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










Publication year range
1.
Antiviral Res ; 226: 105896, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38679167

ABSTRACT

Immune tolerance to the hepatitis B virus (HBV) is crucial for developing chronic hepatitis B, and the HBV surface antigen (HBsAg) produced and secreted in high amounts is regarded as a key contributor. HBsAg is expressed in HBV-infected hepatocytes and those carrying an HBV integration. Whether either HBsAg secretion or the high antigen amount expressed in the liver determines its immunomodulatory properties, however, remains unclear. We, therefore, developed a novel HBV animal model that allowed us to study the role of secreted HBsAg. We introduced a previously described HBs mutation, C65S, abolishing HBsAg secretion into a replication-competent 1.3-overlength HBV genome and used adeno-associated virus vectors to deliver it to the mouse liver. The AAV-HBV established a carrier state of wildtype and C65S mutant HBV, respectively. We investigated antiviral B- and T-cell immunity in the HBV-carrier mice after therapeutic vaccination. Moreover, we compared the effect of a lacking HBsAg secretion with that of an antiviral siRNA. While missing HBsAg secretion allowed for higher levels of detectable anti-HBs antibodies after therapeutic vaccination, it did neither affect antiviral T-cell responses nor intrahepatic HBV gene expression, irrespective of the starting level. A treatment with HBV siRNA restricting viral antigen expression within hepatocytes, however, improved the antiviral efficacy of therapeutic vaccination, irrespective of the ability of HBV to secrete HBsAg. Our data indicate that clearing HBsAg from blood cannot significantly impact HBV persistence or T-cell immunity. This indicates that a restriction of hepatic viral antigen expression will be required to break HBV immunotolerance.

2.
Sci Rep ; 14(1): 5768, 2024 03 08.
Article in English | MEDLINE | ID: mdl-38459123

ABSTRACT

The SARS-CoV-2 pandemic has highlighted the need to better define in-hospital transmissions, a need that extends to all other common infectious diseases encountered in clinical settings. To evaluate how whole viral genome sequencing can contribute to deciphering nosocomial SARS-CoV-2 transmission 926 SARS-CoV-2 viral genomes from 622 staff members and patients were collected between February 2020 and January 2021 at a university hospital in Munich, Germany, and analysed along with the place of work, duration of hospital stay, and ward transfers. Bioinformatically defined transmission clusters inferred from viral genome sequencing were compared to those inferred from interview-based contact tracing. An additional dataset collected at the same time at another university hospital in the same city was used to account for multiple independent introductions. Clustering analysis of 619 viral genomes generated 19 clusters ranging from 3 to 31 individuals. Sequencing-based transmission clusters showed little overlap with those based on contact tracing data. The viral genomes were significantly more closely related to each other than comparable genomes collected simultaneously at other hospitals in the same city (n = 829), suggesting nosocomial transmission. Longitudinal sampling from individual patients suggested possible cross-infection events during the hospital stay in 19.2% of individuals (14 of 73 individuals). Clustering analysis of SARS-CoV-2 whole genome sequences can reveal cryptic transmission events missed by classical, interview-based contact tracing, helping to decipher in-hospital transmissions. These results, in line with other studies, advocate for viral genome sequencing as a pathogen transmission surveillance tool in hospitals.


Subject(s)
COVID-19 , Cross Infection , Humans , SARS-CoV-2/genetics , COVID-19/epidemiology , COVID-19/genetics , Genome, Viral/genetics , Cross Infection/epidemiology , Cross Infection/genetics , Hospitals, University
3.
J Control Release ; 345: 661-674, 2022 05.
Article in English | MEDLINE | ID: mdl-35364120

ABSTRACT

SARS-CoV-2 has been the cause of a global pandemic since 2019 and remains a medical urgency. siRNA-based therapies are a promising strategy to fight viral infections. By targeting a specific region of the viral genome, siRNAs can efficiently downregulate viral replication and suppress viral infection. However, to achieve the desired therapeutic activity, siRNA requires a suitable delivery system. The VIPER (virus-inspired polymer for endosomal release) block copolymer has been reported as promising delivery system for both plasmid DNA and siRNA in the past years. It is composed of a hydrophilic block for condensation of nucleic acids as well as a hydrophobic, pH-sensitive block that, at acidic pH, exposes the membrane lytic peptide melittin, which enhances endosomal escape. In this study, we aimed at developing a formulation for pulmonary administration of siRNA to suppress SARS-CoV-2 replication in lung epithelial cells. After characterizing siRNA/VIPER polyplexes, the activity and safety profile were confirmed in a lung epithelial cell line. To further investigate the activity of the polyplexes in a more sophisticated cell culture system, an air-liquid interface (ALI) culture was established. siRNA/VIPER polyplexes reached the cell monolayer and penetrated through the mucus layer secreted by the cells. Additionally, the activity against wild-type SARS-CoV-2 in the ALI model was confirmed by qRT-PCR. To investigate translatability of our findings, the activity against SARS-CoV-2 was tested ex vivo in human lung explants. Here, siRNA/VIPER polyplexes efficiently inhibited SARS-CoV-2 replication. Finally, we verified the delivery of siRNA/VIPER polyplexes to lung epithelial cells in vivo, which represent the main cellular target of viral infection in the lung. In conclusion, siRNA/VIPER polyplexes efficiently delivered siRNA to lung epithelial cells and mediated robust downregulation of viral replication both in vitro and ex vivo without toxic or immunogenic side effects in vivo, demonstrating the potential of local siRNA delivery as a promising antiviral therapy in the lung.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/therapy , Humans , Lung/metabolism , Polymers/chemistry , RNA, Small Interfering , SARS-CoV-2/genetics , Virus Replication/genetics
4.
Biomolecules ; 12(3)2022 03 18.
Article in English | MEDLINE | ID: mdl-35327662

ABSTRACT

In chronic hepatitis B virus (HBV) infection, virus-specific T cells are scarce and partially dysfunctional. Therapeutic vaccination is a promising strategy to induce and activate new virus-specific T cells. In long-term or high-level HBV carriers, however, therapeutic vaccination by itself may not suffice to cure HBV. One reason is the impairment of antiviral T cells by immune checkpoints. In this study, we used small-interfering RNA (siRNA) in combination with a heterologous prime-boost therapeutic vaccination scheme (TherVacB) to interfere with a major immune checkpoint, the interaction of programmed death protein-1 (PD-1) and its ligand (PDL-1). In mice persistently replicating HBV after infection with an adeno-associated virus harboring the HBV genome, siRNA targeting PD-L1 resulted in a higher functionality of HBV-specific CD8+ T cells after therapeutic vaccination, and allowed for a more sustained antiviral effect and control of HBV in peripheral blood and in the liver. The antiviral effect was more pronounced if PD-L1 was down-regulated during prime than during boost vaccination. Thus, targeting PD-L1 using siRNA is a promising approach to enhance the efficacy of therapeutic vaccination and finally cure HBV.


Subject(s)
Hepatitis B, Chronic , Animals , Antiviral Agents , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes , Hepatitis B virus/genetics , Hepatitis B, Chronic/therapy , Mice , RNA, Small Interfering/genetics , Vaccination
5.
Nucleic Acids Res ; 50(1): 333-349, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34928377

ABSTRACT

A promising approach to tackle the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) could be small interfering (si)RNAs. So far it is unclear, which viral replication steps can be efficiently inhibited with siRNAs. Here, we report that siRNAs can target genomic RNA (gRNA) of SARS-CoV-2 after cell entry, and thereby terminate replication before start of transcription and prevent virus-induced cell death. Coronaviruses replicate via negative sense RNA intermediates using a unique discontinuous transcription process. As a result, each viral RNA contains identical sequences at the 5' and 3' end. Surprisingly, siRNAs were not active against intermediate negative sense transcripts. Targeting common sequences shared by all viral transcripts allowed simultaneous suppression of gRNA and subgenomic (sg)RNAs by a single siRNA. The most effective suppression of viral replication and spread, however, was achieved by siRNAs that targeted open reading frame 1 (ORF1) which only exists in gRNA. In contrast, siRNAs that targeted the common regions of transcripts were outcompeted by the highly abundant sgRNAs leading to an impaired antiviral efficacy. Verifying the translational relevance of these findings, we show that a chemically modified siRNA that targets a highly conserved region of ORF1, inhibited SARS-CoV-2 replication ex vivo in explants of the human lung. Our work encourages the development of siRNA-based therapies for COVID-19 and suggests that early therapy start, or prophylactic application, together with specifically targeting gRNA, might be key for high antiviral efficacy.


Subject(s)
COVID-19/virology , Lung/virology , RNA, Small Interfering , RNA, Viral , SARS-CoV-2/genetics , Virus Replication , 3' Untranslated Regions , Animals , Antiviral Agents/pharmacology , Cell Survival , Databases, Genetic , HEK293 Cells , Humans , Nucleic Acid Conformation , Oligonucleotides , Open Reading Frames , RNA, Small Interfering/metabolism , COVID-19 Drug Treatment
6.
J Hepatol ; 75(1): 64-73, 2021 07.
Article in English | MEDLINE | ID: mdl-33516779

ABSTRACT

BACKGROUND & AIMS: Hypoxia inducible factors (HIFs) are a hallmark of inflammation and are key regulators of hepatic immunity and metabolism, yet their role in HBV replication is poorly defined. HBV replicates in hepatocytes within the liver, a naturally hypoxic organ, however most studies of viral replication are performed under conditions of atmospheric oxygen, where HIFs are inactive. We therefore investigated the role of HIFs in regulating HBV replication. METHODS: Using cell culture, animal models, human tissue and pharmacological agents inhibiting the HIF-prolyl hydroxylases, we investigated the impact of hypoxia on the HBV life cycle. RESULTS: Culturing liver cell-based model systems under low oxygen uncovered a new role for HIFs in binding HBV DNA and activating the basal core promoter, leading to increased pre-genomic RNA and de novo HBV particle secretion. The presence of hypoxia responsive elements among all primate members of the hepadnaviridae highlights an evolutionary conserved role for HIFs in regulating this virus family. CONCLUSIONS: Identifying a role for this conserved oxygen sensor in regulating HBV transcription suggests that this virus has evolved to exploit the HIF signaling pathway to persist in the low oxygen environment of the liver. Our studies show the importance of considering oxygen availability when studying HBV-host interactions and provide innovative routes to better understand and target chronic HBV infection. LAY SUMMARY: Viral replication in host cells is defined by the cellular microenvironment and one key factor is local oxygen tension. Hepatitis B virus (HBV) replicates in the liver, a naturally hypoxic organ. Hypoxia inducible factors (HIFs) are the major sensors of low oxygen; herein, we identify a new role for these factors in regulating HBV replication, revealing new therapeutic targets.


Subject(s)
Hepatitis B virus , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Kruppel-Like Factor 6/metabolism , Oxygen/metabolism , Virus Replication/physiology , Animals , Cellular Microenvironment , Hepadnaviridae/physiology , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology , Host Microbial Interactions , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Liver/metabolism , Signal Transduction , Transcriptional Activation
7.
Adv Healthc Mater ; 10(7): e2001650, 2021 04.
Article in English | MEDLINE | ID: mdl-33506607

ABSTRACT

Acute viral respiratory tract infections (AVRIs) are a major burden on human health and global economy and amongst the top five causes of death worldwide resulting in an estimated 3.9 million lives lost every year. In addition, new emerging respiratory viruses regularly cause outbreaks such as SARS-CoV-1 in 2003, the "Swine flu" in 2009, or most importantly the ongoing SARS-CoV-2 pandemic, which intensely impact global health, social life, and economy. Despite the prevalence of AVRIs and an urgent need, no vaccines-except for influenza-or effective treatments were available at the beginning of the COVID-19 pandemic. However, the innate RNAi pathway offers the ability to develop nucleic acid-based antiviral drugs. siRNA sequences against conserved, essential regions of the viral genome can prevent viral replication. In addition, viral infection can be averted prophylactically by silencing host genes essential for host-viral interactions. Unfortunately, delivering siRNAs to their target cells and intracellular site of action remains the principle hurdle toward their therapeutic use. Currently, siRNA formulations and chemical modifications are evaluated for their delivery. This progress report discusses the selection of antiviral siRNA sequences, delivery techniques to the infection sites, and provides an overview of antiviral siRNAs against respiratory viruses.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , RNA, Small Interfering/therapeutic use , Respiratory Tract Infections/drug therapy , Virus Diseases/drug therapy , Humans , Respiratory Tract Infections/virology , Viruses
9.
Viruses ; 12(11)2020 11 23.
Article in English | MEDLINE | ID: mdl-33238650

ABSTRACT

Several viral factors impact the natural course of hepatitis B virus (HBV) infection, the sensitivity of diagnostic tests, or treatment response to interferon-α and nucleos(t)ide analogues. These factors include the viral genotype and serotype but also mutations affecting the HBV surface antigen, basal core promoter/pre-core region, or reverse transcriptase. However, a comprehensive overview of the distribution of HBV variants between HBV genotypes or different geographical locations is lacking. To address this, we performed an in silico analysis of publicly available HBV full-length genome sequences. We found that not only the serotype frequency but also the majority of clinically relevant mutations are primarily associated with specific genotypes. Distinct mutations enriched in certain world regions are not explained by the local genotype distribution. Two HBV variants previously identified to confer resistance to the nucleotide analogue tenofovir in vitro were not identified, questioning their translational relevance. In summary, our work elucidates the differences in the clinical manifestation of HBV infection observed between genotypes and geographical locations and furthermore helps identify suitable diagnostic tests and therapies.


Subject(s)
Databases, Nucleic Acid , Genetic Variation , Genotype , Global Health , Hepatitis B virus/genetics , Access to Information , Antiviral Agents/pharmacology , Computer Simulation , Geography , Hepatitis B/virology , Hepatitis B virus/classification , Hepatitis B virus/drug effects , Humans , Sequence Analysis, DNA , Tenofovir/pharmacology
10.
Sci Rep ; 10(1): 14101, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32839523

ABSTRACT

Hepatitis B virus (HBV) is the leading cause of hepatocellular carcinoma (HCC) worldwide. The prolyl hydroxylase domain (PHD)-hypoxia inducible factor (HIF) pathway is a key mammalian oxygen sensing pathway and is frequently perturbed by pathological states including infection and inflammation. We discovered a significant upregulation of hypoxia regulated gene transcripts in patients with chronic hepatitis B (CHB) in the absence of liver cirrhosis. We used state-of-the-art in vitro and in vivo HBV infection models to evaluate a role for HBV infection and the viral regulatory protein HBx to drive HIF-signalling. HBx had no significant impact on HIF expression or associated transcriptional activity under normoxic or hypoxic conditions. Furthermore, we found no evidence of hypoxia gene expression in HBV de novo infection, HBV infected human liver chimeric mice or transgenic mice with integrated HBV genome. Collectively, our data show clear evidence of hypoxia gene induction in CHB that is not recapitulated in existing models for acute HBV infection, suggesting a role for inflammatory mediators in promoting hypoxia gene expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/genetics , Hepatitis B, Chronic/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Disease Models, Animal , Female , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress/physiology , Oxygen/metabolism
11.
Mol Cell Proteomics ; 19(9): 1503-1522, 2020 09.
Article in English | MEDLINE | ID: mdl-32591346

ABSTRACT

As the COVID-19 pandemic continues to spread, thousands of scientists around the globe have changed research direction to understand better how the virus works and to find out how it may be tackled. The number of manuscripts on preprint servers is soaring and peer-reviewed publications using MS-based proteomics are beginning to emerge. To facilitate proteomic research on SARS-CoV-2, the virus that causes COVID-19, this report presents deep-scale proteomes (10,000 proteins; >130,000 peptides) of common cell line models, notably Vero E6, Calu-3, Caco-2, and ACE2-A549 that characterize their protein expression profiles including viral entry factors such as ACE2 or TMPRSS2. Using the 9 kDa protein SRP9 and the breast cancer oncogene BRCA1 as examples, we show how the proteome expression data can be used to refine the annotation of protein-coding regions of the African green monkey and the Vero cell line genomes. Monitoring changes of the proteome on viral infection revealed widespread expression changes including transcriptional regulators, protease inhibitors, and proteins involved in innate immunity. Based on a library of 98 stable-isotope labeled synthetic peptides representing 11 SARS-CoV-2 proteins, we developed PRM (parallel reaction monitoring) assays for nano-flow and micro-flow LC-MS/MS. We assessed the merits of these PRM assays using supernatants of virus-infected Vero E6 cells and challenged the assays by analyzing two diagnostic cohorts of 24 (+30) SARS-CoV-2 positive and 28 (+9) negative cases. In light of the results obtained and including recent publications or manuscripts on preprint servers, we critically discuss the merits of MS-based proteomics for SARS-CoV-2 research and testing.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/genetics , Host-Pathogen Interactions/genetics , Pneumonia, Viral/genetics , Proteomics/methods , Viral Proteins/genetics , A549 Cells , Amino Acid Sequence , Angiotensin-Converting Enzyme 2 , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Betacoronavirus/pathogenicity , COVID-19 , Caco-2 Cells , Case-Control Studies , Chlorocebus aethiops , Coronavirus Infections/pathology , Coronavirus Infections/virology , Gene Expression Regulation , Gene Ontology , Humans , Indicators and Reagents , Molecular Sequence Annotation , Open Reading Frames , Pandemics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Proteomics/instrumentation , SARS-CoV-2 , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Recognition Particle/genetics , Signal Recognition Particle/metabolism , Signal Transduction , Vero Cells , Viral Proteins/classification , Viral Proteins/metabolism , Virus Internalization
12.
Gastroenterology ; 158(6): 1762-1775.e9, 2020 05.
Article in English | MEDLINE | ID: mdl-32001321

ABSTRACT

BACKGROUND & AIMS: Hepatitis B virus (HBV) infection persists because the virus-specific immune response is dysfunctional. Therapeutic vaccines might be used to end immune tolerance to the virus in patients with chronic infection, but these have not been effective in patients so far. In patients with chronic HBV infection, high levels of virus antigens might prevent induction of HBV-specific immune responses. We investigated whether knocking down expression levels of HBV antigens in liver might increase the efficacy of HBV vaccines in mice. METHODS: We performed studies with male C57BL/6 mice that persistently replicate HBV (genotype D, serotype ayw)-either from a transgene or after infection with an adeno-associated virus that transferred an overlength HBV genome-and expressed HB surface antigen at levels relevant to patients. Small hairpin or small interfering (si)RNAs against the common 3'-end of all HBV transcripts were used to knock down antigen expression in mouse hepatocytes. siRNAs were chemically stabilized and conjugated to N-acetylgalactosamine to increase liver uptake. Control mice were given either entecavir or non-HBV-specific siRNAs and vaccine components. Eight to 12 weeks later, mice were immunized twice with a mixture of adjuvanted HBV S and core antigen, followed by a modified Vaccinia virus Ankara vector to induce HBV-specific B- and T-cell responses. Serum and liver samples were collected and analyzed for HBV-specific immune responses, liver damage, and viral parameters. RESULTS: In both models of HBV infection, mice that express hepatocyte-specific small hairpin RNAs or that were given subcutaneous injections of siRNAs had reduced levels of HBV antigens, HBV replication, and viremia (1-3 log10 reduction) compared to mice given control RNAs. Vaccination induced production of HBV-neutralizing antibodies and increased numbers and functionality of HBV-specific, CD8+ T cells in mice with low, but not in mice with high, levels of HBV antigen. Mice with initially high titers of HBV and knockdown of HBV antigen expression, but not mice with reduced viremia after administration of entecavir, developed polyfunctional, HBV-specific CD8+ T cells, and HBV was eliminated. CONCLUSIONS: In mice with high levels of HBV replication, knockdown of HBV antigen expression along with a therapeutic vaccination strategy, but not knockdown alone, increased numbers of effector T cells and eliminated the virus. These findings indicate that high titers of virus antigens reduce the efficacy of therapeutic vaccination. Anti-HBV siRNAs and therapeutic vaccines are each being tested in clinical trials-their combination might cure chronic HBV infection.


Subject(s)
Hepatitis B Surface Antigens/genetics , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/therapy , RNA, Small Interfering/administration & dosage , Animals , B-Lymphocytes/immunology , Carrier State/immunology , Carrier State/virology , Combined Modality Therapy/methods , Disease Models, Animal , Female , Gene Knockdown Techniques , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/administration & dosage , Hepatitis B virus/genetics , Hepatitis B virus/isolation & purification , Hepatitis B, Chronic/diagnosis , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Hepatocytes/virology , Humans , Immunization, Secondary , Immunogenicity, Vaccine , Male , Mice , T-Lymphocytes, Cytotoxic/immunology , Virus Replication/genetics , Virus Replication/immunology
13.
Genes (Basel) ; 9(10)2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30326600

ABSTRACT

Hepatitis B virus (HBV) is divided into nine genotypes, A to I. Currently, it remains unclear how the individual genotypes contribute to the estimated 250 million chronic HBV infections. We performed a literature search on HBV genotyping data throughout the world. Over 900 publications were assessed and data were extracted from 213 records covering 125 countries. Using previously published HBV prevalence, and population data, we approximated the number of infections with each HBV genotype per country and the genotype distribution among global chronic HBV infections. We estimated that 96% of chronic HBV infections worldwide are caused by five of the nine genotypes: genotype C is most common (26%), followed by genotype D (22%), E (18%), A (17%) and B (14%). Genotypes F to I together cause less than 2% of global chronic HBV infections. Our work provides an up-to-date analysis of global HBV genotyping data and an initial approach to estimate how genotypes contribute to the global burden of chronic HBV infection. Results highlight the need to provide HBV cell culture and animal models that cover at least genotypes A to E and represent the vast majority of global HBV infections to test novel treatment strategies.

14.
Mol Ther Nucleic Acids ; 12: 242-253, 2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30195763

ABSTRACT

High-capacity adenoviral vectors (HCAdVs) devoid of all coding genes are powerful tools to deliver large DNA cargos into cells. Here HCAdVs were designed to deliver a multiplexed complete CRISPR/Cas9 nuclease system or a complete pair of transcription activator-like effector nucleases (TALENs) directed against the hepatitis B virus (HBV) genome. HBV, which remains a serious global health burden, forms covalently closed circular DNA (cccDNA) as a persistent DNA species in infected cells. This cccDNA promotes the chronic carrier status, and it represents a major hurdle in the treatment of chronic HBV infection. To date, only one study demonstrated viral delivery of a CRISPR/Cas9 system and a single guide RNA (gRNA) directed against HBV by adeno-associated viral (AAV) vectors. The advancement of this study is the co-delivery of multiple gRNA expression cassettes along with the Cas9 expression cassette in one HCAdV. Treatment of HBV infection models resulted in a significant reduction of HBV antigen production and the introduction of mutations into the HBV genome. In the transduction experiments, the HBV genome, including the HBV cccDNA, was degraded by the CRISPR/Cas9 system. In contrast, the combination of two parts of a TALEN pair in one vector could not be proven to yield an active system. In conclusion, we successfully delivered the CRISPR/Cas9 system containing three gRNAs using HCAdV, and we demonstrated its antiviral effect.

15.
Curr Opin Virol ; 24: 38-45, 2017 06.
Article in English | MEDLINE | ID: mdl-28433762

ABSTRACT

Hepatitis B virus (HBV) infection is a global health threat with 240 million chronic carriers at high risk to develop hepatocellular carcinoma. Current antiviral treatment can efficiently control viral replication and reduce liver inflammation, but is still quite far from achieving a cure. Significant progress has been made in understanding the virus life cycle and virus-host interaction in the past few years. With identification of the HBV receptor, cell-culture infection systems have become available that allow drug screening and establishing a pipeline of potential antivirals targeting either viral or host factors. Most of the candidate antivirals summarized in this review are still in preclinical development, but some have already entered or are about to enter early clinical trials.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis B virus/drug effects , Hepatitis B, Chronic/drug therapy , Hepatitis B/drug therapy , Host-Pathogen Interactions , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Carcinoma, Hepatocellular , Hepatitis B/virology , Hepatitis B virus/physiology , Hepatitis B, Chronic/virology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/virology , Virus Replication/drug effects
16.
EMBO Mol Med ; 8(9): 1082-98, 2016 09.
Article in English | MEDLINE | ID: mdl-27473329

ABSTRACT

Hepatitis B virus (HBV) is a promising target for therapies based on RNA interference (RNAi) since it replicates via RNA transcripts that are vulnerable to RNAi silencing. Clinical translation of RNAi technology, however, requires improvements in potency, specificity and safety. To this end, we systematically compared different strategies to express anti-HBV short hairpin RNA (shRNA) in a pre-clinical immunocompetent hepatitis B mouse model. Using recombinant Adeno-associated virus (AAV) 8 vectors for delivery, we either (i) embedded the shRNA in an artificial mi(cro)RNA under a liver-specific promoter; (ii) co-expressed Argonaute-2, a rate-limiting cellular factor whose saturation with excess RNAi triggers can be toxic; or (iii) co-delivered a decoy ("TuD") directed against the shRNA sense strand to curb off-target gene regulation. Remarkably, all three strategies minimised adverse side effects as compared to a conventional shRNA vector that caused weight loss, liver damage and dysregulation of > 100 hepatic genes. Importantly, the novel AAV8 vector co-expressing anti-HBV shRNA and TuD outperformed all other strategies regarding efficiency and persistence of HBV knock-down, thus showing substantial promise for clinical translation.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Hepatitis B/therapy , RNA, Small Interfering/pharmacology , Animals , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Dependovirus/genetics , Disease Models, Animal , Drug Carriers , Drug-Related Side Effects and Adverse Reactions , Gene Expression , Genetic Vectors , Mice , Transduction, Genetic
17.
Gastroenterology ; 150(1): 194-205, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26416327

ABSTRACT

BACKGROUND & AIMS: Viral clearance involves immune cell cytolysis of infected cells. However, studies of hepatitis B virus (HBV) infection in chimpanzees have indicated that cytokines released by T cells also can promote viral clearance via noncytolytic processes. We investigated the noncytolytic mechanisms by which T cells eliminate HBV from infected hepatocytes. METHODS: We performed a cytokine enzyme-linked immunosorbent assay of serum samples from patients with acute and chronic hepatitis B. Liver biopsy specimens were analyzed by in situ hybridization. HepG2-H1.3 cells, HBV-infected HepaRG cells, and primary human hepatocytes were incubated with interferon-γ (IFNγ) or tumor necrosis factor-α (TNF-α), or co-cultured with T cells. We measured markers of HBV replication, including the covalently closed circular DNA (cccDNA). RESULTS: Levels of IFNγ and TNF-α were increased in serum samples from patients with acute vs chronic hepatitis B and controls. In human hepatocytes with stably replicating HBV, as well as in HBV-infected primary human hepatocytes or HepaRG cells, IFNγ and TNF-α each induced deamination of cccDNA and interfered with its stability; their effects were additive. HBV-specific T cells, through secretion of IFNγ and TNF-α, inhibited HBV replication and reduced cccDNA in infected cells without the direct contact required for cytolysis. Blocking IFNγ and TNF-α after T-cell stimulation prevented the loss of cccDNA. Deprivation of cccDNA required activation of nuclear APOBEC3 deaminases by the cytokines. In liver biopsy specimens from patients with acute hepatitis B, but not chronic hepatitis B or controls, hepatocytes expressed APOBEC3A and APOBEC3B. CONCLUSIONS: IFNγ and TNF-α, produced by T cells, reduce levels of HBV cccDNA in hepatocytes by inducing deamination and subsequent cccDNA decay.


Subject(s)
Hepatitis B/metabolism , Interferon-gamma/pharmacology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Virus Replication/drug effects , Antiviral Agents/pharmacology , Cells, Cultured , Coculture Techniques , DNA Replication/drug effects , DNA, Viral/drug effects , DNA, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Hep G2 Cells/immunology , Hep G2 Cells/metabolism , Hepacivirus/metabolism , Hepatitis B/physiopathology , Hepatitis B, Chronic/immunology , Humans , T-Lymphocytes/immunology , Viral Load
18.
Trends Pharmacol Sci ; 35(9): 470-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108320

ABSTRACT

Enveloped viruses pose an important health threat because most of the persistent and many emerging viruses are enveloped. In particular, newly emerging viruses create a need to develop broad-spectrum antivirals, which usually are obtained by targeting host cell factors. Persistent viruses have developed efficient strategies to escape host immune control, and treatment options are limited. Targeting host cell factors essential for virus persistence, or immune-based therapies provide alternative approaches. In this review, we therefore focus on recent developments to generate antivirals targeting host cell factors or immune-based therapeutic approaches to fight infections with enveloped viruses.


Subject(s)
Antiviral Agents/therapeutic use , Virus Diseases/drug therapy , Animals , Antiviral Agents/pharmacology , DNA Viruses , Humans , Immunotherapy , RNA Viruses , Virus Diseases/therapy
19.
Hepatology ; 59(6): 2110-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24425003

ABSTRACT

UNLABELLED: Gene therapy has become an accepted concept for the treatment of a variety of different diseases. In contrast to preclinical models, subjects enrolled in clinical trials, including gene therapy, possess a history of infection with microbes that may influence its safety and efficacy. Especially, viruses that establish chronic infections in the liver, one of the main targets for in vivo gene therapy, raise important concerns. Among them is the hepatitis B virus (HBV), which has chronically infected more than 350 million people worldwide. Here, we investigated the effect of HBV on adeno-associated viral (AAV) vectors, the most frequently applied gene transfer vehicles for in vivo gene therapy. Unexpectedly, we found that HBV greatly improved AAV transduction in cells replicating HBV and identified HBV protein x (HBx) as a key factor. Whereas HBV-positive and -negative cells were indistinguishable with respect to cell-entry efficiency, significantly higher numbers of AAV vector genomes were successfully delivered to the nucleus in the presence of HBV. The HBV-promoting effect was abolished by inhibitors of phosphatidylinositol 3-kinase (PI3K). PI3K was required for efficient trafficking of AAV to the nucleus and was enhanced in HBV-replicating cells and upon HBx expression. Enhancement of AAV transduction was confirmed in vivo using HBV transgenic mice and could successfully be applied to inhibit HBV progeny release. CONCLUSION: Our results demonstrate that acute, as well as chronic, infections with unrelated viruses change the intracellular milieu, thereby likely influencing gene therapy outcomes. In the case of HBV, HBx-mediated enhancement of AAV transduction is an advantage that could be exploited for development of novel treatments of HBV infection.


Subject(s)
Dependovirus/genetics , Genetic Predisposition to Disease , Hepatitis B virus/genetics , Hepatitis B/virology , Animals , Enzyme Activation/genetics , Female , Genetic Vectors , HEK293 Cells , Hepatitis B/genetics , Hepatitis B/therapy , Humans , Interferon-gamma/genetics , Male , Mice , Mice, Transgenic , RNA, Small Interfering/genetics , Trans-Activators/genetics , Transduction, Genetic , Viral Regulatory and Accessory Proteins , Virus Replication/genetics
20.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G181-92, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23139224

ABSTRACT

The endocannabinoid system has been shown to mediate beneficial effects on gastrointestinal inflammation via cannabinoid receptors 1 (CB(1)) and 2 (CB(2)). These receptors have also been reported to activate the MAP kinases p38 and c-Jun NH(2)-terminal kinase (JNK), which are involved in early acinar events leading to acute pancreatitis and induction of proinflammatory cytokines. Our aim was to examine the role of cannabinoid receptor activation in an experimental model of acute pancreatitis and the potential involvement of MAP kinases. Cerulein pancreatitis was induced in wild-type, CB(1)-/-, and MK2-/- mice pretreated with selective cannabinoid receptor agonists or antagonists. Severity of pancreatitis was determined by serum amylase and IL-6 levels, intracellular activation of pancreatic trypsinogen, lung myeloperoxidase activity, pancreatic edema, and histological examinations. Pancreatic lysates were investigated by Western blotting using phospho-specific antibodies against p38 and JNK. Quantitative PCR data, Western blotting experiments, and immunohistochemistry clearly show that CB(1) and CB(2) are expressed in mouse pancreatic acini. During acute pancreatitis, an upregulation especially of CB(2) on apoptotic cells occurred. The unselective CB(1)/CB(2) agonist HU210 ameliorated pancreatitis in wild-type and CB(1)-/- mice, indicating that this effect is mediated by CB(2). Furthermore, blockade of CB(2), not CB(1), with selective antagonists engraved pathology. Stimulation with a selective CB(2) agonist attenuated acute pancreatitis and an increased activation of p38 was observed in the acini. With use of MK2-/- mice, it could be demonstrated that this attenuation is dependent on MK2. Hence, using the MK2-/- mouse model we reveal a novel CB(2)-activated and MAP kinase-dependent pathway that modulates cytokine expression and reduces pancreatic injury and affiliated complications.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cannabinoids/pharmacology , Dronabinol/analogs & derivatives , Intracellular Signaling Peptides and Proteins/metabolism , Pancreas, Exocrine/drug effects , Pancreatitis/prevention & control , Protein Serine-Threonine Kinases/metabolism , Receptor, Cannabinoid, CB2/agonists , p38 Mitogen-Activated Protein Kinases/metabolism , Amylases/blood , Animals , Apoptosis , Blotting, Western , Ceruletide , Disease Models, Animal , Dronabinol/pharmacology , Edema/chemically induced , Edema/enzymology , Edema/prevention & control , Enzyme Activation , Immunohistochemistry , Interleukin-6/blood , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lung/drug effects , Lung/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas, Exocrine/enzymology , Pancreatitis/blood , Pancreatitis/chemically induced , Pancreatitis/enzymology , Pancreatitis/genetics , Peroxidase/metabolism , Phosphorylation , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Trypsinogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...