Network


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

Hotspot


Dive into the research topics where Vanessa Venturi is active.

Publication


Featured researches published by Vanessa Venturi.


Journal of Immunology | 2011

A mechanism for TCR sharing between T cell subsets and individuals revealed by pyrosequencing.

Vanessa Venturi; Máire F. Quigley; Hui Yee Greenaway; Pauline C. Ng; Zachary Ende; Tina McIntosh; Tedi E. Asher; Jorge R. Almeida; Samuel Levy; David A. Price; Miles P. Davenport

The human naive T cell repertoire is the repository of a vast array of TCRs. However, the factors that shape their hierarchical distribution and relationship with the memory repertoire remain poorly understood. In this study, we used polychromatic flow cytometry to isolate highly pure memory and naive CD8+ T cells, stringently defined with multiple phenotypic markers, and used deep sequencing to characterize corresponding portions of their respective TCR repertoires from four individuals. The extent of interindividual TCR sharing and the overlap between the memory and naive compartments within individuals were determined by TCR clonotype frequencies, such that higher-frequency clonotypes were more commonly shared between compartments and individuals. TCR clonotype frequencies were, in turn, predicted by the efficiency of their production during V(D)J recombination. Thus, convergent recombination shapes the TCR repertoire of the memory and naive T cell pools, as well as their interrelationship within and between individuals.


Journal of Experimental Medicine | 2009

Public clonotype usage identifies protective Gag-specific CD8+ T cell responses in SIV infection

David A. Price; Tedi E. Asher; Nancy A. Wilson; Martha Nason; Jason M. Brenchley; Ian Metzler; Vanessa Venturi; Emma Gostick; Pratip K. Chattopadhyay; Mario Roederer; Miles P. Davenport; David I. Watkins; Daniel C. Douek

Despite the pressing need for an AIDS vaccine, the determinants of protective immunity to HIV remain concealed within the complexity of adaptive immune responses. We dissected immunodominant virus-specific CD8+ T cell populations in Mamu-A*01+ rhesus macaques with primary SIV infection to elucidate the hallmarks of effective immunity at the level of individual constituent clonotypes, which were identified according to the expression of distinct T cell receptors (TCRs). The number of public clonotypes, defined as those that expressed identical TCR β-chain amino acid sequences and recurred in multiple individuals, contained within the acute phase CD8+ T cell population specific for the biologically constrained Gag CM9 (CTPYDINQM; residues 181–189) epitope correlated negatively with the virus load set point. This independent molecular signature of protection was confirmed in a prospective vaccine trial, in which clonotype engagement was governed by the nature of the antigen rather than the context of exposure and public clonotype usage was associated with enhanced recognition of epitope variants. Thus, the pattern of antigen-specific clonotype recruitment within a protective CD8+ T cell population is a prognostic indicator of vaccine efficacy and biological outcome in an AIDS virus infection.


Journal of Virology | 2011

High-Functional-Avidity Cytotoxic T Lymphocyte Responses to HLA-B-Restricted Gag-Derived Epitopes Associated with Relative HIV Control

Christoph T. Berger; Nicole Frahm; David A. Price; Beatriz Mothe; Musie Ghebremichael; Kari L. Hartman; Leah M. Henry; Jason M. Brenchley; Laura E. Ruff; Vanessa Venturi; Florencia Pereyra; John Sidney; Alessandro Sette; Bruce D. Walker; Daniel E. Kaufmann; Christian Brander

ABSTRACT Virus-specific cytotoxic T lymphocytes (CTL) with high levels of functional avidity have been associated with viral clearance in hepatitis C virus infection and with enhanced antiviral protective immunity in animal models. However, the role of functional avidity as a determinant of HIV-specific CTL efficacy remains to be assessed. Here we measured the functional avidities of HIV-specific CTL responses targeting 20 different, optimally defined CTL epitopes restricted by 13 different HLA class I alleles in a cohort comprising 44 HIV controllers and 68 HIV noncontrollers. Responses restricted by HLA-B alleles and responses targeting epitopes located in HIV Gag exhibited significantly higher functional avidities than responses restricted by HLA-A or HLA-C molecules (P = 0.0003) or responses targeting epitopes outside Gag (P < 0.0001). The functional avidities of Gag-specific and HLA-B-restricted responses were higher in HIV controllers than in noncontrollers (P = 0.014 and P = 0.018) and were not restored in HIV noncontrollers initiating antiretroviral therapy. T-cell receptor (TCR) analyses revealed narrower TCR repertoires in higher-avidity CTL populations, which were dominated by public TCR sequences in HIV controllers. Together, these data link the presence of high-avidity Gag-specific and HLA-B-restricted CTL responses with viral suppression in vivo and provide new insights into the immune parameters that mediate spontaneous control of HIV infection.


Immunity | 2013

A Molecular Basis for the Control of Preimmune Escape Variants by HIV-Specific CD8(+) T Cells.

Kristin Ladell; Masao Hashimoto; Maria Candela Iglesias; Pascal G. Wilmann; James Edward McLaren; Stephanie Gras; Takayuki Chikata; Nozomi Kuse; Solène Fastenackels; Emma Gostick; John S. Bridgeman; Vanessa Venturi; Zaïna Aït Arkoub; Henri Agut; David van Bockel; Jorge R. Almeida; Laurence Meyer; Alain Venet; Masafumi Takiguchi; Jamie Rossjohn; David A. Price; Victor Appay

The capacity of the immune system to adapt to rapidly evolving viruses is a primary feature of effective immunity, yet its molecular basis is unclear. Here, we investigated protective HIV-1-specific CD8+ T cell responses directed against the immunodominant p24 Gag-derived epitope KK10 (KRWIILGLNK263-272) presented by human leukocyte antigen (HLA)-B∗2705. We found that cross-reactive CD8+ T cell clonotypes were mobilized to counter the rapid emergence of HIV-1 variants that can directly affect T cell receptor (TCR) recognition. These newly recruited clonotypes expressed TCRs that engaged wild-type and mutant KK10 antigens with similar affinities and almost identical docking modes, thereby accounting for their antiviral efficacy in HLA-B∗2705+ individuals. A protective CD8+ T cell repertoire therefore encompasses the capacity to control TCR-accessible mutations, ultimately driving the development of more complex viral escape variants that disrupt antigen presentation.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Sharing of T cell receptors in antigen-specific responses is driven by convergent recombination

Vanessa Venturi; Katherine Kedzierska; David A. Price; Peter C. Doherty; Stephen J. Turner; Miles P. Davenport

Public responses where identical T cell receptors (TCRs) are clonally dominant and shared between different individuals are a common characteristic of CD8+ T cell-mediated immunity. Focusing on TCR sharing, we analyzed ≈3,400 TCR β chains (TCRβs) from mouse CD8+ T cells responding to the influenza A virus DbNP366 and DbPA224 epitopes. Both the “public” DbNP366-specific and “private” DbPA224-specific TCR repertoires contain a high proportion (≈36%) of shared TCRβs, although the numbers of mice sharing TCRβs in each repertoire varies greatly. Sharing of both the TCRβ amino acid and TCRβ nucleotide sequence was negatively correlated with the prevalence of random nucleotide additions in the sequence. However, the extent of TCRβ amino acid sequence sharing among mice was strongly correlated with the level of diversity in the encoding nucleotide sequences, suggesting that a key feature of public TCRs is that they can be made in a variety of ways. Using a computer simulation of random V(D)J recombination, we estimated the relative production frequencies and variety of production mechanisms for TCRβ sequences and found strong correlations with the sharing of both TCRβ amino acid sequences and TCRβ nucleotide sequences. The overall conclusion is that “convergent recombination,” rather than a bias in recombination or subsequent selection, provides the mechanistic basis for TCR sharing between individuals responding to identical peptide plus MHC class I glycoprotein complexes.


Immunology and Cell Biology | 2015

Naive CD8+ T-cell precursors display structured TCR repertoires and composite antigen-driven selection dynamics

Michelle A. Neller; Kristin Ladell; James Edward McLaren; Katherine K. Matthews; Emma Gostick; Johanne M. Pentier; Garry Dolton; Andrea J. A. Schauenburg; Dan Koning; Ana I. Costa; Thomas S. Watkins; Vanessa Venturi; Corey Smith; Rajiv Khanna; Kelly Louise Miners; Mathew Clement; Linda Wooldridge; David R. Cole; Debbie van Baarle; Andrew K. Sewell; Scott R. Burrows; David A. Price; John J. Miles

Basic parameters of the naive antigen (Ag)‐specific T‐cell repertoire in humans remain poorly defined. Systematic characterization of this ‘ground state’ immunity in comparison with memory will allow a better understanding of clonal selection during immune challenge. Here, we used high‐definition cell isolation from umbilical cord blood samples to establish the baseline frequency, phenotype and T‐cell antigen receptor (TCR) repertoire of CD8+ T‐cell precursor populations specific for a range of viral and self‐derived Ags. Across the board, these precursor populations were phenotypically naive and occurred with hierarchical frequencies clustered by Ag specificity. The corresponding patterns of TCR architecture were highly ordered and displayed partial overlap with adult memory, indicating biased structuring of the T‐cell repertoire during Ag‐driven selection. Collectively, these results provide new insights into the complex nature and dynamics of the naive T‐cell compartment.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Nonrandom attrition of the naive CD8+ T-cell pool with aging governed by T-cell receptor:pMHC interactions

Brian D. Rudd; Vanessa Venturi; Gang Li; Partha Samadder; James M. Ertelt; Sing Sing Way; Miles P. Davenport; Janko Nikolich-Žugich

Immunity against new infections declines in the last quartile of life, as do numbers of naive T cells. Peripheral maintenance of naive T cells over the lifespan is necessary because their production drastically declines by puberty, a result of thymic involution. We report that this maintenance is not random in advanced aging. As numbers and diversity of naive CD8+ T cells declined with aging, surviving cells underwent faster rates of homeostatic proliferation, were selected for high T-cell receptor:pMHC avidity, and preferentially acquired “memory-like” phenotype. These high-avidity precursors preferentially responded to infection and exhibited strong antimicrobial function. Thus, T-cell receptor avidity for self-pMHC provides a proofreading mechanism to maintain some of the fittest T cells in the otherwise crumbling naive repertoire, providing a degree of compensation for numerical and diversity defects in old T cells.


Journal of Immunology | 2008

TCR β-Chain Sharing in Human CD8+ T Cell Responses to Cytomegalovirus and EBV

Vanessa Venturi; Hui Yee Chin; Tedi E. Asher; Kristin Ladell; Phillip Scheinberg; Ethan Bornstein; David van Bockel; Anthony D. Kelleher; David A. Price; Miles P. Davenport

The CD8+ TCR repertoires specific for many immunogenic epitopes of CMV and EBV are dominated by a few TCR clonotypes and involve public TCRs that are shared between many MHC-matched individuals. In previous studies, we demonstrated that the observed sharing of epitope-specific TCRβ chains between individuals is strongly associated with TCRβ production frequency, and that a process of convergent recombination facilitates the more efficient production of some TCRβ sequences. In this study, we analyzed a total of 2836 TCRβ sequences from 23 CMV-infected and 10 EBV-infected individuals to investigate the factors that influence the sharing of TCRβ sequences in the CD8+ T cell responses to two immunodominant HLA-A*0201-restricted epitopes from these viruses. The most shared TCRβ amino acid sequences were found to have two features that indicate efficient TCRβ production, as follows: 1) they required fewer nucleotide additions, and 2) they were encoded by a greater variety of nucleotide sequences. We used simulations of random V(D)J recombination to demonstrate that the in silico TCRβ production frequency was predictive of the extent to which both TCRβ nucleotide and amino acid sequences were shared in vivo. These results suggest that TCRβ production frequency plays an important role in the interindividual sharing of TCRβ sequences within CD8+ T cell responses specific for CMV and EBV.


Cell Reports | 2014

CD161 Defines a Transcriptional and Functional Phenotype across Distinct Human T Cell Lineages

Joannah R. Fergusson; Kira E. Smith; Vicki M. Fleming; Neil Rajoriya; Evan W. Newell; Ruth Simmons; Emanuele Marchi; Sophia Björkander; Yu-Hoi Kang; Leo Swadling; Ayako Kurioka; Natasha Sahgal; Helen Lockstone; Dilair Baban; Gordon J. Freeman; Eva Sverremark-Ekström; Mark M. Davis; Miles P. Davenport; Vanessa Venturi; James E. Ussher; Christian B. Willberg; Paul Klenerman

Summary The C-type lectin CD161 is expressed by a large proportion of human T lymphocytes of all lineages, including a population known as mucosal-associated invariant T (MAIT) cells. To understand whether different T cell subsets expressing CD161 have similar properties, we examined these populations in parallel using mass cytometry and mRNA microarray approaches. The analysis identified a conserved CD161++/MAIT cell transcriptional signature enriched in CD161+CD8+ T cells, which can be extended to CD161+ CD4+ and CD161+TCRγδ+ T cells. Furthermore, this led to the identification of a shared innate-like, TCR-independent response to interleukin (IL)-12 plus IL-18 by different CD161-expressing T cell populations. This response was independent of regulation by CD161, which acted as a costimulatory molecule in the context of T cell receptor stimulation. Expression of CD161 hence identifies a transcriptional and functional phenotype, shared across human T lymphocytes and independent of both T cell receptor (TCR) expression and cell lineage.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Convergent recombination shapes the clonotypic landscape of the naïve T-cell repertoire

Máire F. Quigley; Hui Yee Greenaway; Vanessa Venturi; Ross W. B. Lindsay; Kylie M. Quinn; Robert A. Seder; Miles P. Davenport; David A. Price

Adaptive T-cell immunity relies on the recruitment of antigen-specific clonotypes, each defined by the expression of a distinct T-cell receptor (TCR), from an array of naïve T-cell precursors. Despite the enormous clonotypic diversity that resides within the naïve T-cell pool, interindividual sharing of TCR sequences has been observed within mobilized T-cell responses specific for certain peptide–major histocompatibility complex (pMHC) antigens. The mechanisms that underlie this phenomenon have not been fully elucidated, however. A mechanism of convergent recombination has been proposed to account for the occurrence of shared, or “public,” TCRs in specific memory T-cell populations. According to this model, TCR sharing between individuals is directly related to TCR production frequency; this, in turn, is determined on a probabilistic basis by the relative generation efficiency of particular nucleotide and amino acid sequences during the recombination process. Here, we tested the key predictions of convergent recombination in a comprehensive evaluation of the naïve CD8+ TCRβ repertoire in mice. Within defined segments of the naïve CD8+ T-cell repertoire, TCRβ sequences with convergent features were (i) present at higher copy numbers within individual mice and (ii) shared between individual mice. Thus, the naïve CD8+ T-cell repertoire is not flat, but comprises a hierarchy of recurrence rates for individual clonotypes that is determined by relative production frequencies. These findings provide a framework for understanding the early mobilization of public CD8+ T-cell clonotypes, which can exert profound biological effects during acute infectious processes.

Collaboration


Dive into the Vanessa Venturi's collaboration.

Top Co-Authors

Avatar

Miles P. Davenport

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

G Peach

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui Yee Greenaway

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge