Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(47): eadi1735, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37992173

ABSTRACT

Cross-linked elastomers are stretchable materials that typically are not recyclable or biodegradable. Medium-chain-length polyhydroxyalkanoates (mcl-PHAs) are soft and ductile, making these bio-based polymers good candidates for biodegradable elastomers. Elasticity is commonly imparted by a cross-linked network structure, and covalent adaptable networks have emerged as a solution to prepare recyclable thermosets via triggered rearrangement of dynamic covalent bonds. Here, we develop biodegradable and recyclable elastomers by chemically installing the covalent adaptable network within biologically produced mcl-PHAs. Specifically, an engineered strain of Pseudomonas putida was used to produce mcl-PHAs containing pendent terminal alkenes as chemical handles for postfunctionalization. Thiol-ene chemistry was used to incorporate boronic ester (BE) cross-links, resulting in PHA-based vitrimers. mcl-PHAs cross-linked with BE at low density (<6 mole %) affords a soft, elastomeric material that demonstrates thermal reprocessability, biodegradability, and denetworking at end of life. The mechanical properties show potential for applications including adhesives and soft, biodegradable robotics and electronics.


Subject(s)
Polyhydroxyalkanoates , Pseudomonas putida , Polyhydroxyalkanoates/chemistry , Pseudomonas putida/genetics , Elasticity , Elastomers
2.
Sci Rep ; 12(1): 15517, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36109550

ABSTRACT

Coronavirus disease 2019 (COVID-19) continues to significantly impact the global population, thus countermeasure platforms that enable rapid development of therapeutics against variants of SARS-CoV-2 are essential. We report use of a phage display human antibody library approach to rapidly identify neutralizing antibodies (nAbs) against SARS-CoV-2. We demonstrate the binding and neutralization capability of two nAbs, STI-2020 and STI-5041, against the SARS-CoV-2 WA-1 strain as well as the Alpha and Beta variants. STI-2020 and STI-5041 were protective when administered intravenously or intranasally in the golden (Syrian) hamster model of COVID-19 challenged with the WA-1 strain or Beta variant. The ability to administer nAbs intravenously and intranasally may have important therapeutic implications and Phase 1 healthy subjects clinical trials are ongoing.


Subject(s)
COVID-19 , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Cricetinae , Humans , Mesocricetus , Neutralization Tests , SARS-CoV-2
3.
Front Oncol ; 12: 884196, 2022.
Article in English | MEDLINE | ID: mdl-35664753

ABSTRACT

Therapeutic blockade of the CD47/SIRPα axis by small molecules or monoclonal antibodies (mAbs) is a proven strategy to enhance macrophages-mediated anti-tumor activity. However, this strategy has been hampered by elevated on-target toxicities and rapid clearance due to the extensive CD47 expression on normal cells ("antigen sink") such as red blood cells (RBCs). To address these hurdles, we report on the development of STI-6643, an affinity-engineered fully human anti-CD47 IgG4 antibody with negligible binding to normal cells. STI-6643 exhibited no hemagglutination activity on human RBCs at concentrations up to 300 µg/mL yet specifically blocked the CD47/SIPRα interaction. Of particular interest, STI-6643 preserved T cell functionality in vitro and showed significantly lower immune cell depletion in vivo in contrast to three previously published competitor reference anti-CD47 clones Hu5F9, AO-176 and 13H3. In cynomolgus monkeys, STI-6643 was well-tolerated at the highest dose tested (300 mg/kg/week) and provided favorable clinical safety margins. Finally, STI-6643 displayed comparable anti-tumor activity to the high-affinity reference clone Hu5F9 in a RAJI-Fluc xenograft tumor model as monotherapy or in combination with anti-CD20 (rituximab) or anti-CD38 (daratumumab) mAbs. These data suggest that STI-6643 possesses the characteristics of an effective therapeutic candidate given its potent anti-tumor activity and low toxicity profile.

4.
Antiviral Res ; 195: 105185, 2021 11.
Article in English | MEDLINE | ID: mdl-34634289

ABSTRACT

Monoclonal antibodies (mAbs) are emerging as safe and effective therapeutics against SARS-CoV-2. However, variant strains of SARS-CoV-2 have evolved, with early studies showing that some mAbs may not sustain their efficacy in the face of escape mutants. Also, from the onset of the COVID-19 pandemic, concern has been raised about the potential for Fcγ receptor-mediated antibody-dependent enhancement (ADE) of infection. In this study, plaque reduction neutralization assays demonstrated that mAb 1741-LALA neutralizes SARS-CoV-2 strains B.1.351, D614 and D614G. MAbs S1D2-hIgG1 and S1D2-LALA mutant (STI-1499-LALA) did not neutralize B.1.351, but did neutralize SARS-CoV-2 strains D614 and D614G. LALA mutations did not result in substantial differences in neutralizing abilities between clones S1D2-hIgG1 vs STI-1499-LALA. S1D2-hIgG1, STI-1499-LALA, and convalescent plasma showed minimal ability to induce ADE in human blood monocyte-derived macrophages. Further, no differences in pharmacokinetic clearance of S1D2-hIgG1 vs STI-1499-LALA were observed in mice expressing human FcRn. These findings confirm that SARS-CoV-2 has already escaped some mAbs, and identify a mAb candidate that may neutralize multiple SARS-CoV-2 variants. They also suggest that risk of ADE in macrophages may be low with SARS-CoV-2 D614, and LALA Fc change impacts neither viral neutralization nor Ab clearance.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Enhancement , SARS-CoV-2/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chlorocebus aethiops , Humans , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutralization Tests , Spike Glycoprotein, Coronavirus/immunology , Vero Cells
5.
Antimicrob Agents Chemother ; 58(7): 3860-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24777097

ABSTRACT

Novel broad-spectrum antimicrobials are a critical component of a strategy for combating antibiotic-resistant pathogens. In this study, we explored the activity of the broad-spectrum antiviral compound ST-669 for activity against different intracellular bacteria and began a characterization of its mechanism of antimicrobial action. ST-669 inhibits the growth of three different species of chlamydia and the intracellular bacterium Coxiella burnetii in Vero and HeLa cells but not in McCoy (murine) cells. The antichlamydial and anti-C. burnetii activity spectrum was consistent with those observed for tested viruses, suggesting a common mechanism of action. Cycloheximide treatment in the presence of ST-669 abrogated the inhibitory effect, demonstrating that eukaryotic protein synthesis is required for tested activity. Immunofluorescence microscopy demonstrated that different chlamydiae grow atypically in the presence of ST-669, in a manner that suggests the compound affects inclusion formation and organization. Microscopic analysis of cells treated with a fluorescent derivative of ST-669 demonstrated that the compound localized to host cell lipid droplets but not to other organelles or the host cytosol. These results demonstrate that ST-669 affects intracellular growth in a host-cell-dependent manner and interrupts proper development of chlamydial inclusions, possibly through a lipid droplet-dependent process.


Subject(s)
Antiviral Agents/pharmacology , Chlamydia Infections/drug therapy , Chlamydia/drug effects , Inclusion Bodies/microbiology , Lipids/chemistry , Thiourea/pharmacology , Animals , Cells, Cultured , Chlamydia/growth & development , Chlamydia Infections/microbiology , Coxiella burnetii/drug effects , Genome, Bacterial , Humans , Mice
6.
Bioorg Med Chem Lett ; 22(13): 4263-72, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22664128

ABSTRACT

A series of acylthiourea derivatives were designed, synthesized, and evaluated for broad-spectrum antiviral activity with selected viruses from Poxviridae (vaccinia virus) and two different genera of the family Bunyaviridae (Rift Valley fever and La Crosse viruses). A compound selected from a library screen, compound 1, displayed submicromolar antiviral activity against both vaccinia virus (EC(50)=0.25 µM) and La Crosse virus (EC(50)=0.27 µM) in cytopathic effect (CPE) assays. SAR analysis was performed to further improve antiviral potency and to optimize drug-like properties of the initial hits. During our analysis, we identified 26, which was found to be nearly fourfold more potent than 1 against both vaccinia and La Crosse viruses. Selected compounds were further tested to more fully characterize the spectrum of antiviral activity. Many of these possessed single digit micromolar and sub-micromolar antiviral activity against a diverse array of targets, including influenza virus (Orthomyxoviridae), Tacaribe virus (Arenaviridae), and dengue virus (Flaviviridae).


Subject(s)
Antiviral Agents/chemistry , Thiourea/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Arenavirus/drug effects , Dengue Virus/drug effects , La Crosse virus/drug effects , Orthomyxoviridae/drug effects , Structure-Activity Relationship , Thiourea/chemical synthesis , Thiourea/pharmacology , Vaccinia virus/drug effects
7.
Antiviral Res ; 90(1): 70-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21371508

ABSTRACT

Lassa virus (LASV), a member of the Arenaviridae family, causes a viral hemorrhagic fever endemic to West Africa, where as many as 300,000 infections occur per year. Presently, there are no FDA-approved LASV-specific vaccines or antiviral agents, although the antiviral drug ribavirin has shown some efficacy. A recently identified small-molecule inhibitor of arenavirus entry, ST-193, exhibits submicromolar antiviral activity in vitro. To determine the antiviral utility of ST-193 in vivo, we tested the efficacy of this compound in the LASV guinea pig model. Four groups of strain 13 guinea pigs were administered 25 or 80 mg/kg ST-193, 25 mg/kg of ribavirin, or the vehicle by the intraperitoneal (i.p.) route before infection with a lethal dose of LASV, strain Josiah, and continuing once daily for 14 days. Control animals exhibited severe disease, becoming moribund between days 10 and 15 postinfection. ST-193-treated animals exhibited fewer signs of disease and enhanced survival when compared to the ribavirin or vehicle groups. Body temperatures in all groups were elevated by day 9, but returned to normal by day 19 postinfection in the majority of ST-193-treated animals. ST-193 treatment mediated a 2-3-log reduction in viremia relative to vehicle-treated controls. The overall survival rate for the ST-193-treated guinea pigs was 62.5% (10/16) compared with 0% in the ribavirin (0/8) and vehicle (0/7) groups. These data suggest that ST-193 may serve as an improved candidate for the treatment of Lassa fever.


Subject(s)
Antiviral Agents/administration & dosage , Lassa Fever/drug therapy , Animals , Body Temperature , Disease Models, Animal , Female , Guinea Pigs , Injections, Intraperitoneal , Lassa Fever/mortality , Lassa Fever/pathology , Survival Analysis , Viremia/prevention & control
8.
J Virol ; 83(1): 314-28, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18971285

ABSTRACT

In the process of characterizing the requirements for expression of the essential immediate-early transcriptional activator (RTA) encoded by gene 50 of murine gammaherpesvirus 68 (MHV68), a recombinant virus was generated in which the known gene 50 promoter was deleted (G50pKO). Surprisingly, the G50pKO mutant retained the ability to replicate in permissive murine fibroblasts, albeit with slower kinetics than wild-type MHV68. 5'-rapid amplification of cDNA ends analyses of RNA prepared from G50pKO-infected fibroblasts revealed a novel upstream transcription initiation site, which was also utilized during wild-type MHV68 infection of permissive cells. Furthermore, the region upstream of the distal gene 50/RTA transcription initiation site exhibited promoter activity in both permissive NIH 3T12 fibroblasts as well as in the murine macrophage cell line RAW 264.7. In addition, in RAW 264.7 cells the activity of the distal gene 50/RTA promoter was strongly upregulated (>20-fold) by treatment of the cells with lipopolysaccharide. Reverse transcriptase PCR analyses of RNA prepared from Kaposi's sarcoma-associated herpesvirus- and Epstein-Barr virus-infected B-cell lines, following induction of virus reactivation, also revealed the presence of gene 50/RTA transcripts initiating upstream of the known transcription initiation site. The latter argues that alternative initiation of gene 50/RTA transcription is a strategy conserved among murine and human gammaherpesviruses. Infection of mice with the MHV68 G50pKO demonstrated the ability of this mutant virus to establish latency in the spleen and peritoneal exudate cells (PECs). However, the G50pKO mutant was unable to reactivate from latently infected splenocytes and also exhibited a significant reactivation defect from latently infected PECs, arguing in favor of a model where the proximal gene 50/RTA promoter plays a critical role in virus reactivation from latency, particularly from B cells. Finally, analyses of viral genome methylation in the regions upstream of the proximal and distal gene 50/RTA transcription initiation sites revealed that the distal promoter is partially methylated in vivo and heavily methylated in MHV68 latently infected B-cell lines, suggesting that DNA methylation may serve to silence the activity of this promoter during virus latency.


Subject(s)
Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Promoter Regions, Genetic , Rhadinovirus/physiology , Transcription Factors/biosynthesis , Transcription Initiation Site , Transcription, Genetic , Viral Proteins/biosynthesis , Animals , B-Lymphocytes/virology , Cell Line , Cells, Cultured , Female , Fibroblasts/virology , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/genetics , Humans , Macrophages/virology , Mice , Rhadinovirus/genetics , Transcription Factors/genetics , Virus Latency , Virus Replication
9.
J Virol ; 81(21): 11957-71, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17699571

ABSTRACT

Gammaherpesviruses establish lifelong, latent infections in host lymphocytes, during which a limited subset of viral gene products facilitates maintenance of the viral episome. Among the gamma-2-herpesvirus (rhadinovirus) subfamily, this includes expression of the conserved ORF73-encoded LANA proteins. We previously demonstrated by loss-of-function mutagenesis that the murine gammaherpesvirus 68 (MHV68) ORF73 gene product, mLANA, is required for the establishment of latency following intranasal inoculation of mice (N. J. Moorman, D. O. Willer, and S. H. Speck, J. Virol. 77:10295-10303, 2003). mLANA-deficient viruses also exhibited a defect in acute virus replication in the lungs of infected mice. The latter observation led us to examine the role of mLANA in productive viral replication. We assessed the capacity of mLANA-deficient virus (73.Stop) to replicate in cell culture at low multiplicities of infection (MOIs) and found that 73.Stop growth was impaired in murine fibroblasts but not in Vero cells. A recombinant virus expressing an mLANA-green fluorescent protein (GFP) fusion revealed that mLANA is expressed throughout the virus replication cycle. In addition, 73.Stop infection of murine fibroblasts at high MOIs was substantially more cytotoxic than infection with a genetically repaired marker rescue virus (73.MR), a phenotype that correlated with enhanced kinetics of viral gene expression and increased activation of p53. Notably, augmented cell death, viral gene expression, and p53 induction were independent of viral DNA replication. Expression of a mLANA-GFP fusion protein in fibroblasts correlated with both reduced p53 stabilization and reduced cell death following treatment with p53-inducing agonists. In agreement, accentuated cell death associated with 73.Stop infection was reduced in p53-deficient murine embryonic fibroblasts. Additionally, replication of 73.Stop in p53-deficient cells was restored to levels comparable to those of 73.MR. More remarkably, the absence of p53 led to an overall delay in replication for both 73.Stop and 73.MR viruses, which correlated with delayed viral gene expression, indicating a role for p53 in MHV68 replication. Consistent with these findings, the expression of replication-promoting viral genes was positively influenced by p53 overexpression or treatment with the p53 agonist etoposide. Overall, these data demonstrate the importance of mLANA in MHV68 replication and suggest that LANA proteins limit the induction of cellular stress responses to regulate the viral gene expression cascade and limit host cell injury.


Subject(s)
Antigens, Neoplasm/physiology , Neoplasm Proteins/physiology , Tumor Suppressor Protein p53/metabolism , Viral Proteins/genetics , Virus Replication , Animals , Cell Line , Chlorocebus aethiops , Fibroblasts/metabolism , Gammaherpesvirinae , Gene Expression Regulation, Viral , Green Fluorescent Proteins/metabolism , Humans , MART-1 Antigen , Mice , NIH 3T3 Cells , Vero Cells , Viral Proteins/physiology
10.
Virology ; 365(2): 250-9, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17477952

ABSTRACT

Among the distinguishing characteristics of members of the gamma-2 herpesvirus family is the expression of a mammalian D-type cyclin homolog, termed v-cyclin. Murine gammaherpesvirus 68 (gammaHV68) is a gamma2-herpesvirus that can infect inbred and outbred strains of mice, providing a genetic system for the study of gammaherpesvirus pathogenesis. Disruption of the v-cyclin gene of gammaHV68 results in a virus that establishes latency in infected mice to wild-type levels, but is severely attenuated for virus reactivation [van Dyk, L.F., Virgin IV, H.W., Speck, S.H., 2000. J. Virol. 74:7451-7461]. Transcriptional regulation of the gammaHV68 v-cyclin has not been defined. We report here the initial characterization of the v-cyclin transcript expressed in permissive murine fibroblasts. Based on 5' mapping of the v-cyclin transcript, we identified a promoter that is involved in driving v-cyclin expression during virus replication. In addition, we determined that the promoter is responsive to the major viral lytic transactivator, Rta, encoded by orf 50. Using reporter plasmids we have analyzed both basal and Rta-induced v-cyclin promoter activity, initially identifying two regions of the v-cyclin promoter important for both basal and Rta-induced activity. Notably, only one of these regions could be shown to confer Rta responsiveness on a reporter construct containing the hsp70 TATA box. The importance of this region in regulating v-cyclin expression during virus replication was confirmed by introducing these mutations into the context of the viral genome and assessing v-cyclin expression following infection of permissive murine fibroblasts in tissue culture. In addition, we show that mutations that severely cripple Rta-induction of v-cyclin expression did not adversely impact virus reactivation from splenocytes recovered from latently infected mice, indicating that alternatively regulated v-cyclin gene expression is required for virus reactivation.


Subject(s)
Cyclins/biosynthesis , Gammaherpesvirinae/genetics , Gene Expression Regulation, Viral , Promoter Regions, Genetic , Viral Proteins/biosynthesis , Virus Activation/genetics , Virus Replication/genetics , Animals , Artificial Gene Fusion , Base Sequence , Cell Line , Fibroblasts/virology , Gammaherpesvirinae/physiology , Genes, Reporter , Luciferases/biosynthesis , Luciferases/genetics , Mice , Molecular Sequence Data , Mutagenesis , RNA, Messenger/biosynthesis , RNA, Viral/biosynthesis , Regulatory Elements, Transcriptional , Trans-Activators/metabolism , Transcription, Genetic , Virus Activation/physiology , Virus Replication/physiology
11.
J Virol ; 80(4): 2055-62, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16439562

ABSTRACT

Regulation of orf73 (LANA) gene expression is critical to the establishment and maintenance of latency following infection by members of the gamma-2 herpesvirus (rhadinovirus) family. Previous studies of murine gammaherpesvirus 68 (gammaHV68) have demonstrated that loss of LANA function results in a complete failure to establish virus latency in the spleens of laboratory mice. Here we report the characterization of alternatively spliced LANA and v-cyclin (orf72) transcripts encoded by gammaHV68. Similar to other rhadinoviruses, alternative splicing, coupled with alternative 3' processing, of a ca. 16-kb transcriptional unit can lead to expression of either LANA or v-cyclin during gammaHV68 infection. Spliced LANA and v-cyclin transcripts were initially identified from an analysis of the gammaHV68 latently infected B-cell lymphoma cell line S11E, but were also detected during lytic infection of NIH 3T12 fibroblasts. 5' Random amplification of cDNA ends (RACE) analyses identified two distinct promoters, p1 and p2, that drive expression of spliced LANA transcripts. Analysis of p1 and p2, using transiently transfected reporter constructs, mapped the minimal sequences required for promoter activity and demonstrated that both promoters are active in the absence of any viral antigens. Analysis of spliced LANA and v-cyclin transcripts in spleens recovered from latently infected mice at days 16 and 42 postinfection revealed that spliced v-cyclin transcripts can only be detected sporadically, suggesting that these may be associated with cells reactivating from latency. In contrast, spliced LANA transcripts were detected in ca. 1 in 4,000 splenocytes harvested at day 16 postinfection. Notably, based on the frequency of viral genome-positive splenocytes at day 16 postinfection (ca. 1 in 200), only 5 to 10% of viral genome-positive splenocytes express LANA. The failure of the majority of infected splenocytes at day 16 postinfection to express LANA may contribute to the contraction in the frequency of latently infected splenocytes as chronic infection is established, due to failure to maintain the viral episome in proliferating B cells.


Subject(s)
Alternative Splicing , Antigens, Viral/genetics , Cyclins/genetics , Gammaherpesvirinae/genetics , Herpesviridae Infections/virology , Nuclear Proteins/genetics , RNA, Messenger/analysis , Viral Proteins/genetics , Virus Latency , Animals , Base Sequence , Cell Line, Tumor , Cells, Cultured , Mice , Models, Animal , Molecular Sequence Data , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional , RNA, Viral/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
12.
J Virol ; 79(15): 9480-91, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014911

ABSTRACT

Murine gammaherpesvirus 68 (gammaHV68) provides a tractable small animal model with which to study the mechanisms involved in the establishment and maintenance of latency by gammaherpesviruses. Similar to the human gammaherpesvirus Epstein-Barr virus (EBV), gammaHV68 establishes and maintains latency in the memory B-cell compartment following intranasal infection. Here we have sought to determine whether, like EBV infection, gammaHV68 infection in vivo is associated with B-cell proliferation during the establishment of chronic infection. We show that gammaHV68 infection leads to significant splenic B-cell proliferation as late as day 42 postinfection. Notably, gammaHV68 latency was found predominantly in the proliferating B-cell population in the spleen on both days 16 and 42 postinfection. Furthermore, virus reactivation upon ex vivo culture was heavily biased toward the proliferating B-cell population. DNA methyltransferase 1 (Dnmt1) is a critical maintenance methyltransferase which, during DNA replication, maintains the DNA methylation patterns of the cellular genome, a process that is essential for the survival of proliferating cells. To assess whether the establishment of gammaHV68 latency requires B-cell proliferation, we characterized infections of conditional Dnmt1 knockout mice by utilizing a recombinant gammaHV68 that expresses Cre-recombinase (gammaHV68-Cre). In C57BL/6 mice, the gammaHV68-Cre virus exhibited normal acute virus replication in the lungs as well as normal establishment and reactivation from latency. Furthermore, the gammaHV68-Cre virus also replicated normally during the acute phase of infection in the lungs of Dnmt1 conditional mice. However, deletion of the Dnmt1 alleles from gammaHV68-infected cells in vivo led to a severe ablation of viral latency, as assessed on both days 16 and 42 postinfection. Thus, the studies provide direct evidence that the proliferation of latently infected B cells is critical for the establishment of chronic gammaHV68 infection.


Subject(s)
B-Lymphocytes/immunology , Gammaherpesvirinae/physiology , Herpesviridae Infections/immunology , Animals , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics , Time Factors , Virus Latency
SELECTION OF CITATIONS
SEARCH DETAIL
...