Network


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

Hotspot


Dive into the research topics where Ali Zaid is active.

Publication


Featured researches published by Ali Zaid.


Nature | 2011

Different patterns of peripheral migration by memory CD4+ and CD8+ T cells

Thomas Gebhardt; Paul G. Whitney; Ali Zaid; Laura K. Mackay; Andrew G. Brooks; William R. Heath; Francis R. Carbone; Scott N. Mueller

Infections localized to peripheral tissues such as the skin result in the priming of T-cell responses that act to control pathogens. Activated T cells undergo migrational imprinting within the draining lymph nodes, resulting in memory T cells that provide local and systemic protection. Combinations of migrating and resident memory T cells have been implicated in long-term peripheral immunity, especially at the surfaces that form pathogen entry points into the body. However, T-cell immunity consists of separate CD4+ helper T cells and CD8+ killer T cells, with distinct effector and memory programming requirements. Whether these subsets also differ in their ability to form a migrating pool involved in peripheral immunosurveillance or a separate resident population responsible for local infection control has not been explored. Here, using mice, we show key differences in the migration and tissue localization of memory CD4+ and CD8+ T cells following infection of the skin by herpes simplex virus. On resolution of infection, the skin contained two distinct virus-specific memory subsets; a slow-moving population of sequestered CD8+ T cells that were resident in the epidermis and confined largely to the original site of infection, and a dynamic population of CD4+ T cells that trafficked rapidly through the dermis as part of a wider recirculation pattern. Unique homing-molecule expression by recirculating CD4+ T effector-memory cells mirrored their preferential skin-migratory capacity. Overall, these results identify a complexity in memory T-cell migration, illuminating previously unappreciated differences between the CD4+ and CD8+ subsets.


Science | 2016

Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes

Laura K. Mackay; Martina Minnich; Natasja A. M. Kragten; Yang Liao; Benjamin Nota; Cyril Seillet; Ali Zaid; Kevin Man; Simon Preston; David Freestone; Asolina Braun; Erica Wynne-Jones; Felix M. Behr; Regina Stark; Daniel G. Pellicci; Dale I. Godfrey; Gabrielle T. Belz; Marc Pellegrini; Thomas Gebhardt; Meinrad Busslinger; Wei Shi; Francis R. Carbone; René A. W. van Lier; Axel Kallies; Klaas P. J. M. van Gisbergen

Transcription factors define tissue T cells The immune system fights microbial invaders by maintaining multiple lines of defense. For instance, specialized memory T cells [resident memory T cells (Trms)] colonize portals of pathogen entry, such as the skin, lung, and gut, to quickly halt reinfections. Mackay et al. now report that in mice, Trms as well as other tissue-dwelling lymphocyte populations such as natural killer cells share a common transcriptional program driven by the related transcription factors Hobit and Blimp1. Tissue residency and retention of lymphocytes require expression of Hobit and Blimp1, which, among other functions, suppress genes that promote tissue exit. Science, this issue p. 459 Tissue-dwelling lymphocyte populations share a common transcriptional signature. Tissue-resident memory T (Trm) cells permanently localize to portals of pathogen entry, where they provide immediate protection against reinfection. To enforce tissue retention, Trm cells up-regulate CD69 and down-regulate molecules associated with tissue egress; however, a Trm-specific transcriptional regulator has not been identified. Here, we show that the transcription factor Hobit is specifically up-regulated in Trm cells and, together with related Blimp1, mediates the development of Trm cells in skin, gut, liver, and kidney in mice. The Hobit-Blimp1 transcriptional module is also required for other populations of tissue-resident lymphocytes, including natural killer T (NKT) cells and liver-resident NK cells, all of which share a common transcriptional program. Our results identify Hobit and Blimp1 as central regulators of this universal program that instructs tissue retention in diverse tissue-resident lymphocyte populations.


Nature | 2013

A type III effector antagonizes death receptor signalling during bacterial gut infection

Jaclyn S. Pearson; Sze Ong; Catherine L. Kennedy; Michelle Kelly; Keith S. Robinson; Tania Lung; Ashley Mansell; Patrice Riedmaier; Claire Oates; Ali Zaid; Sabrina Mühlen; Valerie F. Crepin; Oliver Marchès; Ching-Seng Ang; Nicholas A. Williamson; Lorraine A. O'Reilly; Aleksandra Bankovacki; Ueli Nachbur; Giuseppe Infusini; Andrew I. Webb; John Silke; Andreas Strasser; Gad Frankel; Elizabeth L. Hartland

Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonize the gut, replicate in host tissues and disseminate to other hosts. Pathogens such as Salmonella, Shigella and enteropathogenic and enterohaemorrhagic (EPEC and EHEC, respectively) Escherichia coli use a type III secretion system (T3SS) to deliver virulence effector proteins into host cells during infection that promote colonization and interfere with antimicrobial host responses. Here we report that the T3SS effector NleB1 from EPEC binds to host cell death-domain-containing proteins and thereby inhibits death receptor signalling. Protein interaction studies identified FADD, TRADD and RIPK1 as binding partners of NleB1. NleB1 expressed ectopically or injected by the bacterial T3SS prevented Fas ligand or TNF-induced formation of the canonical death-inducing signalling complex (DISC) and proteolytic activation of caspase-8, an essential step in death-receptor-induced apoptosis. This inhibition depended on the N-acetylglucosamine transferase activity of NleB1, which specifically modified Arg 117 in the death domain of FADD. The importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing pathogens antagonize death-receptor-induced apoptosis of infected cells, thereby blocking a major antimicrobial host response.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Persistence of skin-resident memory T cells within an epidermal niche.

Ali Zaid; Laura K. Mackay; Azad Rahimpour; Asolina Braun; Marc Veldhoen; Francis R. Carbone; Jonathan H. Manton; William R. Heath; Scott N. Mueller

Significance Tissue-resident memory T cells (TRM) form in the skin where they are retained and can protect against subsequent infection. Using a combination of intravital imaging and mathematical modeling of skin TRM that form after cutaneous herpes simplex virus 1 infection, we reveal that these memory T cells persist at the site of infection for the life of a mouse owing to slow random migration. We also report that TRM compete with dendritic epidermal γδ T cells in skin for local survival signals, suggesting that T cells compete for space within an epidermal niche. Barrier tissues such as the skin contain various populations of immune cells that contribute to protection from infections. These include recently identified tissue-resident memory T cells (TRM). In the skin, these memory CD8+ T cells reside in the epidermis after being recruited to this site by infection or inflammation. In this study, we demonstrate prolonged persistence of epidermal TRM preferentially at the site of prior infection despite sustained migration. Computational simulation of TRM migration within the skin over long periods revealed that the slow rate of random migration effectively constrains these memory cells within the region of skin in which they form. Notably, formation of TRM involved a concomitant local reduction in dendritic epidermal γδ T-cell numbers in the epidermis, indicating that these populations persist in mutual exclusion and may compete for local survival signals. Accordingly, we show that expression of the aryl hydrocarbon receptor, a transcription factor important for dendritic epidermal γδ T-cell maintenance in skin, also contributes to the persistence of skin TRM. Together, these data suggest that skin tissue-resident memory T cells persist within a tightly regulated epidermal T-cell niche.


Lancet Infectious Diseases | 2017

Chikungunya virus: an update on the biology and pathogenesis of this emerging pathogen

Felicity J. Burt; Weiqiang Chen; Jonathan J. Miner; Deborah J. Lenschow; Andres Merits; Esther Schnettler; Alain Kohl; Penny A. Rudd; Adam Taylor; Lara J. Herrero; Ali Zaid; Lisa F. P. Ng; Suresh Mahalingam

Re-emergence of chikungunya virus, a mosquito-transmitted pathogen, is of serious public health concern. In the past 15 years, after decades of infrequent, sporadic outbreaks, the virus has caused major epidemic outbreaks in Africa, Asia, the Indian Ocean, and more recently the Caribbean and the Americas. Chikungunya virus is mainly transmitted by Aedes aegypti mosquitoes in tropical and subtropical regions, but the potential exists for further spread because of genetic adaptation of the virus to Aedes albopictus, a species that thrives in temperate regions. Chikungunya virus represents a substantial health burden to affected populations, with symptoms that include severe joint and muscle pain, rashes, and fever, as well as prolonged periods of disability in some patients. The inflammatory response coincides with raised levels of immune mediators and infiltration of immune cells into infected joints and surrounding tissues. Animal models have provided insights into disease pathology and immune responses. Although host innate and adaptive responses have a role in viral clearance and protection, they can also contribute to virus-induced immune pathology. Understanding the mechanisms of host immune responses is essential for the development of treatments and vaccines. Inhibitory compounds targeting key inflammatory pathways, as well as attenuated virus vaccines, have shown some success in animal models, including an attenuated vaccine strain based on an isolate from La Reunion incorporating an internal ribosome entry sequence that prevents the virus from infecting mosquitoes and a vaccine based on virus-like particles expressing envelope proteins. However, immune correlates of protection, as well as the safety of prophylactic and therapeutic candidates, are important to consider for their application in chikungunya infections. In this Review, we provide an update on chikungunya virus with regard to its epidemiology, molecular virology, virus-host interactions, immunological responses, animal models, and potential antiviral therapies and vaccines.


Nature Communications | 2016

Skin CD4+ memory T cells exhibit combined cluster-mediated retention and equilibration with the circulation

Nicholas Collins; Xiaodong Jiang; Ali Zaid; Bethany L. Macleod; Jane Li; Chang Ook Park; Ashraful Haque; Sammy Bedoui; William R. Heath; Scott N. Mueller; Thomas S. Kupper; Thomas Gebhardt; Francis R. Carbone

Although memory T cells within barrier tissues can persist as permanent residents, at least some exchange with blood. The extent to which this occurs is unclear. Here we show that memory CD4+ T cells in mouse skin are in equilibrium with the circulation at steady state. These cells are dispersed throughout the inter-follicular regions of the dermis and form clusters with antigen presenting cells around hair follicles. After infection or administration of a contact sensitizing agent, there is a sustained increase in skin CD4+ T-cell content, which is confined to the clusters, with a concomitant CCL5-dependent increase in CD4+ T-cell recruitment. Skin CCL5 is derived from CD11b+ cells and CD8+ T cells, with the elimination of the latter decreasing CD4+ T-cell numbers. These results reveal a complex pattern of tissue-retention and equilibration for CD4+ memory T cells in skin, which is altered by infection and inflammation history.


Frontiers in Immunology | 2014

Tissue-resident T cells: dynamic players in skin immunity.

Scott N. Mueller; Ali Zaid; Francis R. Carbone

The skin is a large and complex organ that acts as a critical barrier protecting the body from pathogens in the environment. Numerous heterogeneous populations of immune cells are found within skin, including some that remain resident and others that can enter and exit the skin as part of their migration program. Pathogen-specific CD8+ T cells that persist in the epidermis following infection are a unique population of memory cells with important roles in immune surveillance and protective responses to reinfection. How these tissue-resident memory T cells form in the skin, the signals controlling their persistence and behavior, and the mechanisms by which they mediate local recall responses are just beginning to be elucidated. Here, we discuss recent progress in understanding the roles of these skin-resident T cells and also highlight some of the key unanswered questions that need addressing.


Nature Immunology | 2018

Local proliferation maintains a stable pool of tissue-resident memory T cells after antiviral recall responses

Simone L Park; Ali Zaid; Jyh Liang Hor; Susan N. Christo; Julia E. Prier; Brooke Davies; Yannick O. Alexandre; Julia L. Gregory; Tiffany A. Russell; Thomas Gebhardt; Francis R. Carbone; David C. Tscharke; William R. Heath; Scott N. Mueller; Laura K. Mackay

Although tissue-resident memory T cells (TRM cells) are critical in fighting infection, their fate after local pathogen re-encounter is unknown. Here we found that skin TRM cells engaged virus-infected cells, proliferated in situ in response to local antigen encounter and did not migrate out of the epidermis, where they exclusively reside. As a consequence, secondary TRM cells formed from pre-existing TRM cells, as well as from precursors recruited from the circulation. Newly recruited antigen-specific or bystander TRM cells were generated in the skin without displacement of the pre-existing TRM cell pool. Thus, pre-existing skin TRM cell populations are not displaced after subsequent infections, which enables multiple TRM cell specificities to be stably maintained within the tissue.Mackay, Mueller and colleagues show that tissue-resident memory T cells proliferate in situ in response to local antigen and persist during subsequent antigen encounters.


Journal of Immunology | 2015

Targeting Antigen to Clec9A Primes Follicular Th Cell Memory Responses Capable of Robust Recall

Yu Kato; Ali Zaid; Gayle M. Davey; Scott N. Mueller; Stephen L. Nutt; Dimitra Zotos; David M. Tarlinton; Ken Shortman; Mireille H. Lahoud; William R. Heath; Irina Caminschi

Targeting Ags to dendritic cell (DC) surface receptors can induce a variety of responses depending on the DC type targeted, the receptor targeted, and the adjuvant used. Clec9A (DNGR-1), which is expressed by CD8+ DCs, has been shown to bind F-actin exposed on damaged cells. Targeting Ag to this receptor in mice and nonhuman primates induces strong humoral immunity even in the absence of adjuvant, a process seen for a few select DC receptors. In contrast with other receptors, however, targeting Clec9A induces long-lived, affinity-matured Ab responses that are associated with efficient CD4+ T cell responses shown to possess properties of follicular Th cells (TFH). In this article, we provide definitive evidence that Clec9A targeting promotes the development of TFH by showing that responding CD4 T cells express CXCR5, PD1, the TFH transcription factor Bcl6, and the cytokine IL-21, and that these cells localize to germinal centers. Furthermore, we extend studies from the model Ag OVA to the viral Ag glycoprotein D of HSV-1 and examine the capacity of primed TFH to form functional memory. We show that targeting glycoprotein D to Clec9A even in the absence of adjuvant induced long-lived memory CXCR5+ PD1hi CD4+ T cells that proliferated extensively upon secondary challenge and rapidly developed into effector TFH. This was associated with enhanced germinal center B cell responses and accelerated Ab production. Our study indicates that targeting Ags to Clec9A in the absence of adjuvant routinely generates TFH responses that form long-lived memory capable of robust secondary TFH responses.


The Journal of Infectious Diseases | 2011

Downregulation of Interferon-β in Antibody-Dependent Enhancement of Dengue Viral Infections of Human Macrophages Is Dependent on Interleukin-6

Michael S. Rolph; Ali Zaid; Nestor E. Rulli; Suresh Mahalingam

To the Editor—Dengue virus (DENV) is a major arthropod-borne virus affecting millions across Asia, Africa, and any tropical areas where vector mosquitoes find optimal breeding conditions. A feature of DENV infection is that individuals who have been previously infected with DENV are at increased risk of developing severe dengue hemorrhagic fever (DHF) following subsequent infection. Despite intensive research, the mechanisms underlying disease exacerbation following secondary infections are still unclear. The recent work by Ubol et al identified possible mechanisms to explain how previous dengue infection might lead to disease enhancement [1]. The authors suggested that antibodydependent enhancement (ADE) of infection is a key mechanism in the pathophysiological changes that occur during DHF. ADE in DENV infection was first described in the mid-1970s [2], and has since been described during infection with a broad range of viruses [3]. ADE is believed to involve enhanced virus entry and replication in the

Collaboration


Dive into the Ali Zaid'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

Andreas Suhrbier

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge