Network


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

Hotspot


Dive into the research topics where Summer Park is active.

Publication


Featured researches published by Summer Park.


Nature | 2015

Novel antibody–antibiotic conjugate eliminates intracellular S. aureus

Sophie M. Lehar; Thomas H. Pillow; Min Xu; Leanna Staben; Kimberly Kajihara; Richard Vandlen; Laura DePalatis; Helga Raab; Wouter L. W. Hazenbos; J. Hiroshi Morisaki; Janice Kim; Summer Park; Martine Darwish; Byoung-Chul Lee; Hilda Hernandez; Kelly M. Loyet; Patrick Lupardus; Rina Fong; Donghong Yan; Cecile Chalouni; Elizabeth Luis; Yana Khalfin; Emile Plise; Jonathan Cheong; Joseph P. Lyssikatos; Magnus Strandh; Klaus Koefoed; Peter S. Andersen; John A. Flygare; Man Wah Tan

Staphylococcus aureus is considered to be an extracellular pathogen. However, survival of S. aureus within host cells may provide a reservoir relatively protected from antibiotics, thus enabling long-term colonization of the host and explaining clinical failures and relapses after antibiotic therapy. Here we confirm that intracellular reservoirs of S. aureus in mice comprise a virulent subset of bacteria that can establish infection even in the presence of vancomycin, and we introduce a novel therapeutic that effectively kills intracellular S. aureus. This antibody–antibiotic conjugate consists of an anti-S. aureus antibody conjugated to a highly efficacious antibiotic that is activated only after it is released in the proteolytic environment of the phagolysosome. The antibody–antibiotic conjugate is superior to vancomycin for treatment of bacteraemia and provides direct evidence that intracellular S. aureus represents an important component of invasive infections.


Nature Immunology | 2014

Transcriptional programming of dendritic cells for enhanced MHC class II antigen presentation

Bryan Vander Lugt; Aly A. Khan; Jason A. Hackney; Smita Agrawal; Justin Lesch; Meijuan Zhou; Wyne P. Lee; Summer Park; Min Xu; Jason DeVoss; Chauncey J. Spooner; Cecile Chalouni; Lélia Delamarre; Ira Mellman; Harinder Singh

CD11b+ dendritic cells (DCs) seem to be specialized for presenting antigens via major histocompatibility (MHC) class II complexes to stimulate helper T cells, but the genetic and regulatory basis for this is not established. Conditional deletion of Irf4 resulted in loss of CD11b+ DCs, impaired formation of peptide–MHC class II complexes and defective priming of helper T cells but not of cytotoxic T lymphocyte (CTL) responses. Gene expression and chromatin immunoprecipitation followed by deep sequencing (ChIP-Seq) analyses delineated an IRF4-dependent regulatory module that programs enhanced MHC class II antigen presentation. Expression of the transcription factor IRF4 but not of IRF8 restored the ability of IRF4-deficient DCs to efficiently process and present antigen to MHC class II–restricted T cells and promote helper T cell responses. We propose that the evolutionary divergence of IRF4 and IRF8 facilitated the specialization of DC subsets for distinct modes of antigen presentation and priming of helper T cell versus CTL responses.


The Journal of Infectious Diseases | 2014

Global Gene Expression of Methicillin-resistant Staphylococcus aureus USA300 During Human and Mouse Infection

Shailesh V. Date; Zora Modrusan; Michael S. Lawrence; J. Hiroshi Morisaki; Karen Toy; Ishita M. Shah; Janice Kim; Summer Park; Min Xu; Li Basuino; Liana Chan; Deborah Zeitschel; Henry F. Chambers; Man-Wah Tan; Eric J. Brown; Binh An Diep; Wouter L. W. Hazenbos

Little is known about the expression of methicillin-resistant Staphylococcus aureus (MRSA) genes during infection conditions. Here, we described the transcriptome of the clinical MRSA strain USA300 derived from human cutaneous abscesses, and compared it with USA300 bacteria derived from infected kidneys in a mouse model. Remarkable similarity between the transcriptomes allowed us to identify genes encoding multiple proteases and toxins, and iron- and peptide-transporter molecules, which are upregulated in both infections and are likely important for establishment of infection. We also showed that disruption of the global transcriptional regulators agr and sae prevents in vivo upregulation of many toxins and proteases, protecting mice from lethal infection dose, and hinting at the role of these transcriptional regulators in the pathology of MRSA infection.


PLOS Pathogens | 2016

Two Escape Mechanisms of Influenza A Virus to a Broadly Neutralizing Stalk-Binding Antibody

Ning Chai; Lee R. Swem; Mike Reichelt; Haiyin Chen-Harris; Elizabeth Luis; Summer Park; Ashley E. Fouts; Patrick Lupardus; Thomas D. Wu; Olga Li; Jacqueline McBride; Michael S. Lawrence; Min Xu; Man-Wah Tan

Broadly neutralizing antibodies targeting the stalk region of influenza A virus (IAV) hemagglutinin (HA) are effective in blocking virus infection both in vitro and in vivo. The highly conserved epitopes recognized by these antibodies are critical for the membrane fusion function of HA and therefore less likely to be permissive for virus mutational escape. Here we report three resistant viruses of the A/Perth/16/2009 strain that were selected in the presence of a broadly neutralizing stalk-binding antibody. The three resistant viruses harbor three different mutations in the HA stalk: (1) Gln387Lys; (2) Asp391Tyr; (3) Asp391Gly. The Gln387Lys mutation completely abolishes binding of the antibody to the HA stalk epitope. The other two mutations, Asp391Tyr and Asp391Gly, do not affect antibody binding at neutral pH and only slightly reduce binding at low pH. Interestingly, they enhance the fusion ability of the HA, representing a novel mechanism that allows productive membrane fusion even in the presence of antibody and hence virus escape from antibody neutralization. Therefore, these mutations illustrate two different resistance mechanisms used by IAV to escape broadly neutralizing stalk-binding antibodies. Compared to the wild type virus, the resistant viruses release fewer progeny viral particles during replication and are more sensitive to Tamiflu, suggesting reduced viral fitness.


mAbs | 2016

Preclinical pharmacokinetics of MHAA4549A, a human monoclonal antibody to influenza A virus, and the prediction of its efficacious clinical dose for the treatment of patients hospitalized with influenza A

Priyanka Gupta; Amrita V. Kamath; Summer Park; Henry Chiu; Jeff Lutman; Mauricio Maia; Man-Wah Tan; Min Xu; Lee R. Swem; Rong Deng

ABSTRACT MHAA4549A is a human immunoglobulin G1 (IgG1) monoclonal antibody that binds to a highly conserved epitope on the stalk of influenza A hemagglutinin and blocks the hemagglutinin-mediated membrane fusion in the endosome, neutralizing all known human influenza A strains. Pharmacokinetics (PK) of MHAA4549A and its related antibodies were determined in DBA/2J and Balb-c mice at 5 mg/kg and in cynomolgus monkeys at 5 and 100 mg/kg as a single intravenous dose. Serum samples were analyzed for antibody concentrations using an ELISA and the PK was evaluated using WinNonlin software. Human PK profiles were projected based on the PK in monkeys using species-invariant time method. The human efficacious dose projection was based on in vivo nonclinical pharmacological active doses, exposure in mouse infection models and expected human PK. The PK profiles of MHAA4549A and its related antibody showed a linear bi-exponential disposition in mice and cynomolgus monkeys. In mice, clearance and half-life ranged from 5.77 to 9.98 mL/day/kg and 10.2 to 5.76 days, respectively. In cynomolgus monkeys, clearance and half-life ranged from 4.33 to 4.34 mL/day/kg and 11.3 to 11.9 days, respectively. The predicted clearance in humans was ∼2.60 mL/day/kg. A single intravenous dose ranging from 15 to 45 mg/kg was predicted to achieve efficacious exposure in humans. In conclusion, the PK of MHAA4549A was as expected for a human IgG1 monoclonal antibody that lacks known endogenous host targets. The predicted clearance and projected efficacious doses in humans for MHAA4549A have been verified in a Phase 1 study and Phase 2a study, respectively.


Infection and Immunity | 2012

Mycobacterium marinum SecA2 Promotes Stable Granulomas and Induces Tumor Necrosis Factor Alpha In Vivo

Brigitte Y. Watkins; Shilpa A. Joshi; David A. Ball; Hadley Leggett; Summer Park; Janice Kim; Cary D. Austin; Andres Paler-Martinez; Min Xu; Kenneth H. Downing; Eric J. Brown

ABSTRACT SecA2 is an ATPase present in some pathogenic Gram-positive bacteria, is required for translocation of a limited set of proteins across the cytosolic membrane, and plays an important role in virulence in several bacteria, including mycobacteria that cause diseases such as tuberculosis and leprosy. However, the mechanisms by which SecA2 affects virulence are incompletely understood. To investigate whether SecA2 modulates host immune responses in vivo, we studied Mycobacterium marinum infection in two different hosts: an established zebrafish model and a recently described mouse model. Here we show that M. marinum ΔsecA2 was attenuated for virulence in both host species and SecA2 was needed for normal granuloma numbers and for optimal tumor necrosis factor alpha response in both zebrafish and mice. M. marinum ΔsecA2 was more sensitive to SDS and had unique protrusions from its cell envelope when examined by cryo-electron tomography, suggesting that SecA2 is important for bacterial cell wall integrity. These results provide evidence that SecA2 induces granulomas and is required for bacterial modulation of the host response because it affects the mycobacterial cell envelope.


Nature Communications | 2017

A broadly protective therapeutic antibody against influenza B virus with two mechanisms of action

Ning Chai; Lee R. Swem; Summer Park; Gerald R. Nakamura; Nan Chiang; Alberto Estevez; Rina Fong; Lynn Kamen; Elviza Kho; Mike Reichelt; Zhonghua Lin; Henry Chiu; Elizabeth Skippington; Zora Modrusan; Jeremy Stinson; Min Xu; Patrick Lupardus; Claudio Ciferri; Man-Wah Tan

Influenza B virus (IBV) causes annual influenza epidemics around the world. Here we use an in vivo plasmablast enrichment technique to isolate a human monoclonal antibody, 46B8 that neutralizes all IBVs tested in vitro and protects mice against lethal challenge of all IBVs tested when administered 72 h post infection. 46B8 demonstrates a superior therapeutic benefit over Tamiflu and has an additive antiviral effect in combination with Tamiflu. 46B8 binds to a conserved epitope in the vestigial esterase domain of hemagglutinin (HA) and blocks HA-mediated membrane fusion. After passage of the B/Brisbane/60/2008 virus in the presence of 46B8, we isolated three resistant clones, all harbouring the same mutation (Ser301Phe) in HA that abolishes 46B8 binding to HA at low pH. Interestingly, 46B8 is still able to protect mice against lethal challenge of the mutant viruses, possibly owing to its ability to mediate antibody-dependent cellular cytotoxicity (ADCC).


PLOS ONE | 2015

The Staphylococcus aureus ABC-Type Manganese Transporter MntABC Is Critical for Reinitiation of Bacterial Replication Following Exposure to Phagocytic Oxidative Burst.

Alison Coady; Min Xu; Qui Phung; Tommy K. Cheung; Corey E. Bakalarski; Mary Kate Alexander; Sophie M. Lehar; Janice Kim; Summer Park; Man-Wah Tan; Mireille Nishiyama

Manganese plays a central role in cellular detoxification of reactive oxygen species (ROS). Therefore, manganese acquisition is considered to be important for bacterial pathogenesis by counteracting the oxidative burst of phagocytic cells during host infection. However, detailed analysis of the interplay between bacterial manganese acquisition and phagocytic cells and its impact on bacterial pathogenesis has remained elusive for Staphylococcus aureus, a major human pathogen. Here, we show that a mntC mutant, which lacks the functional manganese transporter MntABC, was more sensitive to killing by human neutrophils but not murine macrophages, unless the mntC mutant was pre-exposed to oxidative stress. Notably, the mntC mutant formed strikingly small colonies when recovered from both type of phagocytic cells. We show that this phenotype is a direct consequence of the inability of the mntC mutant to reinitiate growth after exposure to phagocytic oxidative burst. Transcript and quantitative proteomics analyses revealed that the manganese-dependent ribonucleotide reductase complex NrdEF, which is essential for DNA synthesis and repair, was highly induced in the mntC mutant under oxidative stress conditions including after phagocytosis. Since NrdEF proteins are essential for S. aureus viability we hypothesize that cells lacking MntABC might attempt to compensate for the impaired function of NrdEF by increasing their expression. Our data suggest that besides ROS detoxification, functional manganese acquisition is likely crucial for S. aureus pathogenesis by repairing oxidative damages, thereby ensuring efficient bacterial growth after phagocytic oxidative burst, which is an attribute critical for disseminating and establishing infection in the host.


Key Engineering Materials | 2007

In Vitro Evaluation of Poly ε-Caprolactone/Hydroxyapatite Composite as Scaffolds for Bone Tissue Engineering with Human Bone Marrow Stromal Cells

Su-Jin Heo; Seong-Il Kim; Yong Taek Hyun; Dong Hwa Kim; Hyang Mi Lee; Yeong Maw Hwang; Summer Park; Jung Woog Shin

This study evaluated the potential of the PCL (poly -caprolactone)/HA(Hydroxyapatite) composite materials as a scaffold for bone regeneration. For this, we fabricated scaffolds utilizing salt leaching method. The PCL/HA composite scaffolds were prepared with various HA contents (20wt%, 40wt%, 60 wt %). To ensure the potential for the scaffolds, porosity tests were conducted along with SEM observations. The porosity decreased with the increase of the contents of HA particles. The porosity of the composite with the highest contents of HA was still adoptable (~85%). In addition, the PCL/HA composite scaffolds were evaluated for their ability of osteogenic differentiation with human bone marrow stromal cell (hBMSC) in vitro. Alkaline phosphatase (ALP) activity, markers for osteoblastic differentiation, and total protein contents were evaluated in hBMSCs following 14 days of cultivation. The addition of HA particles enhanced proliferation of hBMSC during the test. Also, the differentiation ability of the cells was increased as HA particles were added. In this study, we concluded that PCL/HA composite scaffolds has great potential as a scaffold for bone tissue engineering.


Nature | 2018

Optimized arylomycins are a new class of Gram-negative antibiotics

Peter A. Smith; Michael F. T. Koehler; Hany S. Girgis; Donghong Yan; Yongsheng Chen; Yuan Chen; James J. Crawford; Matthew Durk; Robert I. Higuchi; Jing Kang; Jeremy Murray; Prasuna Paraselli; Summer Park; Wilson Phung; John Quinn; Tucker C. Roberts; Lionel Rouge; Jacob B. Schwarz; Elizabeth Skippington; John Wai; Min Xu; Zhiyong Yu; Hua Zhang; Man-Wah Tan; Christopher E. Heise

Multidrug-resistant bacteria are spreading at alarming rates, and despite extensive efforts no new class of antibiotic with activity against Gram-negative bacteria has been approved in over fifty years. Natural products and their derivatives have a key role in combating Gram-negative pathogens. Here we report chemical optimization of the arylomycins—a class of natural products with weak activity and limited spectrum—to obtain G0775, a molecule with potent, broad-spectrum activity against Gram-negative bacteria. G0775 inhibits the essential bacterial type I signal peptidase, a new antibiotic target, through an unprecedented molecular mechanism. It circumvents existing antibiotic resistance mechanisms and retains activity against contemporary multidrug-resistant Gram-negative clinical isolates in vitro and in several in vivo infection models. These findings demonstrate that optimized arylomycin analogues such as G0775 could translate into new therapies to address the growing threat of multidrug-resistant Gram-negative infections.Chemical optimization of arylomycins results in an inhibitor of bacterial type I signal peptidase that shows activity both against multidrug-resistant clinical isolates of Gram-negative bacteria in vitro and in several in vivo infection models.

Collaboration


Dive into the Summer Park's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nan Chiang

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge