188 results on '"Iredale JP"'
Search Results
2. Resolving the fibrotic niche of human liver cirrhosis using single-cell transcriptomics
- Author
-
Ramachandran, P, primary, Dobie, R, additional, Wilson-Kanamori, JR, additional, Dora, EF, additional, Henderson, BEP, additional, Taylor, RS, additional, Matchett, KP, additional, Portman, JR, additional, Efremova, M, additional, Vento-Tormo, R, additional, Luu, NT, additional, Weston, CJ, additional, Newsome, PN, additional, Harrison, EM, additional, Mole, DJ, additional, Wigmore, SJ, additional, Iredale, JP, additional, Tacke, F, additional, Pollard, JW, additional, Ponting, CP, additional, Marioni, JC, additional, Teichmann, SA, additional, and Henderson, NC, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Inhibition of apoptosis of activated hepatic stellate cells by tissue inhibitor of metalloproteinase-1 is mediated via effects on matrix metalloproteinase inhibition: implications for reversibility of liver fibrosis
- Author
-
Murphy, FR, Issa, R, Zhou, X, Ratnarajah, S, Nagase, H, Arthur, MJ, Benyon, C, and Iredale, JP
- Abstract
The activated hepatic stellate cell (HSC) is central to liver fibrosis as the major source of collagens I and III and the tissue inhibitors of metalloproteinase-1 (TIMP-1). During spontaneous recovery from liver fibrosis, there is a decrease of TIMP expression, an increase in collagenase activity, and increased apoptosis of HSC, highlighting a potential role for TIMP-1 in HSC survival. In this report, we use tissue culture and in vivo models to demonstrate that TIMP-1 directly inhibits HSC apoptosis. TIMP-1 demonstrated a consistent, significant, and dose-dependent antiapoptotic effect for HSC activated in tissue culture and stimulated to undergo apoptosis by serum deprivation, cycloheximide exposure, and nerve growth factor stimulation. A nonfunctional mutated TIMP-1 (T2G mutant) in which all other domains are conserved did not inhibit apoptosis, indicating that inhibition of apoptosis was mediated through MMP inhibition. Synthetic MMP inhibitors also inhibited HSC apoptosis. Studies of experimental liver cirrhosis demonstrated that persistent expression of TIMP-1 mRNA determined by PCR correlated with persistence of activated HSC quantified by alpha smooth muscle actin staining, while in fibrosis, loss of activated HSC correlated with a reduction in TIMP-1 mRNA. We conclude that TIMP-1 inhibits apoptosis of activated HSC via MMP inhibition.
- Published
- 2016
4. Low-dose dexamethasone as a treatment for women with heavy menstrual bleeding: protocol for response-adaptive randomised placebo-controlled dose-finding parallel group trial (DexFEM)
- Author
-
Warner, P, Weir, CJ, Hansen, CH, Douglas, A, Madhra, M, Hillier, SG, Saunders, PTK, Iredale, JP, Semple, S, Walker, BR, and Critchley, HOD
- Abstract
INTRODUCTION: Heavy menstrual bleeding (HMB) diminishes individual quality-of-life and poses substantial societal burden. In HMB endometrium, inactivation of cortisol (by enzyme 11β hydroxysteroid dehydrogenase type 2 (11βHSD2)), may cause local endometrial glucocorticoid deficiency and hence increased angiogenesis and impaired vasoconstriction. We propose that 'rescue' of luteal phase endometrial glucocorticoid deficiency could reduce menstrual bleeding. METHODS AND ANALYSIS: DexFEM is a double-blind response-adaptive parallel-group placebo-controlled trial in women with HMB (108 to be randomised), with active treatment the potent oral synthetic glucocorticoid dexamethasone, which is relatively resistant to 11βHSD2 inactivation. Participants will be aged over 18 years, with mean measured menstrual blood loss (MBL) for two screening cycles ≥50 mL. The primary outcome is reduction in MBL from screening. Secondary end points are questionnaire assessments of treatment effect and acceptability. Treatment will be for 5 days in the mid-luteal phases of three treatment menstrual cycles. Six doses of low-dose dexamethasone (ranging from 0.2 to 0.9 mg twice daily) will be compared with placebo, to ascertain optimal dose, and whether this has advantage over placebo. Statistical efficiency is maximised by allowing randomisation probabilities to 'adapt' at five points during enrolment phase, based on the response data available so far, to favour doses expected to provide greatest additional information on the dose-response. Bayesian Normal Dynamic Linear Modelling, with baseline MBL included as covariate, will determine optimal dose (re reduction in MBL). Secondary end points will be analysed using generalised dynamic linear models. For each dose for all end points, a 95% credible interval will be calculated for effect versus placebo. ETHICS AND DISSEMINATION: Dexamethasone is widely used and hence well-characterised safety-wise. Ethical approval has been obtained from Scotland A Research Ethics Committee (12/SS/0147). Trial findings will be disseminated via open-access peer-reviewed publications, conferences, clinical networks, public lectures, and our websites. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov NCT01769820; EudractCT 2012-003405-98.
- Published
- 2015
5. Manipulation of liver regeneration with macrophages to influence the hepatic progenitor cell niche
- Author
-
Bird, TG, primary, Boutler, L, additional, Cole, A, additional, Lorenzini, S, additional, Lu, WY, additional, Hay, T, additional, Ridgway, R, additional, Williams, M, additional, Knight, B, additional, Keylock, S Gordon, additional, Wjotacha, D, additional, Jamieson, T, additional, Iredale, JP, additional, Clarke, AR, additional, Sansom, OJ, additional, and Forbes, SJ, additional
- Published
- 2013
- Full Text
- View/download PDF
6. Role of regulatory T cells in murine model of liver fibrosis resolution
- Author
-
Lim, X, primary, Ramachandran, P, additional, and Iredale, JP, additional
- Published
- 2012
- Full Text
- View/download PDF
7. Targeting the cytoskeleton inhibits proliferation of hepatoma cells by suppressing mitogenic signalling
- Author
-
Schrader, J, primary, Gordon-Walker, TT, additional, Fahl, M, additional, Lohse, AW, additional, and Iredale, JP, additional
- Published
- 2012
- Full Text
- View/download PDF
8. The Balance of TIMPs and MMP-2 Determines Hepatic Stellate Cell Fate during Recovery from Liver Fibrosis
- Author
-
Murphy, FR, primary, Zhou, X, additional, Issa, R, additional, Hussain, H, additional, Waung, J, additional, Patel, N, additional, Collins, J, additional, Brew, K, additional, Nagase, H, additional, Arthur, MJP, additional, Benyon, RC, additional, and Iredale, JP, additional
- Published
- 2003
- Full Text
- View/download PDF
9. Inhibition of Apoptosis of Activated Hepatic Stellate Cells by Timp-1 is Mediated via Effects on Mmp Inhibition: Implications for Reversibility of Liver Fibrosis
- Author
-
Murphy, F., primary, Issa, R., additional, Zhou, X., additional, Hussain, H., additional, Ratnarajah, S., additional, Soloway, P., additional, Nagase, H., additional, Mjp, Arthur, additional, Benyon, RC, additional, and Iredale, JP, additional
- Published
- 2002
- Full Text
- View/download PDF
10. The Role of Transforming Growth Factor-β in the Regulation of Matrix Metalloproteinases and Tissue Inhibitors of Metalloproteinases by Pancreatic Stellate Cells: Implications for Chronic Pancreatitis
- Author
-
Shek, FW, primary, Fmj, Walker, additional, Slf, Pender, additional, Chance, V., additional, Williams, EJ, additional, Johnson, CD, additional, Benyon, RC, additional, Iredale, JP, additional, and Fine, DR, additional
- Published
- 2002
- Full Text
- View/download PDF
11. Secretion and Activation of TGF-β Isoforms by Hepatic Stellate Cells and Consequences for Cell Proliferation and Survival
- Author
-
Williams, EJ, primary, Cochrane, BC, additional, Arthur, MJP, additional, Iredale, JP, additional, and Benyon, RC, additional
- Published
- 2001
- Full Text
- View/download PDF
12. Jun Family Proto-Oncogenes Modulate Timp-1 Promoter Activity in Rat Hepatic Stellate Cells
- Author
-
Smart, DE, primary, Mann, DA, additional, and Iredale, JP, additional
- Published
- 2000
- Full Text
- View/download PDF
13. Expression of tissue inhibitor of metalloproteinases 1 and 2 is increased in fibrotic human liver
- Author
-
Benyon, RC, primary, Iredale, JP, additional, Goddard, S, additional, Winwood, PJ, additional, and Arthur, MJ, additional
- Published
- 1996
- Full Text
- View/download PDF
14. Multicentre trial of octreotide versus injection sclerotherapy (IS) for acute variceal haemorrhage
- Author
-
Jenkins, SA, primary, Shields, R, additional, Sutton, R, additional, Kingsnorth, AN, additional, Davies, M, additional, Elias, E, additional, Turnbull, AJ, additional, Bassendine, MF, additional, James, OFW, additional, Iredale, JP, additional, Vyas, SK, additional, and Arthur, MJP, additional
- Published
- 1995
- Full Text
- View/download PDF
15. Tissue Inhibitor of Metalloproteinase-1 is Expressed by Hepatic Lipocytes and Upregulated in Cirrhosis and CCl4-Induced Liver
- Author
-
Iredale, JP, primary, Ferris, WF, primary, Murphy, G, primary, and Arthur, Mjp, primary
- Published
- 1994
- Full Text
- View/download PDF
16. Science, medicine, and the future: cirrhosis: new research provides a basis for rational and targeted treatments.
- Author
-
Iredale JP
- Published
- 2003
- Full Text
- View/download PDF
17. Resolving the fibrotic niche of human liver cirrhosis at single-cell level
- Author
-
Ramachandran, P, Dobie, R, Wilson-Kanamori, Dora, EF, Henderson, BEP, Luu, NT, Portman, Matchett, KP, Brice, M, Marwick, JA, Taylor, RS, Efremova, M, Vento-Tormo, R, Carragher, NO, Kendall, TJ, Fallowfield, JA, Harrison, EM, Mole, DJ, Wigmore, SJ, Newsome, PN, Weston, CJ, Iredale, JP, Tacke, F, Pollard, JW, Ponting, CP, Marioni, JC, Teichmann, SA, and Henderson, NC
- Subjects
Liver Cirrhosis ,Male ,Membrane Glycoproteins ,Receptor, Platelet-Derived Growth Factor alpha ,Macrophages ,Transendothelial and Transepithelial Migration ,Endothelial Cells ,Membrane Proteins ,Receptors, Cell Surface ,Tetraspanin 29 ,3. Good health ,Mice ,Phenotype ,Liver ,Case-Control Studies ,Hepatic Stellate Cells ,Hepatocytes ,Animals ,Humans ,Cell Lineage ,Female ,Receptors, Immunologic ,Single-Cell Analysis ,Duffy Blood-Group System ,Transcriptome - Abstract
Liver cirrhosis is a major cause of death worldwide and is characterized by extensive fibrosis. There are currently no effective antifibrotic therapies available. To obtain a better understanding of the cellular and molecular mechanisms involved in disease pathogenesis and enable the discovery of therapeutic targets, here we profile the transcriptomes of more than 100,000 single human cells, yielding molecular definitions for non-parenchymal cell types that are found in healthy and cirrhotic human liver. We identify a scar-associated TREM2+CD9+ subpopulation of macrophages, which expands in liver fibrosis, differentiates from circulating monocytes and is pro-fibrogenic. We also define ACKR1+ and PLVAP+ endothelial cells that expand in cirrhosis, are topographically restricted to the fibrotic niche and enhance the transmigration of leucocytes. Multi-lineage modelling of ligand and receptor interactions between the scar-associated macrophages, endothelial cells and PDGFRα+ collagen-producing mesenchymal cells reveals intra-scar activity of several pro-fibrogenic pathways including TNFRSF12A, PDGFR and NOTCH signalling. Our work dissects unanticipated aspects of the cellular and molecular basis of human organ fibrosis at a single-cell level, and provides a conceptual framework for the discovery of rational therapeutic targets in liver cirrhosis.
18. The Role of Transforming Growth Factor-ß in the Regulation of Matrix Metalloproteinases and Tissue Inhibitors of Metalloproteinases by Pancreatic Stellate Cells: Implications for Chronic Pancreatitis
- Author
-
Shek, FW, Fmj, Walker, Slf, Pender, Chance, V., Williams, EJ, Johnson, CD, Benyon, RC, Iredale, JP, and Fine, DR
- Published
- 2002
- Full Text
- View/download PDF
19. Secretion and Activation of TGF-ß Isoforms by Hepatic Stellate Cells and Consequences for Cell Proliferation and Survival
- Author
-
Williams, EJ, Cochrane, BC, Arthur, MJP, Iredale, JP, and Benyon, RC
- Published
- 2001
- Full Text
- View/download PDF
20. Characterisation of a stereotypical cellular and extracellular adult liver progenitor cell niche in rodents and diseased human liver
- Author
-
Christopher Bellamy, Rebecca L. Aucott, Luke Boulter, John P. Iredale, Elizabeth Clayton, S. Lorenzini, Mauro Bernardi, Kay Samuel, Malcolm R. Alison, Pietro Andreone, Stuart J. Forbes, Mathew Golding, Thomas G. Bird, Lorenzini S, Bird TG, Clayton E, Bellamy C, Samuel K, Aucott R, Andreone P, Bernardi M, Golding M, Alison MR, Iredale JP, and Forbes SJ.
- Subjects
Male ,Pathology ,medicine.medical_specialty ,CD34 ,HEPATIC PROGENITOR CELL NICHE ,STEM CELLS ,LIVER DAMAGE ,RODENTS ,HUMANS ,Mice, Transgenic ,Biology ,Article ,Mice ,Recurrence ,medicine ,Animals ,Humans ,Progenitor cell ,Myofibroblasts ,Cells, Cultured ,Progenitor ,Interleukin 3 ,Hepatocyte differentiation ,Liver Diseases ,Macrophages ,Stem Cells ,Gastroenterology ,Endothelial Cells ,Hepatitis C, Chronic ,Rats, Inbred F344 ,Cell biology ,Extracellular Matrix ,Liver Regeneration ,Rats ,Endothelial stem cell ,Mice, Inbred C57BL ,Disease Models, Animal ,Phenotype ,Liver ,Hepatocytes ,Female ,Laminin ,Stem cell ,Adult stem cell - Abstract
Background Stem/progenitor cell niches in tissues regulate stem/progenitor cell differentiation and proliferation through local signalling.Objective To examine the composition and formation of stem progenitor cell niches.Methods The composition of the hepatic progenitor cell niche in independent models of liver injury and hepatic progenitor cell activation in rodents and humans was studied. To identify the origin of the progenitor and niche cells, sex-mismatched bone marrow transplants in mice, who had received the choline–ethionine-deficient-diet to induce liver injury and progenitor cell activation, were used. The matrix surrounding the progenitor cells was described by immunohistochemical staining and its functional role controlling progenitor cell behaviour was studied in cell culture experiments using different matrix layers.Results The progenitor cell response in liver injury is intimately surrounded by myofibroblasts and macrophages, and to a lesser extent by endothelial cells. Hepatic progenitor cells are not of bone marrow origin; however, bone marrow-derived cells associate intimately with these cells and are macrophages. Laminin always surrounds the progenitor cells. In vitro studies showed that laminin aids maintenance of progenitor and biliary cell phenotype and promotes their gene expression (Dlk1, Aquaporin 1, γGT) while inhibiting hepatocyte differentiation and gene expression (CEPB/α).Conclusions During liver damage in rodents and humans a stereotypical cellular and laminin niche forms around hepatic progenitor cells. Laminin helps maintenance of undifferentiated progenitor cells. The niche links the intrahepatic progenitor cells with bone marrow-derived cells and links tissue damage with progenitor cell-mediated tissue repair.
- Published
- 2010
- Full Text
- View/download PDF
21. In utero exposures to perfluoroalkyl substances and the human fetal liver metabolome in Scotland: a cross-sectional study.
- Author
-
Hyötyläinen T, McGlinchey A, Salihovic S, Schubert A, Douglas A, Hay DC, O'Shaughnessy PJ, Iredale JP, Shaw S, Fowler PA, and Orešič M
- Subjects
- Adult, Pregnancy, Humans, Female, Male, Cross-Sectional Studies, Metabolome, Scotland, Bile Acids and Salts, Metabolic Diseases, Fluorocarbons adverse effects
- Abstract
Background: Perfluoroalkyl and polyfluoroalkyl substances are classed as endocrine disrupting compounds but continue to be used in many products such as firefighting foams, flame retardants, utensil coatings, and waterproofing of food packaging. Perfluoroalkyl exposure aberrantly modulates lipid, metabolite, and bile acid levels, increasing susceptibility to onset and severity of metabolic diseases, such as diabetes and metabolic dysfunction-associated steatotic liver disease. To date, most studies in humans have focused on perfluoroalkyl-exposure effects in adults. In this study we aimed to show if perfluoroalkyls are present in the human fetal liver and if they have metabolic consequences for the human fetus., Methods: In this cross-sectional study, human fetal livers from elective termination of pregnancies at the Aberdeen Pregnancy Counselling Service, Aberdeen, UK, were analysed by both targeted (bile acids and perfluoroalkyl substances) and combined targeted and untargeted (lipids and polar metabolites) mass spectrometry based metabolomic analyses, as well as with RNA-Seq. Only fetuses from normally progressing pregnancies (determined at ultrasound scan before termination), terminated for non-medical reasons, from women older than 16 years, fluent in English, and between 11 and 21 weeks of gestation were collected. Women exhibiting considerable emotional distress or whose fetuses had anomalies identified at ultrasound scan were excluded. Stringent bioinformatic and statistical methods such as partial correlation network analysis, linear regression, and pathway analysis were applied to this data to investigate the association of perfluoroalkyl exposure with hepatic metabolic pathways., Findings: Fetuses included in this study were collected between Dec 2, 2004, and Oct 27, 2014. 78 fetuses were included in the study: all 78 fetuses were included in the metabolomics analysis (40 female and 38 male) and 57 fetuses were included in the RNA-Seq analysis (28 female and 29 male). Metabolites associated with perfluoroalkyl were identified in the fetal liver and these varied with gestational age. Conjugated bile acids were markedly positively associated with fetal age. 23 amino acids, fatty acids, and sugar derivatives in fetal livers were inversely associated with perfluoroalkyl exposure, and the bile acid glycolithocholic acid was markedly positively associated with all quantified perfluoroalkyl. Furthermore, 7α-hydroxy-4-cholesten-3-one, a marker of bile acid synthesis rate, was strongly positively associated with perfluoroalkyl levels and was detectable as early as gestational week 12., Interpretation: Our study shows direct evidence for the in utero effects of perfluoroalkyl exposure on specific key hepatic products. Our results provide evidence that perfluoroalkyl exposure, with potential future consequences, manifests in the human fetus as early as the first trimester of gestation. Furthermore, the profiles of metabolic changes resemble those observed in perinatal perfluoroalkyl exposures. Such exposures are already linked with susceptibility, initiation, progression, and exacerbation of a wide range of metabolic diseases., Funding: UK Medical Research Council, Horizon Europe Program of the European Union, Seventh Framework Programme of the European Union, NHS Grampian Endowments grants, European Partnership for the Assessment of Risks from Chemicals, Swedish Research Council, Formas, Novo Nordisk Foundation, and the Academy of Finland., Competing Interests: Declaration of interests DCH is a founder, director, and shareholder in Stemnovate and Stimuliver. All other authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
22. Kynurenine monooxygenase regulates inflammation during critical illness and recovery in experimental acute pancreatitis.
- Author
-
Hayes AJ, Zheng X, O'Kelly J, Neyton LPA, Bochkina NA, Uings I, Liddle J, Baillie JK, Just G, Binnie M, Homer NZM, Murray TBJ, Baily J, McGuire K, Skouras C, Garden OJ, Webster SP, Iredale JP, Howie SEM, and Mole DJ
- Subjects
- Mice, Animals, Critical Illness, Multiple Organ Failure, Acute Disease, Mice, Knockout, Inflammation, Kynurenine 3-Monooxygenase genetics, Kynurenine, Pancreatitis
- Abstract
Kynurenine monooxygenase (KMO) blockade protects against multiple organ failure caused by acute pancreatitis (AP), but the link between KMO and systemic inflammation has eluded discovery until now. Here, we show that the KMO product 3-hydroxykynurenine primes innate immune signaling to exacerbate systemic inflammation during experimental AP. We find a tissue-specific role for KMO, where mice lacking Kmo solely in hepatocytes have elevated plasma 3-hydroxykynurenine levels that prime inflammatory gene transcription. 3-Hydroxykynurenine synergizes with interleukin-1β to cause cellular apoptosis. Critically, mice with elevated 3-hydroxykynurenine succumb fatally earlier and more readily to experimental AP. Therapeutically, blockade with the highly selective KMO inhibitor GSK898 rescues the phenotype, reducing 3-hydroxykynurenine and protecting against critical illness and death. Together, our findings establish KMO and 3-hydroxykynurenine as regulators of inflammation and the innate immune response to sterile inflammation. During critical illness, excess morbidity and death from multiple organ failure can be rescued by systemic KMO blockade., Competing Interests: Declaration of interests S.P.W. and D.J.M. are co-founders of Kynos Therapeutics, Ltd. D.J.M. is a board member of Kynos. The University of Edinburgh controls patents WO2015/091647, WO2016/097144, and WO2016/188827, which relate to inhibitors of KMO and include the compound used in this paper., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
23. Low dose dexamethasone as treatment for women with heavy menstrual bleeding: A response-adaptive randomised placebo-controlled dose-finding parallel group trial (DexFEM).
- Author
-
Warner P, Whitaker LHR, Parker RA, Weir CJ, Douglas A, Hansen CH, Madhra M, Hillier SG, Saunders PTK, Iredale JP, Semple S, Slayden OD, Walker BR, and Critchley HOD
- Subjects
- Adult, Dexamethasone therapeutic use, Endometrium blood supply, Female, Glucocorticoids therapeutic use, Humans, Menorrhagia physiopathology, Middle Aged, Vasoconstriction, Dexamethasone administration & dosage, Glucocorticoids administration & dosage, Menorrhagia drug therapy
- Abstract
Background: The symptom of heavy menstrual bleeding (HMB) diminishes quality-of-life for many mid-age women and imposes substantial societal burden. We investigated our hypothesis that HMB reflects impaired endometrial vasoconstriction due to endometrial glucocorticoid deficiency. Does reversing this deficiency, by short-term luteal-phase treatment with exogenous glucocorticoid (dexamethasone), ameliorate HMB?, Methods: In our Bayesian response-adaptive parallel-group placebo-controlled randomised trial, five pre-planned interim analyses used primary outcome data to adjust randomisation probabilities to favour doses providing most dose-response information. Participants with HMB, recruited from Lothian (Scotland) NHS clinics and via community invitations/advertisements, were aged over 18 years; reported regular 21-42 day menstrual cycles; and had measured menstrual blood loss (MBL) averaging ≥ 50 mL over two screening periods. Identically encapsulated placebo, or one of six Dexamethasone doses (0·2 mg, 0·4 mg, 0·5 mg, 0·6 mg, 0·75 mg, 0·9 mg), were taken orally twice-daily over five days in the mid-luteal phase of three menstrual cycles. Participants, investigators, and those measuring outcomes were masked to group assignment. Primary outcome, change in average MBL from screening to 'treatment', was analysed by allocated treatment, for all with data., Trial Registration: ClinicalTrials.gov NCT01769820; EudractCT 2012-003,405-98 FINDINGS: Recruitment lasted 29/01/2014 to 25/09/2017; 176 were screened, 107 randomised and 97 provided primary outcome data (n = 24,5,9,21,8,14,16 in the seven arms, placebo to 1·8 mg total daily active dose). In Bayesian normal dynamic linear modelling, 1·8 mg dexamethasone daily showed a 25 mL greater reduction in MBL from screening, than placebo (95% credible interval 1 to 49 mL), and probability 0·98 of benefit over placebo. Adverse events were reported by 75% (58/77) receiving dexamethasone, 58% (15/26) taking placebo. Three serious adverse events occurred, two during screening, one in a placebo participant. No woman withdrew due to adverse effects., Interpretation: Our adaptive trial in HMB showed that dexamethasone 1·8 mg daily reduced menstrual blood loss. The role of dexamethasone in HMB management deserves further investigation., Funding: UK MRC DCS/DPFS grant MR/J003611/1., Competing Interests: Declaration of Competing Interest PW, CHH, AD, LW & MM, PTKS, SGH, ODS, CJW and RAP have no competing interests to disclose. Other disclosures, all outside the current research study, are: BRW is a paid consultant for Actinogen Medical, an inventor on patents owned by the University of Edinburgh relating to manipulating and monitoring glucocorticoid action; receives royalties as editor of Davidson's Principles & Practice of Medicine published by Elsevier; and an honorarium as Chair of the MRC Population & Systems Medicine. SS receives support from GlaxoSmithKline for his research group. JPI is supported by the UK National Institute of Health Research (NIHR) and is Director of the University Hospitals Bristol Trust and University of Bristol NIHR Biomedical Research Centre. HODC reports clinical research support for laboratory consumables and staff from Bayer AG and she has also provided consultancy advice (but with no personal remuneration) for Bayer AG; Vifor Pharma; Gedeon Richter; Myovant. She receives royalties from Up-to-Date for article on Abnormal Uterine Bleeding., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Prostaglandin E 2 promotes intestinal inflammation via inhibiting microbiota-dependent regulatory T cells.
- Author
-
Crittenden S, Goepp M, Pollock J, Robb CT, Smyth DJ, Zhou Y, Andrews R, Tyrrell V, Gkikas K, Adima A, O'Connor RA, Davies L, Li XF, Yao HX, Ho GT, Zheng X, Mair A, Vermeren S, Qian BZ, Mole DJ, Gerasimidis K, Schwarze JKJ, Breyer RM, Arends MJ, O'Donnell VB, Iredale JP, Anderton SM, Narumiya S, Maizels RM, Rossi AG, Howie SE, and Yao C
- Subjects
- Dinoprostone pharmacology, Humans, Inflammation, Receptors, Prostaglandin E, EP2 Subtype, Gastrointestinal Microbiome, T-Lymphocytes, Regulatory
- Abstract
The gut microbiota fundamentally regulates intestinal homeostasis and disease partially through mechanisms that involve modulation of regulatory T cells (T
regs ), yet how the microbiota-Treg cross-talk is physiologically controlled is incompletely defined. Here, we report that prostaglandin E2 (PGE2 ), a well-known mediator of inflammation, inhibits mucosal Tregs in a manner depending on the gut microbiota. PGE2 through its receptor EP4 diminishes Treg -favorable commensal microbiota. Transfer of the gut microbiota that was modified by PGE2 -EP4 signaling modulates mucosal Treg responses and exacerbates intestinal inflammation. Mechanistically, PGE2 -modified microbiota regulates intestinal mononuclear phagocytes and type I interferon signaling. Depletion of mononuclear phagocytes or deficiency of type I interferon receptor diminishes PGE2 -dependent Treg inhibition. Together, our findings provide emergent evidence that PGE2 -mediated disruption of microbiota-Treg communication fosters intestinal inflammation., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution License 4.0 (CC BY).)- Published
- 2021
- Full Text
- View/download PDF
25. Integration of geoscience frameworks into digital pathology analysis permits quantification of microarchitectural relationships in histological landscapes.
- Author
-
Kendall TJ, Duff CM, Thomson AM, and Iredale JP
- Subjects
- Animals, Disease Models, Animal, Humans, Kidney ultrastructure, Liver ultrastructure, Liver Diseases pathology, Machine Learning, Mice, Organ Specificity, Pancreas ultrastructure, Reproducibility of Results, Software, Thyroid Diseases pathology, Thyroid Gland ultrastructure, Earth Sciences methods, Histological Techniques, Image Processing, Computer-Assisted methods, Pathology, Clinical methods
- Abstract
Although gold-standard histological assessment is subjective it remains central to diagnosis and clinical trial protocols and is crucial for the evaluation of any preclinical disease model. Objectivity and reproducibility are enhanced by quantitative analysis of histological images but current methods require application-specific algorithm training and fail to extract understanding from the histological context of observable features. We reinterpret histopathological images as disease landscapes to describe a generalisable framework defining topographic relationships in tissue using geoscience approaches. The framework requires no user-dependent training to operate on all image datasets in a classifier-agnostic manner but is adaptable and scalable, able to quantify occult abnormalities, derive mechanistic insights, and define a new feature class for machine-learning diagnostic classification. We demonstrate application to inflammatory, fibrotic and neoplastic disease in multiple organs, including the detection and quantification of occult lobular enlargement in the liver secondary to hilar obstruction. We anticipate this approach will provide a robust class of histological data for trial stratification or endpoints, provide quantitative endorsement of experimental models of disease, and could be incorporated within advanced approaches to clinical diagnostic pathology.
- Published
- 2020
- Full Text
- View/download PDF
26. Resolving the fibrotic niche of human liver cirrhosis at single-cell level.
- Author
-
Ramachandran P, Dobie R, Wilson-Kanamori JR, Dora EF, Henderson BEP, Luu NT, Portman JR, Matchett KP, Brice M, Marwick JA, Taylor RS, Efremova M, Vento-Tormo R, Carragher NO, Kendall TJ, Fallowfield JA, Harrison EM, Mole DJ, Wigmore SJ, Newsome PN, Weston CJ, Iredale JP, Tacke F, Pollard JW, Ponting CP, Marioni JC, Teichmann SA, and Henderson NC
- Subjects
- Animals, Case-Control Studies, Cell Lineage, Duffy Blood-Group System metabolism, Endothelial Cells metabolism, Female, Hepatic Stellate Cells cytology, Hepatic Stellate Cells metabolism, Hepatic Stellate Cells pathology, Hepatocytes cytology, Hepatocytes metabolism, Hepatocytes pathology, Humans, Liver cytology, Liver Cirrhosis genetics, Macrophages metabolism, Male, Membrane Glycoproteins metabolism, Membrane Proteins metabolism, Mice, Phenotype, Receptor, Platelet-Derived Growth Factor alpha metabolism, Receptors, Cell Surface metabolism, Receptors, Immunologic metabolism, Tetraspanin 29 metabolism, Transcriptome, Transendothelial and Transepithelial Migration, Endothelial Cells pathology, Liver pathology, Liver Cirrhosis pathology, Macrophages pathology, Single-Cell Analysis
- Abstract
Liver cirrhosis is a major cause of death worldwide and is characterized by extensive fibrosis. There are currently no effective antifibrotic therapies available. To obtain a better understanding of the cellular and molecular mechanisms involved in disease pathogenesis and enable the discovery of therapeutic targets, here we profile the transcriptomes of more than 100,000 single human cells, yielding molecular definitions for non-parenchymal cell types that are found in healthy and cirrhotic human liver. We identify a scar-associated TREM2
+ CD9+ subpopulation of macrophages, which expands in liver fibrosis, differentiates from circulating monocytes and is pro-fibrogenic. We also define ACKR1+ and PLVAP+ endothelial cells that expand in cirrhosis, are topographically restricted to the fibrotic niche and enhance the transmigration of leucocytes. Multi-lineage modelling of ligand and receptor interactions between the scar-associated macrophages, endothelial cells and PDGFRα+ collagen-producing mesenchymal cells reveals intra-scar activity of several pro-fibrogenic pathways including TNFRSF12A, PDGFR and NOTCH signalling. Our work dissects unanticipated aspects of the cellular and molecular basis of human organ fibrosis at a single-cell level, and provides a conceptual framework for the discovery of rational therapeutic targets in liver cirrhosis.- Published
- 2019
- Full Text
- View/download PDF
27. Embryonic mesothelial-derived hepatic lineage of quiescent and heterogenous scar-orchestrating cells defined but suppressed by WT1.
- Author
-
Kendall TJ, Duff CM, Boulter L, Wilson DH, Freyer E, Aitken S, Forbes SJ, Iredale JP, and Hastie ND
- Subjects
- Animals, Cell Lineage, Cicatrix metabolism, Hepatic Stellate Cells metabolism, Liver Cirrhosis metabolism, Mice, Myofibroblasts metabolism, WT1 Proteins metabolism, Cicatrix genetics, Epithelium embryology, Hepatic Stellate Cells cytology, Liver embryology, Liver Cirrhosis genetics, Myofibroblasts cytology, WT1 Proteins genetics
- Abstract
Activated hepatic stellate cells (aHSCs) orchestrate scarring during liver injury, with putative quiescent precursor mesodermal derivation. Here we use lineage-tracing from development, through adult homoeostasis, to fibrosis, to define morphologically and transcriptionally discreet subpopulations of aHSCs by expression of WT1, a transcription factor controlling morphological transitions in organogenesis and adult homoeostasis. Two distinct populations of aHSCs express WT1 after injury, and both re-engage a transcriptional signature reflecting embryonic mesothelial origin of their discreet quiescent adult precursor. WT1-deletion enhances fibrogenesis after injury, through upregulated Wnt-signalling and modulation of genes central to matrix persistence in aHSCs, and augmentation of myofibroblastic transition. The mesothelial-derived lineage demonstrates punctuated phenotypic plasticity through bidirectional mesothelial-mesenchymal transitions. Our findings demonstrate functional heterogeneity of adult scar-orchestrating cells that can be whole-life traced back through specific quiescent adult precursors to differential origin in development, and define WT1 as a paradoxical regulator of aHSCs induced by injury but suppressing scarring.
- Published
- 2019
- Full Text
- View/download PDF
28. Loss of Integrin αvβ8 in Murine Hepatocytes Accelerates Liver Regeneration.
- Author
-
Greenhalgh SN, Matchett KP, Taylor RS, Huang K, Li JT, Saeteurn K, Donnelly MC, Simpson EEM, Pollack JL, Atakilit A, Simpson KJ, Maher JJ, Iredale JP, Sheppard D, and Henderson NC
- Subjects
- Animals, Hepatocytes pathology, Liver pathology, Mice, Mice, Transgenic, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Cell Proliferation, Hepatocytes metabolism, Integrins deficiency, Liver metabolism, Liver Regeneration, Signal Transduction
- Abstract
Recent fate-mapping studies in mice have provided substantial evidence that mature adult hepatocytes are a major source of new hepatocytes after liver injury. In other systems, integrin αvβ8 has a major role in activating transforming growth factor (TGF)-β, a potent inhibitor of hepatocyte proliferation. We hypothesized that depletion of hepatocyte integrin αvβ8 would increase hepatocyte proliferation and accelerate liver regeneration after injury. Using Itgb8
flox/flox ;Alb-Cre mice to deplete hepatocyte αvβ8, after partial hepatectomy, hepatocyte proliferation and liver-to-body weight ratio were significantly increased in Itgb8flox/flox ;Alb-Cre mice compared with control mice. Antibody-mediated blockade of hepatocyte αvβ8 in vitro, with assessment of TGF-β signaling pathways by real-time quantitative PCR array, supported the hypothesis that integrin αvβ8 inhibition alters hepatocyte TGF-β signaling toward a pro-regenerative phenotype. A diethylnitrosamine-induced model of hepatocellular carcinoma, used to examine the possibility that this pro-proliferative phenotype might be oncogenic, revealed no difference in either tumor number or size between Itgb8flox/flox ;Alb-Cre and control mice. Immunohistochemistry for integrin αvβ8 in healthy and injured human liver demonstrated that human hepatocytes express integrin αvβ8. Depletion of hepatocyte integrin αvβ8 results in increased hepatocyte proliferation and accelerated liver regeneration after partial hepatectomy in mice. These data demonstrate that targeting integrin αvβ8 may represent a promising therapeutic strategy to drive liver regeneration in patients with a broad range of liver diseases., (Copyright © 2019 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
29. An efficient method to isolate Kupffer cells eliminating endothelial cell contamination and selective bias.
- Author
-
Lynch RW, Hawley CA, Pellicoro A, Bain CC, Iredale JP, and Jenkins SJ
- Subjects
- Animals, Endothelial Cells, Mice, Flow Cytometry methods, Kupffer Cells
- Abstract
Multicolor flow cytometry and cell sorting are powerful immunologic tools for the study of hepatic mϕ, yet there is no consensus on the optimal method to prepare liver homogenates for these analyses. Using a combination of mϕ and endothelial cell reporter mice, flow cytometry, and confocal imaging, we have shown that conventional flow-cytometric strategies for identification of Kupffer cells (KCs) leads to inclusion of a significant proportion of CD31
hi endothelial cells. These cells were present regardless of the method used to prepare cells for flow cytometry and represented endothelium tightly adhered to remnants of KC membrane. Antibodies to endothelial markers, such as CD31, were vital for their exclusion. This result brings into focus recently published microarray datasets that identify high expression of endothelial cell-associated genes by KCs compared with other tissue-resident mϕ. Our studies also revealed significant and specific loss of KCs among leukocytes with commonly used isolation methods that led to enrichment of proliferating and monocyte-derived mϕ. Hence, we present an optimal method to generate high yields of liver myeloid cells without bias for cell type or contamination with endothelial cells., (©2018 The Authors. Society for Leukocyte Biology Published by Wiley Periodicals, Inc.)- Published
- 2018
- Full Text
- View/download PDF
30. TGFβ inhibition restores a regenerative response in acute liver injury by suppressing paracrine senescence.
- Author
-
Bird TG, Müller M, Boulter L, Vincent DF, Ridgway RA, Lopez-Guadamillas E, Lu WY, Jamieson T, Govaere O, Campbell AD, Ferreira-Gonzalez S, Cole AM, Hay T, Simpson KJ, Clark W, Hedley A, Clarke M, Gentaz P, Nixon C, Bryce S, Kiourtis C, Sprangers J, Nibbs RJB, Van Rooijen N, Bartholin L, McGreal SR, Apte U, Barry ST, Iredale JP, Clarke AR, Serrano M, Roskams TA, Sansom OJ, and Forbes SJ
- Subjects
- Animals, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Disease Models, Animal, Hepatocytes metabolism, Hepatocytes pathology, Humans, Liver pathology, Macrophages metabolism, Male, Mice, Inbred C57BL, Necrosis, Signal Transduction, Transforming Growth Factor beta metabolism, Cellular Senescence, Liver injuries, Liver physiopathology, Liver Regeneration, Paracrine Communication, Transforming Growth Factor beta antagonists & inhibitors
- Abstract
Liver injury results in rapid regeneration through hepatocyte proliferation and hypertrophy. However, after acute severe injury, such as acetaminophen poisoning, effective regeneration may fail. We investigated how senescence may underlie this regenerative failure. In human acute liver disease, and murine models, p21-dependent hepatocellular senescence was proportionate to disease severity and was associated with impaired regeneration. In an acetaminophen injury mouse model, a transcriptional signature associated with the induction of paracrine senescence was observed within 24 hours and was followed by one of impaired proliferation. In mouse genetic models of hepatocyte injury and senescence, we observed transmission of senescence to local uninjured hepatocytes. Spread of senescence depended on macrophage-derived transforming growth factor-β1 (TGFβ1) ligand. In acetaminophen poisoning, inhibition of TGFβ receptor 1 (TGFβR1) improved mouse survival. TGFβR1 inhibition reduced senescence and enhanced liver regeneration even when delivered beyond the therapeutic window for treating acetaminophen poisoning. This mechanism, in which injury-induced senescence impairs liver regeneration, is an attractive therapeutic target for developing treatments for acute liver failure., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
31. 11Beta-hydroxysteroid dehydrogenase-1 deficiency or inhibition enhances hepatic myofibroblast activation in murine liver fibrosis.
- Author
-
Zou X, Ramachandran P, Kendall TJ, Pellicoro A, Dora E, Aucott RL, Manwani K, Man TY, Chapman KE, Henderson NC, Forbes SJ, Webster SP, Iredale JP, Walker BR, and Michailidou Z
- Subjects
- Animals, Disease Models, Animal, Hepatocytes, Male, Mice, Mice, Inbred C57BL, 11-beta-Hydroxysteroid Dehydrogenases antagonists & inhibitors, 11-beta-Hydroxysteroid Dehydrogenases deficiency, Liver Cirrhosis etiology, Myofibroblasts physiology
- Abstract
A hallmark of chronic liver injury is fibrosis, with accumulation of extracellular matrix orchestrated by activated hepatic stellate cells (HSCs). Glucocorticoids limit HSC activation in vitro, and tissue glucocorticoid levels are amplified by 11beta-hydroxysteroid dehydrogenase-1 (11βHSD1). Although 11βHSD1 inhibitors have been developed for type 2 diabetes mellitus and improve diet-induced fatty liver in various mouse models, effects on the progression and/or resolution of liver injury and consequent fibrosis have not been characterized. We have used the reversible carbon tetrachloride-induced model of hepatocyte injury and liver fibrosis to show that in two models of genetic 11βHSD1 deficiency (global, Hsd11b1
-/- , and hepatic myofibroblast-specific, Hsd11b1fl/fl /Pdgfrb-cre) 11βHSD1 pharmacological inhibition in vivo exacerbates hepatic myofibroblast activation and liver fibrosis. In contrast, liver injury and fibrosis in hepatocyte-specific Hsd11b1fl/fl /albumin-cre mice did not differ from that of controls, ruling out 11βHSD1 deficiency in hepatocytes as the cause of the increased fibrosis. In primary HSC culture, glucocorticoids inhibited expression of the key profibrotic genes Acta2 and Col1α1, an effect attenuated by the 11βHSD1 inhibitor [4-(2-chlorophenyl-4-fluoro-1-piperidinyl][5-(1H-pyrazol-4-yl)-3-thienyl]-methanone. HSCs from Hsd11b1-/- and Hsd11b1fl/fl /Pdgfrb-cre mice expressed higher levels of Acta2 and Col1α1 and were correspondingly more potently activated. In vivo [4-(2-chlorophenyl-4-fluoro-1-piperidinyl][5-(1H-pyrazol-4-yl)-3-thienyl]-methanone administration prior to chemical injury recapitulated findings in Hsd11b1-/- mice, including greater fibrosis., Conclusion: 11βHSD1 deficiency enhances myofibroblast activation and promotes initial fibrosis following chemical liver injury; hence, the effects of 11βHSD1 inhibitors on liver injury and repair are likely to be context-dependent and deserve careful scrutiny as these compounds are developed for chronic diseases including metabolic syndrome and dementia. (Hepatology 2018;67:2167-2181)., (© 2017 The Authors. Hepatology published by Wiley Periodicals, Inc. on behalf of American Association for the Study of Liver Diseases.)- Published
- 2018
- Full Text
- View/download PDF
32. The STAT3-IL-10-IL-6 Pathway Is a Novel Regulator of Macrophage Efferocytosis and Phenotypic Conversion in Sterile Liver Injury.
- Author
-
Campana L, Starkey Lewis PJ, Pellicoro A, Aucott RL, Man J, O'Duibhir E, Mok SE, Ferreira-Gonzalez S, Livingstone E, Greenhalgh SN, Hull KL, Kendall TJ, Vernimmen D, Henderson NC, Boulter L, Gregory CD, Feng Y, Anderton SM, Forbes SJ, and Iredale JP
- Subjects
- Adoptive Transfer, Animals, Apoptosis immunology, Humans, Liver pathology, Macrophages transplantation, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Necrosis immunology, Regeneration physiology, Zebrafish embryology, Interleukin-10 metabolism, Interleukin-6 metabolism, Liver injuries, Liver Cirrhosis pathology, Macrophages immunology, Phagocytosis immunology, STAT3 Transcription Factor metabolism
- Abstract
The disposal of apoptotic bodies by professional phagocytes is crucial to effective inflammation resolution. Our ability to improve the disposal of apoptotic bodies by professional phagocytes is impaired by a limited understanding of the molecular mechanisms that regulate the engulfment and digestion of the efferocytic cargo. Macrophages are professional phagocytes necessary for liver inflammation, fibrosis, and resolution, switching their phenotype from proinflammatory to restorative. Using sterile liver injury models, we show that the STAT3-IL-10-IL-6 axis is a positive regulator of macrophage efferocytosis, survival, and phenotypic conversion, directly linking debris engulfment to tissue repair., (Copyright © 2018 by The American Association of Immunologists, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
33. MDR1 deficiency impairs mitochondrial homeostasis and promotes intestinal inflammation.
- Author
-
Ho GT, Aird RE, Liu B, Boyapati RK, Kennedy NA, Dorward DA, Noble CL, Shimizu T, Carter RN, Chew ETS, Morton NM, Rossi AG, Sartor RB, Iredale JP, and Satsangi J
- Subjects
- Animals, Disease Models, Animal, Genetic Predisposition to Disease, Homeostasis, Humans, Metabolic Detoxication, Phase I genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Reactive Oxygen Species metabolism, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Colitis genetics, Inflammation genetics, Inflammatory Bowel Diseases genetics, Intestines immunology, Mitochondria physiology, Superoxide Dismutase genetics
- Abstract
The multidrug resistance-1 (MDR1) gene encodes an ATP-dependent efflux transporter that is highly expressed in the colon. In mice, loss of MDR1 function results in colitis with similarities to human inflammatory bowel diseases (IBD). Here, we show that MDR1 has an unexpected protective role for the mitochondria where MDR1 deficiency results in mitochondrial dysfunction with increased mitochondrial reactive oxygen species (mROS) driving the development of colitis. Exogenous induction of mROS accelerates, while inhibition attenuates colitis in vivo; these effects are amplified in MDR1 deficiency. In human IBD, MDR1 is negatively correlated to SOD2 gene expression required for mROS detoxification. To provide direct evidential support, we deleted intestinal SOD2 gene in mice and showed an increased susceptibility to colitis. We exploited the genome-wide association data sets and found many (∼5%) of IBD susceptibility genes with direct roles in regulating mitochondria homeostasis. As MDR1 primarily protects against xenotoxins via its efflux function, our findings implicate a distinct mitochondrial toxin+genetic susceptibility interaction leading to mitochondrial dysfunction, a novel pathogenic mechanism that could offer many new therapeutic opportunities for IBD.
- Published
- 2018
- Full Text
- View/download PDF
34. Detecting drug-target binding in cells using fluorescence-activated cell sorting coupled with mass spectrometry analysis.
- Author
-
Wilson K, Webster SP, Iredale JP, Zheng X, Homer NZ, Pham NT, Auer M, and Mole DJ
- Subjects
- Flow Cytometry, Fluorescence, HEK293 Cells, Humans, Mass Spectrometry, Biological Assay methods, Enzyme Inhibitors pharmacology, Kynurenine 3-Monooxygenase antagonists & inhibitors, Kynurenine 3-Monooxygenase genetics, Kynurenine 3-Monooxygenase metabolism
- Abstract
The assessment of drug-target engagement for determining the efficacy of a compound inside cells remains challenging, particularly for difficult target proteins. Existing techniques are more suited to soluble protein targets. Difficult target proteins include those with challenging in vitro solubility, stability or purification properties that preclude target isolation. Here, we report a novel technique that measures intracellular compound-target complex formation, as well as cellular permeability, specificity and cytotoxicity-the toxicity-affinity-permeability-selectivity (TAPS) technique. The TAPS assay is exemplified here using human kynurenine 3-monooxygenase (KMO), a challenging intracellular membrane protein target of significant current interest. TAPS confirmed target binding of known KMO inhibitors inside cells. We conclude that the TAPS assay can be used to facilitate intracellular hit validation on most, if not all intracellular drug targets.
- Published
- 2017
- Full Text
- View/download PDF
35. Erratum to: Modelling foetal exposure to maternal smoking using hepatoblasts from pluripotent stem cells.
- Author
-
Lucendo-Villarin B, Filis P, Swortwood MJ, Huestis MA, Meseguer-Ripolles J, Cameron K, Iredale JP, O'Shaughnessy PJ, Fowler PA, and Hay DC
- Abstract
During manuscript proofing, the following sentence was not deleted in the section "Results" at the end of the paragraph: "Both male and female hepatocytes responded in a similar fashion to cotinine, whereas male hepatocyte function was more sensitive to chrysene, fluorene and naphthalene than female hepatocytes".
- Published
- 2017
- Full Text
- View/download PDF
36. Modelling foetal exposure to maternal smoking using hepatoblasts from pluripotent stem cells.
- Author
-
Lucendo-Villarin B, Filis P, Swortwood MJ, Huestis MA, Meseguer-Ripolles J, Cameron K, Iredale JP, O'Shaughnessy PJ, Fowler PA, and Hay DC
- Subjects
- Adenosine Triphosphate metabolism, Cell Differentiation, Cells, Cultured, Cotinine toxicity, Embryonic Stem Cells cytology, Embryonic Stem Cells drug effects, Female, Humans, Male, Pluripotent Stem Cells cytology, Polycyclic Aromatic Hydrocarbons toxicity, Sex Factors, alpha-Fetoproteins metabolism, Hepatocytes cytology, Hepatocytes drug effects, Pluripotent Stem Cells drug effects, Smoking adverse effects
- Abstract
The liver is a dynamic organ which is both multifunctional and highly regenerative. A major role of the liver is to process both endo and xenobiotics. Cigarettes are an example of a legal and widely used drug which can cause major health problems for adults and constitute a particular risk to the foetus, if the mother smokes during pregnancy. Cigarette smoke contains a complex mixture of thousands of different xenobiotics, including nicotine and polycyclic aromatic hydrocarbons. These affect foetal development in a sex-specific manner, inducing sex-dependant molecular responses in different organs. To date, the effect of maternal smoking on the foetal liver has been studied in vitro using cell lines, primary tissue and animal models. While these models have proven to be useful, poor cell phenotype, tissue scarcity, batch-to-batch variation and species differences have led to difficulties in data extrapolation toward human development. Therefore, in this study we have employed hepatoblasts, derived from pluripotent stem cells, to model the effects of xenobiotics from cigarette smoke on human hepatocyte development. Highly pure hepatocyte populations (>90%) were produced in vitro and exposed to factors present in cigarette smoke. Analysis of ATP levels revealed that, independent of the sex, the majority of smoking derivatives tested individually did not deplete ATP levels below 50%. However, following exposure to a cocktail of smoking derivatives, ATP production fell below 50% in a sex-dependent manner. This was paralleled by a loss metabolic activity and secretory ability in both female and male hepatocytes. Interestingly, cell depletion was less pronounced in female hepatocytes, whereas caspase activation was ~twofold greater, indicating sex differences in cell death upon exposure to the smoking derivatives tested.
- Published
- 2017
- Full Text
- View/download PDF
37. αv integrins on mesenchymal cells regulate skeletal and cardiac muscle fibrosis.
- Author
-
Murray IR, Gonzalez ZN, Baily J, Dobie R, Wallace RJ, Mackinnon AC, Smith JR, Greenhalgh SN, Thompson AI, Conroy KP, Griggs DW, Ruminski PG, Gray GA, Singh M, Campbell MA, Kendall TJ, Dai J, Li Y, Iredale JP, Simpson H, Huard J, Péault B, and Henderson NC
- Subjects
- Animals, Apoptosis, Cell Movement, Cells, Cultured, Collagen metabolism, Fibrosis, Genotype, Humans, Male, Mesenchymal Stem Cells cytology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Receptor, Platelet-Derived Growth Factor beta metabolism, Recombinant Proteins metabolism, Integrin alphaV metabolism, Muscle, Skeletal pathology, Myocardium pathology, Receptor, Platelet-Derived Growth Factor beta genetics
- Abstract
Mesenchymal cells expressing platelet-derived growth factor receptor beta (PDGFRβ) are known to be important in fibrosis of organs such as the liver and kidney. Here we show that PDGFRβ
+ cells contribute to skeletal muscle and cardiac fibrosis via a mechanism that depends on αv integrins. Mice in which αv integrin is depleted in PDGFRβ+ cells are protected from cardiotoxin and laceration-induced skeletal muscle fibrosis and angiotensin II-induced cardiac fibrosis. In addition, a small-molecule inhibitor of αv integrins attenuates fibrosis, even when pre-established, in both skeletal and cardiac muscle, and improves skeletal muscle function. αv integrin blockade also reduces TGFβ activation in primary human skeletal muscle and cardiac PDGFRβ+ cells, suggesting that αv integrin inhibitors may be effective for the treatment and prevention of a broad range of muscle fibroses.- Published
- 2017
- Full Text
- View/download PDF
38. In search of a small molecule agonist of the relaxin receptor RXFP1 for the treatment of liver fibrosis.
- Author
-
McBride A, Hoy AM, Bamford MJ, Mossakowska DE, Ruediger MP, Griggs J, Desai S, Simpson K, Caballero-Hernandez I, Iredale JP, Pell T, Aucott RL, Holmes DS, Webster SP, and Fallowfield JA
- Subjects
- Biopsy, Cells, Cultured, Enzyme Activators chemical synthesis, Enzyme Activators pharmacology, High-Throughput Screening Assays, Humans, Liver Cirrhosis pathology, Drug Discovery methods, Drug Evaluation, Preclinical methods, Enzyme Activators isolation & purification, Liver Cirrhosis drug therapy, Receptors, G-Protein-Coupled agonists, Receptors, Peptide agonists
- Abstract
The peptide hormone human relaxin-2 (H2-RLX) has emerged as a potential therapy for cardiovascular and fibrotic diseases, but its short in vivo half-life is an obstacle to long-term administration. The discovery of ML290 demonstrated that it is possible to identify small molecule agonists of the cognate G-protein coupled receptor for H2-RLX (relaxin family peptide receptor-1 (RXFP1)). In our efforts to generate a new medicine for liver fibrosis, we sought to identify improved small molecule functional mimetics of H2-RLX with selective, full agonist or positive allosteric modulator activity against RXFP1. First, we confirmed expression of RXFP1 in human diseased liver. We developed a robust cellular cAMP reporter assay of RXFP1 signaling in HEK293 cells transiently expressing RXFP1. A high-throughput screen did not identify further specific agonists or positive allosteric modulators of RXFP1, affirming the low druggability of this receptor. As an alternative approach, we generated novel ML290 analogues and tested their activity in the HEK293-RXFP1 cAMP assay and the human hepatic cell line LX-2. Differences in activity of compounds on cAMP activation compared with changes in expression of fibrotic markers indicate the need to better understand cell- and tissue-specific signaling mechanisms and their disease-relevant phenotypes in order to enable drug discovery.
