Network


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

Hotspot


Dive into the research topics where Dmitriy Zamarin is active.

Publication


Featured researches published by Dmitriy Zamarin.


PLOS Pathogens | 2007

A Single Mutation in the PB1-F2 of H5N1 (HK/97) and 1918 Influenza A Viruses Contributes to Increased Virulence

Gina Conenello; Dmitriy Zamarin; Lucy A. Perrone; Terrence M. Tumpey; Peter Palese

The proapoptotic PB1-F2 protein of influenza A viruses has been shown to contribute to pathogenesis in the mouse model. Expression of full-length PB1-F2 increases the pathogenesis of the influenza A virus, causing weight loss, slower viral clearance, and increased viral titers in the lungs. After comparing viruses from the Hong Kong 1997 H5N1 outbreak, one amino acid change (N66S) was found in the PB1-F2 sequence at position 66 that correlated with pathogenicity. This same amino acid change (N66S) was also found in the PB1-F2 protein of the 1918 pandemic A/Brevig Mission/18 virus. Two isogenic recombinant chimeric viruses were created with an influenza A/WSN/33 virus background containing the PB1 segment from the HK/156/97: WH and WH N66S. In mice infected with WH N66S virus there was increased pathogenicity as measured by weight loss and decreased survival, and a 100-fold increase in virus replication when compared to mice infected with the WH virus. The 1918 pandemic strain A/Brevig Mission/18 was reconstructed with a pathogenicity-reducing mutation in PB1-F2 (S66N). The resultant 1918 S66N virus was attenuated in mice having a 3-log lower 50% lethal dose and caused less morbidity and mortality in mice than the wild-type virus. Viral lung titers were also decreased in 1918 S66N–infected mice compared with wild-type 1918 virus–infected mice. In addition, both viruses with an S at position 66 (WH N66S and wt 1918) induced elevated levels of cytokines in the lungs of infected mice. Together, these data show that a single amino acid substitution in PB1-F2 can result in increased viral pathogenicity and could be one of the factors contributing to the high lethality seen with the 1918 pandemic virus.


Journal of Virology | 2005

A Single Amino Acid Substitution in 1918 Influenza Virus Hemagglutinin Changes Receptor Binding Specificity

Laurel Glaser; James Stevens; Dmitriy Zamarin; Ian A. Wilson; Adolfo García-Sastre; Terrence M. Tumpey; Christopher F. Basler; Jeffery K. Taubenberger; Peter Palese

ABSTRACT The receptor binding specificity of influenza viruses may be important for host restriction of human and avian viruses. Here, we show that the hemagglutinin (HA) of the virus that caused the 1918 influenza pandemic has strain-specific differences in its receptor binding specificity. The A/South Carolina/1/18 HA preferentially binds the α2,6 sialic acid (human) cellular receptor, whereas the A/New York/1/18 HA, which differs by only one amino acid, binds both the α2,6 and the α2,3 sialic acid (avian) cellular receptors. Compared to the conserved consensus sequence in the receptor binding site of avian HAs, only a single amino acid at position 190 was changed in the A/New York/1/18 HA. Mutation of this single amino acid back to the avian consensus resulted in a preference for the avian receptor.


Journal of Experimental Medicine | 2013

Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4

Rikke B. Holmgaard; Dmitriy Zamarin; David H. Munn; Jedd D. Wolchok; James P. Allison

Indoleamine 2,3-dioxygenase suppresses infiltration and accumulation of tumor-reactive T cells in the context of anti–CTLA-4 immunotherapy and attenuates the anti-tumor efficacy.


PLOS Pathogens | 2005

Influenza virus PB1-F2 protein induces cell death through mitochondrial ANT3 and VDAC1.

Dmitriy Zamarin; Adolfo García-Sastre; Xiaoyao Xiao; Rong Wang; Peter Palese

The influenza virus PB1-F2 is an 87-amino acid mitochondrial protein that previously has been shown to induce cell death, although the mechanism of apoptosis induction has remained unclear. In the process of characterizing its mechanism of action we found that the viral PB1-F2 protein sensitizes cells to apoptotic stimuli such as tumor necrosis factor alpha, as demonstrated by increased cleavage of caspase 3 substrates in PB1-F2-expressing cells. Moreover, treatment of purified mouse liver mitochondria with recombinant PB1-F2 protein resulted in cytochrome c release, loss of the mitochondrial membrane potential, and enhancement of tBid-induced mitochondrial permeabilization, suggesting a possible mechanism for the observed cellular sensitization to apoptosis. Using glutathione-S-transferase pulldowns with subsequent mass spectrometric analysis, we identified the mitochondrial interactors of the PB1-F2 protein and showed that the viral protein uniquely interacts with the inner mitochondrial membrane adenine nucleotide translocator 3 and the outer mitochondrial membrane voltage-dependent anion channel 1, both of which are implicated in the mitochondrial permeability transition during apoptosis. Consistent with this interaction, blockers of the permeability transition pore complex (PTPC) inhibited PB1-F2-induced mitochondrial permeabilization. Based on our findings, we propose a model whereby the proapoptotic PB1-F2 protein acts through the mitochondrial PTPC and may play a role in the down-regulation of the host immune response to infection.


Science Translational Medicine | 2014

Localized Oncolytic Virotherapy Overcomes Systemic Tumor Resistance to Immune Checkpoint Blockade Immunotherapy

Dmitriy Zamarin; Rikke B. Holmgaard; Sumit K. Subudhi; Joon Seok Park; Mena Mansour; Peter Palese; Taha Merghoub; Jedd D. Wolchok; James P. Allison

Intratumoral treatment with an oncolytic virus results in inflammatory infiltrates in distant tumors and sensitizes the tumors to immune-mediated rejection in the setting of immunotherapy with CTLA-4 blocking antibodies. Immune Cells and a Virus Teaming Up to Fight Cancer Immunotherapy, or harnessing the patient’s own immune system to help fight cancer, is becoming increasingly popular as researchers discover newer and more successful approaches focused on different aspects of the immune system. Two of these approaches include oncolytic viruses and therapies that block immune checkpoints and thus stimulate the antitumor activity of T cells. One virus that is known to have oncolytic activity is Newcastle disease virus (NDV), an avian virus that is not pathogenic in humans. Although previous studies have successfully demonstrated the antitumor effects of NDV, these were thought to require direct injection of this virus into every tumor, thus greatly limiting its effectiveness against metastatic disease. Now, Zamarin and coauthors used mouse models of cancer with multiple tumor sites to demonstrate that NDV can be used even in the setting of distant spread of disease. Although the virus had to be injected into a tumor to have any effect, the subsequent tumor killing was not limited to the injected tumor. Additional masses that were distant in space and time, such as tumors implanted at different body sites and at later time points, were targeted by the immune system in animals that had been treated with NDV injection into the primary tumor. This immunostimulatory effect of NDV was particularly pronounced when the virotherapy was combined with immune checkpoint blockade by an anti–CTLA-4 antibody. The current study was performed in mice bearing tumors derived from established cell lines, and the findings will need to be confirmed in the setting of spontaneously arising metastatic tumors. Nevertheless, the results of this work raise the tantalizing possibility that two therapies known to be safe for human use could be combined and work with the immune system to eradicate tumors throughout the body. Preexisting lymphocytic infiltration of tumors is associated with superior prognostic outcomes in a variety of cancers. Recent studies also suggest that lymphocytic responses may identify patients more likely to benefit from therapies targeting immune checkpoints, suggesting that therapeutic efficacy of immune checkpoint blockade can be enhanced through strategies that induce tumor inflammation. To achieve this effect, we explored the immunotherapeutic potential of oncolytic Newcastle disease virus (NDV). We find that localized intratumoral therapy of B16 melanoma with NDV induces inflammatory responses, leading to lymphocytic infiltrates and antitumor effect in distant (nonvirally injected) tumors without distant virus spread. The inflammatory effect coincided with distant tumor infiltration with tumor-specific CD4+ and CD8+ T cells, which was dependent on the identity of the virus-injected tumor. Combination therapy with localized NDV and systemic CTLA-4 blockade led to rejection of preestablished distant tumors and protection from tumor rechallenge in poorly immunogenic tumor models, irrespective of tumor cell line sensitivity to NDV-mediated lysis. Therapeutic effect was associated with marked distant tumor infiltration with activated CD8+ and CD4+ effector but not regulatory T cells, and was dependent on CD8+ cells, natural killer cells, and type I interferon. Our findings demonstrate that localized therapy with oncolytic NDV induces inflammatory immune infiltrates in distant tumors, making them susceptible to systemic therapy with immunomodulatory antibodies, which provides a strong rationale for investigation of such combination therapies in the clinic.


Journal of Virology | 2006

Influenza A Virus PB1-F2 Protein Contributes to Viral Pathogenesis in Mice

Dmitriy Zamarin; Mila Brum Ortigoza; Peter Palese

ABSTRACT The influenza virus PB1-F2 protein is a novel protein previously shown to be involved in induction of cell death. Here we characterize the expression and the function of the protein within the context of influenza viral infection in tissue culture and a mouse model. We show that the C-terminal region of the protein can be expressed from a downstream initiation codon and is capable of interaction with the full-length protein. Using this knowledge, we generated influenza viruses knocked out for the expression of PB1-F2 protein and its downstream truncation products. Knocking out the PB1-F2 protein had no effect on viral replication in tissue culture but diminished virus pathogenicity and mortality in mice. The viruses replicated to similar levels in mouse lungs by day 3 postinfection, suggesting that the knockout did not impair viral replication. However, while the PB1-F2 knockout viruses were cleared after day 5, the wild-type viruses were detectable in mouse lungs until day 7, implying that expression of PB1-F2 resulted in delayed clearance of the viruses by the host immune system. Based on our findings and on the fact that the PB1 genomic segment was always newly introduced into some pandemic influenza viruses of the last century, we speculate that the PB1-F2 protein plays an important role in pathogenesis of influenza virus infection and may be an important contributor to pathogenicity of pandemic influenza viruses.


Journal of Virology | 2005

Attenuation of Equine Influenza Viruses through Truncations of the NS1 Protein

Michelle Quinlivan; Dmitriy Zamarin; Adolfo García-Sastre; Ann Cullinane; Thomas M. Chambers; Peter Palese

ABSTRACT Equine influenza is a common disease of the horse, causing significant morbidity worldwide. Here we describe the establishment of a plasmid-based reverse genetics system for equine influenza virus. Utilizing this system, we generated three mutant viruses encoding carboxy-terminally truncated NS1 proteins. We have previously shown that a recombinant human influenza virus lacking the NS1 gene (delNS1) could only replicate in interferon (IFN)-incompetent systems, suggesting that the NS1 protein is responsible for IFN antagonist activity. Contrary to previous findings with human influenza virus, we found that in the case of equine influenza virus, the length of the NS1 protein did not correlate with the level of attenuation of that virus. With equine influenza virus, the mutant virus with the shortest NS1 protein turned out to be the least attenuated. We speculate that the basis for attenuation of the equine NS1 mutant viruses generated is related to their level of NS1 protein expression. Our findings show that the recombinant mutant viruses are impaired in their ability to inhibit IFN production in vitro and they do not replicate as efficiently as the parental recombinant strain in embryonated hen eggs, in MDCK cells, or in vivo in a mouse model. Therefore, these attenuated mutant NS1 viruses may have potential as candidates for a live equine influenza vaccine.


Journal of Virology | 2005

Nuclear Localization of the Nipah Virus W Protein Allows for Inhibition of both Virus- and Toll-Like Receptor 3-Triggered Signaling Pathways

Megan L. Shaw; Washington B. Cárdenas; Dmitriy Zamarin; Peter Palese; Christopher F. Basler

ABSTRACT The Nipah virus V and W proteins, which are encoded by the P gene via RNA editing, have a common N-terminal domain but unique C-terminal domains. They localize to the cytoplasm and nucleus, respectively, and have both been shown to function as inhibitors of JAK/STAT signaling. Here we report that V and W proteins also block virus activation of the beta interferon (IFN-β) promoter and the IFN regulatory factor 3 (IRF3)-responsive IFN-stimulated gene 54 promoter. Surprisingly, only W protein shows strong inhibition of promoter activation in response to stimulation of Toll-like receptor 3 (TLR3) by extracellular double-stranded RNA. This activity is dependent on the nuclear localization of W protein. Within the unique C-terminal domain of W protein, we have identified a nuclear localization signal (NLS) that requires basic residues at positions 439, 440, and 442. This NLS is responsible for mediating the preferential interaction of W protein with karyopherin-α 3 and karyopherin-α 4. Nuclear localization of W protein therefore enables it to target both virus and TLR3 pathways, whereas the cytoplasmic V protein is restricted to inhibiting the virus pathway. We propose that this discrepancy is in part due to the V protein being less able to block signaling in response to the kinase, TBK-1, whereas both V and W can prevent promoter activation in response to IKKε. We demonstrate that, when the TLR3 pathway is stimulated, the levels of phosphorylated IRF3 are reduced in the presence of W protein but not V protein, confirming the differential effects of these proteins and illustrating that W protein-mediated inhibition is due to a loss of active IRF3.


Future Microbiology | 2012

Oncolytic Newcastle Disease Virus for cancer therapy: old challenges and new directions

Dmitriy Zamarin; Peter Palese

Newcastle disease virus (NDV) is an avian paramyxovirus, which has been demonstrated to possess significant oncolytic activity against mammalian cancers. This review summarizes the research leading to the elucidation of the mechanisms of NDV-mediated oncolysis, as well as the development of novel oncolytic agents through the use of genetic engineering. Clinical trials utilizing NDV strains and NDV-based autologous tumor cell vaccines will expand our knowledge of these novel anticancer strategies and will ultimately result in the successful use of the virus in the clinical setting.


Journal of Virology | 2011

Oncolytic Specificity of Newcastle Disease Virus Is Mediated by Selectivity for Apoptosis-Resistant Cells

Mena Mansour; Peter Palese; Dmitriy Zamarin

ABSTRACT Newcastle disease virus (NDV) is a negative-sense RNA virus that has been shown to possess oncolytic activity. NDVs selective replication in tumor cells has been previously suggested to be due to the lack of a proper antiviral response in these cells. Here we demonstrate that NDV possesses oncolytic activity in tumor cells capable of a robust type I interferon (IFN) response, suggesting that another mechanism underlies NDVs tumor specificity. We show that the oncolytic selectivity of NDV for tumor cells is dependent upon tumor cell resistance to apoptosis. Utilizing the human non-small-cell lung cancer cell line A549 overexpressing the antiapoptotic protein Bcl-xL, we show significant enhancement of oncolytic activity and NDV replication. Interestingly, while the Bcl-xL-overexpressing cells were resistant to apoptotic stimuli induced by chemotherapeutic agents and early viral replication, during the subsequent viral cycles, we observed a paradoxical increase in apoptosis in response to NDV. The increased oncolytic activity seen was secondary to enhanced viral replication and syncytium formation. The induction of a type I IFN response was enhanced in Bcl-xL cells. Overall, these findings propose a new mechanism for cancer cell specificity for NDV, making it an attractive anticancer agent for chemoresistant tumors with enhanced antiapoptotic activity.

Collaboration


Dive into the Dmitriy Zamarin's collaboration.

Top Co-Authors

Avatar

Peter Palese

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jedd D. Wolchok

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Taha Merghoub

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yuman Fong

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Carol Aghajanian

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Adolfo García-Sastre

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Rikke B. Holmgaard

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jacob Ricca

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jason A. Konner

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Paul Sabbatini

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge