candidate. The compound caused a consistent decrease in viremia in vivo using the chimeric mouse model of HCV infection.


Subject(s)
Antiviral Agents/chemical synthesis , Hepacivirus/drug effects , Indoles/chemical synthesis , Oxazocines/chemical synthesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Administration, Oral , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Biological Availability , Cell Line, Tumor , Crystallography, X-Ray , Dogs , Hepacivirus/enzymology , Hepacivirus/physiology , Humans , Indoles/pharmacokinetics , Indoles/pharmacology , Macaca mulatta , Mice , Mice, SCID , Mice, Transgenic , Models, Molecular , Molecular Structure , Oxazocines/pharmacokinetics , Oxazocines/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Viremia/drug therapy , Viremia/virology , Virus Replication/drug effects
14.
ChemMedChem ; 4(10): 1695-713, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19672916

ABSTRACT

Infections caused by the hepatitis C virus (HCV) are a significant world health problem for which novel therapies are in urgent demand. The NS5B polymerase of HCV is responsible for the replication of viral RNA and has been a prime target in the search for novel treatment options. We had discovered allosteric finger-loop inhibitors based on a thieno[3,2-b]pyrrole scaffold as an alternative to the related indole inhibitors. Optimization of the thienopyrrole series led to several N-acetamides with submicromolar potency in the cell-based replicon assay, but they lacked oral bioavailability in rats. By linking the N4-position to the ortho-position of the C5-aryl group, we were able to identify the tetracyclic thienopyrrole 40, which displayed a favorable pharmacokinetic profile in rats and dogs and is equipotent with recently disclosed finger-loop inhibitors based on an indole scaffold.


Subject(s)
Antiviral Agents/pharmacology , Azocines/pharmacology , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Pyrroles/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Azocines/chemistry , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Hepacivirus/enzymology , Humans , Protein Conformation , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Rats , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
15.
J Med Chem ; 52(16): 5217-27, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19877603

ABSTRACT

The hepatitis C virus (HCV) NS5B RNA-dependent RNA polymerase (RdRp) plays a central role in virus replication. NS5B has no functional equivalent in mammalian cells and, as a consequence, is an attractive target for inhibition. Herein, we present 1H-benzo[de]isoquinoline-1,3(2H)-diones as a new series of selective inhibitors of HCV NS5B polymerase. The HTS hit 1 shows submicromolar potency in two different HCV replicons (1b and 2b) and displays no activity on other polymerases (HIV-RT, Polio-pol, GBV-b-pol). These inhibitors act during the pre-elongation phase by binding to NS5B non-nucleoside binding site Thumb Site II as demonstrated by crystal structure of compound 1 with the DeltaC55-1b and DeltaC21-2b enzymes and by mutagenesis studies. SAR in this new series reveals inhibitors, such as 20, with low micromolar activity in the HCV replicon and with good activity/toxicity window in cells.


Subject(s)
Antiviral Agents/chemical synthesis , Isoquinolines/chemical synthesis , Viral Nonstructural Proteins/antagonists & inhibitors , Administration, Oral , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Binding Sites , Biological Availability , Cell Line, Tumor , Crystallography, X-Ray , Drug Resistance, Viral , Genotype , Hepacivirus/genetics , Hepacivirus/physiology , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Isoquinolines/chemistry , Isoquinolines/pharmacology , Models, Molecular , Molecular Structure , Mutation , Rats , Replicon/drug effects , Structure-Activity Relationship , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication
16.
Cell Res ; 17(3): 195-211, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17325692

ABSTRACT

Histone deacetylases (HDACs) and histone acetyl transferases (HATs) are two counteracting enzyme families whose enzymatic activity controls the acetylation state of protein lysine residues, notably those contained in the N-terminal extensions of the core histones. Acetylation of histones affects gene expression through its influence on chromatin conformation. In addition, several non-histone proteins are regulated in their stability or biological function by the acetylation state of specific lysine residues. HDACs intervene in a multitude of biological processes and are part of a multiprotein family in which each member has its specialized functions. In addition, HDAC activity is tightly controlled through targeted recruitment, protein-protein interactions and post-translational modifications. Control of cell cycle progression, cell survival and differentiation are among the most important roles of these enzymes. Since these processes are affected by malignant transformation, HDAC inhibitors were developed as antineoplastic drugs and are showing encouraging efficacy in cancer patients.


Subject(s)
Histone Deacetylases/physiology , Molecular Biology/methods , Neoplasms/therapy , Transcription, Genetic , Acetylation , Animals , Histone Deacetylase Inhibitors , Histone Deacetylases/classification , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Models, Biological , Models, Molecular
17.
EMBO Rep ; 8(9): 879-84, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17721440

ABSTRACT

Histone deacetylases (HDACs)-an enzyme family that deacetylates histones and non-histone proteins-are implicated in human diseases such as cancer, and the first-generation of HDAC inhibitors are now in clinical trials. Here, we report the 2.0 A resolution crystal structure of a catalytically inactive HDAC8 active-site mutant, Tyr306Phe, bound to an acetylated peptidic substrate. The structure clarifies the role of active-site residues in the deacetylation reaction and substrate recognition. Notably, the structure shows the unexpected role of a conserved residue at the active-site rim, Asp 101, in positioning the substrate by directly interacting with the peptidic backbone and imposing a constrained cis-conformation. A similar interaction is observed in a new hydroxamate inhibitor-HDAC8 structure that we also solved. The crucial role of Asp 101 in substrate and inhibitor recognition was confirmed by activity and binding assays of wild-type HDAC8 and Asp101Ala, Tyr306Phe and Asp101Ala/Tyr306Phe mutants.


Subject(s)
Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Aspartic Acid/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Protein Structure, Secondary , Structure-Activity Relationship , Substrate Specificity
18.
Archaea ; 1(6): 411-23, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16243781

ABSTRACT

The recombinant amidase from the hyperthermophylic archaeon Sulfolobus solfataricus (SSAM) a signature amidase, was cloned, purified and characterized. The enzyme is active on a large number of aliphatic and aromatic amides over the temperature range 60-95 degrees C and at pH values between 4.0 and 9.5, with an optimum at pH 5.0. The recombinant enzyme is in the form of a dimer of about 110 kD that reversibly associates into an octamer in a pH-dependent reaction. The pH dependence of the state of association was studied using gel permeation chromatography, analytical ultracentrifugation and dynamic light scattering techniques. At pH 7.0 all three techniques show the presence of two species, in about equal amounts, which is compatible with the existence of a dimeric and an octameric form. In decreasing pH, the dimers formed the octameric species and in increasing pH, the octameric species was converted to dimers. Above pH 8.0, only dimers were present, below pH 3.0 only octamers were present. The association of dimers into octamers decreased in non-polar solvents and increased with temperature. A mutant (Y41C) was obtained that did not show this behavior.


Subject(s)
Amidohydrolases/metabolism , Archaeal Proteins/metabolism , Sulfolobus solfataricus/enzymology , Amides/metabolism , Amidohydrolases/chemistry , Amidohydrolases/genetics , Amino Acid Sequence , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Hydrogen-Ion Concentration , Models, Molecular , Molecular Sequence Data , Molecular Weight , Recombinant Proteins/biosynthesis , Sequence Alignment , Substrate Specificity , Sulfolobus solfataricus/genetics , Temperature
19.
J Biol Chem ; 280(33): 29765-70, 2005 Aug 19.
Article in English | MEDLINE | ID: mdl-15955819

ABSTRACT

The hepatitis C virus (HCV) polymerase is required for replication of the viral genome and is a key target for therapeutic intervention against HCV. We have determined the crystal structures of the HCV polymerase complexed with two indole-based allosteric inhibitors at 2.3- and 2.4-Angstroms resolution. The structures show that these inhibitors bind to a site on the surface of the thumb domain. A cyclohexyl and phenyl ring substituents, bridged by an indole moiety, fill two closely spaced pockets, whereas a carboxylate substituent forms a salt bridge with an exposed arginine side chain. Interestingly, in the apoenzyme, the inhibitor binding site is occupied by a small alpha-helix at the tip of the N-terminal loop that connects the fingers and thumb domains. Thus, these molecules inhibit the enzyme by preventing formation of intramolecular contacts between these two domains and consequently precluding their coordinated movements during RNA synthesis. Our structures identify a novel mechanism by which a new class of allosteric inhibitors inhibits the HCV polymerase and open the way to the development of novel antiviral agents against this clinically relevant human pathogen.


Subject(s)
Enzyme Inhibitors/pharmacology , Viral Nonstructural Proteins/chemistry , Allosteric Site , Amino Acid Sequence , Antiviral Agents/pharmacology , Binding Sites , Molecular Sequence Data , Protein Conformation , Viral Nonstructural Proteins/antagonists & inhibitors
20.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 1): 146-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14684912

ABSTRACT

TraM is a 11.4 kDa protein involved in the control of the conjugal transfer of Agrobacterium tumefaciens Ti plasmids by quorum-sensing. TraM was overexpressed and purified from Escherichia coli. This protein binds to the transcriptional regulator TraR, abolishing its function. Size-exclusion chromatography and dynamic light scattering show that the recombinant protein has an apparent molecular weight of 30 kDa in solution. Crystals have been obtained of both native and selenomethionine-substituted TraM by the vapour-diffusion method. Crystals diffract to 1.67 A and belong to the space group P2(1)2(1)2, with unit-cell parameters a = 76.43, b = 47.09, c = 47.46 A and two molecules in the asymmetric unit. A two-wavelength MAD data set for the selenomethionine-substituted form has been collected to a resolution of 2.0 A. The selenium substructure (five out of six possible sites) has been solved using direct methods.


Subject(s)
Agrobacterium tumefaciens/chemistry , Bacterial Proteins/chemistry , Agrobacterium tumefaciens/genetics , Amino Acid Sequence , Bacterial Proteins/genetics , Chromatography, Gel , Cloning, Molecular , Crystallization , Crystallography, X-Ray , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/genetics , Light , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Scattering, Radiation , Selenomethionine/chemistry , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL