LIVIVO - Das Suchportal für Lebenswissenschaften

switch to English language
Erweiterte Suche

Ihre letzten Suchen

  1. AU="Kalachikov, Sergey"
  2. AU="Das, Tilak"
  3. AU="Bessat, Cécile"
  4. AU="Galina Velikova"
  5. AU="Greene, Sharrell"
  6. AU="Chen, Kallie J"
  7. AU="Schwab, Jörg O."
  8. AU="Ke Chen"
  9. AU="Hewei Liang"
  10. AU="Abreu, Cristina"
  11. AU="Mamani Ortiz, Yercin"
  12. AU="Castro, Lucíola de Fátima Albuquerque Almeida Peixoto"
  13. AU="Šimůnek, Tomáš"
  14. AU="Ong, Lizhen"
  15. AU="Chai, Chaoqing"
  16. AU="Maheswaran Kesavan"
  17. AU="Mehta, Mrunal"
  18. AU=Paredes Sergio D
  19. AU=Ghosh Nilanjan AU=Ghosh Nilanjan
  20. AU="Hofmann, Alexander"
  21. AU="Radici, Marco"
  22. AU="Noro, Fabrizia"
  23. AU="Wang, Jianzhao"
  24. AU="Divya Jeyam"
  25. AU="Wolf, Lisette"
  26. AU="Marjanovic, Nemanja Despot"
  27. AU="Jitxin, Lim"

Suchergebnis

Treffer 1 - 10 von insgesamt 49

Suchoptionen

  1. Artikel: Identifying Structural Features of Nucleotide Analogues to Overcome SARS-CoV-2 Exonuclease Activity

    Wang, Xuanting / Tao, Chuanjuan / Morozova, Irina / Kalachikov, Sergey / Li, Xiaoxu / Kumar, Shiv / Russo, James J. / Ju, Jingyue

    Viruses. 2022 June 28, v. 14, no. 7

    2022  

    Abstract: With the recent global spread of new SARS-CoV-2 variants, there remains an urgent need to develop effective and variant-resistant oral drugs. Recently, we reported in vitro results validating the use of combination drugs targeting both the SARS-CoV-2 RNA- ...

    Abstract With the recent global spread of new SARS-CoV-2 variants, there remains an urgent need to develop effective and variant-resistant oral drugs. Recently, we reported in vitro results validating the use of combination drugs targeting both the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) and proofreading exonuclease (ExoN) as potential COVID-19 therapeutics. For the nucleotide analogues to be efficient SARS-CoV-2 inhibitors, two properties are required: efficient incorporation by RdRp and substantial resistance to excision by ExoN. Here, we have selected and evaluated nucleotide analogues with a variety of structural features for resistance to ExoN removal when they are attached at the 3′ RNA terminus. We found that dideoxynucleotides and other nucleotides lacking both 2′- and 3′-OH groups were most resistant to ExoN excision, whereas those possessing both 2′- and 3′-OH groups were efficiently removed. We also found that the 3′-OH group in the nucleotide analogues was more critical than the 2′-OH for excision by ExoN. Since the functionally important sequences in Nsp14/10 are highly conserved among all SARS-CoV-2 variants, these identified structural features of nucleotide analogues offer invaluable insights for designing effective RdRp inhibitors that can be simultaneously efficiently incorporated by the RdRp and substantially resist ExoN excision. Such newly developed RdRp terminators would be good candidates to evaluate their ability to inhibit SARS-CoV-2 in cell culture and animal models, perhaps combined with additional exonuclease inhibitors to increase their overall effectiveness.
    Schlagwörter COVID-19 infection ; RNA ; RNA-directed RNA polymerase ; Severe acute respiratory syndrome coronavirus 2 ; animals ; cell culture ; excision ; nucleotides
    Sprache Englisch
    Erscheinungsverlauf 2022-0628
    Erscheinungsort Multidisciplinary Digital Publishing Institute
    Dokumenttyp Artikel
    ZDB-ID 2516098-9
    ISSN 1999-4915
    ISSN 1999-4915
    DOI 10.3390/v14071413
    Datenquelle NAL Katalog (AGRICOLA)

    Zusatzmaterialien

    Kategorien

  2. Artikel ; Online: Identifying Structural Features of Nucleotide Analogues to Overcome SARS-CoV-2 Exonuclease Activity.

    Wang, Xuanting / Tao, Chuanjuan / Morozova, Irina / Kalachikov, Sergey / Li, Xiaoxu / Kumar, Shiv / Russo, James J / Ju, Jingyue

    Viruses

    2022  Band 14, Heft 7

    Abstract: With the recent global spread of new SARS-CoV-2 variants, there remains an urgent need to develop effective and variant-resistant oral drugs. Recently, we reported in vitro results validating the use of combination drugs targeting both the SARS-CoV-2 RNA- ...

    Abstract With the recent global spread of new SARS-CoV-2 variants, there remains an urgent need to develop effective and variant-resistant oral drugs. Recently, we reported in vitro results validating the use of combination drugs targeting both the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) and proofreading exonuclease (ExoN) as potential COVID-19 therapeutics. For the nucleotide analogues to be efficient SARS-CoV-2 inhibitors, two properties are required: efficient incorporation by RdRp and substantial resistance to excision by ExoN. Here, we have selected and evaluated nucleotide analogues with a variety of structural features for resistance to ExoN removal when they are attached at the 3' RNA terminus. We found that dideoxynucleotides and other nucleotides lacking both 2'- and 3'-OH groups were most resistant to ExoN excision, whereas those possessing both 2'- and 3'-OH groups were efficiently removed. We also found that the 3'-OH group in the nucleotide analogues was more critical than the 2'-OH for excision by ExoN. Since the functionally important sequences in Nsp14/10 are highly conserved among all SARS-CoV-2 variants, these identified structural features of nucleotide analogues offer invaluable insights for designing effective RdRp inhibitors that can be simultaneously efficiently incorporated by the RdRp and substantially resist ExoN excision. Such newly developed RdRp terminators would be good candidates to evaluate their ability to inhibit SARS-CoV-2 in cell culture and animal models, perhaps combined with additional exonuclease inhibitors to increase their overall effectiveness.
    Mesh-Begriff(e) Animals ; Antiviral Agents/therapeutic use ; COVID-19 ; Exonucleases ; Nucleotides/chemistry ; RNA, Viral/genetics ; SARS-CoV-2
    Chemische Substanzen Antiviral Agents ; Nucleotides ; RNA, Viral ; Exonucleases (EC 3.1.-)
    Sprache Englisch
    Erscheinungsdatum 2022-06-28
    Erscheinungsland Switzerland
    Dokumenttyp Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2516098-9
    ISSN 1999-4915 ; 1999-4915
    ISSN (online) 1999-4915
    ISSN 1999-4915
    DOI 10.3390/v14071413
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  3. Artikel: Functional Genomic Analysis of Amphetamine Sensitivity in

    Karam, Caline S / Williams, Brenna L / Morozova, Irina / Yuan, Qiaoping / Panarsky, Rony / Zhang, Yuchao / Hodgkinson, Colin A / Goldman, David / Kalachikov, Sergey / Javitch, Jonathan A

    Frontiers in psychiatry

    2022  Band 13, Seite(n) 831597

    Abstract: Abuse of psychostimulants, including amphetamines (AMPHs), is a major public health problem with profound psychiatric, medical, and psychosocial complications. The actions of these drugs at the dopamine transporter (DAT) play a critical role in their ... ...

    Abstract Abuse of psychostimulants, including amphetamines (AMPHs), is a major public health problem with profound psychiatric, medical, and psychosocial complications. The actions of these drugs at the dopamine transporter (DAT) play a critical role in their therapeutic efficacy as well as their liability for abuse and dependence. To date, however, the mechanisms that mediate these actions are not well-understood, and therapeutic interventions for AMPH abuse have been limited. Drug exposure can induce broad changes in gene expression that can contribute to neuroplasticity and effect long-lasting changes in neuronal function. Identifying genes and gene pathways perturbed by drug exposure is essential to our understanding of the molecular basis of drug addiction. In this study, we used
    Sprache Englisch
    Erscheinungsdatum 2022-02-18
    Erscheinungsland Switzerland
    Dokumenttyp Journal Article
    ZDB-ID 2564218-2
    ISSN 1664-0640
    ISSN 1664-0640
    DOI 10.3389/fpsyt.2022.831597
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  4. Artikel ; Online: Sofosbuvir terminated RNA is more resistant to SARS-CoV-2 proofreader than RNA terminated by Remdesivir.

    Jockusch, Steffen / Tao, Chuanjuan / Li, Xiaoxu / Chien, Minchen / Kumar, Shiv / Morozova, Irina / Kalachikov, Sergey / Russo, James J / Ju, Jingyue

    Scientific reports

    2020  Band 10, Heft 1, Seite(n) 16577

    Abstract: SARS-CoV-2 is responsible for COVID-19, resulting in the largest pandemic in over a hundred years. After examining the molecular structures and activities of hepatitis C viral inhibitors and comparing hepatitis C virus and coronavirus replication, we ... ...

    Abstract SARS-CoV-2 is responsible for COVID-19, resulting in the largest pandemic in over a hundred years. After examining the molecular structures and activities of hepatitis C viral inhibitors and comparing hepatitis C virus and coronavirus replication, we previously postulated that the FDA-approved hepatitis C drug EPCLUSA (Sofosbuvir/Velpatasvir) might inhibit SARS-CoV-2. We subsequently demonstrated that Sofosbuvir triphosphate is incorporated by the relatively low fidelity SARS-CoV and SARS-CoV-2 RNA-dependent RNA polymerases (RdRps), serving as an immediate polymerase reaction terminator, but not by a host-like high fidelity DNA polymerase. Other investigators have since demonstrated the ability of Sofosbuvir to inhibit SARS-CoV-2 replication in lung and brain cells; additionally, COVID-19 clinical trials with EPCLUSA and with Sofosbuvir plus Daclatasvir have been initiated in several countries. SARS-CoV-2 has an exonuclease-based proofreader to maintain the viral genome integrity. Any effective antiviral targeting the SARS-CoV-2 RdRp must display a certain level of resistance to this proofreading activity. We report here that Sofosbuvir terminated RNA resists removal by the exonuclease to a substantially higher extent than RNA terminated by Remdesivir, another drug being used as a COVID-19 therapeutic. These results offer a molecular basis supporting the current use of Sofosbuvir in combination with other drugs in COVID-19 clinical trials.
    Mesh-Begriff(e) Adenosine Monophosphate/analogs & derivatives ; Adenosine Monophosphate/chemistry ; Adenosine Monophosphate/pharmacology ; Adenosine Monophosphate/therapeutic use ; Alanine/analogs & derivatives ; Alanine/chemistry ; Alanine/pharmacology ; Alanine/therapeutic use ; Antiviral Agents/chemistry ; Antiviral Agents/pharmacology ; Antiviral Agents/therapeutic use ; Betacoronavirus/drug effects ; Betacoronavirus/enzymology ; COVID-19 ; Coronavirus Infections/drug therapy ; Coronavirus Infections/virology ; Coronavirus RNA-Dependent RNA Polymerase ; Drug Discovery/methods ; Drug Repositioning/methods ; Exonucleases/metabolism ; Hepacivirus/drug effects ; Hepacivirus/enzymology ; Hepatitis C/drug therapy ; Hepatitis C/virology ; Humans ; Pandemics ; Pneumonia, Viral/drug therapy ; Pneumonia, Viral/virology ; Prodrugs/pharmacology ; Prodrugs/therapeutic use ; RNA, Viral/chemistry ; RNA, Viral/drug effects ; RNA, Viral/metabolism ; RNA-Dependent RNA Polymerase/antagonists & inhibitors ; RNA-Dependent RNA Polymerase/metabolism ; SARS-CoV-2 ; Sofosbuvir/chemistry ; Sofosbuvir/pharmacology ; Sofosbuvir/therapeutic use ; Viral Nonstructural Proteins/antagonists & inhibitors ; Viral Nonstructural Proteins/metabolism ; Virus Replication/drug effects
    Chemische Substanzen Antiviral Agents ; Prodrugs ; RNA, Viral ; Viral Nonstructural Proteins ; remdesivir (3QKI37EEHE) ; Adenosine Monophosphate (415SHH325A) ; Coronavirus RNA-Dependent RNA Polymerase (EC 2.7.7.48) ; NSP12 protein, SARS-CoV-2 (EC 2.7.7.48) ; RNA-Dependent RNA Polymerase (EC 2.7.7.48) ; Exonucleases (EC 3.1.-) ; Alanine (OF5P57N2ZX) ; Sofosbuvir (WJ6CA3ZU8B)
    Schlagwörter covid19
    Sprache Englisch
    Erscheinungsdatum 2020-10-06
    Erscheinungsland England
    Dokumenttyp Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2615211-3
    ISSN 2045-2322 ; 2045-2322
    ISSN (online) 2045-2322
    ISSN 2045-2322
    DOI 10.1038/s41598-020-73641-9
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  5. Artikel ; Online: Charge-neutral morpholino microarrays for nucleic acid analysis.

    Qiao, Wanqiong / Kalachikov, Sergey / Liu, Yatao / Levicky, Rastislav

    Analytical biochemistry

    2012  Band 434, Heft 2, Seite(n) 207–214

    Abstract: A principal challenge in microarray experiments is to facilitate hybridization between probe strands on the array with complementary target strands from solution while suppressing any competing interactions that the probes and targets may experience. ... ...

    Abstract A principal challenge in microarray experiments is to facilitate hybridization between probe strands on the array with complementary target strands from solution while suppressing any competing interactions that the probes and targets may experience. Synthetic DNA analogs, whose hybridization to targets can exhibit qualitatively different dependence on experimental conditions than for nucleic acid probes, open up an attractive alternative for improving selectivity of array hybridization. Morpholinos (MOs), a class of uncharged DNA analogs, are investigated as microarray probes instead of DNA. MO microarrays were fabricated by contact printing of amino-modified probes onto aldehyde slides. In addition to covalent immobilization, MOs were found to efficiently immobilize through physical adsorption; such physically adsorbed probes could be removed by post-printing washes with surfactant solutions. Hybridization of double-stranded DNA targets to MO microarrays revealed a hybridization maximum at intermediate ionic strengths. The decline in hybridization at lower ionic strengths was attributed to an electrostatic barrier accumulated from hybridized DNA targets, whereas at higher ionic strengths it was attributed to stabilization of target secondary structure in solution. These trends, which illustrate ionic strength tuning of forming on-array relative to solution secondary structure, were supported by a stability analysis of MO/DNA and DNA/DNA duplexes in solution.
    Mesh-Begriff(e) Chemistry Techniques, Analytical/methods ; DNA/analysis ; DNA/chemistry ; Molecular Probes/chemistry ; Morpholinos/chemistry ; Oligonucleotide Array Sequence Analysis ; Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ; Surface Properties
    Chemische Substanzen Molecular Probes ; Morpholinos ; DNA (9007-49-2)
    Sprache Englisch
    Erscheinungsdatum 2012-12-12
    Erscheinungsland United States
    Dokumenttyp Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 1110-1
    ISSN 1096-0309 ; 0003-2697
    ISSN (online) 1096-0309
    ISSN 0003-2697
    DOI 10.1016/j.ab.2012.12.001
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  6. Artikel ; Online: Sofosbuvir Terminated RNA is More Resistant to SARS-CoV-2 Proofreader than RNA Terminated by Remdesivir

    Jockusch, Steffen / Tao, Chuanjuan / Li, Xiaoxu / Chien, Minchen / Kumar, Shiv / Morozova, Irina / Kalachikov, Sergey / Russo, James J. / Ju, Jingyue

    bioRxiv

    Abstract: SARS-CoV-2 is responsible for COVID-19, resulting in the largest pandemic in over a hundred years. After examining the molecular structures and activities of hepatitis C viral inhibitors and comparing hepatitis C virus and coronavirus replication, we ... ...

    Abstract SARS-CoV-2 is responsible for COVID-19, resulting in the largest pandemic in over a hundred years. After examining the molecular structures and activities of hepatitis C viral inhibitors and comparing hepatitis C virus and coronavirus replication, we previously postulated that the FDA-approved hepatitis C drug EPCLUSA (Sofosbuvir/Velpatasvir) might inhibit SARS-CoV-2. We subsequently demonstrated that Sofosbuvir triphosphate is incorporated by the relatively low fidelity SARS-CoV and SARS-CoV-2 RNA-dependent RNA polymerases (RdRps), serving as an immediate polymerase reaction terminator, but not by a host-like high fidelity DNA polymerase. Other investigators have since demonstrated the ability of Sofosbuvir to inhibit SARS-CoV-2 replication in lung and brain cells; additionally, COVID-19 clinical trials with EPCLUSA and with Sofosbuvir plus Daclatasvir have been initiated in several countries. SARS-CoV-2 has an exonuclease-based proofreader to maintain the viral genome integrity. Any effective antiviral targeting the SARS-CoV-2 RdRp must display a certain level of resistance to this proofreading activity. We report here that Sofosbuvir terminated RNA resists removal by the exonuclease to a substantially higher extent than RNA terminated by Remdesivir, another drug being used as a COVID-19 therapeutic. These results offer a molecular basis supporting the current use of Sofosbuvir in combination with other drugs in COVID-19 clinical trials.
    Schlagwörter covid19
    Sprache Englisch
    Erscheinungsdatum 2020-08-10
    Verlag Cold Spring Harbor Laboratory
    Dokumenttyp Artikel ; Online
    DOI 10.1101/2020.08.07.242156
    Datenquelle COVID19

    Kategorien

  7. Artikel ; Online: Sofosbuvir Terminated RNA is More Resistant to SARS-CoV-2 Proofreader than RNA Terminated by Remdesivir

    Jockusch, Steffen / Tao, Chuanjuan / Li, Xiaoxu / Chien, Minchen / Kumar, Shiv / Morozova, Irina / Kalachikov, Sergey / Russo, James J. / Ju, Jingyue

    bioRxiv

    Abstract: SARS-CoV-2 is responsible for COVID-19, resulting in the largest pandemic in over a hundred years. After examining the molecular structures and activities of hepatitis C viral inhibitors and comparing hepatitis C virus and coronavirus replication, we ... ...

    Abstract SARS-CoV-2 is responsible for COVID-19, resulting in the largest pandemic in over a hundred years. After examining the molecular structures and activities of hepatitis C viral inhibitors and comparing hepatitis C virus and coronavirus replication, we previously postulated that the FDA-approved hepatitis C drug EPCLUSA (Sofosbuvir/Velpatasvir) might inhibit SARS-CoV-2.1 We subsequently demonstrated that Sofosbuvir triphosphate is incorporated by the relatively low fidelity SARS-CoV and SARS-CoV-2 RNA-dependent RNA polymerases (RdRps), serving as an immediate polymerase reaction terminator, but not by a host-like high fidelity DNA polymerase.2,3 Other investigators have since demonstrated the ability of Sofosbuvir to inhibit SARS-CoV-2 replication in lung and brain cells;4,5 additionally, COVID-19 clinical trials with EPCLUSA6 and with Sofosbuvir plus Daclatasvir7 have been initiated in several countries. SARS-CoV-2 has an exonuclease-based proofreader to maintain the viral genome integrity.8 Any effective antiviral targeting the SARS-CoV-2 RdRp must display a certain level of resistance to this proofreading activity. We report here that Sofosbuvir terminated RNA resists removal by the exonuclease to a substantially higher extent than RNA terminated by Remdesivir, another drug being used as a COVID-19 therapeutic. These results offer a molecular basis supporting the current use of Sofosbuvir in combination with other drugs in COVID-19 clinical trials.
    Schlagwörter covid19
    Verlag BioRxiv; WHO
    Dokumenttyp Artikel ; Online
    DOI 10.1101/2020.08.07.242156
    Datenquelle COVID19

    Kategorien

  8. Artikel ; Online: Nucleotide analogues as inhibitors of SARS-CoV Polymerase.

    Ju, Jingyue / Li, Xiaoxu / Kumar, Shiv / Jockusch, Steffen / Chien, Minchen / Tao, Chuanjuan / Morozova, Irina / Kalachikov, Sergey / Kirchdoerfer, Robert N / Russo, James J

    Pharmacology research & perspectives

    2020  Band 8, Heft 6, Seite(n) e00674

    Abstract: SARS-CoV-2, a member of the coronavirus family, has caused a global public health emergency. Based on our analysis of hepatitis C virus and coronavirus replication, and the molecular structures and activities of viral inhibitors, we previously reasoned ... ...

    Abstract SARS-CoV-2, a member of the coronavirus family, has caused a global public health emergency. Based on our analysis of hepatitis C virus and coronavirus replication, and the molecular structures and activities of viral inhibitors, we previously reasoned that the FDA-approved hepatitis C drug EPCLUSA (Sofosbuvir/Velpatasvir) should inhibit coronaviruses, including SARS-CoV-2. Here, using model polymerase extension experiments, we demonstrate that the active triphosphate form of Sofosbuvir is incorporated by low-fidelity polymerases and SARS-CoV RNA-dependent RNA polymerase (RdRp), and blocks further incorporation by these polymerases; the active triphosphate form of Sofosbuvir is not incorporated by a host-like high-fidelity DNA polymerase. Using the same molecular insight, we selected 3'-fluoro-3'-deoxythymidine triphosphate and 3'-azido-3'-deoxythymidine triphosphate, which are the active forms of two other anti-viral agents, Alovudine and AZT (an FDA-approved HIV/AIDS drug) for evaluation as inhibitors of SARS-CoV RdRp. We demonstrate the ability of two of these HIV reverse transcriptase inhibitors to be incorporated by SARS-CoV RdRp where they also terminate further polymerase extension. Given the 98% amino acid similarity of the SARS-CoV and SARS-CoV-2 RdRps, we expect these nucleotide analogues would also inhibit the SARS-CoV-2 polymerase. These results offer guidance to further modify these nucleotide analogues to generate more potent broad-spectrum anti-coronavirus agents.
    Mesh-Begriff(e) Antiviral Agents/pharmacology ; Betacoronavirus/drug effects ; Betacoronavirus/enzymology ; COVID-19 ; Carbamates/pharmacology ; Coronavirus Infections/drug therapy ; Coronavirus Infections/virology ; Dideoxynucleotides/pharmacology ; Drug Combinations ; Heterocyclic Compounds, 4 or More Rings/pharmacology ; Humans ; Pandemics ; Pneumonia, Viral/drug therapy ; Pneumonia, Viral/virology ; RNA-Dependent RNA Polymerase/antagonists & inhibitors ; SARS-CoV-2 ; Sofosbuvir/pharmacology ; Thymine Nucleotides/pharmacology ; Zidovudine/analogs & derivatives ; Zidovudine/pharmacology
    Chemische Substanzen Antiviral Agents ; Carbamates ; Dideoxynucleotides ; Drug Combinations ; Heterocyclic Compounds, 4 or More Rings ; Thymine Nucleotides ; sofosbuvir-velpatasvir drug combination ; 3'-fluorothymidine-5'-triphosphate (40026-13-9) ; Zidovudine (4B9XT59T7S) ; zidovudine triphosphate (6RGF96R053) ; RNA-Dependent RNA Polymerase (EC 2.7.7.48) ; Sofosbuvir (WJ6CA3ZU8B)
    Schlagwörter covid19
    Sprache Englisch
    Erscheinungsdatum 2020-10-02
    Erscheinungsland United States
    Dokumenttyp Journal Article
    ZDB-ID 2740389-0
    ISSN 2052-1707 ; 2052-1707
    ISSN (online) 2052-1707
    ISSN 2052-1707
    DOI 10.1002/prp2.674
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  9. Artikel ; Online: Contribution of the Opioid System to the Antidepressant Effects of Fluoxetine.

    Carazo-Arias, Elena / Nguyen, Phi T / Kass, Marley / Jee, Hyun Jung / Nautiyal, Katherine M / Magalong, Valerie / Coie, Lilian / Andreu, Valentine / Gergues, Mark M / Khalil, Huzefa / Akil, Huda / Arcego, Danusa Mar / Meaney, Michael / Anacker, Christoph / Samuels, Benjamin A / Pintar, John E / Morozova, Irina / Kalachikov, Sergey / Hen, Rene

    Biological psychiatry

    2022  Band 92, Heft 12, Seite(n) 952–963

    Abstract: Background: Selective serotonin reuptake inhibitors such as fluoxetine have a limited treatment efficacy. The mechanism by which some patients respond to fluoxetine while others do not remains poorly understood, limiting treatment effectiveness. We have ...

    Abstract Background: Selective serotonin reuptake inhibitors such as fluoxetine have a limited treatment efficacy. The mechanism by which some patients respond to fluoxetine while others do not remains poorly understood, limiting treatment effectiveness. We have found the opioid system to be involved in the responsiveness to fluoxetine treatment in a mouse model for anxiety- and depressive-like behavior.
    Methods: We analyzed gene expression changes in the dentate gyrus of mice chronically treated with corticosterone and fluoxetine. After identifying a subset of genes of interest, we studied their expression patterns in relation to treatment responsiveness. We further characterized their expression through in situ hybridization and the analysis of a single-cell RNA sequencing dataset. Finally, we behaviorally tested mu and delta opioid receptor knockout mice in the novelty suppressed feeding test and the forced swim test after chronic corticosterone and fluoxetine treatment.
    Results: Chronic fluoxetine treatment upregulates proenkephalin expression in the dentate gyrus, and this upregulation is associated with treatment responsiveness. The expression of several of the most significantly upregulated genes, including proenkephalin, is localized to an anatomically and transcriptionally specialized subgroup of mature granule cells in the dentate gyrus. We have also found that the delta opioid receptor contributes to some, but not all, of the behavioral effects of fluoxetine.
    Conclusions: These data indicate that the opioid system is involved in the antidepressant effects of fluoxetine, and this effect may be mediated through the upregulation of proenkephalin in a subpopulation of mature granule cells.
    Mesh-Begriff(e) Mice ; Animals ; Fluoxetine/pharmacology ; Analgesics, Opioid/pharmacology ; Corticosterone ; Receptors, Opioid, delta/genetics ; Antidepressive Agents/pharmacology ; Antidepressive Agents/therapeutic use ; Selective Serotonin Reuptake Inhibitors/pharmacology ; Mice, Knockout
    Chemische Substanzen Fluoxetine (01K63SUP8D) ; Analgesics, Opioid ; Corticosterone (W980KJ009P) ; Receptors, Opioid, delta ; Antidepressive Agents ; Serotonin Uptake Inhibitors
    Sprache Englisch
    Erscheinungsdatum 2022-06-14
    Erscheinungsland United States
    Dokumenttyp 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.
    ZDB-ID 209434-4
    ISSN 1873-2402 ; 0006-3223
    ISSN (online) 1873-2402
    ISSN 0006-3223
    DOI 10.1016/j.biopsych.2022.05.030
    Datenquelle MEDical Literature Analysis and Retrieval System OnLINE

    Zusatzmaterialien

    Kategorien

  10. Artikel ; Online: Nucleotide Analogues as Inhibitors of SARS-CoV Polymerase

    Ju, Jingyue / Li, Xiaoxu / Kumar, Shiv / Jockusch, Steffen / Chien, Minchen / Tao, Chuanjuan / Morozova, Irina / Kalachikov, Sergey / Kirchdoerfer, Robert N. / Russo, James J.

    bioRxiv

    Abstract: SARS-CoV-2, a member of the coronavirus family, has caused a global public health emergency.1 Based on our analysis of hepatitis C virus and coronavirus replication, and the molecular structures and activities of viral inhibitors, we previously reasoned ... ...

    Abstract SARS-CoV-2, a member of the coronavirus family, has caused a global public health emergency.1 Based on our analysis of hepatitis C virus and coronavirus replication, and the molecular structures and activities of viral inhibitors, we previously reasoned that the FDA-approved heptatitis C drug EPCLUSA (Sofosbuvir/Velpatasvir) should inhibit coronaviruses, including SARS-CoV-2.2 Here, using model polymerase extension experiments, we demonstrate that the activated triphosphate form of Sofosbuvir is incorporated by low-fidelity polymerases and SARS-CoV RNA-dependent RNA polymerase (RdRp), and blocks further incorporation by these polymerases; the activated triphosphate form of Sofosbuvir is not incorporated by a host-like high-fidelity DNA polymerase. Using the same molecular insight, we selected two other anti-viral agents, Alovudine and AZT (an FDA approved HIV/AIDS drug) for evaluation as inhibitors of SARS-CoV RdRp. We demonstrate the ability of two HIV reverse transcriptase inhibitors, 3’-fluoro-3’-deoxythymidine triphosphate and 3’-azido-3’-deoxythymidine triphosphate (the active triphosphate forms of Alovudine and AZT), to be incorporated by SARS-CoV RdRp where they also terminate further polymerase extension. Given the 98% amino acid similarity of the SARS-CoV and SARS-CoV-2 RdRps, we expect these nucleotide analogues would also inhibit the SARS-CoV-2 polymerase. These results offer guidance to further modify these nucleotide analogues to generate more potent broad-spectrum anti-coronavirus agents.
    Schlagwörter covid19
    Verlag BioRxiv; WHO
    Dokumenttyp Artikel ; Online
    DOI 10.1101/2020.03.12.989186
    Datenquelle COVID19

    Kategorien

Zum Seitenanfang