Network


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

Hotspot


Dive into the research topics where Chad E. Mire is active.

Publication


Featured researches published by Chad E. Mire.


Nature | 2015

Lipid nanoparticle siRNA treatment of Ebola virus Makona infected nonhuman primates

Emily P. Thi; Chad E. Mire; Amy Lee; Joan B. Geisbert; Joy Z. Zhou; Krystle N. Agans; Nicholas M. Snead; Daniel J. Deer; Trisha R. Barnard; Karla A. Fenton; Ian Maclachlan; Thomas W. Geisbert

The current outbreak of Ebola virus in West Africa is unprecedented, causing more cases and fatalities than all previous outbreaks combined, and has yet to be controlled. Several post-exposure interventions have been employed under compassionate use to treat patients repatriated to Europe and the United States. However, the in vivo efficacy of these interventions against the new outbreak strain of Ebola virus is unknown. Here we show that lipid-nanoparticle-encapsulated short interfering RNAs (siRNAs) rapidly adapted to target the Makona outbreak strain of Ebola virus are able to protect 100% of rhesus monkeys against lethal challenge when treatment was initiated at 3 days after exposure while animals were viraemic and clinically ill. Although all infected animals showed evidence of advanced disease including abnormal haematology, blood chemistry and coagulopathy, siRNA-treated animals had milder clinical features and fully recovered, while the untreated control animals succumbed to the disease. These results represent the first, to our knowledge, successful demonstration of therapeutic anti-Ebola virus efficacy against the new outbreak strain in nonhuman primates and highlight the rapid development of lipid-nanoparticle-delivered siRNA as a countermeasure against this highly lethal human disease.


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.


PLOS Neglected Tropical Diseases | 2012

Recombinant Vesicular Stomatitis Virus Vaccine Vectors Expressing Filovirus Glycoproteins Lack Neurovirulence in Nonhuman Primates

Chad E. Mire; Andrew D. Miller; Angela Carville; Susan V. Westmoreland; Joan B. Geisbert; Keith G. Mansfield; Heinz Feldmann; Lisa E. Hensley; Thomas W. Geisbert

The filoviruses, Marburg virus and Ebola virus, cause severe hemorrhagic fever with high mortality in humans and nonhuman primates. Among the most promising filovirus vaccines under development is a system based on recombinant vesicular stomatitis virus (rVSV) that expresses an individual filovirus glycoprotein (GP) in place of the VSV glycoprotein (G). The main concern with all replication-competent vaccines, including the rVSV filovirus GP vectors, is their safety. To address this concern, we performed a neurovirulence study using 21 cynomolgus macaques where the vaccines were administered intrathalamically. Seven animals received a rVSV vector expressing the Zaire ebolavirus (ZEBOV) GP; seven animals received a rVSV vector expressing the Lake Victoria marburgvirus (MARV) GP; three animals received rVSV-wild type (wt) vector, and four animals received vehicle control. Two of three animals given rVSV-wt showed severe neurological symptoms whereas animals receiving vehicle control, rVSV-ZEBOV-GP, or rVSV-MARV-GP did not develop these symptoms. Histological analysis revealed major lesions in neural tissues of all three rVSV-wt animals; however, no significant lesions were observed in any animals from the filovirus vaccine or vehicle control groups. These data strongly suggest that rVSV filovirus GP vaccine vectors lack the neurovirulence properties associated with the rVSV-wt parent vector and support their further development as a vaccine platform for human use.


Nature | 2015

Single-dose attenuated Vesiculovax vaccines protect primates against Ebola Makona virus

Chad E. Mire; Demetrius Matassov; Joan B. Geisbert; Theresa Latham; Krystle N. Agans; Rong Xu; Ayuko Ota-Setlik; Michael A. Egan; Karla A. Fenton; David K. Clarke; John H. Eldridge; Thomas W. Geisbert

The family Filoviridae contains three genera, Ebolavirus (EBOV), Marburg virus, and Cuevavirus. Some members of the EBOV genus, including Zaire ebolavirus (ZEBOV), can cause lethal haemorrhagic fever in humans. During 2014 an unprecedented ZEBOV outbreak occurred in West Africa and is still ongoing, resulting in over 10,000 deaths, and causing global concern of uncontrolled disease. To meet this challenge a rapid-acting vaccine is needed. Many vaccine approaches have shown promise in being able to protect nonhuman primates against ZEBOV. In response to the current ZEBOV outbreak several of these vaccines have been fast tracked for human use. However, it is not known whether any of these vaccines can provide protection against the new outbreak Makona strain of ZEBOV. One of these approaches is a first-generation recombinant vesicular stomatitis virus (rVSV)-based vaccine expressing the ZEBOV glycoprotein (GP) (rVSV/ZEBOV). To address safety concerns associated with this vector, we developed two candidate, further-attenuated rVSV/ZEBOV vaccines. Both attenuated vaccines produced an approximately tenfold lower vaccine-associated viraemia compared to the first-generation vaccine and both provided complete, single-dose protection of macaques from lethal challenge with the Makona outbreak strain of ZEBOV.


Science Translational Medicine | 2014

Marburg virus infection in nonhuman primates: Therapeutic treatment by lipid-encapsulated siRNA.

Emily P. Thi; Chad E. Mire; Raul Ursic-Bedoya; Joan B. Geisbert; Amy C. H. Lee; Krystle N. Agans; Marjorie Robbins; Daniel J. Deer; Karla A. Fenton; Ian Maclachlan; Thomas W. Geisbert

Lipid nanoparticle delivery of anti-MARV nucleoprotein–targeting small interfering RNA can treat Marburg virus in nonhuman primates after symptom onset. Medicating Marburg Marburg virus (MARV) and the closely related filovirus Ebola virus cause severe and often fatal hemorrhagic fever; however, there are currently no vaccines or drugs approved for human use to treat these devastating infections. Thi et al. now report that a lipid-encapsulated siRNA can treat MARV in nonhuman primates after symptom onset. They use lipid nanoparticles to deliver siRNA targeting the MARV nucleoprotein to treated animals at various time points after virus exposure. All animals that received the therapy survived MARV infection, including those that were treated 3 days after infection—a stage when animals are viremic and demonstrate the first clinical signs of disease. Therefore, this approach holds promise as a strategy to treat filovirus infection in humans. Marburg virus (MARV) and the closely related filovirus Ebola virus cause severe and often fatal hemorrhagic fever (HF) in humans and nonhuman primates with mortality rates up to 90%. There are no vaccines or drugs approved for human use, and no postexposure treatment has completely protected nonhuman primates against MARV-Angola, the strain associated with the highest rate of mortality in naturally occurring human outbreaks. Studies performed with other MARV strains assessed candidate treatments at times shortly after virus exposure, before signs of disease are detectable. We assessed the efficacy of lipid nanoparticle (LNP) delivery of anti-MARV nucleoprotein (NP)–targeting small interfering RNA (siRNA) at several time points after virus exposure, including after the onset of detectable disease in a uniformly lethal nonhuman primate model of MARV-Angola HF. Twenty-one rhesus monkeys were challenged with a lethal dose of MARV-Angola. Sixteen of these animals were treated with LNP containing anti-MARV NP siRNA beginning at 30 to 45 min, 1 day, 2 days, or 3 days after virus challenge. All 16 macaques that received LNP-encapsulated anti-MARV NP siRNA survived infection, whereas the untreated or mock-treated control subjects succumbed to disease between days 7 and 9 after infection. These results represent the successful demonstration of therapeutic anti–MARV-Angola efficacy in nonhuman primates and highlight the substantial impact of an LNP-delivered siRNA therapeutic as a countermeasure against this highly lethal human disease.


Cell Reports | 2014

The Marburg Virus VP24 Protein Interacts with Keap1 to Activate the Cytoprotective Antioxidant Response Pathway.

Megan R. Edwards; Britney Johnson; Chad E. Mire; Wei Xu; Reed S. Shabman; Lauren N. Speller; Daisy W. Leung; Thomas W. Geisbert; Gaya K. Amarasinghe; Christopher F. Basler

Kelch-like ECH-associated protein 1 (Keap1) is a ubiquitin E3 ligase specificity factor that targets transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) for ubiquitination and degradation. Disrupting Keap1-Nrf2 interaction stabilizes Nrf2, resulting in Nrf2 nuclear accumulation, binding to antioxidant response elements (AREs), and transcription of cytoprotective genes. Marburg virus (MARV) is a zoonotic pathogen that likely uses bats as reservoir hosts. We demonstrate that MARV protein VP24 (mVP24) binds the Kelch domain of either human or bat Keap1. This binding is of high affinity and 1:1 stoichiometry and activates Nrf2. Modeling based on the Zaire ebolavirus (EBOV) VP24 (eVP24) structure identified in mVP24 an acidic loop (K-loop) critical for Keap1 interaction. Transfer of the K-loop to eVP24, which otherwise does not bind Keap1, confers Keap1 binding and Nrf2 activation, and infection by MARV, but not EBOV, activates ARE gene expression. Therefore, MARV targets Keap1 to activate Nrf2-induced cytoprotective responses during infection.


Applied and Environmental Microbiology | 2004

Lead Precipitation by Vibrio harveyi: Evidence for Novel Quorum-Sensing Interactions

Chad E. Mire; Jeanette A. Tourjee; William F. O'Brien; Kandalam V. Ramanujachary; Gregory B. Hecht

ABSTRACT Three pleiotropic, quorum sensing-defective Vibrio harveyi mutants were observed to precipitate soluble Pb2+ as an insoluble compound. The compound was purified and subjected to X-ray diffraction and elemental analyses. These assays identified the precipitated compound as Pb9(PO4)6, an unusual and complex lead phosphate salt that is produced synthetically at temperatures of ca. 200°C. Regulation of the precipitation phenotype was also examined. Introduction of a luxO::kan allele into one of the mutants abolished lead precipitation, indicating that the well-characterized autoinducer 1 (AI1)-AI2 quorum-sensing system can block lead precipitation in dense cell populations. Interestingly, the V. harveyi D1 mutant, a strain defective for secretion of both AI1 and AI2, was shown to be an effective trans inhibitor of lead precipitation. This suggests that a previously undescribed V. harveyi autoinducer, referred to as AI3, can also negatively regulate lead precipitation. Experiments with heterologous bacterial populations demonstrated that many different species are capable of trans regulating the V. harveyi lead precipitation phenotype. Moreover, one of the V. harveyi mutants in this study exhibited little or no response to intercellular signals from other V. harveyi inocula but was quite responsive to some of the heterologous bacteria. Based on these observations, we propose that V. harveyi carries at least one quorum sensor that is specifically dedicated to receiving cross-species communication.


Journal of Clinical Investigation | 2015

Aerosolized Ebola vaccine protects primates and elicits lung-resident T cell responses

Michelle Meyer; Tania Garron; Ndongala Michel Lubaki; Chad E. Mire; Karla A. Fenton; Curtis Klages; Gene G. Olinger; Thomas W. Geisbert; Peter L. Collins; Alexander Bukreyev

Direct delivery of aerosolized vaccines to the respiratory mucosa elicits both systemic and mucosal responses. This vaccine strategy has not been tested for Ebola virus (EBOV) or other hemorrhagic fever viruses. Here, we examined the immunogenicity and protective efficacy of an aerosolized human parainfluenza virus type 3-vectored vaccine that expresses the glycoprotein (GP) of EBOV (HPIV3/EboGP) delivered to the respiratory tract. Rhesus macaques were vaccinated with aerosolized HPIV3/EboGP, liquid HPIV3/EboGP, or an unrelated, intramuscular, Venezuelan equine encephalitis replicon vaccine expressing EBOV GP. Serum and mucosal samples from aerosolized HPIV3/EboGP recipients exhibited high EBOV-specific IgG, IgA, and neutralizing antibody titers, which exceeded or equaled titers observed in liquid recipients. The HPIV3/EboGP vaccine induced an EBOV-specific cellular response that was greatest in the lungs and yielded polyfunctional CD8+ T cells, including a subset that expressed CD103 (αE integrin), and CD4+ T helper cells that were predominately type 1. The magnitude of the CD4+ T cell response was greater in aerosol vaccinees. The HPIV3/EboGP vaccine produced a more robust cell-mediated and humoral immune response than the systemic replicon vaccine. Moreover, 1 aerosol HPIV3/EboGP dose conferred 100% protection to macaques exposed to EBOV. Aerosol vaccination represents a useful and feasible vaccination mode that can be implemented with ease in a filovirus disease outbreak situation.


Science Translational Medicine | 2014

Therapeutic treatment of Nipah virus infection in nonhuman primates with a neutralizing human monoclonal antibody.

Thomas W. Geisbert; Chad E. Mire; Joan B. Geisbert; Yee Peng Chan; Krystle N. Agans; Friederike Feldmann; Karla A. Fenton; Zhongyu Zhu; Dimiter S. Dimitrov; Dana P. Scott; Katharine N. Bossart; Heinz Feldmann; Christopher C. Broder

A monoclonal antibody against Nipah virus has in vivo efficacy in a nonhuman primate. Time’s Up for Nipah Virus A human antibody may be the key for treating a deadly viral infection. Geisbert et al. report that a human monoclonal antibody to the emerging zoonotic paramyxovirus Nipah virus (NiV) can treat nonhuman primates at the onset of clinical symptoms. Fruit bats are the natural reservoir for NiV. However, NiV can infect a broad range of mammalian species, and exposed pigs may serve as an intermediary for human infection, which is frequently fatal. There is currently no approved human treatment for NiV, and previous studies have only shown efficacy after infection—but before the onset of clinical symptoms. Now, a human monoclonal antibody has resulted in survival in all animals treated, even after the development of clinical symptoms. This antibody represents a potential new therapeutic strategy for this emerging viral infection. Nipah virus (NiV) is an emerging zoonotic paramyxovirus that causes severe and often fatal disease in pigs and humans. There are currently no vaccines or treatments approved for human use. Studies in small-animal models of NiV infection suggest that antibody therapy may be a promising treatment. However, most studies have assessed treatment at times shortly after virus exposure before animals show signs of disease. We assessed the efficacy of a fully human monoclonal antibody, m102.4, at several time points after virus exposure including at the onset of clinical illness in a uniformly lethal nonhuman primate model of NiV disease. Sixteen African green monkeys (AGMs) were challenged intratracheally with a lethal dose of NiV, and 12 animals were infused twice with m102.4 (15 mg/kg) beginning at either 1, 3, or 5 days after virus challenge and again about 2 days later. The presence of viral RNA, infectious virus, and/or NiV-specific immune responses demonstrated that all subjects were infected after challenge. All 12 AGMs that received m102.4 survived infection, whereas the untreated control subjects succumbed to disease between days 8 and 10 after infection. AGMs in the day 5 treatment group exhibited clinical signs of disease, but all animals recovered by day 16. These results represent the successful therapeutic in vivo efficacy by an investigational drug against NiV in a nonhuman primate and highlight the potential impact that a monoclonal antibody can have on a highly pathogenic zoonotic human disease.


Mbio | 2014

Deep Sequencing Identifies Noncanonical Editing of Ebola and Marburg Virus RNAs in Infected Cells

Reed S. Shabman; Omar J. Jabado; Chad E. Mire; Timothy B. Stockwell; Megan R. Edwards; Milind Mahajan; Thomas W. Geisbert; Christopher F. Basler

ABSTRACT Deep sequencing of RNAs produced by Zaire ebolavirus (EBOV) or the Angola strain of Marburgvirus (MARV-Ang) identified novel viral and cellular mechanisms that diversify the coding and noncoding sequences of viral mRNAs and genomic RNAs. We identified previously undescribed sites within the EBOV and MARV-Ang mRNAs where apparent cotranscriptional editing has resulted in the addition of non-template-encoded residues within the EBOV glycoprotein (GP) mRNA, the MARV-Ang nucleoprotein (NP) mRNA, and the MARV-Ang polymerase (L) mRNA, such that novel viral translation products could be produced. Further, we found that the well-characterized EBOV GP mRNA editing site is modified at a high frequency during viral genome RNA replication. Additionally, editing hot spots representing sites of apparent adenosine deaminase activity were found in the MARV-Ang NP 3′-untranslated region. These studies identify novel filovirus-host interactions and reveal production of a greater diversity of filoviral gene products than was previously appreciated. IMPORTANCE This study identifies novel mechanisms that alter the protein coding capacities of Ebola and Marburg virus mRNAs. Therefore, filovirus gene expression is more complex and diverse than previously recognized. These observations suggest new directions in understanding the regulation of filovirus gene expression. This study identifies novel mechanisms that alter the protein coding capacities of Ebola and Marburg virus mRNAs. Therefore, filovirus gene expression is more complex and diverse than previously recognized. These observations suggest new directions in understanding the regulation of filovirus gene expression.

Collaboration


Dive into the Chad E. Mire's collaboration.

Top Co-Authors

Avatar

Thomas W. Geisbert

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Krystle N. Agans

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Joan B. Geisbert

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Karla A. Fenton

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Robert W. Cross

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Daniel J. Deer

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Viktoriya Borisevich

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Heinz Feldmann

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael A. Whitt

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge