Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Priya Luthra is active.

Publication


Featured researches published by Priya Luthra.


Science | 2015

Infectious disease. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency

Michael J. Ciancanelli; Sarah X.L. Huang; Priya Luthra; Hannah Garner; Yuval Itan; Stefano Volpi; Fabien G. Lafaille; Céline Trouillet; Mirco Schmolke; Randy A. Albrecht; Elisabeth Israelsson; Hye Kyung Lim; Melina Casadio; Tamar Hermesh; Lazaro Lorenzo; Lawrence W. Leung; Vincent Pedergnana; Bertrand Boisson; Satoshi Okada; Capucine Picard; Benedicte Ringuier; Françoise Troussier; Damien Chaussabel; Laurent Abel; Isabelle Pellier; Luigi D. Notarangelo; Adolfo García-Sastre; Christopher F. Basler; Frederic Geissmann; Shen-Ying Zhang

A genetic cause for severe influenza Although chicken soup and plenty of rest get most kids through an influenza virus infection, some require hospitalization. Ciancanelli et al. report on one child who suffered severely from influenza because of null mutations in the gene for transcription factor IRF7. Cells isolated from this patient could not make enough secreted antiviral proteins, called interferons, to halt viral replication. The requirement for IRF7 seems quite specific, because this patient recovers normally from other common childhood viral infections. Science, this issue p. 448 A mutation that reduces antiviral interferons underlies certain cases of severe influenza in children. Severe influenza disease strikes otherwise healthy children and remains unexplained. We report compound heterozygous null mutations in IRF7, which encodes the transcription factor interferon regulatory factor 7, in an otherwise healthy child who suffered life-threatening influenza during primary infection. In response to influenza virus, the patient’s leukocytes and plasmacytoid dendritic cells produced very little type I and III interferons (IFNs). Moreover, the patient’s dermal fibroblasts and induced pluripotent stem cell (iPSC)–derived pulmonary epithelial cells produced reduced amounts of type I IFN and displayed increased influenza virus replication. These findings suggest that IRF7-dependent amplification of type I and III IFNs is required for protection against primary infection by influenza virus in humans. They also show that severe influenza may result from single-gene inborn errors of immunity.


Cell Host & Microbe | 2013

Mutual Antagonism between the Ebola Virus VP35 Protein and the RIG-I Activator PACT Determines Infection Outcome

Priya Luthra; Parameshwaran Ramanan; Chad E. Mire; Carla Weisend; Yoshimi Tsuda; Benjamin Yen; Gai Liu; Daisy W. Leung; Thomas W. Geisbert; Hideki Ebihara; Gaya K. Amarasinghe; Christopher F. Basler

The cytoplasmic pattern recognition receptor RIG-I is activated by viral RNA and induces type I IFN responses to control viral replication. The cellular dsRNA binding protein PACT can also activate RIG-I. To counteract innate antiviral responses, some viruses, including Ebola virus (EBOV), encode proteins that antagonize RIG-I signaling. Here, we show that EBOV VP35 inhibits PACT-induced RIG-I ATPase activity in a dose-dependent manner. The interaction of PACT with RIG-I is disrupted by wild-type VP35, but not by VP35 mutants that are unable to bind PACT. In addition, PACT-VP35 interaction impairs the association between VP35 and the viral polymerase, thereby diminishing viral RNA synthesis and modulating EBOV replication. PACT-deficient cells are defective in IFN induction and are insensitive to VP35 function. These data support a model in which the VP35-PACT interaction is mutually antagonistic and plays a fundamental role in determining the outcome of EBOV infection.


Biochemistry | 2013

Development of RNA Aptamers Targeting Ebola Virus VP35

Jennifer M. Binning; Tianjiao Wang; Priya Luthra; Reed S. Shabman; Dominika Borek; Gai Liu; Wei Xu; Daisy W. Leung; Christopher F. Basler; Gaya K. Amarasinghe

Viral protein 35 (VP35), encoded by filoviruses, is a multifunctional dsRNA binding protein that plays important roles in viral replication, innate immune evasion, and pathogenesis. The multifunctional nature of these proteins also presents opportunities to develop countermeasures that target distinct functional regions. However, functional validation and the establishment of therapeutic approaches toward such multifunctional proteins, particularly for nonenzymatic targets, are often challenging. Our previous work on filoviral VP35 proteins defined conserved basic residues located within its C-terminal dsRNA binding interferon (IFN) inhibitory domain (IID) as important for VP35 mediated IFN antagonism and viral polymerase cofactor functions. In the current study, we used a combination of structural and functional data to determine regions of Ebola virus (EBOV) VP35 (eVP35) to target for aptamer selection using SELEX. Select aptamers, representing, two distinct classes, were further characterized based on their interaction properties to eVP35 IID. These results revealed that these aptamers bind to distinct regions of eVP35 IID with high affinity (10-50 nM) and specificity. These aptamers can compete with dsRNA for binding to eVP35 and disrupt the eVP35-nucleoprotein (NP) interaction. Consistent with the ability to antagonize the eVP35-NP interaction, select aptamers can inhibit the function of the EBOV polymerase complex reconstituted by the expression of select viral proteins. Taken together, our results support the identification of two aptamers that bind filoviral VP35 proteins with high affinity and specificity and have the capacity to potentially function as filoviral VP35 protein inhibitors.


Journal of Molecular Biology | 2014

In Silico Derived Small Molecules Bind the Filovirus VP35 Protein and Inhibit Its Polymerase Cofactor Activity

Craig S. Brown; Michael S. Lee; Daisy W. Leung; Tianjiao Wang; Wei Xu; Priya Luthra; Manu Anantpadma; Reed S. Shabman; Lisa Melito; Karen S. MacMillan; Dominika Borek; Zbyszek Otwinowski; Parameshwaran Ramanan; Alisha Stubbs; Dayna S. Peterson; Jennifer M. Binning; Marco Tonelli; Mark A. Olson; Robert A. Davey; Joseph M. Ready; Christopher F. Basler; Gaya K. Amarasinghe

The Ebola virus (EBOV) genome only encodes a single viral polypeptide with enzymatic activity, the viral large (L) RNA-dependent RNA polymerase protein. However, currently, there is limited information about the L protein, which has hampered the development of antivirals. Therefore, antifiloviral therapeutic efforts must include additional targets such as protein-protein interfaces. Viral protein 35 (VP35) is multifunctional and plays important roles in viral pathogenesis, including viral mRNA synthesis and replication of the negative-sense RNA viral genome. Previous studies revealed that mutation of key basic residues within the VP35 interferon inhibitory domain (IID) results in significant EBOV attenuation, both in vitro and in vivo. In the current study, we use an experimental pipeline that includes structure-based in silico screening and biochemical and structural characterization, along with medicinal chemistry, to identify and characterize small molecules that target a binding pocket within VP35. NMR mapping experiments and high-resolution x-ray crystal structures show that select small molecules bind to a region of VP35 IID that is important for replication complex formation through interactions with the viral nucleoprotein (NP). We also tested select compounds for their ability to inhibit VP35 IID-NP interactions in vitro as well as VP35 function in a minigenome assay and EBOV replication. These results confirm the ability of compounds identified in this study to inhibit VP35-NP interactions in vitro and to impair viral replication in cell-based assays. These studies provide an initial framework to guide development of antifiloviral compounds against filoviral VP35 proteins.


Nature microbiology | 2017

Dengue virus NS2B protein targets cGAS for degradation and prevents mitochondrial DNA sensing during infection

Sebastian Aguirre; Priya Luthra; Maria Teresa Sánchez-Aparicio; Ana M. Maestre; Jenish R. Patel; Francise Lamothe; Anthony C. Fredericks; Shashank Tripathi; Tongtong Zhu; Jessica Pintado-Silva; Laurence G. Webb; Dabeiba Bernal-Rubio; Alexander Solovyov; Benjamin D. Greenbaum; Viviana Simon; Christopher F. Basler; Lubbertus C. F. Mulder; Adolfo García-Sastre; Ana Fernandez-Sesma

During the last few decades, the global incidence of dengue virus (DENV) has increased dramatically, and it is now endemic in more than 100 countries. To establish a productive infection in humans, DENV uses different strategies to inhibit or avoid the host innate immune system. Several DENV proteins have been shown to strategically target crucial components of the type I interferon system. Here, we report that the DENV NS2B protease cofactor targets the DNA sensor cyclic GMP-AMP synthase (cGAS) for lysosomal degradation to avoid the detection of mitochondrial DNA during infection. Such degradation subsequently results in the inhibition of type I interferon production in the infected cell. Our data demonstrate a mechanism by which cGAS senses cellular damage upon DENV infection.


Journal of Virology | 2015

Ebola Virus VP35 Interaction with Dynein LC8 Regulates Viral RNA Synthesis

Priya Luthra; David S. Jordan; Daisy W. Leung; Gaya K. Amarasinghe; Christopher F. Basler

ABSTRACT Ebola virus VP35 inhibits alpha/beta interferon production and functions as a viral polymerase cofactor. Previously, the 8-kDa cytoplasmic dynein light chain (LC8) was demonstrated to interact with VP35, but the functional consequences were unclear. Here we demonstrate that the interaction is direct and of high affinity and that binding stabilizes the VP35 N-terminal oligomerization domain and enhances viral RNA synthesis. Mutational analysis demonstrates that VP35 interaction is required for the functional effects of LC8.


Mbio | 2017

Topoisomerase II inhibitors induce DNA damage-dependent interferon responses circumventing ebola virus immune evasion

Priya Luthra; Sebastian Aguirre; Benjamin C. Yen; Colette Pietzsch; Maria Teresa Sánchez-Aparicio; Bersabeh Tigabu; Lorraine K. Morlock; Adolfo García-Sastre; Daisy W. Leung; Noelle S. Williams; Ana Fernandez-Sesma; Alexander Bukreyev; Christopher F. Basler

ABSTRACT Ebola virus (EBOV) protein VP35 inhibits production of interferon alpha/beta (IFN) by blocking RIG-I-like receptor signaling pathways, thereby promoting virus replication and pathogenesis. A high-throughput screening assay, developed to identify compounds that either inhibit or bypass VP35 IFN-antagonist function, identified five DNA intercalators as reproducible hits from a library of bioactive compounds. Four, including doxorubicin and daunorubicin, are anthracycline antibiotics that inhibit topoisomerase II and are used clinically as chemotherapeutic drugs. These compounds were demonstrated to induce IFN responses in an ATM kinase-dependent manner and to also trigger the DNA-sensing cGAS-STING pathway of IFN induction. These compounds also suppress EBOV replication in vitro and induce IFN in the presence of IFN-antagonist proteins from multiple negative-sense RNA viruses. These findings provide new insights into signaling pathways activated by important chemotherapy drugs and identify a novel therapeutic approach for IFN induction that may be exploited to inhibit RNA virus replication. IMPORTANCE Ebola virus and other emerging RNA viruses are significant but unpredictable public health threats. Therapeutic approaches with broad-spectrum activity could provide an attractive response to such infections. We describe a novel assay that can identify small molecules that overcome Ebola virus-encoded innate immune evasion mechanisms. This assay identified as hits cancer chemotherapeutic drugs, including doxorubicin. Follow-up studies provide new insight into how doxorubicin induces interferon (IFN) responses, revealing activation of both the DNA damage response kinase ATM and the DNA sensor cGAS and its partner signaling protein STING. The studies further demonstrate that the ATM and cGAS-STING pathways of IFN induction are a point of vulnerability not only for Ebola virus but for other RNA viruses as well, because viral innate immune antagonists consistently fail to block these signals. These studies thereby define a novel avenue for therapeutic intervention against emerging RNA viruses. Ebola virus and other emerging RNA viruses are significant but unpredictable public health threats. Therapeutic approaches with broad-spectrum activity could provide an attractive response to such infections. We describe a novel assay that can identify small molecules that overcome Ebola virus-encoded innate immune evasion mechanisms. This assay identified as hits cancer chemotherapeutic drugs, including doxorubicin. Follow-up studies provide new insight into how doxorubicin induces interferon (IFN) responses, revealing activation of both the DNA damage response kinase ATM and the DNA sensor cGAS and its partner signaling protein STING. The studies further demonstrate that the ATM and cGAS-STING pathways of IFN induction are a point of vulnerability not only for Ebola virus but for other RNA viruses as well, because viral innate immune antagonists consistently fail to block these signals. These studies thereby define a novel avenue for therapeutic intervention against emerging RNA viruses.


Journal of Virology | 2017

The host E3-Ubiquitin ligase TRIM6 ubiquitinates the Ebola virus VP35 protein and promotes virus replication

Preeti Bharaj; Colm Atkins; Priya Luthra; Maria Isabel Giraldo; Brian E. Dawes; Lisa Miorin; Jeffrey R. Johnson; Nevan J. Krogan; Christopher F. Basler; Alexander N. Freiberg; Ricardo Rajsbaum

ABSTRACT Ebola virus (EBOV), a member of the Filoviridae family, is a highly pathogenic virus that causes severe hemorrhagic fever in humans and is responsible for epidemics throughout sub-Saharan, central, and West Africa. The EBOV genome encodes VP35, an important viral protein involved in virus replication by acting as an essential cofactor of the viral polymerase as well as a potent antagonist of the host antiviral type I interferon (IFN-I) system. By using mass spectrometry analysis and coimmunoprecipitation assays, we show here that VP35 is ubiquitinated on lysine 309 (K309), a residue located on its IFN antagonist domain. We also found that VP35 interacts with TRIM6, a member of the E3-ubiquitin ligase tripartite motif (TRIM) family. We recently reported that TRIM6 promotes the synthesis of unanchored K48-linked polyubiquitin chains, which are not covalently attached to any protein, to induce efficient antiviral IFN-I-mediated responses. Consistent with this notion, VP35 also associated noncovalently with polyubiquitin chains and inhibited TRIM6-mediated IFN-I induction. Intriguingly, we also found that TRIM6 enhances EBOV polymerase activity in a minigenome assay and TRIM6 knockout cells have reduced replication of infectious EBOV, suggesting that VP35 hijacks TRIM6 to promote EBOV replication through ubiquitination. Our work provides evidence that TRIM6 is an important host cellular factor that promotes EBOV replication, and future studies will focus on whether TRIM6 could be targeted for therapeutic intervention against EBOV infection. IMPORTANCE EBOV belongs to a family of highly pathogenic viruses that cause severe hemorrhagic fever in humans and other mammals with high mortality rates (40 to 90%). Because of its high pathogenicity and lack of licensed antivirals and vaccines, EBOV is listed as a tier 1 select-agent risk group 4 pathogen. An important mechanism for the severity of EBOV infection is its suppression of innate immune responses. The EBOV VP35 protein contributes to pathogenesis, because it serves as an essential cofactor of the viral polymerase as well as a potent antagonist of innate immunity. However, how VP35 function is regulated by host cellular factors is poorly understood. Here, we report that the host E3-ubiquitin ligase TRIM6 promotes VP35 ubiquitination and is important for efficient virus replication. Therefore, our study identifies a new host factor, TRIM6, as a potential target in the development of antiviral drugs against EBOV.


Antiviral Research | 2018

A high throughput screen identifies benzoquinoline compounds as inhibitors of Ebola virus replication

Priya Luthra; Jue Liang; Colette Pietzsch; Sudip Khadka; Megan R. Edwards; Shuguang Wei; Sampriti De; Bruce A. Posner; Alexander Bukreyev; Joseph M. Ready; Christopher F. Basler

Abstract Ebola virus (EBOV) is an enveloped negative‐sense, single‐stranded RNA virus of the filovirus family that causes severe disease in humans. Approved therapies for EBOV disease are lacking. EBOV RNA synthesis is carried out by a virus‐encoded complex with RNA‐dependent RNA polymerase activity that is required for viral propagation. This complex and its activities are therefore potential antiviral targets. To identify potential lead inhibitors of EBOV RNA synthesis, a library of small molecule compounds was screened against a previously established assay of EBOV RNA synthesis, the EBOV minigenome assay (MGA), in 384 well microplate format. The screen identified 56 hits that inhibited EBOV MGA activity by more than 70% while exhibiting less than 20% cell cytotoxicity. Inhibitory chemical scaffolds included angelicin derivatives, derivatives of the antiviral compound GSK983 and benzoquinolines. Structure‐activity relationship (SAR) studies of the benzoquinoline scaffold produced ˜50 analogs and led to identification of an optimized compound, SW456, with a submicromolar IC50 in the EBOV MGA and antiviral activity against infectious EBOV in cell culture. The compound was also active against a MGA for another deadly filovirus, Marburg virus. It also exhibited antiviral activity towards a negative‐sense RNA virus from the rhabdovirus family, vesicular stomatitis virus, and a positive‐sense RNA virus, Zika virus. Overall, these data demonstrate the potential of the EBOV MGA to identify anti‐EBOV compounds and identifies the benzoquinoline series as a broad‐spectrum antiviral lead. HighlightsAn Ebola virus minigenome assay and a novel counterscreen were used to screen a 200,000 compound library.The screen identified a benzoquinoline scaffold as an inhibitor of Ebola virus RNA synthesis.SAR studies indicate that the full three‐ring structure of the heterocycle is essential for inhibition of the minigenome assay.An optimized compound, SW456, exhibits broad spectrum activity against filoviruses, vesicular stomatitis virus and Zika virus.


PLOS ONE | 2017

The Ebola virus VP35 protein binds viral immunostimulatory and host RNAs identified through deep sequencing

Kari A. Dilley; Alexander A. Voorhies; Priya Luthra; Vinita Puri; Timothy B. Stockwell; Hernan Lorenzi; Christopher F. Basler; Reed S. Shabman

Ebola virus and Marburg virus are members of the Filovirdae family and causative agents of hemorrhagic fever with high fatality rates in humans. Filovirus virulence is partially attributed to the VP35 protein, a well-characterized inhibitor of the RIG-I-like receptor pathway that triggers the antiviral interferon (IFN) response. Prior work demonstrates the ability of VP35 to block potent RIG-I activators, such as Sendai virus (SeV), and this IFN-antagonist activity is directly correlated with its ability to bind RNA. Several structural studies demonstrate that VP35 binds short synthetic dsRNAs; yet, there are no data that identify viral immunostimulatory RNAs (isRNA) or host RNAs bound to VP35 in cells. Utilizing a SeV infection model, we demonstrate that both viral isRNA and host RNAs are bound to Ebola and Marburg VP35s in cells. By deep sequencing the purified VP35-bound RNA, we identified the SeV copy-back defective interfering (DI) RNA, previously identified as a robust RIG-I activator, as the isRNA bound by multiple filovirus VP35 proteins, including the VP35 protein from the West African outbreak strain (Makona EBOV). Moreover, RNAs isolated from a VP35 RNA-binding mutant were not immunostimulatory and did not include the SeV DI RNA. Strikingly, an analysis of host RNAs bound by wild-type, but not mutant, VP35 revealed that select host RNAs are preferentially bound by VP35 in cell culture. Taken together, these data support a model in which VP35 sequesters isRNA in virus-infected cells to avert RIG-I like receptor (RLR) activation.

Collaboration


Dive into the Priya Luthra's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daisy W. Leung

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Gaya K. Amarasinghe

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Reed S. Shabman

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Gai Liu

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Megan R. Edwards

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adolfo García-Sastre

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Alexander Bukreyev

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Colette Pietzsch

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge