20 results on '"Ager, Ann"'
Search Results
2. Homing to solid cancers: a vascular checkpoint in adoptive cell therapy using CAR T-cells.
- Author
-
Ager, Ann, Watson, H. Angharad, Wehenkel, Sophie C., and Mohammed, Rebar N.
- Subjects
- *
CANCER diagnosis , *T cells , *CHEMOKINE receptors , *CELLULAR therapy , *CELL adhesion molecules , *THERAPEUTICS - Abstract
The success of adoptive T-cell therapies for the treatment of cancer patients depends on transferred Tlymphocytes finding and infiltrating cancerous tissues. For intravenously transferred T-cells, this means leaving the bloodstream (extravasation) from tumour blood vessels. In inflamed tissues, a key event in extravasation is the capture, rolling and arrest of T-cells inside blood vessels which precedes transmigration across the vessel wall and entry into tissues. This depends on co-ordinated signalling of selectins, integrins and chemokine receptors on T-cells by their respective ligands which are up-regulated on inflamed blood vessels. Clinical data and experimental studies in mice suggest that tumour blood vessels are anergic to inflammatory stimuli and the recruitment of cytotoxic CD8+ T-lymphocytes is not very efficient. Interestingly, and somewhat counter-intuitively, anti-angiogenic therapy can promote CD8+ T-cell infiltration of tumours and increase the efficacy of adoptive CD8+ T-cell therapy. Rather than inhibit tumour angiogenesis, anti-angiogenic therapy 'normalizes' (matures) tumour blood vessels by promoting pericyte recruitment, increasing tumour blood vessel perfusion and sensitizing tumour blood vessels to inflammatory stimuli. A number of different approaches are currently being explored to increase recruitment by manipulating the expression of homing-associated molecules on T-cells and tumour blood vessels. Future studies should address whether these approaches improve the efficacy of adoptive T-cell therapies for solid, vascularized cancers in patients. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
3. Inflammation: Border crossings.
- Author
-
Ager, Ann
- Subjects
- *
INFLAMMATION , *LEUCOCYTES - Abstract
Focuses on the involvement of inflammatory cells and blood vessels during the inflammation process. Role of blood vessels in accommodating and propelling migrating inflammatory cells; Emigration of leukocytes from postcapillary venules into veins; Regulation of the permeability of the endothelial layer.
- Published
- 2003
- Full Text
- View/download PDF
4. Understanding high endothelial venules: Lessons for cancer immunology.
- Author
-
Ager, Ann and May, Michael J
- Subjects
- *
ENDOTHELIAL cells , *LYMPHOCYTES , *LYMPH nodes , *AUTOIMMUNITY , *CANCER cells , *CELLULAR immunity - Abstract
High endothelial venules (HEVs) are blood vessels especially adapted for lymphocyte trafficking which are normally found in secondary lymphoid organs such as lymph nodes (LN) and Peyer's patches. It has long been known that HEVs develop in non-lymphoid organs during chronic inflammation driven by autoimmunity, infection or allografts. More recently, HEVs have been observed in solid, vascularized tumors and their presence correlated with reduced tumor size and improved patient outcome. It is proposed that newly formed HEV promote antitumor immunity by recruiting naive lymphocytes into the tumor, thus allowing the local generation of cancerous tissue-destroying lymphocytes. Understanding how HEVs develop and function are therefore important to unravel their role in human cancers. In LN, HEVs develop during embryonic and early post-natal life and are actively maintained by the LN microenvironment. Systemic blockade of lymphotoxin-β receptor leads to HEV de-differentiation, but the LN components that induce HEV differentiation have remained elusive. Recent elegant studies using gene-targeted mice have demonstrated clearly that triggering the lymphotoxin-β receptor in endothelial cells (EC) induces the differentiation of HEV and that CD11c+dendritic cells play a crucial role in this process. It will be important to determine whether lymphotoxin-β receptor-dependent signaling in EC drives the development of HEV during tumorigenesis and which cells have HEV-inducer properties. This may reveal therapeutic approaches to promote HEV neogenesis and determine the impact of newly formed HEV on tumor immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
5. Lymphocyte-vascular endothelial cell interactions in the immune response.
- Author
-
Ager, Ann
- Subjects
- *
CELL communication , *CELLULAR control mechanisms , *IMMUNE response , *IMMUNOLOGY , *LEUCOCYTES , *LYMPHOCYTES - Abstract
The article presents information on lymphocite-vascular endothelial cell interactions in the immune response. A critical event in the immune response is the regulation of lymphocyte migration from the blood by vascular endothelial cells (EC) Immune responses are initiated following contact between lymphocytes and antigen on the surface of specialized antigen presenting cells in lymphoid organs. Lymphocyte extravasation is a complex event which involves several distinct stages, including binding to the luminal surface of EC and migration across the endothelial lining and the basal lamina which constitute the vessel wall.
- Published
- 1993
6. Drug Inhibition of Redox Factor-1 Restores Hypoxia-Driven Changes in Tuberous Sclerosis Complex 2 Deficient Cells.
- Author
-
Champion, Jesse D., Dodd, Kayleigh M., Lam, Hilaire C., Alzahrani, Mohammad A. M., Seifan, Sara, Rad, Ellie, Scourfield, David Oliver, Fishel, Melissa L., Calver, Brian L., Ager, Ann, Henske, Elizabeth P., Davies, David Mark, Kelley, Mark R., and Tee, Andrew R.
- Subjects
- *
IN vitro studies , *CELL culture , *WESTERN immunoblotting , *OXIDATION-reduction reaction , *HYPOXEMIA , *TUBEROUS sclerosis , *PHOSPHORYLATION - Abstract
Simple Summary: Tuberous sclerosis complex (TSC) is a genetic disease where patients are predisposed to tumors and neurological complications. Current therapies for this disease are not fully curative. We aimed to explore novel drug targets and therapies that could further benefit TSC patients. This work uncovered a novel pathway that drives disease in TSC cell models involving redox factor-1 (Ref-1). Ref-1 is a protein that turns on several key transcription factors that collectively promote tumor growth and survival through direct redox signaling. Processes regulated by Ref-1 include angiogenesis, inflammation, and metabolic transformation. Therefore, this work reveals a new drug target, where inhibitors of Ref-1 could have an additional benefit compared to current drug therapies. Therapies with the mechanistic target of rapamycin complex 1 (mTORC1) inhibitors are not fully curative for tuberous sclerosis complex (TSC) patients. Here, we propose that some mTORC1-independent disease facets of TSC involve signaling through redox factor-1 (Ref-1). Ref-1 possesses a redox signaling activity that stimulates the transcriptional activity of STAT3, NF-kB, and HIF-1α, which are involved in inflammation, proliferation, angiogenesis, and hypoxia, respectively. Here, we demonstrate that redox signaling through Ref-1 contributes to metabolic transformation and tumor growth in TSC cell model systems. In TSC2-deficient cells, the clinically viable Ref-1 inhibitor APX3330 was effective at blocking the hyperactivity of STAT3, NF-kB, and HIF-1α. While Ref-1 inhibitors do not inhibit mTORC1, they potently block cell invasion and vasculature mimicry. Of interest, we show that cell invasion and vasculature mimicry linked to Ref-1 redox signaling are not blocked by mTORC1 inhibitors. Metabolic profiling revealed that Ref-1 inhibitors alter metabolites associated with the glutathione antioxidant pathway as well as metabolites that are heavily dysregulated in TSC2-deficient cells involved in redox homeostasis. Therefore, this work presents Ref-1 and associated redox-regulated transcription factors such as STAT3, NF-kB, and HIF-1α as potential therapeutic targets to treat TSC, where targeting these components would likely have additional benefits compared to using mTORC1 inhibitors alone. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. High endothelial venules: Help or hindrance in the quest for antitumor immunity?
- Author
-
Gallimore, Awen, Godkin, Andrew, and Ager, Ann
- Subjects
- *
ENDOTHELIAL cells , *ANTINEOPLASTIC agents , *IMMUNE response , *BLOOD vessels , *NEOPLASTIC cell transformation - Abstract
Recently, a number of studies have documented the presence of high endothelial venules in both mouse and human tumors. The significance of these highly specialized blood vessels within neoplastic lesions, and notably their capacity to influence anticancer immune responses and tumor progression, remains to be fully understood. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
8. Primary breast tumours but not lung metastases induce protective anti-tumour immune responses after Treg-depletion.
- Author
-
Hughes, Ellyn, Lauder, Sarah N., Smart, Kathryn, Bloom, Anja, Scott, Jake, Jones, Emma, Somerville, Michelle, Browne, Molly, Blainey, Andrew, Godkin, Andrew, Ager, Ann, and Gallimore, Awen
- Subjects
- *
IMMUNE response , *SUPPRESSOR cells , *TUMORS , *CANCER , *METASTASIS - Abstract
Although metastatic disease is responsible for the majority of cancer deaths, tests of novel immunotherapies in mouse tumour models often focus on primary tumours without determining whether these therapies also target metastatic disease. This study examined the impact of depleting Foxp3+ regulatory T cells (Treg), on lung metastases, using a mouse model of breast cancer. After Treg-depletion, generation of an immune response to the primary tumour was a critical determinant for limiting development of metastasis. Indeed, resection of the primary tumour abrogated any effect of Treg-depletion on metastases. In addition, whilst the immune response, generated by the primary tumour, prevented metastases development, it had little impact on controlling established disease. Collectively, the data indicate that metastatic cells in the lung are not controlled by immune responses induced by the primary tumour. These findings indicate that targeting Tregs alone will not suffice for treating lung metastases. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
9. High endothelial venules are associated with microsatellite instability, hereditary background and immune evasion in colorectal cancer.
- Author
-
Pfuderer, Pauline L., Ballhausen, Alexej, Seidler, Florian, Stark, Hans-Jürgen, Grabe, Niels, Frayling, Ian M., Ager, Ann, von Knebel Doeberitz, Magnus, Kloor, Matthias, and Ahadova, Aysel
- Abstract
Background: Microsatellite-unstable (MSI) tumours show a high load of mutational neoantigens, as a consequence of DNA mismatch repair deficiency. Consequently, MSI tumours commonly present with dense immune infiltration and develop immune evasion mechanisms. Whether improved lymphocyte recruitment contributes to the pronounced immune infiltration in MSI tumours is unknown. We analysed the density of high endothelial venules (HEV) and postcapillary blood vessels specialised for lymphocyte trafficking, in MSI colorectal cancers (CRC).Methods: HEV density was determined by immunohistochemical staining of FFPE tissue sections from MSI (n = 48) and microsatellite-stable (MSS, n = 35) CRCs. Associations with clinical and pathological variables were analysed.Results: We found elevated HEV densities in MSI compared with MSS CRCs (median 0.049 vs 0.000 counts/mm2, respectively, p = 0.0002), with the highest densities in Lynch syndrome MSI CRCs. Dramatically elevated HEV densities were observed in B2M-mutant Lynch syndrome CRCs, pointing towards a link between lymphocyte recruitment and immune evasion (median 0.485 vs 0.0885 counts/mm2 in B2M-wild-type tumours, p = 0.0237).Conclusions: Our findings for the first time indicate a significant contribution of lymphocyte trafficking in immune responses against MSI CRC, particularly in the context of Lynch syndrome. High HEV densities in B2M-mutant tumours underline the significance of immunoediting during tumour evolution. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
10. Peptide mimic for influenza vaccination using nonnatural combinatorial chemistry.
- Author
-
Miles, John J., Mai Ping Tan, Dolton, Garry, Edwards, Emily S. J., Galloway, Sarah A. E., Laugel, Bruno, Clement, Mathew, Makinde, Julia, Ladell, Kristin, Matthews, Katherine K., Watkins, Thomas S., Tungatt, Katie, Yide Wong, Han Siean Lee, Clark, Richard J., Pentier, Johanne M., Attaf, Meriem, Lissina, Anya, Ager, Ann, and Gallimore, Awen
- Subjects
- *
INFLUENZA vaccines , *POLYPEPTIDES , *T cells , *INFLUENZA viruses , *GASTRIC acid - Abstract
Polypeptide vaccines effectively activate human T cells but suffer from poor biological stability, which confines both transport logistics and in vivo therapeutic activity. Synthetic biology has the potential to address these limitations through the generation of highly stable antigenic "mimics" using subunits that do not exist in the natural world. We developed a platform based on D-amino acid combinatorial chemistry and used this platform to reverse engineer a fully artificial CD8+ T cell agonist that mirrored the immunogenicity profile of a native epitope blueprint from influenza virus. This nonnatural peptide was highly stable in human serum and gastric acid, reflecting an intrinsic resistance to physical and enzymatic degradation. In vitro, the synthetic agonist stimulated and expanded an archetypal repertoire of polyfunctional human influenza virus-specific CD8+ T cells. In vivo, specific responses were elicited in naive humanized mice by subcutaneous vaccination, conferring protection from subsequent lethal influenza challenge. Moreover, the synthetic agonist was immunogenic after oral administration. This proof-of-concept study highlights the power of synthetic biology to expand the horizons of vaccine design and therapeutic delivery. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
11. TMEFF2 shedding is regulated by oxidative stress and mediated by ADAMs and transmembrane serine proteases implicated in prostate cancer.
- Author
-
Gaweł‐Bęben, Katarzyna, Ali, Nazim, Ellis, Vincent, Velasco, Gloria, Poghosyan, Zaruhi, Ager, Ann, and Knäuper, Vera
- Subjects
- *
PROSTATE cancer & genetics , *OXIDATIVE stress , *SERINE proteinases , *MATRIPTASE , *MEMBRANE proteins - Abstract
Abstract: TMEFF2 is a type I transmembrane protein with two follistatin (FS) and one EGF‐like domain over‐expressed in prostate cancer; however its biological role in prostate cancer development and progression remains unclear, which may, at least in part, be explained by its proteolytic processing. The extracellular part of TMEFF2 (TMEFF2‐ECD) is cleaved by ADAM17 and the membrane‐retained fragment is further processed by the gamma‐secretase complex. TMEFF2 shedding is increased with cell crowding, a condition associated with the tumour microenvironment, which was mediated by oxidative stress signalling, requiring jun‐kinase (JNK) activation. Moreover, we have identified that TMEFF2 is also a novel substrate for other proteases implicated in prostate cancer, including two ADAMs (ADAM9 and ADAM12) and the type II transmembrane serine proteinases (TTSPs) matriptase‐1 and hepsin. Whereas cleavage by ADAM9 and ADAM12 generates previously identified TMEFF2‐ECD, proteolytic processing by matriptase‐1 and hepsin produced TMEFF2 fragments, composed of TMEFF2‐ECD or FS and/or EGF‐like domains as well as novel membrane retained fragments. Differential TMEFF2 processing from a single transmembrane protein may be a general mechanism to modulate transmembrane protein levels and domains, dependent on the repertoire of ADAMs or TTSPs expressed by the target cell. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
12. Purity of transferred CD8+ T cells is crucial for safety and efficacy of combinatorial tumor immunotherapy in the absence of SHP-1.
- Author
-
Watson, H Angharad, Dolton, Garry, Ohme, Julia, Ladell, Kristin, Vigar, Miriam, Wehenkel, Sophie, Hindley, James, Mohammed, Rebar N, Miners, Kelly, Luckwell, Rhys A, Price, David A, Matthews, R James, and Ager, Ann
- Abstract
Adoptive transfer of tumor‐specific cytotoxic T cells is a promising advance in cancer therapy. Similarly, checkpoint inhibition has shown striking clinical results in some patients. Here we combine adoptive cell transfer with ablation of the checkpoint protein Src homology 2‐domain‐containing phosphatase 1 (SHP‐1, Ptpn6). Naturally occurring motheaten mice lack SHP‐1 and do not survive weaning due to extensive immunopathology. To circumvent this limitation, we created a novel SHP‐1null mouse that is viable up to 12 weeks of age by knocking out IL1r1. Using this model, we demonstrate that the absence of SHP‐1 augments the ability of adoptively transferred CD8+ T cells to control tumor growth. This therapeutic effect was only observed in situations where T‐cell numbers were limited, analogous to clinical settings. However, adoptive transfer of non‐CD8+ SHP‐1null hematopoietic cells resulted in lethal motheaten‐like pathology, indicating that systemic inhibition of SHP‐1 could have serious adverse effects. Despite this caveat, our findings support the development of SHP‐1 inhibition strategies in human T cells to complement adoptive transfer therapies in the clinic. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
13. SHP-1: the next checkpoint target for cancer immunotherapy?
- Author
-
Watson, H. Angharad, Wehenkel, Sophie, Matthews, James, and Ager, Ann
- Subjects
- *
IMMUNOTHERAPY , *CYTOTOXIC T cells , *CANCER education , *INCURABLE diseases , *PROTEIN-tyrosine kinases , *ANTIGEN receptors - Abstract
The immense power of the immune system is harnessed in healthy individuals by a range of negative regulatory signals and checkpoints. Manipulating these checkpoints through inhibition has resulted in striking immune-mediated clearance of otherwise untreatable tumours and metastases; unfortunately, not all patients respond to treatment with the currently available inhibitors of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1). Combinatorial studies using both anti-CTLA-4 and anti-PD-1 demonstrate synergistic effects of targeting multiple checkpoints, paving the way for other immune checkpoints to be targeted. Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) is a widely expressed inhibitory protein tyrosine phosphatase (PTP). In T-cells, it is a negative regulator of antigen-dependent activation and proliferation. It is a cytosolic protein, and therefore not amenable to antibody-mediated therapies, but its role in activation and proliferation makes it an attractive target for genetic manipulation in adoptive transfer strategies, such as chimeric antigen receptor (CAR) T-cells. This review will discuss the potential value of SHP-1 inhibition in future tumour immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
14. A distinct chemokine axis does not account for enrichment of Foxp3+ CD4+ T cells in carcinogen-induced fibrosarcomas.
- Author
-
Ondondo, Beatrice, Colbeck, Emily, Jones, Emma, Smart, Kathryn, Lauder, Sarah N., Hindley, James, Godkin, Andrew, Moser, Bernhard, Ager, Ann, and Gallimore, Awen
- Subjects
- *
CHEMOKINES , *FORKHEAD transcription factors , *CD4 antigen , *T cells , *CARCINOGENS , *FIBROSARCOMA , *TUMOR immunology - Abstract
The frequency of CD4+ Foxp3+ regulatory T (Treg) cells is often significantly increased in the blood of tumour-bearing mice and people with cancer. Moreover, Treg cell frequencies are often higher in tumours compared with blood and lymphoid organs. We wished to determine whether certain chemokines expressed within the tumour mass selectively recruit Treg cells, thereby contributing to their enrichment within the tumour-infiltrating lymphocyte pool. To achieve this goal, the chemokine profile of carcinogen-induced fibrosarcomas was determined, and the chemokine receptor expression profiles of both CD4+ Foxp3− and CD4+ Foxp3+ T cells were compared. These analyses revealed that the tumours are characterized by expression of inflammatory chemokines ( CCL2, CCL5, CCL7, CCL8, CCL12, CXCL9, CXCL10 and CX3 CL1), reflected by an enrichment of activated Foxp3− and Foxp3+ T cells expressing T helper type 1-associated chemokine receptors. Notably, we found that CXCR3+ T cells were significantly enriched in the tumours although curiously we found no evidence that CXCR3 was required for their recruitment. Instead, CXCR3 marks a population of activated Foxp3− and Foxp3+ T cells, which use multiple and overlapping ligand receptor pairs to guide their migration to tumours. Collectively, these data indicate that enrichment of Foxp3+ cells in tumours characterized by expression of inflammatory chemokines, does not occur via a distinct chemokine axis, thus selective chemokine blockade is unlikely to represent a meaningful therapeutic strategy for preventing Treg cell accumulation in tumours. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
15. Enhancement of T Cell Responses as a Result of Synergy between Lower Doses of Radiation and T Cell Stimulation.
- Author
-
Spary, Lisa K., Al-Taei, Saly, Salimu, Josephine, Cook, Alexander D., Ager, Ann, Angharad Watson, H., Clayton, Aled, Staffurth, John, Mason, Malcolm D., and Tabi, Zsuzsanna
- Subjects
- *
T cells , *LYMPHOCYTES , *MEDICAL technology , *RADIOLOGY , *RADIOBIOLOGY - Abstract
As a side effect of cancer radiotherapy, immune cells receive varying doses of radiation. Whereas high doses of radiation (>10 Gy) can lead to lymphopenia, lower radiation doses (2-4 Gy) represent a valid treatment option in some hematological cancers, triggering clinically relevant immunological changes. Based on our earlier observations, we hypothesized that lower radiation doses have a direct positive effect on T cells. In this study, we show that 0.6-2.4 Gy radiation enhances proliferation and IFN-γ production of PBMC or purified T cells induced by stimulation via the TCR. Radiation with 1.2 Gy also lowered T cell activation threshold and broadened the Th1 cytokine profile. Although radiation alone did not activate T cells, when followed by TCR stimulation, ERK1/2 and Akt phosphorylation increased above that induced by stimulation alone. These changes were followed by an early increase in glucose uptake. Naive (CD45RA+) or memory (CD45RA-) T cell responses to stimulation were boosted at similar rates by radiation. Whereas increased Ag-specific cytotoxic activity of a CD8+ T cell line manifested in a 4-h assay (10- 20% increase), highly significant (5- to 10-fold) differences in cytokine production were detected in 6-d Ag-stimulation assays of PBMC, probably as a net outcome of death of nonstimulated and enhanced response of Ag-stimulated T cells. T cells from patients receiving pelvic radiation (2.2-2.75 Gy) also displayed increased cytokine production when stimulated in vitro. We report in this study enhanced T cell function induced by synergistic radiation treatment, with potential physiological significance in a wide range of T cell responses. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
16. T-Cell Trafficking Facilitated by High Endothelial Venules Is Required for Tumor Control after Regulatory T-Cell Depletion.
- Author
-
Hindley, James P., Jones, Emma, Smart, Kathryn, Bridgeman, Hayley, Lauder, Sarah N., Ondondo, Beatrice, Cutting, Scott, Ladell, Kristin, Wynn, Katherine K., Withers, David, Price, David A., Ager, Ann, Godkin, Andrew J., and Gallimore, Awen M.
- Subjects
- *
T cells , *TUMOR growth , *ENDOTHELIAL cells , *CANCER immunotherapy , *CELL proliferation , *CANCER - Abstract
The evolution of immune blockades in tumors limits successful antitumor immunity, but the mechanisms underlying this process are not fully understood. Depletion of regulatory T cells (Treg), a T-cell subset that dampens excessive inflammatory and autoreactive responses, can allow activation of tumor-specific T cells. However, cancer immunotherapy studies have shown that a persistent failure of activated lymphocytes to infiltrate tumors remains a fundamental problem. In evaluating this issue, we found that despite an increase in T-cell activation and proliferation following Treg depletion, there was no significant association with tumor growth rate. In contrast, there was a highly significant association between low tumor growth rate and the extent of T-cell infiltration. Further analyses revealed a total concordance between low tumor growth rate, high T-cell infiltration, and the presence of high endothelial venules (HEV). HEV are blood vessels normally found in secondary lymphoid tissue where they are specialized for lymphocyte recruitment. Thus, our findings suggest that Treg depletion may promote HEV neogenesis, facilitating increased lymphocyte infiltration and destruction of the tumor tissue. These findings are important as they point to a hitherto unidentified role of Tregs, the manipulation of which may refine strategies for more effective cancer immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
17. Phosphatidylinositol-3-OH kinase and nutrient-sensing mTOR pathways control T lymphocyte trafficking.
- Author
-
Sinclair, Linda V, Finlay, David, Feijoo, Carmen, Cornish, Georgina H, Gray, Alex, Ager, Ann, Okkenhaug, Klaus, Hagenbeek, Thijs J, Spits, Hergen, and Cantrell, Doreen A
- Abstract
Phosphatidylinositol-3-OH kinase (PI(3)K) and the nutrient sensor mTOR are evolutionarily conserved regulators of cell metabolism. Here we show that PI(3)K and mTOR determined the repertoire of adhesion and chemokine receptors expressed by T lymphocytes. The key lymph node–homing receptors CD62L (L-selectin) and CCR7 were highly expressed on naive T lymphocytes but were downregulated after immune activation. CD62L downregulation occurred through ectodomain proteolysis and suppression of gene transcription. The p110δ subunit of PI(3)K controlled CD62L proteolysis through mitogen-activated protein kinases, whereas control of CD62L transcription by p110δ was mediated by mTOR through regulation of the transcription factor KLF2. PI(3)K-mTOR nutrient-sensing pathways also determined expression of the chemokine receptor CCR7 and regulated lymphocyte trafficking in vivo. Hence, lymphocytes use PI(3)K and mTOR to match metabolism and trafficking. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
18. Mutagenesis of the Ezrin-Radixin-Moesin Binding Domain of L-selectin Tail Affects Shedding, Microvillar Positioning, and Leukocyte Tethering.
- Author
-
Ivetic, Aleksander, Florey, Oliver, Deka, Jürgen, Haskard, Dorian O., Ager, Ann, and Ridley, Anne J.
- Subjects
- *
CELL adhesion molecules , *CELL adhesion , *LEUCOCYTES , *KILLER cells , *MUTAGENESIS , *GENETIC mutation , *BIOMOLECULES , *LIGANDS (Biochemistry) - Abstract
L-selectin is a cell adhesion molecule that mediates the initial capture (tethering) and subsequent rolling of leukocytes along ligands expressed on endothelial cells. We have previously identified ezrin and moesin as novel binding partners of the 17-amino acid L-selectin tail, but the biological role of this interaction is not known. Here we identify two basic amino acid residues within the L-selectin tail that are required for binding to ezrin-radixinmoesin (ERM) proteins: arginine 357 and lysine 362. L-selectin mutants defective for ERM binding show reduced localization to microvilli and decreased phorbol 12-myristate 13-acetate-induced shedding of the L-selectin ectodomain. Cells expressing these L-selectin mutants have reduced tethering to the L-selectin ligand P-selectin glycoprotein ligand-1, but rolling velocity on P-selectin glycoprotein ligand-1 is not affected. These results suggest that ERM proteins are required for microvillar positioning of L-selectin and that this is important both for leukocyte tethering and L-selectin shedding. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
19. High endothelial venules are rare in colorectal cancers but accumulate in extra-tumoral areas with disease progression.
- Author
-
Bento, Diana Costa, Jones, Emma, Junaid, Syed, Tull, Justyna, Williams, Geraint T, Godkin, Andrew, Ager, Ann, and Gallimore, Awen
- Subjects
- *
T cells , *COLON cancer , *RECTAL cancer , *ENDOTHELIAL cells , *LYMPHOCYTES - Abstract
Prolonged patient survival after surgical resection, is associated with a higher cytotoxic and memory T cell density within colorectal cancers (CRC). High endothelial venules (HEVs) are specialized blood vessels present in secondary lymphoid organs (SLO) that allow ingress of naïve and central memory T cells from the blood. It has been proposed that HEVs in tumors might serve as a similar route of entry for lymphocytes into the tumor and result in an improved prognosis. The present study aimed to characterize HEVs and their microenvironment in resected tumors from colorectal cancer patients (n= 62). We observed HEVs in association with lymphoid aggregates in 49 out of 62 patients. However, these HEV+lymphoid aggregates were largely at the invasive margin of the tumor and although there was an association with lymphocytes and HEVs at the invasive margin (p= 0.002) there was only a very weak association with tumor infiltrating lymphocytes. Indeed, lymphoid aggregates were associated with more advanced disease (Dukes’ stage C) and did not indicate a favorable prognosis. [ABSTRACT FROM PUBLISHER]
- Published
- 2015
- Full Text
- View/download PDF
20. Corrigendum: Phosphatidylinositol-3-OH kinase and nutrient-sensing mTOR pathways control T lymphocyte trafficking.
- Author
-
Sinclair, Linda V, Finlay, David, Feijoo, Carmen, Cornish, Georgina H, Gray, Alex, Ager, Ann, Okkenhaug, Klaus, Hagenbeek, Thijs J, Spits, Hergen, and Cantrell, Doreen A
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.