Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Viruses ; 13(9)2021 09 01.
Article in English | MEDLINE | ID: mdl-34578326

ABSTRACT

The rapid spread of the pandemic caused by the SARS-CoV-2 virus has created an unusual situation, with rapid searches for compounds to interfere with the biological processes exploited by the virus. Doxycycline, with its pleiotropic effects, including anti-viral activity, has been proposed as a therapeutic candidate for COVID-19 and about twenty clinical trials have started since the beginning of the pandemic. To gain information on the activity of doxycycline against SARS-CoV-2 infection and clarify some of the conflicting clinical data published, we designed in vitro binding tests and infection studies with a pseudotyped virus expressing the spike protein, as well as a clinically isolated SARS-CoV-2 strain. Doxycycline inhibited the transduction of the pseudotyped virus in Vero E6 and HEK-293 T cells stably expressing human receptor angiotensin-converting enzyme 2 but did not affect the entry and replication of SARS-CoV-2. Although this conclusion is apparently disappointing, it is paradigmatic of an experimental approach aimed at developing an integrated multidisciplinary platform which can shed light on the mechanisms of action of potential anti-COVID-19 compounds. To avoid wasting precious time and resources, we believe very stringent experimental criteria are needed in the preclinical phase, including infectivity studies with clinically isolated SARS-CoV-2, before moving on to (futile) clinical trials.


Subject(s)
COVID-19/virology , Host-Pathogen Interactions , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Virus Physiological Phenomena/drug effects , Virus Replication/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/metabolism , Cell Cycle , Chlorocebus aethiops , Doxycycline/pharmacology , HEK293 Cells , Humans , Protein Binding , SARS-CoV-2/ultrastructure , Spike Glycoprotein, Coronavirus , Transduction, Genetic , Vero Cells
2.
Virology ; 552: 94-106, 2021 01 02.
Article in English | MEDLINE | ID: mdl-33120225

ABSTRACT

Drugs against flaviviruses such as dengue (DENV) and Zika (ZIKV) virus are urgently needed. We previously demonstrated that three fluoroquinolones, ciprofloxacin, enoxacin, and difloxacin, suppress replication of six flaviviruses. To investigate the barrier to resistance and mechanism(s) of action of these drugs, DENV-4 was passaged in triplicate in HEK-293 cells in the presence or absence of each drug. Resistance to ciprofloxacin was detected by the seventh passage and to difloxacin by the tenth, whereas resistance to enoxacin did not occur within ten passages. Two putative resistance-conferring mutations were detected in the envelope gene of ciprofloxacin and difloxacin-resistant DENV-4. In the absence of ciprofloxacin, ciprofloxacin-resistant viruses sustained a significantly higher viral titer than control viruses in HEK-293 and HuH-7 cells and resistant viruses were more stable than control viruses at 37 °C. These results suggest that the mechanism of action of ciprofloxacin and difloxacin involves interference with virus binding or entry.


Subject(s)
Biological Evolution , Dengue Virus/drug effects , Dengue Virus/physiology , Dengue/virology , Fluoroquinolones/pharmacology , Genetic Fitness/drug effects , Virus Physiological Phenomena/drug effects , Adaptation, Biological , Animals , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Ciprofloxacin/analogs & derivatives , Ciprofloxacin/pharmacology , Drug Resistance, Viral , Enoxacin/pharmacology , HEK293 Cells , Host Microbial Interactions , Humans , Mutation , Vero Cells , Viral Envelope/physiology
3.
Hum Vaccin Immunother ; 16(12): 2992-3000, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32755425

ABSTRACT

The current Coronavirus Disease 2019 (COVID-19) pandemic is causing great alarm around the world. The pathogen for COVID-19 - severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) - is the seventh known coronavirus to cause pneumonia in humans. While much remains unknown about SARS-CoV-2, physicians and researchers have begun to publish relevant findings, and much evidence is available on coronaviruses previously circulating in human and animal populations. In this review, we situate COVID-19 in its context as a transboundary viral disease, and provide a comprehensive discussion focused on the discovery, spread, virology, pathogenesis, and clinical features of this disease, its causative coronaviral pathogen, and approaches to combating the disease through immunotherapies and other treatments and vaccine development. An epidemiological survey revealed a potentially large number of asymptomatic SARS-CoV-2 carriers within the population, which may hamper efforts against COVID-19. Finally, we emphasize that vaccines against SARS-CoV-2, which may be developed by 2021, will be essential for prevention of COVID-19.


Subject(s)
COVID-19/immunology , COVID-19/prevention & control , Drug Development/methods , Immunotherapy/trends , Virus Physiological Phenomena/immunology , Animals , COVID-19/epidemiology , Drug Development/trends , Humans , Immunotherapy/methods , Virus Physiological Phenomena/drug effects
4.
Article in English | MEDLINE | ID: mdl-31681621

ABSTRACT

Regulation of RNA homeostasis or "RNAstasis" is a central step in eukaryotic gene expression. From transcription to decay, cellular messenger RNAs (mRNAs) associate with specific proteins in order to regulate their entire cycle, including mRNA localization, translation and degradation, among others. The best characterized of such RNA-protein complexes, today named membraneless organelles, are Stress Granules (SGs) and Processing Bodies (PBs) which are involved in RNA storage and RNA decay/storage, respectively. Given that SGs and PBs are generally associated with repression of gene expression, viruses have evolved different mechanisms to counteract their assembly or to use them in their favor to successfully replicate within the host environment. In this review we summarize the current knowledge about the viral regulation of SGs and PBs, which could be a potential novel target for the development of broad-spectrum antiviral therapies.


Subject(s)
Host-Pathogen Interactions , Organelles , Virus Diseases/metabolism , Virus Diseases/virology , Virus Physiological Phenomena , Animals , Cytoplasmic Granules , Gene Expression Regulation , Gene Expression Regulation, Viral , Host-Pathogen Interactions/genetics , Humans , Organelles/metabolism , Organelles/virology , Signal Transduction , Stress, Physiological , Virus Diseases/genetics , Virus Physiological Phenomena/drug effects , Virus Replication , Viruses/classification , Viruses/drug effects , Viruses/genetics
5.
FEMS Microbiol Ecol ; 95(8)2019 08 01.
Article in English | MEDLINE | ID: mdl-31314089

ABSTRACT

Subsurface environments hold the largest reservoir of microbes in the biosphere. They play essential roles in transforming nutrients, degrading contaminants and recycling organic matter. Here, we propose a previously unrecognised fundamental microbial process that influences aquifer bioremediation dynamics and that applies to all microbial communities. In contrast to previous models, our proposed Piggyback-the-Persistent (PtP) mechanism occurs when viruses become more dominated by those exhibiting temperate rather than lytic lifestyles driven by persistent chemicals (in our case chlorinated-hydrocarbon pollutants) that provide long-term carbon sources and that refocus the aquifer carbon cycle, thus altering the microbial community. In this ultra-oligotrophic system, the virus:microbial ratio (VMR) ranges from below the detection limit of 0.0001 to 0.6, well below the common aquatic range of 3-10. Shortest-average-path network analysis revealed VMR and trichlorethene (TCE) as nodes through which ecosystem information and biomass most efficiently pass. Novel network rearrangement revealed a hierarchy of Kill-the-Winner (KtW), Piggyback-the-Winner (PtW) and PtP nodes. We propose that KtW, PtW and PtP occur simultaneously as competing strategies, with their relative importance depending on conditions at a particular time and location with unusual nutrient sources, such as TCE, appearing to contribute to a shift in this balance between viral mechanisms.


Subject(s)
Bacteria/virology , Groundwater/microbiology , Trichloroethylene/metabolism , Virus Physiological Phenomena/drug effects , Viruses/growth & development , Bacteria/growth & development , Bacteria/metabolism , Biodegradation, Environmental , Biomass , Ecosystem , Hydrocarbons/metabolism , Viral Load
6.
Pathog Dis ; 77(2)2019 03 01.
Article in English | MEDLINE | ID: mdl-30980658

ABSTRACT

The quasispecies dynamics of viral populations (continuous generation of variant genomes and competition among them) has as one of its frequent consequences variations in overall multiplication capacity, a major component of viral fitness. This parameter has multiple implications for viral pathogenesis and viral disease control, some of them unveiled thanks to deep sequencing of viral populations. Darwinian fitness is an old concept whose quantification dates back to the early developments of population genetics. It was later applied to viruses (mainly to RNA viruses) to quantify relative multiplication capacities of individual mutant clones or complex populations. The present article reviews the fitness concept and its relevance for the understanding of the adaptive dynamics of viruses in constant and changing environments. Many studies have addressed the fitness cost of escape mutations (to antibodies, cytotoxic T cells or inhibitors) as an influence on the efficacy of antiviral interventions. Here, we summarize the evidence that the basal fitness level can be a determinant of inhibitor resistance.


Subject(s)
Genetic Fitness , Host-Pathogen Interactions , Virus Diseases/virology , Virus Physiological Phenomena , Adaptation, Biological , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Biological Evolution , Drug Resistance, Viral , Genetic Fitness/drug effects , Humans , Mutation , Virus Diseases/drug therapy , Virus Diseases/epidemiology , Virus Physiological Phenomena/drug effects
7.
Oral Dis ; 25(3): 652-669, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29656422

ABSTRACT

The oral cavity harbors different taxonomic groups, the evolutionary coexistence of which develops the oral ecosystem. These resident microorganisms can alter the balance between the physiologic and pathologic conditions that affect the host, both locally and systemically. This highly sophisticated nature of the oral cavity poses a significant therapeutic challenge. Numerous human and animal studies have been conducted to potentiate the efficacy and competence of current treatments of pathologic conditions as well as to develop novel therapeutic modalities. One of these studies is the use of the potent antimicrobial agent lactoferrin (LF), which was originally derived from the host immune system. LF is an 80-kDa glycoprotein that has a free iron sequestration mechanism with evident antimicrobial, anti-tumor, and immunomodulatory properties. A wide range of active peptides have been isolated from the N-terminal region of LF, which possess antimicrobial activities. In this review, we discuss the role of LF and LF-derived peptides under a heterogeneous group of oral and maxillofacial conditions, including bacterial, fungal, viral infections; head and neck cancers; xerostomia; and implantology-bone-related manifestations.


Subject(s)
Bacteria/drug effects , Lactoferrin/pharmacology , Mouth Neoplasms/prevention & control , Peptides/pharmacology , Periodontal Diseases/microbiology , Animals , Candida albicans/drug effects , Carcinogenesis/drug effects , Dental Caries/microbiology , Dental Caries/prevention & control , Humans , Lactoferrin/genetics , Lactoferrin/therapeutic use , Peptides/therapeutic use , Polymorphism, Single Nucleotide , Streptococcus mutans/drug effects , Virus Physiological Phenomena/drug effects
8.
EBioMedicine ; 39: 591-602, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30553752

ABSTRACT

Type 2 diabetes (T2D) has become a global epidemic. Although several drugs are available to manage T2D, problems associated with person-to-person variability in drug efficacy and potential side-effects remain unresolved. Owing to the emerging role of the gut microbiome in obesity and T2D, the interaction between gut microbes and anti-diabetic drugs and its influence on drugs' functions remains of immediate research interest. On one hand, drugs can manipulate gut microbiome composition and metabolic capacity. Conversely, the metabolic activities of the microbiome and its metabolites can also influence drug metabolism and effects. Hence, understanding this bi-directional drug-microbiome interaction and how it influences the clinical outcomes of antidiabetic drugs can pave the way to develop next-generation strategies to ameliorate diabetes. This review presents evidences demonstrating the putative interactions between anti-diabetic drugs and the gut microbiome, and discusses the potential of microbiome modulators to manipulate drug-microbiome interactions and the drug metabolism.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Microbiome/drug effects , Hypoglycemic Agents/pharmacology , Archaea/drug effects , Archaea/physiology , Bacterial Physiological Phenomena/drug effects , Diabetes Mellitus, Type 2/metabolism , Fungi/drug effects , Fungi/physiology , Gene Expression Regulation/drug effects , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Virus Physiological Phenomena/drug effects
9.
Viruses ; 10(11)2018 11 01.
Article in English | MEDLINE | ID: mdl-30388805

ABSTRACT

To complement traditional antivirals, natural compounds that act via host targets and present high barriers to resistance are of increasing interest. In the work reported here, we detected that homoharringtonine (HHT) presents effective antiviral activity. HHT completely inhibited infections of vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), and porcine epidemic diarrhea virus (PEDV) at concentrations of 50, 100, and 500 nM in cell cultures, respectively. Treatment with HHT at doses of 0.05 or 0.2 mg/kg significantly reduced viral load and relieved severe symptoms in PEDV- or NDV-infected animals. HHT treatment, however, moderately inhibited avian influenza virus (AIV) infection, suggesting its potent antiviral action is restricted to a number of classes of RNA viruses. In this study, we also observed that HHT actively inhibited herpes simplex virus type 1 (HSV-1) replication with a 50% inhibitory concentration (IC50) of 139 nM; the treatment with HHT at 1000 nM led to reductions of three orders of magnitude. Moreover, HHT antagonized the phosphorylation level of endogenous and exogenous eukaryotic initiation factor 4E (p-eIF4E), which might regulate the selective translation of specific messenger RNA (mRNA). HHT provides a starting point for further progress toward the clinical development of broad-spectrum antivirals.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , Homoharringtonine/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/chemistry , Biological Products/chemistry , Cells, Cultured , Chick Embryo , Dose-Response Relationship, Drug , Homoharringtonine/chemistry , Humans , Microbial Sensitivity Tests , Molecular Structure , Swine , Transcription Factors/metabolism , Viral Load , Viral Plaque Assay , Virus Physiological Phenomena/drug effects , Viruses/drug effects
10.
Arch Microbiol ; 200(6): 841-846, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29846759

ABSTRACT

Virus infections are the root cause of epidemics in the world. Vaccines and antiviral agents have been the two important methods to control viral diseases; in recent times, RNA-mediated therapeutics and prevention have received much attention. In this review, we provide an overview of the current information regarding the use of vaccines, antiviral agents, and RNA-mediated methods in controlling or preventing viral infections. We stress specifically on the potential of existing RNA-mediated methods in clinical applications.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery/trends , Virus Diseases/virology , Viruses/drug effects , Animals , Humans , RNA, Viral/genetics , Virus Diseases/drug therapy , Virus Physiological Phenomena/drug effects , Viruses/genetics
11.
Int J Biol Macromol ; 115: 77-82, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29654857

ABSTRACT

Viral infectious diseases are seriously endangering human health. In the search for effective antiviral drugs, people have found that polysaccharides have good antiviral activity. As an effective and low-toxic antiviral component, polysaccharides have broad prospects for medicinal use and are deserved for further study. Herein, the antiviral activity and action mechanisms of polysaccharides and their various derivatives were summed up and analyzed.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Polysaccharides/chemistry , Polysaccharides/pharmacology , Animals , Humans , Immunity/drug effects , Virus Physiological Phenomena/drug effects
12.
Biotechnol Adv ; 36(3): 557-576, 2018.
Article in English | MEDLINE | ID: mdl-29292156

ABSTRACT

Despite successful vaccination programs and effective treatments for some viral infections, humans are still losing the battle with viruses. Persisting human pandemics, emerging and re-emerging viruses, and evolution of drug-resistant strains impose continuous search for new antiviral drugs. A combination of detailed information about the molecular organization of viruses and progress in molecular biology and computer technologies has enabled rational antivirals design. Initial step in establishing efficacy of new antivirals is based on simple methods assessing inhibition of the intended target. We provide here an overview of biochemical and cell-based assays evaluating the activity of inhibitors of clinically important viruses.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Virus Physiological Phenomena/drug effects , Animals , Capsid/drug effects , Capsid/metabolism , High-Throughput Screening Assays/methods , Host-Pathogen Interactions/drug effects , Humans , Virus Internalization/drug effects , Virus Replication/drug effects
13.
Front Med ; 11(4): 462-470, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29159596

ABSTRACT

The emergence of new pathogens, such as severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and Ebola virus, poses serious challenges to global public health and highlights the urgent need for novel antiviral approaches. Monoclonal antibodies (mAbs) have been successfully used to treat various diseases, particularly cancer and immunological disorders. Antigen-specific mAbs have been isolated using several different approaches, including hybridoma, transgenic mice, phage display, yeast display, and single B-cell isolation. Consequently, an increasing number of mAbs, which exhibit high potency against emerging viruses in vitro and in animal models of infection, have been developed. In this paper, we summarize historical trends and recent developments in mAb discovery, compare the advantages and disadvantages of various approaches to mAb production, and discuss the potential use of such strategies for the development of antivirals against emerging diseases. We also review the application of recently developed human mAbs against SARS-CoV, MERS-CoV, and Ebola virus and discuss prospects for the development of mAbs as therapeutic agents against emerging viral diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antiviral Agents/pharmacology , Communicable Diseases, Emerging/drug therapy , Virus Diseases , Virus Physiological Phenomena , Communicable Diseases, Emerging/virology , Drug Discovery/trends , Humans , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Physiological Phenomena/drug effects , Virus Physiological Phenomena/immunology
14.
Front Med ; 11(4): 471-479, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29159597

ABSTRACT

Several universal influenza virus vaccine candidates based on eliciting antibodies against the hemagglutinin stalk domain are in development. Typically, these vaccines induce responses that target group 1 or group 2 hemagglutinins with little to no cross-group reactivity and protection. Similarly, the majority of human anti-stalk monoclonal antibodies that have been isolated are directed against group 1 or group 2 hemagglutinins with very few that bind to hemagglutinins of both groups. Here we review what is known about the human humoral immune response to vaccination and infection with H7 subtype influenza viruses on a polyclonal and monoclonal level. It seems that unlike vaccination with H5 hemagglutinin, which induces antibody responses mostly restricted to the group 1 stalk domain, H7 exposure induces both group 2 and cross-group antibody responses. A better understanding of this phenomenon and the underlying mechanisms might help to develop future universal influenza virus vaccine candidates.


Subject(s)
Antibodies, Monoclonal/pharmacology , Influenza A Virus, H7N9 Subtype/drug effects , Influenza Vaccines/pharmacology , Influenza, Human , Drug Discovery/trends , Humans , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Virus Physiological Phenomena/drug effects , Virus Physiological Phenomena/immunology
15.
Front Med ; 11(4): 449-461, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29170916

ABSTRACT

In recent years, unexpected outbreaks of infectious diseases caused by emerging and re-emerging viruses have become more frequent, which is possibly due to environmental changes. These outbreaks result in the loss of life and economic hardship. Vaccines and therapeutics should be developed for the prevention and treatment of infectious diseases. In this review, we summarize and discuss the latest progress in the development of small-molecule viral inhibitors against highly pathogenic coronaviruses, including severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome coronavirus, Ebola virus, and Zika virus. These viruses can interfere with the specific steps of viral life cycle by blocking the binding between virus and host cells, disrupting viral endocytosis, disturbing membrane fusion, and interrupting viral RNA replication and translation, thereby demonstrating potent therapeutic effect against various emerging and re-emerging viruses. We also discuss some general strategies for developing small-molecule viral inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Communicable Diseases, Emerging/drug therapy , Virus Diseases/drug therapy , Virus Physiological Phenomena/drug effects , Communicable Diseases, Emerging/virology , Drug Discovery/trends , Humans , RNA, Small Nuclear/antagonists & inhibitors
16.
J Biol Chem ; 292(32): 13284-13295, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28588024

ABSTRACT

Lytic infection by the Epstein-Barr virus (EBV) poses numerous health risks, such as infectious mononucleosis and lymphoproliferative disorder. Proteins in the bromodomain and extraterminal (BET) family regulate multiple stages of viral life cycles and provide promising intervention targets. Synthetic small molecules can bind to the bromodomains and disrupt function by preventing recognition of acetylated lysine substrates. We demonstrate that JQ1 and other BET inhibitors block two different steps in the sequential cascade of the EBV lytic cycle. BET inhibitors prevent expression of the viral immediate-early protein BZLF1. JQ1 alters transcription of genes controlled by the host protein BACH1, and BACH1 knockdown reduces BZLF1 expression. BET proteins also localize to the lytic origin of replication (OriLyt) genetic elements, and BET inhibitors prevent viral late gene expression. There JQ1 reduces BRD4 recruitment during reactivation to preclude replication initiation. This represents a rarely observed dual mode of action for drugs.


Subject(s)
Antiviral Agents/pharmacology , Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Fanconi Anemia Complementation Group Proteins/antagonists & inhibitors , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/drug effects , Nuclear Proteins/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Acetylation , Azepines/pharmacology , Basic-Leucine Zipper Transcription Factors/chemistry , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Cycle Proteins , Cell Line , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions/drug effects , Humans , Lysine/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Protein Transport/drug effects , RNA Interference , Replication Origin/drug effects , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Triazoles/pharmacology , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Activation/drug effects , Virus Physiological Phenomena/drug effects
17.
Biochem J ; 474(10): 1559-1577, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28446620

ABSTRACT

Infections by the human immunodeficiency virus type 1 (HIV-1), the causative agent of the acquired immunodeficiency syndrome (AIDS), are still totaling an appalling 36.7 millions worldwide, with 1.1 million AIDS deaths/year and a similar number of yearly new infections. All this, in spite of the discovery of HIV-1 as the AIDS etiological agent more than 30 years ago and the introduction of an effective combinatorial antiretroviral therapy (cART), able to control disease progression, more than 20 years ago. Although very effective, current cART is plagued by the emergence of drug-resistant viral variants and most of the efforts in the development of novel direct-acting antiviral agents (DAAs) against HIV-1 have been devoted toward the fighting of resistance. In this review, rather than providing a detailed listing of all the drugs and the corresponding resistance mutations, we aim, through relevant examples, at presenting to the general reader the conceptual shift in the approaches that are being taken to overcome the viral resistance hurdle. From the classic 'running faster' strategy, based on the development of novel DAAs active against the mutant viruses selected by the previous drugs and/or presenting to the virus a high genetic barrier toward the development of resilience, to a 'jumping higher' approach, which looks at the cell, rather than the virus, as a source of valuable drug targets, in order to make the cellular environment non-permissive toward the replication of both wild-type and mutated viruses.


Subject(s)
Anti-HIV Agents/therapeutic use , Drug Design , Drug Resistance, Multiple, Viral , Drug Therapy, Combination , HIV Infections/drug therapy , HIV-1/drug effects , Models, Biological , Animals , Anti-HIV Agents/adverse effects , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Antiretroviral Therapy, Highly Active/adverse effects , CCR5 Receptor Antagonists/chemistry , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Drug Therapy, Combination/adverse effects , HIV Infections/metabolism , HIV Infections/virology , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , HIV-1/genetics , HIV-1/growth & development , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Human Immunodeficiency Virus Proteins/antagonists & inhibitors , Human Immunodeficiency Virus Proteins/chemistry , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Molecular Structure , Molecular Targeted Therapy , Mutation , Protein Conformation , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/therapeutic use , Virus Physiological Phenomena/drug effects , Virus Replication/drug effects
18.
Arch Bronconeumol ; 53(1): 19-26, 2017 Jan.
Article in English, Spanish | MEDLINE | ID: mdl-27519544

ABSTRACT

Influenza is a very common contagious disease that carries significant morbidity and mortality. Treatment with antiviral drugs is available, which if administered early, can reduce the risk of severe complications. However, many virus types develop resistance to those drugs, leading to a notable loss of efficacy. There has been great interest in the development of new drugs to combat this disease. A wide range of drugs has shown anti-influenza activity, but they are not yet available for use in the clinic. Many of these target viral components, which others are aimed at elements in the host cell which participate in the viral cycle. Modulating host components is a strategy which minimizes the development of resistance, since host components are not subject to the genetic variability of the virus. The main disadvantage is the risk of treatment-related side effects. The aim of this review is to describe the main pharmacological agents currently available and new drugs in the pipeline with potential benefit in the treatment of influenza.


Subject(s)
Antiviral Agents/therapeutic use , Influenza, Human/drug therapy , Antiviral Agents/pharmacology , Clinical Trials as Topic , Humans , Influenza A virus/drug effects , Influenza B virus/drug effects , MAP Kinase Signaling System/drug effects , Viral Proteins/antagonists & inhibitors , Virus Physiological Phenomena/drug effects
19.
Curr Drug Targets ; 17(8): 942-53, 2016.
Article in English | MEDLINE | ID: mdl-25882224

ABSTRACT

Intracellular pathogens are known to manipulate host cell regulatory pathways to establish an optimal environment for their growth and survival. Pathogens employ active mechanisms to hijack host cell metabolism and acquire existing nutrient and energy store. The role of the cellular energy sensor AMP-activated protein kinase (AMPK) in the regulation of cellular energy homeostasis is well documented. Here, we highlight recent advances showing the importance of AMPK signaling in pathogen-host interactions. Pathogens interact with AMPK by a variety of mechanisms aimed at reprogramming host cell metabolism to their own benefit. Stimulation of AMPK activity provides an efficient process to rapidly adapt pathogen metabolism to the major nutritional changes often encountered during the different phases of infection. However, inhibition of AMPK is also used by pathogens to manipulate innate host response, indicating that AMPK appears relevant to restriction of pathogen infection. We also document the effects of pharmacological AMPK modulators on pathogen proliferation and survival. This review illustrates intricate pathogen-AMPK interactions that may be exploited to the development of novel anti-pathogen therapies.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bacterial Physiological Phenomena , Communicable Diseases/enzymology , Parasites/physiology , Virus Physiological Phenomena , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , Animals , Anti-Infective Agents/pharmacology , Bacterial Physiological Phenomena/drug effects , Communicable Diseases/microbiology , Communicable Diseases/parasitology , Energy Metabolism , Enzyme Activation , Homeostasis , Host-Pathogen Interactions , Humans , Immunity, Innate , Parasites/drug effects , Phosphorylation , Protein Interaction Maps , Signal Transduction , Virus Physiological Phenomena/drug effects
20.
Protein Pept Lett ; 22(6): 497-503, 2015.
Article in English | MEDLINE | ID: mdl-25925771

ABSTRACT

Ribosome inactivating proteins (RIPs) are a family of proteins produced by plants, bacteria and fungi. RIPs have specific N-glycosidase activity, and they cleave a specific glycosidic bond in a universally conserved stem and loop structure within the large ribosomal RNA of all organisms. Saporin, a cytotoxic RIP from the plant Saponaria officinalis has been earlier shown to manifest its cytotoxicity by a combination of its N-glycosidase and apoptosis inducing activities. Saporin, along with many other RIPs also has strong inhibitory activity towards HIV integrase. In the current study, using two in vitro model systems, it is established that saporin inhibits propagation of HIV-1 in host cells. Saporin also showed a potent anti-HIV-1 integrase activity in vitro. Using three active site mutants of saporin, which respectively lack N-glycosidase, apoptosis inducing or both activities, it is shown that saporin's in vitro anti-HIV-1 integrase activity is independent of its N-glycosidase activity. However, for the anti-HIV activity of saporin, the apoptosis inducing activity is important.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Integrase/drug effects , HIV-1/drug effects , Recombinant Proteins/pharmacology , Ribosome Inactivating Proteins, Type 1/pharmacology , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Apoptosis/drug effects , Escherichia coli/genetics , HIV Core Protein p24/metabolism , HIV Infections , HeLa Cells , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosome Inactivating Proteins, Type 1/chemistry , Ribosome Inactivating Proteins, Type 1/genetics , Ribosome Inactivating Proteins, Type 1/metabolism , Saponaria/genetics , Saporins , Virus Physiological Phenomena/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...