Network


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

Hotspot


Dive into the research topics where Sian Lax is active.

Publication


Featured researches published by Sian Lax.


Blood | 2012

Pericytes promote selective vessel regression to regulate vascular patterning

N Simonavicius; M. Ashenden; A van Weverwijk; Sian Lax; David L. Huso; Christopher D. Buckley; I J Huijbers; Helen Yarwood; Clare M. Isacke

Blood vessel networks form in a 2-step process of sprouting angiogenesis followed by selective branch regression and stabilization of remaining vessels. Pericytes are known to function in stabilizing blood vessels, but their role in vascular sprouting and selective vessel regression is poorly understood. The endosialin (CD248) receptor is expressed by pericytes associated with newly forming but not stable quiescent vessels. In the present study, we used the Endosialin(-/-) mouse as a means to uncover novel roles for pericytes during the process of vascular network formation. We demonstrate in a postnatal retina model that Endosialin(-/-) mice have normal vascular sprouting but are defective in selective vessel regression, leading to increased vessel density. Examination of the Endosialin(-/-) mouse tumor vasculature revealed an equivalent phenotype, indicating that pericytes perform a hitherto unidentified function to promote vessel destabilization and regression in vivo in both physiologic and pathologic angiogenesis. Mechanistically, Endosialin(-/-) mice have no defect in pericyte recruitment. Rather, endosialin binding to an endothelial associated, but not a pericyte associated, basement membrane component induces endothelial cell apoptosis and detachment. The results of the present study advance our understanding of pericyte biology and pericyte/endothelial cell cooperation during vascular patterning and have implications for the design of both pro- and antiangiogenic therapies.


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.


PLOS ONE | 2008

A Novel Role for PECAM-1 (CD31) in Regulating Haematopoietic Progenitor Cell Compartmentalization between the Peripheral Blood and Bone Marrow

Ewan A. Ross; Sylvie Freeman; Yan Zhao; Tarvinder S. Dhanjal; Emma J. Ross; Sian Lax; Zubair Ahmed; Tie Zheng Hou; Neena Kalia; Stuart Egginton; Gerard B. Nash; Steve P. Watson; Jon Frampton; Christopher D. Buckley

Although the expression of PECAM-1 (CD31) on vascular and haematopoietic cells within the bone marrow microenvironment has been recognized for some time, its physiological role within this niche remains unexplored. In this study we show that PECAM-1 influences steady state hematopoietic stem cell (HSC) progenitor numbers in the peripheral blood but not the bone marrow compartment. PECAM-1−/− mice have higher levels of HSC progenitors in the blood compared to their littermate controls. We show that PECAM-1 is required on both progenitors and bone marrow vascular cells in order for efficient transition between the blood and bone marrow to occur. We have identified key roles for PECAM-1 in both the regulation of HSC migration to the chemokine CXCL12, as well as maintaining levels of the matrix degrading enzyme MMP-9 in the bone marrow vascular niche. Using intravital microscopy and adoptive transfer of either wild type (WT) or PECAM-1−/− bone marrow precursors, we demonstrate that the increase in HSC progenitors in the blood is due in part to a reduced ability to migrate from blood to the bone marrow vascular niche. These findings suggest a novel role for PECAM-1 as a regulator of resting homeostatic progenitor cell numbers in the blood


Thorax | 2015

Vitamin D deficiency contributes directly to the acute respiratory distress syndrome (ARDS)

Rachel Dancer; Dhruv Parekh; Sian Lax; Vijay D'Souza; Shengxing Zheng; Chris R. Bassford; Daniel Park; Domokos Bartis; Rahul Mahida; Alice M Turner; Elizabeth Sapey; Wenbin Wei; Babu Naidu; Paul M. Stewart; William D. Fraser; Kenneth B. Christopher; Mark S. Cooper; Fang Gao; David M. Sansom; Adrian R. Martineau; Gavin D. Perkins; David R Thickett

Rationale Vitamin D deficiency has been implicated as a pathogenic factor in sepsis and intensive therapy unit mortality but has not been assessed as a risk factor for acute respiratory distress syndrome (ARDS). Causality of these associations has never been demonstrated. Objectives To determine if ARDS is associated with vitamin D deficiency in a clinical setting and to determine if vitamin D deficiency in experimental models of ARDS influences its severity. Methods Human, murine and in vitro primary alveolar epithelial cell work were included in this study. Findings Vitamin D deficiency (plasma 25(OH)D levels <50 nmol/L) was ubiquitous in patients with ARDS and present in the vast majority of patients at risk of developing ARDS following oesophagectomy. In a murine model of intratracheal lipopolysaccharide challenge, dietary-induced vitamin D deficiency resulted in exaggerated alveolar inflammation, epithelial damage and hypoxia. In vitro, vitamin D has trophic effects on primary human alveolar epithelial cells affecting >600 genes. In a clinical setting, pharmacological repletion of vitamin D prior to oesophagectomy reduced the observed changes of in vivo measurements of alveolar capillary damage seen in deficient patients. Conclusions Vitamin D deficiency is common in people who develop ARDS. This deficiency of vitamin D appears to contribute to the development of the condition, and approaches to correct vitamin D deficiency in patients at risk of ARDS should be developed. Trial registration UKCRN ID 11994.


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 | 2011

The stromal cell antigen CD248 (endosialin) is expressed on naive CD8+ human T cells and regulates proliferation

Deborah Hardie; Mj Baldwin; Amy Naylor; Oj Haworth; Tiezheng Hou; Sian Lax; Stephen Curnow; Nick Willcox; John R. MacFadyen; Clare M. Isacke; Christopher D. Buckley

CD248 (endosialin) is a transmembrane glycoprotein that is dynamically expressed on pericytes and fibroblasts during tissue development, tumour neovascularization and inflammation. Its role in tissue remodelling is associated with increased stromal cell proliferation and migration. We show that CD248 is also uniquely expressed by human, but not mouse (C57BL/6), CD8+ naive T cells. CD248 is found only on CD8+ CCR7+ CD11alow naive T cells and on CD8 single‐positive T cells in the thymus. Transfection of the CD248 negative T‐cell line MOLT‐4 with CD248 cDNA surprisingly reduced cell proliferation. Knock‐down of CD248 on naive CD8 T cells increased cell proliferation. These data demonstrate opposing functions for CD248 on haematopoietic (CD8+) versus stromal cells and suggests that CD248 helps to maintain naive CD8+ human T cells in a quiescent state.


Critical Care Medicine | 2017

Vitamin D deficiency in human and murine sepsis

Dhruv Parekh; Jaimin Patel; Aaron Scott; Sian Lax; Rachel Dancer; Vijay D'Souza; Hannah Greenwood; William D. Fraser; Fang Gao; Elizabeth Sapey; Gavin D. Perkins; David R Thickett

Objectives: Vitamin D deficiency has been implicated as a pathogenic factor in sepsis and ICU mortality but causality of these associations has not been demonstrated. To determine whether sepsis and severe sepsis are associated with vitamin D deficiency and to determine whether vitamin D deficiency influences the severity of sepsis. Design, Setting, and Patients: Sixty-one patients with sepsis and severe sepsis from two large U.K. hospitals and 20 healthy controls were recruited. Murine models of cecal ligation and puncture and intratracheal lipopolysaccharide were undertaken in normal and vitamin D deficient mice to address the issue of causality. Measurements and Main Results: Patients with severe sepsis had significantly lower concentrations of 25-hydroxyvitamin D3 than patients with either mild sepsis or age-matched healthy controls (15.7 vs 49.5 vs 66.5 nmol/L; p = 0.0001). 25-hydroxyvitamin D3 concentrations were significantly lower in patients who had positive microbiologic culture than those who were culture negative (p = 0.0023) as well as those who died within 30 days of hospital admission (p = 0.025). Vitamin D deficiency in murine sepsis was associated with increased peritoneal (p = 0.037), systemic (p = 0.019), and bronchoalveolar lavage (p = 0.011) quantitative bacterial culture. This was associated with reduced local expression of the cathelicidin-related antimicrobial peptide as well as evidence of defective macrophage phagocytosis (p = 0.029). In the intratracheal lipopolysaccharide model, 1,500 IU of intraperitoneal cholecalciferol treatment 6 hours postinjury reduced alveolar inflammation, cellular damage, and hypoxia. Conclusions: Vitamin D deficiency is common in severe sepsis. This appears to contribute to the development of the condition in clinically relevant murine models and approaches to correct vitamin D deficiency in patients with sepsis should be developed.


The Lancet | 2014

Vitamin D deficiency and bacterial load in a murine model of sepsis-induced lung injury

Dhruv Parekh; Sian Lax; Rachel Dancer; Gavin D. Perkins; David R Thickett

Abstract Background We have previously shown that patients with acute lung injury are severely vitamin D deficient. Several studies have reported a high prevalence of vitamin D deficiency in critically ill patients with sepsis, associated with increased morbidity and mortality, but whether this association is cause or effect is unknown. Bacteraemic sepsis is more common in the winter months when serum vitamin D concentrations are lowest. The purpose of this study was to investigate the local and systemic effects of vitamin D deficiency in a murine model of sepsis-induced lung injury where we can predictably time the initiating insult. Methods We fed eight wild-type C57BL/6 mice a diet completely devoid of vitamin D for 6 weeks to induce severe vitamin D deficiency (serum concentration 9 nmol/L) and compared them with seven mice fed a vitamin D sufficient diet (42 nmol/L). We used caecal ligation and puncture (CLP) to establish sepsis. Animals were culled 16 h after CLP, and blood, peritoneal lavage fluid (PLF), and bronchoalveloar lavage fluid (BALF) were collected. Cell infiltrates were assessed by flow cytometry. Fluid protein concentrations were measured, and tissue protein permeability index was calculated as the ratio between fluid and serum protein. Bacterial load was evaluated as colony-forming units (CFU) per ml after 24 h incubation on appropriate media. Statistical analysis was done with Minitab 16 statistical software. Findings Vitamin D deficient mice had significantly increased bacterial load compared with dietary sufficient mice in BALF, blood, and PLF (BALF median 2·51 × 10 3 CFU per mL [IQR 0·65 × 10 3 –1097 ×10 3 ] vs 0·51 ×10 3 [0·0–1·15 × 10 3 ], p=0·038; blood 66·1 × 10 3 [1·93 × 10 3 –175 × 10 3 ] vs 0·56 × 10 3 [0·0–5·56 × 10 3 ], p=0·042; and PLF 336 × 10 3 [52 × 10 3 –115 × 10 5 ] vs 8·22 × 10 3 [1·54 ×10 3 –20·6 × 10 3 ], p=0·005). BALF protein permeability index was higher in deficient mice than in sufficient mice (median 3·3 [IQR 2·61–4·64] vs 1·8 [1·22–2·09], p=0·0003) but there was no difference in cell numbers recruited to the lung. PLF protein permeability index was also higher in the deficient group than in sufficient mice (46·8 [28·1–59·3] vs 27·2 [14·2–37·3], p=0·05), with an associated increase in neutrophils recruited to the peritoneum (p=0·04). Interpretation Vitamin D deficiency significantly increased the bacterial load systemically, locally, and within the lung in a murine model of peritonitis. This increase was associated with a rise in tissue permeability locally and within the lung. These data support pre-existing vitamin D deficiency as a determinant of the severity of bacteraemic sepsis and might account for some of the seasonal variations observed in the incidence of sepsis. Funding UK Medical Research Council.


Thorax | 2013

S98 Vitamin D deficiency increases bacterial load in a murine model of sepsis-induced lung injury

D Parekh; Rca Dancer; Sian Lax; Gavin D. Perkins; Thickett

Introduction We have previously shown that patients with Acute Lung Injury (ALI) are severely vitamin D deficient. Several studies have reported a high prevalence of vitamin D deficiency in critically ill patients with sepsis, associated with increased morbidity and mortality but whether this is cause or effect is unknown. Bacteraemic sepsis is more common in the winter months when vitamin D levels are lowest. The purpose of this study was to investigate the local and systemic effects of vitamin D deficiency in a murine model of sepsis-induced lung injury where we can predictably time the initiating insult. Methods We fed 8 wild-type C57Bl/6 mice a diet completely devoid of vitamin D for 6 weeks to induce severe vitamin D deficiency (9 nmol/l) and compared to 7 mice fed a vitamin D sufficient diet (42 nmol/l). Caecal ligation and puncture (CLP) was used to establish sepsis. Animals were culled 16h after CLP and blood, peritoneal lavage fluid (PLF) and bronchoalveloar lavage fluid (BALF) were collected. Cell infiltrates were assessed by flow cytometry. Fluid protein levels were measured and tissue protein permeability index (PPI) was calculated as the ratio between fluid and serum protein. Bacterial load was evaluated as colony-forming units (CFU) after 24h incubation on appropriate media. Results Vitamin D deficient mice had increased bacterial load in BALF, blood and PLF compared to dietary sufficient mice. BALF protein permeability index was higher in deficient compared to sufficient mice but there was no difference in cell numbers recruited to the lung. PLF protein permeability index was also increased in the deficient group compared to sufficient mice with an associated significant increase in neutrophils recruited to the peritoneum. (See Table 1) Abstract S98 Table 1. Differences between dietary deficient and sufficient mice post CLP induced sepsis. Data is expressed as median values. Sufficient (n = 7) Deficient (n = 8) p-value Bacteria (CFU x103) BALFBloodPLF 0.510.288.22 2.5166.1336.6 0.0380.0190.005 PPI (x1000) BALFPLF 1.8227.2 3.3046.9 0.00030.05 Neutrophil Number BALFPLF (x106) 64.12.11 27.94.60 0.1830.04 Conclusion Vitamin D deficiency significantly increases the bacterial load both systemically, locally and within the lung in a murine model of peritonitis. This is associated with an increase in tissue permeability locally and within the lung. These data support pre-existing vitamin D deficiency as a determinant of the severity of bacteraemic sepsis and may account for some of the seasonal variations observed in the incidence of sepsis.


BMJ Open Respiratory Research | 2017

Effect of anti-podoplanin antibody administration during lipopolysaccharide-induced lung injury in mice

Sian Lax; Julie Rayes; David R Thickett; Steve P. Watson

Introduction Acute respiratory distress syndrome (ARDS) is a devastating pulmonary condition in the critically ill patient. A therapeutic intervention is yet to be found that can prevent progression to ARDS. We recently demonstrated that the interaction between podoplanin expressed on inflammatory alveolar macrophages (iAMs) and its endogenous ligand, platelet C-type lectin-like 2 (CLEC-2), protects against exaggerated lung inflammation during a mouse model of ARDS. In this study, we aim to investigate the therapeutic use of a crosslinking/activating anti-podoplanin antibody (α-PDPN, clone 8.1.1) during lipopolysaccharide (LPS)-induced lung inflammation in mice. Methods Intravenous administration of α-PDPN was performed 6 hours after intratracheal LPS in wildtype, C57Bl/6 mice. Lung function decline was measured by pulse oximetry as well as markers of local inflammation including bronchoalveolar lavage neutrophilia and cytokine/chemokine expression. In parallel, alveolar macrophages were isolated and cultured in vitro from haematopoietic-specific podoplanin-deficient mice (Pdpnfl/flVAV1cre+) and floxed-only controls treated with or without LPS in the presence or absence of α-PDPN. Results Lung function decline as well as alveolar neutrophil recruitment was significantly decreased in mice treated with the crosslinking/activating α-PDPN in vivo. Furthermore, we demonstrate that, in vitro, activation of podoplanin on iAMs regulates their secretion of proinflammatory cytokines and chemokines. Conclusions These data confirm the importance of the CLEC-2–podoplanin pathway during intratracheal (IT)-LPS and demonstrate the beneficial effect of targeting podoplanin during IT-LPS in mice possibly via modulation of local cytokine/chemokine expression. Moreover, these data suggest that podoplanin-targeted therapies may have a beneficial effect in patients at risk of developing ARDS.

Collaboration


Dive into the Sian Lax's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dhruv Parekh

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rachel Dancer

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ewan A. Ross

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

Masao Takata

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge