Network


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

Hotspot


Dive into the research topics where Jessica Hitchcock is active.

Publication


Featured researches published by Jessica Hitchcock.


Infection and Immunity | 2011

SadA, a Trimeric Autotransporter from Salmonella enterica Serovar Typhimurium, Can Promote Biofilm Formation and Provides Limited Protection against Infection

Dhaarini Raghunathan; Timothy J. Wells; Faye C. Morris; Robert K. Shaw; Saeeda Bobat; Sarah E. Peters; Gavin K. Paterson; Karina Tveen Jensen; Denisse L. Leyton; Jessica M. A. Blair; Douglas F. Browning; John Pravin; Adriana Flores-Langarica; Jessica Hitchcock; Claudia T. P. Moraes; Roxane M.F. Piazza; Duncan J. Maskell; Mark A. Webber; Robin C. May; Calman A. MacLennan; Laura J. V. Piddock; Adam F. Cunningham; Ian R. Henderson

ABSTRACT Salmonella enterica is a major cause of morbidity worldwide and mortality in children and immunocompromised individuals in sub-Saharan Africa. Outer membrane proteins of Salmonella are of significance because they are at the interface between the pathogen and the host, they can contribute to adherence, colonization, and virulence, and they are frequently targets of antibody-mediated immunity. In this study, the properties of SadA, a purported trimeric autotransporter adhesin of Salmonella enterica serovar Typhimurium, were examined. We demonstrated that SadA is exposed on the Salmonella cell surface in vitro and in vivo during infection of mice. Expression of SadA resulted in cell aggregation, biofilm formation, and increased adhesion to human intestinal Caco-2 epithelial cells. Immunization of mice with folded, full-length, purified SadA elicited an IgG response which provided limited protection against bacterial challenge. When anti-SadA IgG titers were enhanced by administering alum-precipitated protein, a modest additional protection was afforded. Therefore, despite SadA having pleiotropic functions, it is not a dominant, protective antigen for antibody-mediated protection against Salmonella.


Journal of Immunology | 2012

Systemic Flagellin Immunization Stimulates Mucosal CD103+ Dendritic Cells and Drives Foxp3+ Regulatory T Cell and IgA Responses in the Mesenteric Lymph Node

Adriana Flores-Langarica; Jennifer L. Marshall; Jessica Hitchcock; Charlotte N. Cook; Jonathan Jobanputra; Saeeda Bobat; Ewan A. Ross; Ruth E. Coughlan; Ian R. Henderson; Satoshi Uematsu; Shizuo Akira; Adam F. Cunningham

Mucosal immunity is poorly activated after systemic immunization with protein Ags. Nevertheless, induction of mucosal immunity in such a manner would be an attractive and simple way to overcome the intrinsic difficulties in delivering Ag to such sites. Flagellin from Salmonella enterica serovar Typhimurium (FliC) can impact markedly on host immunity, in part via its recognition by TLR5. In this study, we show that systemic immunization with soluble FliC (sFliC) drives distinct immune responses concurrently in the mesenteric lymph nodes (MLN) and the spleen after i.p. and s.c. immunization. In the MLN, but not the spleen, sFliC drives a TLR5-dependent recruitment of CD103+ dendritic cells (DCs), which correlates with a diminution in CD103+ DC numbers in the lamina propria. In the MLN, CD103+ DCs carry Ag and are the major primers of endogenous and transgenic T cell priming. A key consequence of these interactions with CD103+ DCs in the MLN is an increase in local regulatory T cell differentiation. In parallel, systemic sFliC immunization results in a pronounced switching of FliC-specific B cells to IgA in the MLN but not elsewhere. Loss of TLR5 has more impact on MLN than splenic Ab responses, reflected in an ablation of IgA, but not IgG, serum Ab titers. Therefore, systemic sFliC immunization targets CD103+ DCs and drives distinct mucosal T and B cell responses. This offers a potential “Trojan horse” approach to modulate mucosal immunity by systemically immunizing with sFliC.


Nature Medicine | 2015

Homeostatic regulation of T cell trafficking by a B cell-derived peptide is impaired in autoimmune and chronic inflammatory disease

Myriam Chimen; Helen M. McGettrick; Bonita H. R. Apta; Sahithi J. Kuravi; Clara M. Yates; Amy Kennedy; Arjun Odedra; Mohammed Alassiri; Matthew J. Harrison; Ashley Martin; Francesca Barone; Saba Nayar; Jessica Hitchcock; Adam F. Cunningham; Karim Raza; Andrew Filer; David A. Copland; Andrew D. Dick; Joseph Robinson; Neena Kalia; Lucy S. K. Walker; Christopher D. Buckley; Gerard B. Nash; Parth Narendran; G. Ed Rainger

During an inflammatory response, lymphocyte recruitment into tissue must be tightly controlled because dysregulated trafficking contributes to the pathogenesis of chronic disease. Here we show that during inflammation and in response to adiponectin, B cells tonically inhibit T cell trafficking by secreting a peptide (PEPITEM) proteolytically derived from 14.3.3 zeta delta (14.3.3.ζδ) protein. PEPITEM binds cadherin-15 on endothelial cells, promoting synthesis and release of sphingosine-1 phosphate, which inhibits trafficking of T cells without affecting recruitment of other leukocytes. Expression of adiponectin receptors on B cells and adiponectin-induced PEPITEM secretion wanes with age, implying immune senescence of the pathway. Additionally, these changes are evident in individuals with type 1 diabetes or rheumatoid arthritis, and circulating PEPITEM in patient serum is reduced compared to that of healthy age-matched donors. In both diseases, tonic inhibition of T cell trafficking across inflamed endothelium is lost. Control of patient T cell trafficking is re-established by treatment with exogenous PEPITEM. Moreover, in animal models of peritonitis, hepatic ischemia-reperfusion injury, Salmonella infection, uveitis and Sjögrens syndrome, PEPITEM reduced T cell recruitment into inflamed tissues.


Journal of Immunology | 2012

The Capsular Polysaccharide Vi from Salmonella Typhi Is a B1b Antigen

Jennifer L. Marshall; Adriana Flores-Langarica; Robert A. Kingsley; Jessica Hitchcock; Ewan A. Ross; Constantino López-Macías; Jeremy H. Lakey; Laura B. Martin; Kai-Michael Toellner; Calman A. MacLennan; Ian C. M. MacLennan; Ian R. Henderson; Gordon Dougan; Adam F. Cunningham

Vaccination with purified capsular polysaccharide Vi Ag from Salmonella typhi can protect against typhoid fever, although the mechanism for its efficacy is not clearly established. In this study, we have characterized the B cell response to this vaccine in wild-type and T cell–deficient mice. We show that immunization with typhoid vi polysaccharide vaccine rapidly induces proliferation in B1b peritoneal cells, but not in B1a cells or marginal zone B cells. This induction of B1b proliferation is concomitant with the detection of splenic Vi-specific Ab-secreting cells and protective Ab in Rag1-deficient B1b cell chimeras generated by adoptive transfer-induced specific Ab after Vi immunization. Furthermore, Ab derived from peritoneal B cells is sufficient to confer protection against Salmonella that express Vi Ag. Expression of Vi by Salmonella during infection did not inhibit the development of early Ab responses to non-Vi Ags. Despite this, the protection conferred by immunization of mice with porin proteins from Salmonella, which induce Ab-mediated protection, was reduced postinfection with Vi-expressing Salmonella, although protection was not totally abrogated. This work therefore suggests that, in mice, B1b cells contribute to the protection induced by Vi Ag, and targeting non-Vi Ags as subunit vaccines may offer an attractive strategy to augment current Vi-based vaccine strategies.


Journal of Clinical Investigation | 2015

Inflammation drives thrombosis after Salmonella infection via CLEC-2 on platelets

Jessica Hitchcock; Charlotte N. Cook; Saeeda Bobat; Ewan A. Ross; Adriana Flores-Langarica; Kate L. Lowe; Mahmood Khan; C. Coral Dominguez-Medina; Sian Lax; Manuela Carvalho-Gaspar; Stefan G. Hubscher; G. Ed Rainger; Mark Cobbold; Christopher D. Buckley; Timothy J. Mitchell; Andrea M. Mitchell; Nick D. Jones; N. van Rooijen; Daniel Kirchhofer; Ian R. Henderson; David H. Adams; Steve P. Watson; Adam F. Cunningham

Thrombosis is a common, life-threatening consequence of systemic infection; however, the underlying mechanisms that drive the formation of infection-associated thrombi are poorly understood. Here, using a mouse model of systemic Salmonella Typhimurium infection, we determined that inflammation in tissues triggers thrombosis within vessels via ligation of C-type lectin-like receptor-2 (CLEC-2) on platelets by podoplanin exposed to the vasculature following breaching of the vessel wall. During infection, mice developed thrombi that persisted for weeks within the liver. Bacteria triggered but did not maintain this process, as thrombosis peaked at times when bacteremia was absent and bacteria in tissues were reduced by more than 90% from their peak levels. Thrombus development was triggered by an innate, TLR4-dependent inflammatory cascade that was independent of classical glycoprotein VI-mediated (GPVI-mediated) platelet activation. After infection, IFN-γ release enhanced the number of podoplanin-expressing monocytes and Kupffer cells in the hepatic parenchyma and perivascular sites and absence of TLR4, IFN-γ, or depletion of monocytic-lineage cells or CLEC-2 on platelets markedly inhibited the process. Together, our data indicate that infection-driven thrombosis follows local inflammation and upregulation of podoplanin and platelet activation. The identification of this pathway offers potential therapeutic opportunities to control the devastating consequences of infection-driven thrombosis without increasing the risk of bleeding.


European Journal of Immunology | 2011

T-zone localized monocyte-derived dendritic cells promote Th1 priming to Salmonella

Adriana Flores-Langarica; Jennifer L. Marshall; Saeeda Bobat; Elodie Mohr; Jessica Hitchcock; Ewan A. Ross; Ruth E. Coughlan; Mahmood Khan; Nico van Rooijen; Ian R. Henderson; Ian C. M. MacLennan; Adam F. Cunningham

Control of intracellular Salmonella infection requires Th1 priming and IFN‐γ production. Here, we show that efficient Th1 priming after Salmonella infection requires CD11c+CD11bhiF4/80+ monocyte‐derived dendritic cells (moDCs). In non‐infected spleens, moDCs are absent from T‐cell zones (T zones) of secondary lymphoid tissues, but by 24 h post‐infection moDCs are readily discernible in these sites. The accumulation of moDCs is more dependent upon bacterial viability than bacterial virulence. Kinetic studies showed that moDCs were necessary to prime but not sustain Th1 responses, while ex vivo studies showed that antigen‐experienced moDCs were sufficient to induce T‐cell proliferation and IFN‐γ production via a TNF‐α‐dependent mechanism. Importantly, moDCs and cDCs when co‐cultured induced superior Th1 differentiation than either subset alone, and this activity was independent of TNF‐α. Thus, optimal Th1 development to Salmonella requires the rapid accumulation of moDCs within T zones and their collaboration with cDCs.


Journal of Immunology | 2012

Thymic Function Is Maintained during Salmonella-Induced Atrophy and Recovery

Ewan A. Ross; Ruth E. Coughlan; Adriana Flores-Langarica; Sian Lax; Julia Nicholson; Guillaume E. Desanti; Jennifer L. Marshall; Saeeda Bobat; Jessica Hitchcock; Andrea J. White; William E. Jenkinson; Mahmood Khan; Ian R. Henderson; Gareth G. Lavery; Christopher D. Buckley; Graham Anderson; Adam F. Cunningham

Thymic atrophy is a frequent consequence of infection with bacteria, viruses, and parasites and is considered a common virulence trait between pathogens. Multiple reasons have been proposed to explain this atrophy, including premature egress of immature thymocytes, increased apoptosis, or thymic shutdown to prevent tolerance to the pathogen from developing. The severe loss in thymic cell number can reflect an equally dramatic reduction in thymic output, potentially reducing peripheral T cell numbers. In this study, we examine the relationship between systemic Salmonella infection and thymic function. During infection, naive T cell numbers in peripheral lymphoid organs increase. Nevertheless, this occurs despite a pronounced thymic atrophy caused by viable bacteria, with a peak 50-fold reduction in thymocyte numbers. Thymic atrophy is not dependent upon homeostatic feedback from peripheral T cells or on regulation of endogenous glucocorticoids, as demonstrated by infection of genetically altered mice. Once bacterial numbers fall, thymocyte numbers recover, and this is associated with increases in the proportion and proliferation of early thymic progenitors. During atrophy, thymic T cell maturation is maintained, and single-joint TCR rearrangement excision circle analysis reveals there is only a modest fall in recent CD4+ thymic emigrants in secondary lymphoid tissues. Thus, thymic atrophy does not necessarily result in a matching dysfunctional T cell output, and thymic homeostasis can constantly adjust to systemic infection to ensure that naive T cell output is maintained.


Immunology | 2010

Ligation of CD11c during vaccination promotes germinal centre induction and robust humoral responses without adjuvant

Ann L. White; Alison L. Tutt; Sonya James; Kevin A. Wilkinson; Fernanda Castro; Sandra V. Dixon; Jessica Hitchcock; Mahmood Khan; Aymen Al-Shamkhani; Adam F. Cunningham; Martin J. Glennie

In this study, we investigated the mouse dendritic cell (DC) receptor, complement receptor 4 (CR4; CD11c/CD18), as an immunotarget for triggering humoral immunity. Comparison of antibody titres generated against a panel of 13 anti‐antigen‐presenting cell receptor monoclonal antibodies, with or without conjugated ovalbumin (OVA), revealed uniquely rapid and robust responses following CR4 targeting, with antibody titres approaching 1 : 100 000 7 days after a single dose of antigen. Furthermore, using just 100 ng OVA conjugated to anti‐CD11c Fab′, we generated anti‐OVA titres greater than those produced by a 100‐fold higher dose of OVA in complete Freund’s adjuvant at day 28. These anti‐OVA antibody titres were sustained and could be boosted further with targeted OVA on day 21. Investigations to explain this vaccine potency showed that, in addition to targeting splenic DC, anti‐CDl1c antibodies delivered a powerful adjuvant effect and could boost humoral immunity against OVA even when the OVA was targeted to other molecules on DC, such as major histocompatibility complex class II, CD11a and CD11b. However, interestingly, this adjuvant effect was lost if OVA was targeted to other cells such as B cells via CD21 or CD19. The adjuvant effect was mediated through a marked enhancement of both germinal centre and extrafollicular plasma cell formation in responding spleens. These results demonstrate that anti‐CD11c monoclonal antibody can both target antigen and act as a powerful adjuvant for rapid and sustained antibody responses. They also point to an interesting role for CR4 on DC in triggering B cells during humoral immunity.


Journal of Immunology | 2011

CD31 is required on CD4+ T cells to promote T cell survival during Salmonella infection

Ewan A. Ross; Ruth E. Coughlan; Adriana Flores-Langarica; Saeeda Bobat; Jennifer L. Marshall; Khiyam Hussain; James E.G. Charlesworth; Nikita Abhyankar; Jessica Hitchcock; Cristina Gil; Constantino López-Macías; Ian R. Henderson; Mahmood Khan; Steve P. Watson; Ian C. M. MacLennan; C Buckley; Adam F. Cunningham

Hematopoietic cells constitutively express CD31/PECAM1, a signaling adhesion receptor associated with controlling responses to inflammatory stimuli. Although expressed on CD4+ T cells, its function on these cells is unclear. To address this, we have used a model of systemic Salmonella infection that induces high levels of T cell activation and depends on CD4+ T cells for resolution. Infection of CD31-deficient (CD31KO) mice demonstrates that these mice fail to control infection effectively. During infection, CD31KO mice have diminished numbers of total CD4+ T cells and IFN-γ–secreting Th1 cells. This is despite a higher proportion of CD31KO CD4+ T cells exhibiting an activated phenotype and an undiminished capacity to prime normally and polarize to Th1. Reduced numbers of T cells reflected the increased propensity of naive and activated CD31KO T cells to undergo apoptosis postinfection compared with wild-type T cells. Using adoptive transfer experiments, we show that loss of CD31 on CD4+ T cells alone is sufficient to account for the defective CD31KO T cell accumulation. These data are consistent with CD31 helping to control T cell activation, because in its absence, T cells have a greater propensity to become activated, resulting in increased susceptibility to become apoptotic. The impact of CD31 loss on T cell homeostasis becomes most pronounced during severe, inflammatory, and immunological stresses such as those caused by systemic Salmonella infection. This identifies a novel role for CD31 in regulating CD4 T cell homeostasis.


European Journal of Immunology | 2014

Resolving Salmonella infection reveals dynamic and persisting changes in murine bone marrow progenitor cell phenotype and function.

Ewan A. Ross; Adriana Flores-Langarica; Saeeda Bobat; Ruth E. Coughlan; Jennifer L. Marshall; Jessica Hitchcock; Charlotte N. Cook; Manuela Carvalho-Gaspar; Andrea M. Mitchell; Mary Clarke; Paloma García; Mark Cobbold; Timothy J. Mitchell; Ian R. Henderson; Nick D. Jones; Graham Anderson; Christopher D. Buckley; Adam F. Cunningham

The generation of immune cells from BM precursors is a carefully regulated process. This is essential to limit the potential for oncogenesis and autoimmunity yet protect against infection. How infection modulates this is unclear. Salmonella can colonize systemic sites including the BM and spleen. This resolving infection has multiple IFN‐γ‐mediated acute and chronic effects on BM progenitors, and during the first week of infection IFN‐γ is produced by myeloid, NK, NKT, CD4+ T cells, and some lineage‐negative cells. After infection, the phenotype of BM progenitors rapidly but reversibly alters, with a peak ∼30‐fold increase in Sca‐1hi progenitors and a corresponding loss of Sca‐1lo/int subsets. Most strikingly, the capacity of donor Sca‐1hi cells to reconstitute an irradiated host is reduced; the longer donor mice are exposed to infection, and Sca‐1hic‐kitint cells have an increased potential to generate B1a‐like cells. Thus, Salmonella can have a prolonged influence on BM progenitor functionality not directly related to bacterial persistence. These results reflect changes observed in leucopoiesis during aging and suggest that BM functionality can be modulated by life‐long, periodic exposure to infection. Better understanding of this process could offer novel therapeutic opportunities to modulate BM functionality and promote healthy aging.

Collaboration


Dive into the Jessica Hitchcock's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Saeeda Bobat

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ewan A. Ross

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mahmood Khan

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge