Latest Research

Explore our faculty's areas of research and the latest publications from each lab.

Bayer-Santos Lab

About The Bayer-Santos Lab

The Bayer-Santos lab is focused on (micro)biological conflicts and the weapons used by bacteria to target competitors. The group currently uses Salmonella and a specialized contractile nanoweapon called type VI secretion system (T6SS) to understand how enteric pathogens overcome colonization resistance imposed by members of the microbiota and establish an infection. The effectors secreted by the T6SS comprise a pool of proteins with interesting new biochemical activities, providing opportunities for several interdisciplinary projects encompassing microbiology, cell biology, biochemistry, structural biology and bioinformatic.

Research

2023

2022

2021

2020

Contreras Lab

About The Contreras Lab

The Contreras Lab's research combines biomolecular engineering, genetic studies and computational modeling to understand molecular features that lead to the specific recognition and interaction of regulatory RNAs and proteins. They apply fundamental concepts that emerge from experimental (and computational) work to develop novel applications that could beneficially impact human health and biotechnology. Overall research interests:

  • Transcriptional regulation
  • Regulatory RNA networks
  • Epitranscriptomics
  • RNA-protein interactions and RNA-protein folding
  • Novel molecular tools for intracellular studies of RNA-metabolism
  • Diagnostics for RNA-related diseases
  • Molecular mechanisms of stress-responses

Research

2024

2023

2022

2021

2020 

Croyle Lab

About The Croyle Lab

To date, viral vectors have held the most promise as vehicles for gene therapy because they are capable of delivering genes to certain tissues with high efficiency and establishing stable transgene expression for significant periods of time. However, routine use of viruses for therapeutic purposes is significantly limited by the innate immune response against capsid proteins, viral gene products and the therapeutic transgene. Recombinant viral preparations must also be extremely pure for clinical use. In this form, however, they often exhibit poor physical stability. Research in the Croyle Laboratory focuses on the development of methods to reduce the immune response and associated toxicity associated with recombinant viruses and methods to evaluate the physical stability of viral vectors during processing and purification. The primary vectors under investigation are adenoviruses, adeno-associated viruses and lentiviruses. Students in the Croyle Lab are exposed to cutting edge, interdisciplinary research relevant to the fields of cell biology, virology and immunology, with basic skills in pharmaceutics and drug delivery also emphasized. Projects address basic research problems and sharpen skills in hypothesis development and open-ended problem solving. Application of research techniques to clinical settings is also emphasized. Specific projects include:

  • Biochemical Modification of Viruses to Evade the Immune Response
  • Effect of Recombinant Viruses on Hepatic, Renal and Intestinal Drug Metabolism
  • Vaccination Strategies for Rapid Induction of Immunity Against Dangerous Pathogens
  • Production, Processing & Physical Stability of Recombinant Viruses


Research


2024


2023 


2022


2021


2020

Davies Lab

About The Davies Lab

The Davies Lab investigates protein-based antibiotics and bacteria-host interactions. Current projects include:

  • Discovering synthetic peptide antibiotics: Davies Lab researchers are exploring the sequence-structure space of peptide antibiotics and how peptides with distinct antibacterial and in vivo properties are distributed across this landscape. 
  • Developing antibacterial antibodies: the Davies Lab is developing single-chain antibodies that can target and/or directly kill specific bacteria. We are selecting and enhancing scaffolds to support these activities and investigating potential bacterial targets.
  • Characterizing bacterial secretion systems and secreted proteins: the Lab explores bacterial secretion systems and secreted proteins as sources of antibacterial scaffolds and routes to control bacterial growth and host interactions.
  • Designing bacteria-host interactions: the Davies Lab is investigating and repurposing microbe-microbe and microbe-host interactions to develop bacteria that can influence human health.
     

Research


2024


2023 


2022


2021


2020

Dudley Lab

About The Dudley Lab


Mouse mammary tumor virus (MMTV) is a retrovirus that induces mammary carcinomas and T-cell lymphomas in mice by insertional mutagenesis. The Dudley Lab recently discovered a novel viral protein, Rem, which is involved in the nuclear export and expression of intron-containing viral mRNAs. These results are exciting because MMTV serves as a mouse model for study of another retrovirus, human immunodeficiency virus (HIV), which causes AIDS. The Lab's recent results suggest that Rem has a very unusual trafficking pattern within mammalian cells. Prior to nuclear entry, Rem appears to enter the endoplasmic reticulum (ER), where it is partially glycosylated, and cleaved by signal peptidase. Cleavage appears to yield an HIV Rev-like gene product, SP, as well as a unique product (Rem-CT) of unknown function. Mutations that prevent the correct processing and glycosylation of Rem interfere with SP activity in reporter assays. Rem trafficking through the ER is required for Rem processing and function in the nucleus after signal peptidase cleavage and retrotranslocation of the N-terminal SP out of the ER. Retrotranslocation is associated with endoplasmic reticulum-associated degradation (ERAD). ERAD is a poorly understood cellular process that is responsible for disposal of misfolded proteins. Numerous human diseases, including cancer and neurogeneration, show defects in ERAD. Recent exciting data indicate that the Rem C-terminus has a separate function in intrinsic immunity.

Finally, the Dudley Lab is in the process of developing vectors for gene therapy of breast cancer. Their studies have allowed the extensive mapping of the MMTV genome, which has been evolutionarily selected for optimal expression in the mammary gland. Elimination of viral genes, introduction of reporter genes, and manipulation of tissue-specific promoter elements should enable us to develop and test new vectors for safety and efficacy in mice. The Dudey Lab's goal is to provide more specific and less toxic treatments for human breast cancer.
 

Research


2023 


2022


2021


2020

Ehrlich Lab

About The Ehrlich Lab

The Ehrlich Lab's major research goals are to identify the cellular and molecular interactions between developing T cells and the surrounding thymic stromal cells that govern generation of a diverse, non-autoreactive, and non-malignant T cell pool throughout the lifespan. 

The Lab is currently focused on:

  • Identifying cellular and molecular mechanisms, such as chemokine-driven chemotaxis, that contribute to T cell self-tolerance by promoting thymocyte medullary entry and/or interactions with antigen presenting cells.
  • Determining how age-associated changes in the thymus throughout the lifespan impact thymic stromal cellularity and function, thymocyte development, and the establishment of central tolerance.
  • Identifying molecular mediators of interactions between T cell acute lymphoblastic leukemia cells and the surrounding tumor microenvironment that promote leukemia growth.
  • Identifying variation in immune responses in COVID-19 patients across the lifespan (pediatric to geriatric) that correlate with differential disease severity.
  • Determining if SARS-CoV-2 infections predispose patients to autoimmunity.
     

Research


2024

  • Gygi JP, Maguire C, Patel RK, Shinde P, Konstorum A, Shannon CP, Xu L, Hoch A, Jayavelu ND, Haddad EK; IMPACC Network; Reed EF, Kraft M, McComsey GA, Metcalf JP, Ozonoff A, Esserman D, Cairns CB, Rouphael N, Bosinger SE, Kim- Schulze S, Krammer F, Rosen LB, van Bakel H, Wilson M, Eckalbar WL, Maecker HT, Langelier CR, Steen H, Altman MC, Montgomery RR, Levy O, Melamed E, Pulendran B, Diray-Arce J, Smolen KK, Fragiadakis GK, Becker PM, Sekaly RP, Ehrlich LI, Fourati S, Peters B, Kleinstein SH, Guan L. Integrated longitudinal multiomics study identifies immune programs associated with acute COVID-19 severity and mortality. J Clin Invest. 2024 May 1;134(9):e176640. PMID: 38690733; PMCID: PMC11060740.
  • Ozonoff A, Jayavelu ND, Liu S, Melamed E, Milliren CE, Qi J, Geng LN, McComsey GA, Cairns CB, Baden LR, Schaenman J, Shaw AC, Samaha H, Seyfert- Margolis V, Krammer F, Rosen LB, Steen H, Syphurs C, Dandekar R, Shannon CP, Sekaly RP, Ehrlich LIR, Corry DB, Kheradmand F, Atkinson MA, Brakenridge SC, Higuita NIA, Metcalf JP, Hough CL, Messer WB, Pulendran B, Nadeau KC, Davis MM, Sesma AF, Simon V, van Bakel H, Kim-Schulze S, Hafler DA, Levy O, Kraft M, Bime C, Haddad EK, Calfee CS, Erle DJ, Langelier CR, Eckalbar W, Bosinger SE; IMPACC Network; Peters B, Kleinstein SH, Reed EF, Augustine AD, Diray-Arce J, Maecker HT, Altman MC, Montgomery RR, Becker PM, Rouphael N. Features of acute COVID-19 associated with post-acute sequelae of SARS-CoV-2 phenotypes: results from the IMPACC study. Nat Commun. 2024 Jan 3;15(1):216. PMID: 38172101; PMCID: PMC10764789.
  • Siles N, Schuler M, Maguire C, Amengor D, Nguyen A, Wilen R, Rogers J, Bazzi S, Caslin B, DiPasquale C, Abigania M, Olson E, Creaturo J, Hurley K, Triplett TA, Rousseau JF, Strakowski SM, Wylie D, Maynard J, Ehrlich LIR, Melamed E. SARS-CoV-2 Humoral Immune Responses in Convalescent Individuals Over 12 Months Reveal Severity-Dependent Antibody Dynamics. medRxiv [Preprint]. 2023 Dec 7:2023.12.05.23299462. PMID: 38106077; PMCID: PMC10723498.
  • Ozonoff A, Schaenman J, Jayavelu ND, Milliren CE, Calfee CS, Cairns CB, Kraft M, Baden LR, Shaw AC, Krammer F, van Bakel H, Esserman DA, Liu S, Sesma AF, Simon V, Hafler DA, Montgomery RR, Kleinstein SH, Levy O, Bime C, Haddad EK, Erle DJ, Pulendran B, Nadeau KC, Davis MM, Hough CL, Messer WB, Agudelo Higuita NI, Metcalf JP, Atkinson MA, Brakenridge SC, Corry D, Kheradmand F, Ehrlich LIR, Melamed E, McComsey GA, Sekaly R, Diray-Arce J, Peters B, Augustine AD, Reed EF, Altman MC, Becker PM, Rouphael N; IMPACC Study Group Members. Corrigendum to "Phenotypes of disease severity in a cohort of hospitalized COVID-19 patients: results from the IMPACC study". [eBioMedicine 83 (2022) 104208]. EBioMedicine. 2023 Dec;98:104860. Epub 2023 Nov 1. Erratum for: EBioMedicine. 2022 Sep;83:104208. PMID: 37918220; PMCID: PMC10643088.


2023 


2022


2021


2020

Ellington Lab

About The Ellington Lab

Throughout its existence, the Ellington Lab has generated impactful new technologies and approaches.  Currently, they are working in several different areas, including: 

  • Developing novel genetic codes and synthetic organisms based on engineering the translation apparatus.  
  • Developing novel point-of-care diagnostics based on isothermal amplification and DNA computation.  
  • Developing new methods for scalable assembly of protein architectures and programmed biomaterials.  
     

Research

2024

2023

2022

2021

2020

Georgiou Lab

About The Georgiou Lab

Since 2009 the Georgiou laboratory has been focused on the discovery and development of protein therapeutics and on the analysis of adaptive immune responses.  Current projects include:

  • Engineering and preclinical/clinical development of human enzyme therapeutics for the treatment of solid tumors and leukemias.
  • Development of methods for the molecular-level identification of the human antibody repertoire in blood and in secretions.
  • Analysis of the antibody repertoire elicited by viral infection (e.g. HIV-1) and vaccination to aid the development of more efficient vaccination stategies.
  • Engineering of antibody therapeutics displaying enhanced ability to recruit cytotoxic leukocytes and blood proteins (complement) for the effective clearance of pathogens.

Research

2024

2023 


2022


2021


2020

 

Gordon Lab

About The Gordon Lab

As bacteria continue to be treated with antibiotics, we are witnessing the emergence of antibiotic-resistant pathogens capable of forming "biofilms" that further increase its immune system and antibiotic resistance. The Gordon Lab utilizes physics to attack biological problems from a unique perspective. Whereas most scientists studying biofilms have microbiology backgrounds, physics training prompts Gordon Lab researchers to ask different questions and use unique approaches. Rather than studying individual cells, Dr. Gordon and her team study "biofilms," or many interacting bacteria in a structural grouping, to understand how the structures of infections affect their function and characteristics.

The Gordon Lab's approach to infections' structure is uncovering answers that reveal how many cells of bacteria operate when functioning as a community. Current research focuses on Pseudomonas aeruginosa, a bacterium known to infect individuals with compromised immune systems. Pseudomonas is the 4th most commonly acquired infection in hospitals and is particularly dangerous, often deadly, in patients suffering from diabetes or cystic fibrosis.

Research

2024

2023 


2022


2021


2020

Harshey Lab

About The Harshey Lab

The Harshey Lab studies two different areas. The first is the repair of Mu DNA transposition events. Transposition is not complete until short gaps created in the target on either side of the transposed element are repaired, yet how this happens is still not known. The repair assays the Harshey Lab is developing will be important for obtaining insights into this integral aspect of transposition, and should present a potent new target for drug development. Thus, knowledge of the repair process is not only necessary to fully understand the biology of transposition, but can be potentially used to block deleterious transposition events.

The second area Harshey researchers study is the flagellar motor as a sensor. In E. coli and Salmonella, chemosensory information from the environment is detected by chemoreceptors and transduced to the flagellar motor, modulating its CW/CCW bias, and enabling the bacterium to seek optimal habitats. Here, the motor is at the output end of the sensory response. Certain responses between the motor and bacterium play important roles in bacterial infection, surface colonization, persistence and pathogenesis. The Harshey Lab is currently elucidating this sensory mechanism. Lab scientists are also studying why swarming bacteria have a higher tolerance to antibiotics, and are using their knowledge of key motility mechanisms to design new antimicrobial targets.
 

Research


2024

  • Bhattacharyya S, Bhattarai N, Pfannenstiel DM, Wilkins B, Singh A, Harshey RM. Iron Memory in E. coli. bioRxiv [Preprint]. 2023 May 20:2023.05.19.541523. Update in: Proc Natl Acad Sci U S A. 2023 Nov 28;120(48):e2309082120. PMID: 37609133; PMCID: PMC10441380.
     

2023 


2022


2021


2020

Huibregtse Lab

About The Huibregtse Lab

The Huibregtse Lab studies the biochemistry of the ubiquitin proteolysis system and ISG15, a ubiquitin-like modifier involved in innate immune responses to viral and microbial infections. The Lab's current major iprojects include:

  • The mechanism and function of ISG15 conjugation.
  • ISG15's second biochemical function as an extracellular signaling protein.  
  • The role of the ubiquitin system in protein quality control, particularly the role of co-Translational Ubiquitination (CTU).  


Research


2024

2023 


2021


2020

Keiler Lab

About The Keiler Lab

The Keiler Lab's mission is to understand how protein quality control is maintained during stress responses and homeostasis. Most of our work addresses how stalled ribosomes are rescued by trans-translation and alternative pathways. Our goal is to characterize the fundamental biochemistry, genetics, and cell biology of these systems, and to use this knowledge to develop antibiotics and tools for basic research.

Research

2023

2022

2021

2020

  • Senges CHR, Stepanek JJ, Wenzel M, Raatschen N, Ay Ü, Märtens Y, Prochnow P, Vázquez Hernández M, Yayci A, Schubert B, Janzing NBM, Warmuth HL, Kozik M, Bongard J, Alumasa JN, Albada B, Penkova M, Lukežič T, Sorto NA, Lorenz N, Miller RG, Zhu B, Benda M, Stülke J, Schäkermann S, Leichert LI, Scheinpflug K, Brötz-Oesterhelt H, Hertweck C, Shaw JT, Petković H, Brunel JM, Keiler KC, Metzler-Nolte N, Bandow JE. Comparison of Proteomic Responses as Global Approach to Antibiotic Mechanism of Action Elucidation. Antimicrob Agents Chemother. 2020 Dec 16;65(1):e01373-20. PMID: 33046497; PMCID: PMC7927858.
Lu Lab

About The Lu Lab

The Lu Lab's research interests lie at the interface between chemistry and biology. Specific areas of current interests include a) computational design and engineering of functional metalloenzymes as environmentally benign catalysts in renewable energy generation and pharmaceuticals; b) Fundamental understanding of DNAzymes and their applications in environmental monitoring, medical diagnostics, gene editing and targeted drug delivery; and c) Employing principles from biology for directed assembly of nanomaterials with controlled morphologies and its applications in imaging and medicine.

Current projects of interests include a) computational design of artificial metalloenzymes employing machine learning methods for solar energy transfer, (bio)fuel cells, COreduction and nitrogen fixation; b) design of selective sensors and imaging agents for metal ions and metabolites to advance metallomics and metabolomics for medical diagnosis and imaging in neurodegenerative diseases and cancers; and c) developing novel gene-editing methods to improve sequence fidelity and gene target accessibility.


Research


2024

 

2023 


2022


2021


2020

Mavridou Lab

About The Mavridou Lab

The Mavridou Lab is interested in the biology of Gram-negative bacteria and more specifically in processes that occur in the extra-cytoplasmic environment. The Lab's approaches span from the fundamental to the applied and researchers currently focus on three main research directions:

  • Understanding the involvement of disulfide bond formation in mechanisms of virulence and adaptation in key Gram-negative bacterial pathogens;
  • Investigating whether proteins involved in cell envelope proteostasis could be potential targets for the treatment of antibiotic-resistant bacteria;
  • Studying the dynamics of bacterial communities and more specifically the competitive interactions between toxin-producing strains encountered in polymicrobial environments.


Research

 

2023 


2022


2021


2020

Maynard Lab

About The Maynard Lab

The Maynard Lab develops protein therapeutics to address unmet medical needs in infectious diseases. These proteins aim to directly interfere in disease progression or augment essential immune system activities. This work involves design of proteins with therapeutic potential, production in recombinant expression systems, biophysical and biochemical analyses to elucidate the molecular basis of activity and, ultimately, in vitro and in vivo experiments to evaluate a protein's therapeutic potential. 

Maynard Lab researchers look at the following:

  • Advanced antibody therapeutics: Discovery and design of antibodies endowed with novel capabilities, such as antibodies with conditional activity at the site of disease and antibodies that resist pathogen efforts to evade capture.
  • Vaccines that outsmart pathogens: Using what we learn from antibodies that prevent disease, especially antibodies that protect despite a pathogen's efforts to evade the immune system, we aim to design vaccine antigens that induce potently protective antibody responses. 
  • T cell-based therapeutics: Redirect cellular immune responses towards virally-infected cells through manipulation of T cell receptor-peptide MHC interactions.


Research

2023 


2022


2021


2020 

McLellan Lab

About The McLellan Lab

The human body is under constant attack from bacteria, fungi, parasites, and viruses, all of which express proteins that are needed to establish infection and evade the human immune system. The McLellan Lab seeks to obtain structural information on these proteins and their interactions with host macromolecules and translate this knowledge into the rational development of therapeutic interventions such as small-molecule inhibitors, protective antibodies and stabilized vaccine immunogens. These efforts are highly collaborative and involve domestic and international investigators from academia, government, and industry. The McLellan Lab focuses on the following:
 

  • Coronaviridae: Coronoviruses, such as SARS-CoV and MERS-CoV, are an important element of the McLellan Lab's research. One of their main goals is to provide a unifying structural framework for the function of coronavirus spike (S) proteins, including the mechanisms of receptor-induced triggering of membrane fusion. They are also leveraging the structural and mechanistic information to develop broadly protective antibodies and vaccines to combat current and emerging pathogenic coronaviruses.
     
  • Pneumoviridae: Understanding the entry process of Pneumoviruses, such as human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV), into host cells at a molecular level is another major focus of their work. They also have on-going projects involving structure-based vaccine design, antibody isolation and characterization, and development of next-generation small-molecule fusion inhibitors.

Research

2024

2023

2022

2021

2020

Mondy Research

2023


2022


2021

Molineux Lab

About The Mollineux Lab

The major research interest is understanding how nucleic acids pass through lipid bilayers. The Mollineux Lab's model system is bacteriophage T7, which uses three different motor proteins to transport its DNA into the cell at the initiation of infection. The phage ejects proteins into the cell that make a channel for DNA transport across the cell envelope and constitute the first molecular motor. Lab researchers are characterizing this channel and motor with the aim of reconstructing a DNA translocation system in vitro. The second and third motors are RNA polymerases that transport the T7 genome into the cell by transcription. The Lab collaborates with structural biologists who use electron cryo-tomography to obtain extremely high-resolution structures of intermediates of different phages during infection. 

Other interests include various host-parasite interactions; currently Mollineux Lab researchers are determining the molecular basis for exclusion of T7 by cells containing the F plasmid, evaluating phages for use in phage therapy protocols, using phages as models in evolutionary genetics, and phage genomics. A more specific project underway is to determine the mechanism by which transcription of the T7 genome is switched from catalysis by E. coli to the phage RNAP. Two T7 proteins: gp0.7 and gp2, are involved; in their absence the phage undergoes an abortive infection where the phage genome is degraded. The Mollineux Lab is analyzing rare mutants that bypass the requirement for these proteins, anticipating that its researchers will uncover a novel regulatory genetic network for phage development.


Research


2024

 

2019 

Moran Lab

About The Moran Lab


The Moran Lab's long-term interests are in the biology of symbiosis, particularly that between multicellular hosts and microbes. Symbioses are central in the evolution of complexity, have evolved many times and are critical to the lifestyles of many animals and plants and also to whole ecosystems, in which symbiotic organisms are key players. The primary reason that symbiosis research is suddenly active, after decades at the margins of mainstream biology, is that DNA technology and genomics give us enormous new ability to discover symbiont diversity, and more significantly, to reveal how microbial metabolic capabilities contribute to the functioning of hosts and biological communities.

Examples of the Moran Lab's ongoing projects include:

  • Phylogenetic, genomic and experimental studies of previously unstudied insect symbioses.
  • Study of aphid genes underlying carotenoid biosynthesis.
  • Experiments on the interdependent metabolic functioning of Buchnera and aphids.
  • Reconstruction of the evolutionary changes in genomes of bacterial symbionts.
  • Experimental investigations of facultative symbionts of aphids.
  • Studies of the functions, evolution and ecology of the bacteria living in the guts of honey bees and bumble bees.

Research

2024

2023 

2022

2021

2020

Ochman Lab

About the Ochman Lab


The Ochman Lab applies experimental, comparative and computational approaches to examine the evolution and adaptation of microbial genomes. Due to their complex and varied interactions as pathogens and commensal constituents of mammalian hosts, the Lab's work focuses on genome evolution within enteric bacteria, including E. coliShigella and Salmonella. These studies fall into four general areas:

  • Dynamics of bacterial genomes
  • Origins and roles of novel bacterial traits
  • Phylogenetic classification and the history of bacterial genomes
  • Genomic analysis of diversity within bacterial communities

Research

2024

2023 

2022 

2021

2020

Payne Lab

About The Payne Lab


The Payne Lab's studies focus on the genetics and regulation of iron acquisition systems and other virulence factors of the intestinal pathogens Shigella flexneri and Vibrio cholerae. The Lab's work combines biochemistry and genetics to determine how the essential metals iron and manganese are transported and utilized in V. cholerae and S. flexneri. Iron transport systems are of particular interest to Payne Lab researchers because iron acquisition in the human host is critical to microbial virulence. Specifically they are studying the bacterial Feo system for ferrous iron transport. Additionally, Lab scientists use in vitro models combined, such as enteroids, with genetic analyses to determine the role of host lipids and carbon metabolism in the pathogenesis of S. flexneri. Regarding S. flexneri, they also examine interspecies interactions between S. flexneri and normal members of human gut microbiota to determine how they  influence in S. flexneri growth and virulence. Finally, The Payne Lab is also exploring the environmental regulation of critical virulence factors by the RNA binding protein CsrA in V. cholerae.


Research

2024


2023


2022


2021


2020

Smyth Lab

About The Smyth Lab


The Smyth Lab research composes of Drug Delivery, Formulation Science, and Pharmaceutical Engineering. Researchers focus on the development of novel methods for drug delivery including inhalation, nasal, transdermal, ophthalmic, and oral delivery systems for a variety of diseases. Translation of these technologies to the clinic is the long-term goal of the Lab and is supported by developing a mechanistic understanding of the complex physical and biological systems.


Research


2024


2023 


2022


2021


2020

Sullivan Lab

About The Sullivan Lab


The discovery of silencing non-coding RNAs has dramatically changed our understanding of the regulation of gene expression. This has opened up a new area in the regulation of the immune response. The Sullivan Lab's research focuses on understanding the role of non-coding RNAs in virus infection and host defense pathways. Researchers work with an array of diverse virus families including DNA tumor viruses and RNA viruses, as well as the enzyme DUSP11. DUSP11 is an RNA phosphatase that helps to regulate the innate immune response. The Sullivan Lab's goals are several-fold:

  • To understand the functions of viral and host-encoded non-coding RNAs and how they contribute to viral lifecycle and pathogenesis
  • To explore the functions and regulation of immunogenic non-coding RNAs
  • To uncover new mechanisms of gene regulation utilized by viruses and the host
  • To use viruses as "molecular divining rods" to probe for new classes of host defense pathways.


Research


2024


2023


2022


2021


2020

Woznica Lab

About the Woznica Lab

The Woznica Lab investigates the molecular mechanisms by which choanoflagellates, the closest living relatives of animals, interact with their microbial world. Woznica Lab researchers study how choanoflagellates recognize and respond to bacteria and viruses in order to discover mechanisms of choanoflagellate immunity, and gain insight into the evolution of eukaryotic and animal immune pathways.

Research

2024

2021

2020

COVID-19 Research

Several members and affiliates of the LaMontagne Center have been publishing research related to COVID-19.
 

LCID Member Papers
In scientific journals, preprints, etc.

2024
  • Liang CY, Raju S, Liu Z, Li Y, Asthagiri Arunkumar G, Case JB, Scheaffer SM, Zost SJ, Acreman CM, Gagne M, Andrew SF, Carvalho Dos Anjos DC, Foulds KE, McLellan JS, Crowe JE Jr, Douek DC, Whelan SPJ, Elbashir SM, Edwards DK, Diamond MS. Imprinting of serum neutralizing antibodies by Wuhan-1 mRNA vaccines. Nature. 2024 May 15. Epub ahead of print. PMID: 38749479.
  • Gygi JP, Maguire C, Patel RK, Shinde P, Konstorum A, Shannon CP, Xu L, Hoch A, Jayavelu ND, Haddad EK; IMPACC Network; Reed EF, Kraft M, McComsey GA, Metcalf JP, Ozonoff A, Esserman D, Cairns CB, Rouphael N, Bosinger SE, Kim- Schulze S, Krammer F, Rosen LB, van Bakel H, Wilson M, Eckalbar WL, Maecker HT, Langelier CR, Steen H, Altman MC, Montgomery RR, Levy O, Melamed E, Pulendran B, Diray-Arce J, Smolen KK, Fragiadakis GK, Becker PM, Sekaly RP, Ehrlich LI, Fourati S, Peters B, Kleinstein SH, Guan L. Integrated longitudinal multiomics study identifies immune programs associated with acute COVID-19 severity and mortality. J Clin Invest. 2024 May 1;134(9):e176640. PMID: 38690733; PMCID: PMC11060740.
  • Hsieh CL, Leist SR, Miller EH, Zhou L, Powers JM, Tse AL, Wang A, West A, Zweigart MR, Schisler JC, Jangra RK, Chandran K, Baric RS, McLellan JS. Prefusion-stabilized SARS-CoV-2 S2-only antigen provides protection against SARS-CoV-2 challenge. Nat Commun. 2024 Feb 20;15(1):1553. PMID: 38378768; PMCID: PMC10879192.
  • Ozonoff A, Jayavelu ND, Liu S, Melamed E, Milliren CE, Qi J, Geng LN, McComsey GA, Cairns CB, Baden LR, Schaenman J, Shaw AC, Samaha H, Seyfert- Margolis V, Krammer F, Rosen LB, Steen H, Syphurs C, Dandekar R, Shannon CP, Sekaly RP, Ehrlich LIR, Corry DB, Kheradmand F, Atkinson MA, Brakenridge SC, Higuita NIA, Metcalf JP, Hough CL, Messer WB, Pulendran B, Nadeau KC, Davis MM, Sesma AF, Simon V, van Bakel H, Kim-Schulze S, Hafler DA, Levy O, Kraft M, Bime C, Haddad EK, Calfee CS, Erle DJ, Langelier CR, Eckalbar W, Bosinger SE; IMPACC Network; Peters B, Kleinstein SH, Reed EF, Augustine AD, Diray-Arce J, Maecker HT, Altman MC, Montgomery RR, Becker PM, Rouphael N. Features of acute COVID-19 associated with post-acute sequelae of SARS-CoV-2 phenotypes: results from the IMPACC study. Nat Commun. 2024 Jan 3;15(1):216. PMID: 38172101; PMCID: PMC10764789.
  • Johnson NV, Wall SC, Kramer KJ, Holt CM, Periasamy S, Richardson S, Suryadevara N, Andreano E, Paciello I, Pierleoni G, Piccini G, Huang Y, Ge P, Allen JD, Uno N, Shiakolas AR, Pilewski KA, Nargi RS, Sutton RE, Abu-Shmais AA, Parks R, Haynes BF, Carnahan RH, Crowe JE Jr, Montomoli E, Rappuoli R, Bukreyev A, Ross TM, Sautto GA, McLellan JS, Georgiev IS. Discovery and Characterization of a Pan-betacoronavirus S2-binding antibody. bioRxiv [Preprint]. 2024 Jan 16:2024.01.15.575741. PMID: 38293237; PMCID: PMC10827111.
2023

 

2022
2021
2020