Network


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

Hotspot


Dive into the research topics where John Pham is active.

Publication


Featured researches published by John Pham.


Nature | 2017

T cells from patients with Parkinson’s disease recognize α-synuclein peptides

David Sulzer; Roy N. Alcalay; Francesca Garretti; Lucien J. Cote; Ellen Kanter; Julian Agin-Liebes; Christopher Liong; Curtis McMurtrey; William H. Hildebrand; Xiaobo Mao; Valina L. Dawson; Ted M. Dawson; Carla Oseroff; John Pham; John Sidney; Myles B.C. Dillon; Chelsea Carpenter; Daniela Weiskopf; E. Phillips; S. Mallal; Bjoern Peters; April Frazier; Cecilia S. Lindestam Arlehamn; Alessandro Sette

Genetic studies have shown the association of Parkinson’s disease with alleles of the major histocompatibility complex. Here we show that a defined set of peptides that are derived from α-synuclein, a protein aggregated in Parkinson’s disease, act as antigenic epitopes displayed by these alleles and drive helper and cytotoxic T cell responses in patients with Parkinson’s disease. These responses may explain the association of Parkinson’s disease with specific major histocompatibility complex alleles.Genetic studies associate Parkinson’s disease with alleles of the major histocompatibility complex1–3. We find that a defined set of peptides derived from α-synuclein, a protein aggregated in Parkinson’s disease4, act as antigenic epitopes displayed by these alleles and drive helper and cytotoxic T cell responses in Parkinson’s disease patients. These responses may explain the association of Parkinson’s disease with alleles of the acquired immune system.


Journal of Virology | 2017

Prior Dengue Virus Exposure Shapes T Cell Immunity to Zika Virus in Humans

Alba Grifoni; John Pham; John Sidney; Patrick H. O'Rourke; Sinu Paul; Bjoern Peters; Sheridan R Martini; Aruna Dharshan De Silva; Michael J. Ricciardi; Diogo M. Magnani; Cassia G. T. Silveira; Alvino Maestri; Priscilla R. Costa; Luzia Maria de-Oliveira-Pinto; Elzinandes Leal de Azeredo; Paulo Vieira Damasco; E. Phillips; S. Mallal; Aravinda M. de Silva; Matthew Collins; Anna P. Durbin; Sean A. Diehl; Cristhiam Cerpas; Angel Balmaseda; Guillermina Kuan; Josefina Coloma; Eva Harris; James E. Crowe; Mars Stone; Phillip J. Norris

ABSTRACT While progress has been made in characterizing humoral immunity to Zika virus (ZIKV) in humans, little is known regarding the corresponding T cell responses to ZIKV. Here, we investigate the kinetics and viral epitopes targeted by T cells responding to ZIKV and address the critical question of whether preexisting dengue virus (DENV) T cell immunity modulates these responses. We find that memory T cell responses elicited by prior infection with DENV or vaccination with tetravalent dengue attenuated vaccines (TDLAV) recognize ZIKV-derived peptides. This cross-reactivity is explained by the sequence similarity of the two viruses, as the ZIKV peptides recognized by DENV-elicited memory T cells are identical or highly conserved in DENV and ZIKV. DENV exposure prior to ZIKV infection also influences the timing and magnitude of the T cell response. ZIKV-reactive T cells in the acute phase of infection are detected earlier and in greater magnitude in DENV-immune patients. Conversely, the frequency of ZIKV-reactive T cells continues to rise in the convalescent phase in DENV-naive donors but declines in DENV-preexposed donors, compatible with more efficient control of ZIKV replication and/or clearance of ZIKV antigen. The quality of responses is also influenced by previous DENV exposure, and ZIKV-specific CD8 T cells from DENV-preexposed donors selectively upregulated granzyme B and PD1, unlike DENV-naive donors. Finally, we discovered that ZIKV structural proteins (E, prM, and C) are major targets of both the CD4 and CD8 T cell responses, whereas DENV T cell epitopes are found primarily in nonstructural proteins. IMPORTANCE The issue of potential ZIKV and DENV cross-reactivity and how preexisting DENV T cell immunity modulates Zika T cell responses is of great relevance, as the two viruses often cocirculate and Zika virus has been spreading in geographical regions where DENV is endemic or hyperendemic. Our data show that memory T cell responses elicited by prior infection with DENV recognize ZIKV-derived peptides and that DENV exposure prior to ZIKV infection influences the timing, magnitude, and quality of the T cell response. Additionally, we show that ZIKV-specific responses target different proteins than DENV-specific responses, pointing toward important implications for vaccine design against this global threat.


Clinical & Experimental Allergy | 2015

Definition of a pool of epitopes that recapitulates the T cell reactivity against major house dust mite allergens

Denise Hinz; Carla Oseroff; John Pham; John Sidney; Bjoern Peters; Alessandro Sette

Allergens from house dust mites (HDM) are a common cause of asthma. Der p and Der f from Dermatophagoides sp. are strong immunogens in humans. Allergen extracts are used to study T helper (Th2) cell responses to HDM, which are implicated in the development and regulation of allergic disease.


Clinical & Experimental Allergy | 2016

Sequence conservation predicts T cell reactivity against ragweed allergens

John Pham; Carla Oseroff; Denise Hinz; John Sidney; Sinu Paul; Jason Greenbaum; Randi Vita; E. Phillips; S. Mallal; Bjoern Peters; Alessandro Sette

Ragweed is a major cause of seasonal allergy, affecting millions of people worldwide. Several allergens have been defined based on IgE reactivity, but their relative immunogenicity in terms of T cell responses has not been studied.


Clinical & Experimental Allergy | 2017

Immunoproteomic analysis of house dust mite antigens reveals distinct classes of dominant T cell antigens according to function and serological reactivity

Carla Oseroff; Lars Harder Christensen; Luise Westernberg; John Pham; Jerome Lane; Sinu Paul; Jason Greenbaum; Thomas Stranzl; G. Lund; Ilka Hoof; Jens Holm; Peter Adler Würtzen; K. Meno; April Frazier; Véronique Schulten; Peter S. Andersen; Bjoern Peters; Alessandro Sette

House dust mite (HDM) allergens are a common cause of allergy and allergic asthma. A comprehensive analysis of proteins targeted by T cells, which are implicated in the development and regulation of allergic disease independent of their antibody reactivity, is still lacking.


PLOS Neglected Tropical Diseases | 2017

Ontogeny of the B- and T-cell response in a primary Zika virus infection of a dengue-naive individual during the 2016 outbreak in Miami, FL

Michael J. Ricciardi; Diogo M. Magnani; Alba Grifoni; Young-Chan Kwon; Martin J. Gutman; Nathan D. Grubaugh; Karthik Gangavarapu; Mark Sharkey; Cassia G. T. Silveira; Varian K. Bailey; Núria Pedreño-Lopez; Lucas Gonzalez-Nieto; Helen S. Maxwell; Aline Domingues; Mauricio A. Martins; John Pham; Daniela Weiskopf; John D. Altman; Esper G. Kallas; Kristian G. Andersen; Mario Stevenson; Paola Lichtenberger Lichtenberger; Hyeryun Choe; Stephen S. Whitehead; Alessandro Sette; David I. Watkins

Zika virus (ZIKV) is a mosquito-borne flavivirus of significant public health concern. In the summer of 2016, ZIKV was first detected in the contiguous United States. Here we present one of the first cases of a locally acquired ZIKV infection in a dengue-naïve individual. We collected blood from a female with a maculopapular rash at day (D) 5 and D7 post onset of symptoms (POS) and we continued weekly blood draws out to D148 POS. To establish the ontogeny of the immune response against ZIKV, lymphocytes and plasma were analyzed in a longitudinal fashion. The plasmablast response peaked at D7 POS (19.6% of CD19+ B-cells) and was undetectable by D15 POS. ZIKV-specific IgM was present at D5 POS, peaked between D15 and D21 POS, and subsequently decreased. The ZIKV-specific IgG response, however, was not detected until D15 POS and continued to increase after that. Interestingly, even though the patient had never been infected with dengue virus (DENV), cross-reactive IgM and IgG binding against each of the four DENV serotypes could be detected. The highest plasma neutralization activity against ZIKV peaked between D15 and D21 POS, and even though DENV binding antibodies were present in the plasma of the patient, there was neither neutralization nor antibody dependent enhancement (ADE) of DENV. Interestingly, ADE against ZIKV arose at D48 POS and continued until the end of the study. CD4+ and CD8+ T-cells recognized ZIKV-NS2A and ZIKV-E, respectively. The tetramer positive CD8+ T-cell response peaked at D21 POS with elevated levels persisting for months. In summary, this is the first study to establish the timing of the ontogeny of the immune response against ZIKV.


Immunology | 2018

Development of a strategy and computational application to select candidate protein analogues with reduced HLA binding and immunogenicity

Sandeep Kumar Dhanda; Alba Grifoni; John Pham; Kerrie Vaughan; John Sidney; Bjoern Peters; Alessandro Sette

Unwanted immune responses against protein therapeutics can reduce efficacy or lead to adverse reactions. T‐cell responses are key in the development of such responses, and are directed against immunodominant regions within the protein sequence, often associated with binding to several allelic variants of HLA class II molecules (promiscuous binders). Herein, we report a novel computational strategy to predict ‘de‐immunized’ peptides, based on previous studies of erythropoietin protein immunogenicity. This algorithm (or method) first predicts promiscuous binding regions within the target protein sequence and then identifies residue substitutions predicted to reduce HLA binding. Further, this method anticipates the effect of any given substitution on flanking peptides, thereby circumventing the creation of nascent HLA‐binding regions. As a proof‐of‐principle, the algorithm was applied to Vatreptacog α, an engineered Factor VII molecule associated with unintended immunogenicity. The algorithm correctly predicted the two immunogenic peptides containing the engineered residues. As a further validation, we selected and evaluated the immunogenicity of seven substitutions predicted to simultaneously reduce HLA binding for both peptides, five control substitutions with no predicted reduction in HLA‐binding capacity, and additional flanking region controls. In vitro immunogenicity was detected in 21·4% of the cultures of peptides predicted to have reduced HLA binding and 11·4% of the flanking regions, compared with 46% for the cultures of the peptides predicted to be immunogenic. This method has been implemented as an interactive application, freely available online at http://tools.iedb.org/deimmunization/.


Annals of Allergy Asthma & Immunology | 2016

Immunodominance in allergic T-cell reactivity to Japanese cedar in different geographic cohorts

Carla Oseroff; John Pham; April Frazier; Denise Hinz; John Sidney; Sinu Paul; Jason Greenbaum; Randi Vita; Bjoern Peters; Véronique Schulten; Alessandro Sette

BACKGROUND Japanese cedar (JC) pollen is a common trigger for allergic rhinitis in Japan. Pollen proteins targeted by IgE, including Cry j 1 and Cry j 2, and isoflavone reductase (IFR) have been identified. OBJECTIVE To compare antigen-specific IgE titers and T-cell responses to JC pollen-derived extract and peptides in cohorts with high and low pollen exposure. METHODS Peripheral blood mononuclear cells from JC pollen allergic or nonallergic patients who have lived in Japan for at least 1 year and JC pollen allergic patients who have never been to Japan were tested for T-cell responses against JC pollen extract and peptide pools derived from Cry j 1, Cry j 2, or IFR. T-cell reactivity was assessed by interleukin 5 and interferon γ production by ELISPOT. RESULTS JC pollen-specific T-cell reactivity and IgE titers were significantly higher in the allergic compared with the nonallergic Japanese cohort, which was also associated with different patterns of polysensitization. Interestingly, a significant overlap was observed in the hierarchy of the T-cell epitopes in the allergic Japanese cohort compared with the allergic non-Japanese cohort. In all 3 cohorts, T-cell reactivity was dominantly directed against peptides from the major allergens Cry j 1 and 2, with few T-cell responses detected against IFR. CONCLUSION Our studies identify common denominators of T-cell reactivity in patient populations with different sensitization patterns, suggesting that generally applicable immunotherapeutic approaches might be developed irrespective of exposure modality.


PLOS ONE | 2018

Peanut-specific T cell responses in patients with different clinical reactivity

Giovanni Birrueta; Victoria Tripple; John Pham; Monali Manohar; Eddie A. James; William W. Kwok; Kari C. Nadeau; Alessandro Sette; Bjoern Peters; Véronique Schulten

Whole extract or allergen-specific IgE testing has become increasingly popular in the diagnosis of peanut allergy. However, much less is known about T cell responses in peanut allergy and how it relates to different clinical phenotypes. CD4+ T cells play a major role in the pathophysiology of peanut allergy as well as tolerance induction during oral desensitization regimens. We set out to characterize and phenotype the T cell responses and their targets in peanut sensitized patients. Using PBMC from peanut-allergic and non-allergic patients, we mapped T cell epitopes for three major peanut allergens, Ara h 1, 2 and 3 (27 from Ara h 1, 4 from Ara h 2 and 43 from Ara h 3) associated with release of IFNγ (representative Th1 cytokine) and IL5 (representative Th2 cytokine). A pool containing 19 immunodominant peptides, selected to account for 60% of the total Ara h 1-3-specific T cell response in allergics, but only 20% in non-allergics, was shown to discriminate T cell responses in peanut-sensitized, symptomatic vs non-symptomatic individuals more effectively than peanut extract. This pool elicited positive T cell responses above a defined threshold in 12/15 sensitized, symptomatic patients, whereas in the sensitized but non-symptomatic cohort only, 4/14 reacted. The reactivity against this peptide pool in symptomatic patients was dominated by IL-10, IL-17 and to a lesser extend IL-5. For four distinct epitopes, HLA class II restrictions were determined, enabling production of tetrameric reagents. Tetramer staining in four donors (2 symptomatic, 2 non-symptomatic) revealed a trend for increased numbers of peanut epitope-specific T cells in symptomatic patients compared to non-symptomatic patients, which was associated with elevated CRTh2 expression whereas cells from non-symptomatic patients exhibited higher levels of Integrin β7 expression. Our results demonstrate differences in T cell response magnitude, epitope specificity and phenotype between symptomatic and non-symptomatic peanut-sensitized patients. In addition to IgE reactivity, analysis of peanut-specific T cells may be useful to improve our understanding of different clinical manifestations in peanut allergy.


Human Immunology | 2018

Sequence-based HLA-A, B, C, DP, DQ, and DR typing of 496 adults from San Diego, California, USA

Eugene Moore; Alba Grifoni; Daniela Weiskopf; Véronique Schulten; Cecilia S. Lindestam Arlehamn; Michael A. Angelo; John Pham; S. Leary; John Sidney; David H. Broide; April Frazier; E. Phillips; S. Mallal; Steven J. Mack; Alessandro Sette

DNA sequence-based typing at the HLA-A, -B, -C, -DPB1, -DQA1, -DQB1, and -DRB1 loci was performed on 496 healthy adult donors from San Diego, California, to characterize allele frequencies in support of studies of T cell responses to common allergens. Deviations from Hardy Weinberg proportions were detected at each locus except A and C. Several alleles were found in more than 15% of individuals, including the class II alleles DPB1∗02:01, DPB1∗04:01, DQA1∗01:02, DQA1∗05:01, DQB1∗03:01, and the class I allele A∗02:01. Genotype data will be available in the Allele Frequencies Net Database (AFND 3562).

Collaboration


Dive into the John Pham's collaboration.

Top Co-Authors

Avatar

Alessandro Sette

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Bjoern Peters

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

John Sidney

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Carla Oseroff

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Véronique Schulten

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

April Frazier

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sinu Paul

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alba Grifoni

La Jolla Institute for Allergy and Immunology

View shared research outputs
Researchain Logo
Decentralizing Knowledge