LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 13

Search options

  1. Article ; Online: Polyamine-Linked Cholesterol Incorporation in Rift Valley Fever Virus Particles Promotes Infectivity.

    Mastrodomenico, Vincent / LoMascolo, Natalie J / Cruz-Pulido, Yazmin E / Cunha, Christina R / Mounce, Bryan C

    ACS infectious diseases

    2022  Volume 8, Issue 8, Page(s) 1439–1448

    Abstract: Viruses rely on an array of cellular metabolites to replicate and form progeny virions. One set of these molecules, polyamines, are small aliphatic molecules, which are abundant in most cells, that support virus infection; however, the precise roles of ... ...

    Abstract Viruses rely on an array of cellular metabolites to replicate and form progeny virions. One set of these molecules, polyamines, are small aliphatic molecules, which are abundant in most cells, that support virus infection; however, the precise roles of polyamines in virus infection remain incompletely understood. Recent work demonstrated that polyamine metabolism supports cellular cholesterol synthesis through translation of the key transcription factor SREBP2. Here, we show that the bunyavirus Rift Valley fever virus (RVFV) relies on both cholesterol and polyamines for virus infection. Depletion of cellular cholesterol or interruption of cholesterol trafficking negatively impacts RVFV infection. Cholesterol is incorporated into RVFV virions and mediates their infectivity in a polyamine-dependent manner; we find that the virus derived from polyamine-depleted cells lacks cholesterol within the virion membrane. Conversely, we find that virion-associated cholesterol is linked to the incorporation of spermidine within the virion. Our prior work demonstrated that polyamines facilitate pH-mediated fusion and genome release, which may be a consequence of cholesterol depletion within virions. Thus, our work highlights the metabolic connection between polyamines and cholesterol synthesis to impact bunyavirus infection. These data demonstrate the connectedness between cellular metabolic pathways and reveal potential avenues of therapeutic intervention.
    MeSH term(s) Animals ; Cholesterol ; Polyamines ; Rift Valley fever virus/genetics ; Virion/genetics
    Chemical Substances Polyamines ; Cholesterol (97C5T2UQ7J)
    Language English
    Publishing date 2022-07-05
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ISSN 2373-8227
    ISSN (online) 2373-8227
    DOI 10.1021/acsinfecdis.2c00071
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Persistent Coxsackievirus B3 Infection in Pancreatic Ductal Cells

    Mastrodomenico, Vincent / LoMascolo, Natalie J / Firpo, Mason R / Villanueva Guzman, Maria Del Mar / Zaporowski, Adam / Mounce, Bryan C

    mSphere

    2023  Volume 8, Issue 3, Page(s) e0003623

    Abstract: Picornaviruses infect a wide variety of cell ... ...

    Abstract Picornaviruses infect a wide variety of cell types
    MeSH term(s) Animals ; Chlorocebus aethiops ; Humans ; Vero Cells ; Enterovirus B, Human/genetics ; Persistent Infection ; Polyamines/metabolism ; Enterovirus/physiology ; Coxsackievirus Infections/metabolism ; Coxsackievirus Infections/pathology
    Chemical Substances Polyamines
    Language English
    Publishing date 2023-04-25
    Publishing country United States
    Document type Journal Article
    ISSN 2379-5042
    ISSN (online) 2379-5042
    DOI 10.1128/msphere.00036-23
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Peptidomimetic Oligomers Targeting Membrane Phosphatidylserine Exhibit Broad Antiviral Activity.

    Tate, Patrick M / Mastrodomenico, Vincent / Cunha, Christina / McClure, Joshua / Barron, Annelise E / Diamond, Gill / Mounce, Bryan C / Kirshenbaum, Kent

    ACS infectious diseases

    2023  Volume 9, Issue 8, Page(s) 1508–1522

    Abstract: The development of durable new antiviral therapies is challenging, as viruses can evolve rapidly to establish resistance and attenuate therapeutic efficacy. New compounds that selectively target conserved viral features are attractive therapeutic ... ...

    Abstract The development of durable new antiviral therapies is challenging, as viruses can evolve rapidly to establish resistance and attenuate therapeutic efficacy. New compounds that selectively target conserved viral features are attractive therapeutic candidates, particularly for combating newly emergent viral threats. The innate immune system features a sustained capability to combat pathogens through production of antimicrobial peptides (AMPs); however, these AMPs have shortcomings that can preclude clinical use. The essential functional features of AMPs have been recapitulated by peptidomimetic oligomers, yielding effective antibacterial and antifungal agents. Here, we show that a family of AMP mimetics, called peptoids, exhibit direct antiviral activity against an array of enveloped viruses, including the key human pathogens Zika, Rift Valley fever, and chikungunya viruses. These data suggest that the activities of peptoids include engagement and disruption of viral membrane constituents. To investigate how these peptoids target lipid membranes, we used liposome leakage assays to measure membrane disruption. We found that liposomes containing phosphatidylserine (PS) were markedly sensitive to peptoid treatment; in contrast, liposomes formed exclusively with phosphatidylcholine (PC) showed no sensitivity. In addition, chikungunya virus containing elevated envelope PS was more susceptible to peptoid-mediated inactivation. These results indicate that peptoids mimicking the physicochemical characteristics of AMPs act through a membrane-specific mechanism, most likely through preferential interactions with PS. We provide the first evidence for the engagement of distinct viral envelope lipid constituents, establishing an avenue for specificity that may enable the development of a new family of therapeutics capable of averting the rapid development of resistance.
    MeSH term(s) Animals ; Humans ; Antiviral Agents/pharmacology ; Peptidomimetics/pharmacology ; Phosphatidylserines ; Liposomes ; Peptoids/pharmacology ; Peptoids/chemistry ; Zika Virus ; Zika Virus Infection
    Chemical Substances Antiviral Agents ; Peptidomimetics ; Phosphatidylserines ; Liposomes ; Peptoids
    Language English
    Publishing date 2023-08-02
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ISSN 2373-8227
    ISSN (online) 2373-8227
    DOI 10.1021/acsinfecdis.3c00063
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Ribavirin Induces Polyamine Depletion via Nucleotide Depletion to Limit Virus Replication.

    Tate, Patrick M / Mastrodomenico, Vincent / Mounce, Bryan C

    Cell reports

    2019  Volume 28, Issue 10, Page(s) 2620–2633.e4

    Abstract: Common antivirals include nucleoside or nucleotide analogs with base prodrugs. The antiviral ribavirin, a US Food and Drug Administration (FDA)-approved nucleoside antimetabolite, halts guanine production, mutagenizes viral genomes, and activates ... ...

    Abstract Common antivirals include nucleoside or nucleotide analogs with base prodrugs. The antiviral ribavirin, a US Food and Drug Administration (FDA)-approved nucleoside antimetabolite, halts guanine production, mutagenizes viral genomes, and activates interferon signaling. Here, we find that ribavirin induces spermidine-spermine N1-acetyltransferase (SAT1), a polyamine catabolic enzyme. Polyamines are small, positively charged molecules involved in cellular functions such as transcription and translation. Previous work showed that SAT1 activation and polyamine depletion interfere with RNA virus replication. We show ribavirin depletes polyamines via SAT1, in conjunction with its known mechanisms. SAT1 transcripts, protein, and activity are induced in a dose-dependent manner, which depletes polyamine levels and reduces viral titers. Inhibition of SAT1 activity, pharmacologically or genetically, reduces ribavirin's effectiveness against three virus infection models. Additionally, ribavirin-mediated polyamine depletion results from nucleotide pool depletion. These data demonstrate another mechanism of ribavirin that inform its clinical effectiveness, which may provide insight for improved therapies.
    MeSH term(s) Acetyltransferases/metabolism ; Cell Line, Tumor ; Guanosine/metabolism ; HEK293 Cells ; Humans ; Interferon Type I/metabolism ; Nucleotides/metabolism ; Polyamines/metabolism ; Ribavirin/chemistry ; Ribavirin/pharmacology ; Transcription, Genetic/drug effects ; Virus Replication/drug effects
    Chemical Substances Interferon Type I ; Nucleotides ; Polyamines ; Guanosine (12133JR80S) ; Ribavirin (49717AWG6K) ; Acetyltransferases (EC 2.3.1.-) ; diamine N-acetyltransferase (EC 2.3.1.57)
    Language English
    Publishing date 2019-09-01
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2019.07.099
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: A non-canonical sensing pathway mediates Plasmodium adaptation to amino acid deficiency.

    Marreiros, Inês M / Marques, Sofia / Parreira, Ana / Mastrodomenico, Vincent / Mounce, Bryan C / Harris, Chantal T / Kafsack, Björn F / Billker, Oliver / Zuzarte-Luís, Vanessa / Mota, Maria M

    Communications biology

    2023  Volume 6, Issue 1, Page(s) 205

    Abstract: Eukaryotes have canonical pathways for responding to amino acid (AA) availability. Under AA-limiting conditions, the TOR complex is repressed, whereas the sensor kinase GCN2 is activated. While these pathways have been highly conserved throughout ... ...

    Abstract Eukaryotes have canonical pathways for responding to amino acid (AA) availability. Under AA-limiting conditions, the TOR complex is repressed, whereas the sensor kinase GCN2 is activated. While these pathways have been highly conserved throughout evolution, malaria parasites are a rare exception. Despite auxotrophic for most AA, Plasmodium does not have either a TOR complex nor the GCN2-downstream transcription factors. While Ile starvation has been shown to trigger eIF2α phosphorylation and a hibernation-like response, the overall mechanisms mediating detection and response to AA fluctuation in the absence of such pathways has remained elusive. Here we show that Plasmodium parasites rely on an efficient sensing pathway to respond to AA fluctuations. A phenotypic screen of kinase knockout mutant parasites identified nek4, eIK1 and eIK2-the last two clustering with the eukaryotic eIF2α kinases-as critical for Plasmodium to sense and respond to distinct AA-limiting conditions. Such AA-sensing pathway is temporally regulated at distinct life cycle stages, allowing parasites to actively fine-tune replication and development in response to AA availability. Collectively, our data disclose a set of heterogeneous responses to AA depletion in malaria parasites, mediated by a complex mechanism that is critical for modulating parasite growth and survival.
    MeSH term(s) Amino Acids/deficiency ; eIF-2 Kinase/genetics ; eIF-2 Kinase/metabolism ; Phosphorylation ; Phosphotransferases/metabolism ; Plasmodium/enzymology ; Plasmodium/genetics
    Chemical Substances Amino Acids ; eIF-2 Kinase (EC 2.7.11.1) ; Phosphotransferases (EC 2.7.-)
    Language English
    Publishing date 2023-02-21
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ISSN 2399-3642
    ISSN (online) 2399-3642
    DOI 10.1038/s42003-023-04566-y
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Targeting Polyamines Inhibits Coronavirus Infection by Reducing Cellular Attachment and Entry.

    Firpo, Mason R / Mastrodomenico, Vincent / Hawkins, Grant M / Prot, Matthieu / Levillayer, Laura / Gallagher, Tom / Simon-Loriere, Etienne / Mounce, Bryan C

    ACS infectious diseases

    2020  Volume 7, Issue 6, Page(s) 1423–1432

    Abstract: Coronaviruses first garnered widespread attention in 2002 when the severe acute respiratory syndrome coronavirus (SARS-CoV) emerged from bats in China and rapidly spread in human populations. Since then, Middle East respiratory syndrome coronavirus (MERS- ...

    Abstract Coronaviruses first garnered widespread attention in 2002 when the severe acute respiratory syndrome coronavirus (SARS-CoV) emerged from bats in China and rapidly spread in human populations. Since then, Middle East respiratory syndrome coronavirus (MERS-CoV) emerged and still actively infects humans. The recent SARS-CoV-2 outbreak and the resulting disease (coronavirus disease 2019, COVID19) have rapidly and catastrophically spread and highlighted significant limitations to our ability to control and treat infection. Thus, a basic understanding of entry and replication mechanisms of coronaviruses is necessary to rationally evaluate potential antivirals. Here, we show that polyamines, small metabolites synthesized in human cells, facilitate coronavirus replication and the depletion of polyamines with FDA-approved molecules significantly reduces coronavirus replication. We find that diverse coronaviruses, including endemic and epidemic coronaviruses, exhibit reduced attachment and entry into polyamine-depleted cells. We further demonstrate that several molecules targeting the polyamine biosynthetic pathway are antiviral
    MeSH term(s) Antiviral Agents/pharmacology ; COVID-19 ; Humans ; Middle East Respiratory Syndrome Coronavirus ; Polyamines ; SARS-CoV-2
    Chemical Substances Antiviral Agents ; Polyamines
    Keywords covid19
    Language English
    Publishing date 2020-10-06
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ISSN 2373-8227
    ISSN (online) 2373-8227
    DOI 10.1021/acsinfecdis.0c00491
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Virion-Associated Polyamines Transmit with Bunyaviruses to Maintain Infectivity and Promote Entry.

    Mastrodomenico, Vincent / Esin, Jeremy J / Qazi, Shefah / Khomutov, Maxim A / Ivanov, Alexander V / Mukhopadhyay, Suchetana / Mounce, Bryan C

    ACS infectious diseases

    2020  Volume 6, Issue 9, Page(s) 2490–2501

    Abstract: Viruses require host cell metabolites to productively infect, and the mechanisms by which viruses usurp these molecules are diverse. One group of cellular metabolites important in virus infection is the polyamines, small positively charged molecules ... ...

    Abstract Viruses require host cell metabolites to productively infect, and the mechanisms by which viruses usurp these molecules are diverse. One group of cellular metabolites important in virus infection is the polyamines, small positively charged molecules involved in cell cycle, translation, and nucleic acid metabolism, among other cellular functions. Polyamines support replication of diverse viruses, and they are important for processes such as transcription, translation, and viral protein enzymatic activity. Rift Valley fever virus (RVFV) is a negative and ambisense RNA virus that requires polyamines to produce infectious particles. In polyamine depleted conditions, noninfectious particles are produced that interfere with virus replication and stimulate immune signaling. Here, we find that RVFV relies on virion-associated polyamines to maintain infectivity and enhance viral entry. We show that RVFV replication is facilitated by a limited set of polyamines and that spermidine and closely related molecules associate with purified virions and transmit from cell to cell during infection. Virion-associated spermidine maintains virion infectivity, as virions devoid of polyamines rapidly lose infectivity and are temperature sensitive. Further, virions without polyamines bind to cells but exhibit a defect in entry, requiring more acidic conditions than virions containing spermidine. These data highlight a unique role for polyamines, and spermidine particularly, to maintain virus infectivity. Further, these studies are the first to identify polyamines associated with RVFV virions. Targeting polyamines represents a promising antiviral strategy, and this work highlights a new mechanism by which we can inhibit virus replication through FDA-approved polyamine depleting pharmaceuticals.
    MeSH term(s) Animals ; Polyamines ; Rift Valley fever virus ; Viral Proteins ; Virion ; Virus Replication
    Chemical Substances Polyamines ; Viral Proteins
    Language English
    Publishing date 2020-08-12
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ISSN 2373-8227
    ISSN (online) 2373-8227
    DOI 10.1021/acsinfecdis.0c00402
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Tools for Rapid Genetic Engineering of Vibrio fischeri.

    Visick, Karen L / Hodge-Hanson, Kelsey M / Tischler, Alice H / Bennett, Allison K / Mastrodomenico, Vincent

    Applied and environmental microbiology

    2018  Volume 84, Issue 14

    Abstract: ... Vibrio ... ...

    Abstract Vibrio fischeri
    MeSH term(s) Aliivibrio fischeri/genetics ; Cellulose ; Cloning, Molecular ; DNA Nucleotidyltransferases ; Gene Deletion ; Gene Expression Regulation, Bacterial ; Genes, Bacterial/genetics ; Genetic Engineering/methods ; Mutagenesis, Insertional ; Mutation ; Promoter Regions, Genetic ; Quorum Sensing ; Symbiosis
    Chemical Substances Cellulose (9004-34-6) ; DNA Nucleotidyltransferases (EC 2.7.7.-) ; FLP recombinase (EC 2.7.7.-)
    Language English
    Publishing date 2018-07-02
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 223011-2
    ISSN 1098-5336 ; 0099-2240
    ISSN (online) 1098-5336
    ISSN 0099-2240
    DOI 10.1128/AEM.00850-18
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article: Targeting Polyamines Inhibits Coronavirus Infection by Reducing Cellular Attachment and Entry

    Firpo, Mason R / Mastrodomenico, Vincent / Hawkins, Grant M / Prot, Matthieu / Levillayer, Laura / Gallagher, Tom / Simon-Loriere, Etienne / Mounce, Bryan C

    ACS infect. dis

    Abstract: Coronaviruses first garnered widespread attention in 2002 when the severe acute respiratory syndrome coronavirus (SARS-CoV) emerged from bats in China and rapidly spread in human populations. Since then, Middle East respiratory syndrome coronavirus (MERS- ...

    Abstract Coronaviruses first garnered widespread attention in 2002 when the severe acute respiratory syndrome coronavirus (SARS-CoV) emerged from bats in China and rapidly spread in human populations. Since then, Middle East respiratory syndrome coronavirus (MERS-CoV) emerged and still actively infects humans. The recent SARS-CoV-2 outbreak and the resulting disease (coronavirus disease 2019, COVID19) have rapidly and catastrophically spread and highlighted significant limitations to our ability to control and treat infection. Thus, a basic understanding of entry and replication mechanisms of coronaviruses is necessary to rationally evaluate potential antivirals. Here, we show that polyamines, small metabolites synthesized in human cells, facilitate coronavirus replication and the depletion of polyamines with FDA-approved molecules significantly reduces coronavirus replication. We find that diverse coronaviruses, including endemic and epidemic coronaviruses, exhibit reduced attachment and entry into polyamine-depleted cells. We further demonstrate that several molecules targeting the polyamine biosynthetic pathway are antiviral in vitro. In sum, our data suggest that polyamines are critical to coronavirus replication and represent a highly promising drug target in the current and any future coronavirus outbreaks.
    Keywords covid19
    Publisher WHO
    Document type Article
    Note WHO #Covidence: #786550
    Database COVID19

    Kategorien

  10. Article ; Online: Polyamine Depletion Inhibits Bunyavirus Infection via Generation of Noninfectious Interfering Virions.

    Mastrodomenico, Vincent / Esin, Jeremy J / Graham, Marion L / Tate, Patrick M / Hawkins, Grant M / Sandler, Zachary J / Rademacher, David J / Kicmal, Thomas M / Dial, Courtney N / Mounce, Bryan C

    Journal of virology

    2019  Volume 93, Issue 14

    Abstract: Several host and viral processes contribute to forming infectious virions. Polyamines are small host molecules that play diverse roles in viral replication. We previously demonstrated that polyamines are crucial for RNA viruses; however, the mechanisms ... ...

    Abstract Several host and viral processes contribute to forming infectious virions. Polyamines are small host molecules that play diverse roles in viral replication. We previously demonstrated that polyamines are crucial for RNA viruses; however, the mechanisms by which polyamines function remain unknown. Here, we investigated the role of polyamines in the replication of the bunyaviruses Rift Valley fever virus (vaccine strain MP-12) and La Crosse virus (LACV). We found that polyamine depletion did not impact viral RNA or protein accumulation, despite significant decreases in titer. Viral particles demonstrated no change in morphology, size, or density. Thus, polyamine depletion promotes the formation of noninfectious particles. These particles interfere with virus replication and stimulate innate immune responses. We extended this phenotype to Zika virus; however, coxsackievirus did not similarly produce noninfectious particles. In sum, polyamine depletion results in the accumulation of noninfectious particles that interfere with replication and stimulate immune signaling, with important implications for targeting polyamines therapeutically, as well as for vaccine strategies.
    MeSH term(s) Biogenic Polyamines/immunology ; Bunyaviridae Infections/genetics ; Bunyaviridae Infections/immunology ; Bunyaviridae Infections/pathology ; Cell Line, Tumor ; Defective Viruses/physiology ; Encephalitis Virus, California/physiology ; Humans ; Rift Valley fever virus/physiology ; Virion/physiology ; Virus Replication/immunology
    Chemical Substances Biogenic Polyamines
    Language English
    Publishing date 2019-06-28
    Publishing country United States
    Document type Journal Article
    ZDB-ID 80174-4
    ISSN 1098-5514 ; 0022-538X
    ISSN (online) 1098-5514
    ISSN 0022-538X
    DOI 10.1128/JVI.00530-19
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top