- Published
- 2017
- Full Text
- View/download PDF
39. Serelaxin as a potential treatment for renal dysfunction in cirrhosis: Preclinical evaluation and results of a randomized phase 2 trial.
- Author
-
Snowdon VK, Lachlan NJ, Hoy AM, Hadoke PW, Semple SI, Patel D, Mungall W, Kendall TJ, Thomson A, Lennen RJ, Jansen MA, Moran CM, Pellicoro A, Ramachandran P, Shaw I, Aucott RL, Severin T, Saini R, Pak J, Yates D, Dongre N, Duffield JS, Webb DJ, Iredale JP, Hayes PC, and Fallowfield JA
- Subjects
- Adolescent, Adult, Aged, Animals, Female, Glomerular Filtration Rate drug effects, Humans, Kidney blood supply, Male, Middle Aged, Rats, Rats, Sprague-Dawley, Recombinant Proteins pharmacology, Recombinant Proteins therapeutic use, Regional Blood Flow drug effects, Scotland, Young Adult, Kidney drug effects, Kidney Diseases drug therapy, Liver Cirrhosis drug therapy, Relaxin pharmacology, Relaxin therapeutic use
- Abstract
Background: Chronic liver scarring from any cause leads to cirrhosis, portal hypertension, and a progressive decline in renal blood flow and renal function. Extreme renal vasoconstriction characterizes hepatorenal syndrome, a functional and potentially reversible form of acute kidney injury in patients with advanced cirrhosis, but current therapy with systemic vasoconstrictors is ineffective in a substantial proportion of patients and is limited by ischemic adverse events. Serelaxin (recombinant human relaxin-2) is a peptide molecule with anti-fibrotic and vasoprotective properties that binds to relaxin family peptide receptor-1 (RXFP1) and has been shown to increase renal perfusion in healthy human volunteers. We hypothesized that serelaxin could ameliorate renal vasoconstriction and renal dysfunction in patients with cirrhosis and portal hypertension., Methods and Findings: To establish preclinical proof of concept, we developed two independent rat models of cirrhosis that were characterized by progressive reduction in renal blood flow and glomerular filtration rate and showed evidence of renal endothelial dysfunction. We then set out to further explore and validate our hypothesis in a phase 2 randomized open-label parallel-group study in male and female patients with alcohol-related cirrhosis and portal hypertension. Forty patients were randomized 1:1 to treatment with serelaxin intravenous (i.v.) infusion (for 60 min at 80 μg/kg/d and then 60 min at 30 μg/kg/d) or terlipressin (single 2-mg i.v. bolus), and the regional hemodynamic effects were quantified by phase contrast magnetic resonance angiography at baseline and after 120 min. The primary endpoint was the change from baseline in total renal artery blood flow. Therapeutic targeting of renal vasoconstriction with serelaxin in the rat models increased kidney perfusion, oxygenation, and function through reduction in renal vascular resistance, reversal of endothelial dysfunction, and increased activation of the AKT/eNOS/NO signaling pathway in the kidney. In the randomized clinical study, infusion of serelaxin for 120 min increased total renal arterial blood flow by 65% (95% CI 40%, 95%; p < 0.001) from baseline. Administration of serelaxin was safe and well tolerated, with no detrimental effect on systemic blood pressure or hepatic perfusion. The clinical study's main limitations were the relatively small sample size and stable, well-compensated population., Conclusions: Our mechanistic findings in rat models and exploratory study in human cirrhosis suggest the therapeutic potential of selective renal vasodilation using serelaxin as a new treatment for renal dysfunction in cirrhosis, although further validation in patients with more advanced cirrhosis and renal dysfunction is required., Trial Registration: ClinicalTrials.gov NCT01640964.
- Published
- 2017
- Full Text
- View/download PDF
40. Regression of Liver Fibrosis.
- Author
-
Campana L and Iredale JP
- Subjects
- Animals, Disease Models, Animal, Extracellular Matrix chemistry, Hepatic Stellate Cells physiology, Hepatocytes physiology, Humans, Immunity, Cellular, Liver metabolism, Liver Cirrhosis chemically induced, Liver Cirrhosis immunology, Liver Cirrhosis pathology, Macrophage Activation, Macrophages physiology, Metalloproteases metabolism, Liver cytology, Liver Cirrhosis therapy
- Abstract
Liver fibrosis is the final common pathway of chronic or iterative liver damage. Advanced chronic fibrosis is described as cirrhosis with a loss of architecture and attendant functional failure and the development of life-threatening complications. However, compelling evidence from rodent models and human studies indicates that if the injury is removed liver fibrosis is reversible. Hepatocytes, activated hepatic stellate cells, endothelial and immune cells, particularly macrophages, cooperate in the establishment and resolution of liver fibrosis. Here the authors provide a short overview of the mechanisms regulating the profibrotic and proresolution response, with the aim of highlighting potential new therapeutic targets. Liver disease is a major unmet medical need; currently, the sole approaches are the withdrawal of the injurious stimulus and liver transplantation. The authors conclude with a brief review of the feasibility of macrophage-based cell therapy for liver fibrosis., (Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.)
- Published
- 2017
- Full Text
- View/download PDF
41. Liver Fibrosis: Understanding the Dynamics of Bidirectional Wound Repair to Inform the Design of Markers and Therapies.
- Author
-
Iredale JP, Pellicoro A, and Fallowfield JA
- Subjects
- Animals, Humans, Liver Cirrhosis metabolism, Molecular Targeted Therapy, Myofibroblasts pathology, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled metabolism, Relaxin metabolism, Biomarkers metabolism, Liver Cirrhosis pathology, Liver Cirrhosis therapy
- Abstract
Background: Liver fibrosis is the final common pathway of iterative or chronic liver damage. When it reaches an advanced stage, cirrhosis develops and this indicates a loss of normal liver architecture, disruption of normal blood flow, the development of nodules of proliferative hepatocytes and consequent functional failure. Cirrhosis is also associated with life-threatening complications. Importantly, this dysregulation of blood flow involves changes resulting from both architectural disruption and a dynamic rise in portal pressure mediated in part by the contraction of myofibroblasts - the major cell mediator of the fibrogenic process - which are derived from hepatic stellate cells (HSC) and termed activated HSC (myofibroblast/activated HSC). Key Messages: There is now compelling data from both rodent and human models that liver fibrosis is bidirectional. By studying models of progressive and regressing liver fibrosis it has been possible to identify mediators that may be therapeutic targets. Arguably, by identifying the mediators of spontaneous resolution that result in a return of normal architecture, the attributes of a highly effective antifibrotic or pro resolution therapy can be defined. Among these key attributes, understanding the balance of matrix-degrading enzymes and their inhibitors (the metalloproteinases and Tissue inhibitors of metalloproteinases (TIMPs), respectively) led to the identification of both therapeutic targets and the value of TIMP-1 as a serum marker of fibrosis. Furthermore, the action of a potential therapeutic agent, relaxin (RLN) acting through its cognate G-protein coupled receptor (RXFP1) on the surface of activated HSC shows promise. Activation of the RLN-RXFP1-mediated pathway can be detected in vivo by measuring the dynamic contractility of activated HSC and the consequent changes in portal pressure and blood flow as a responsive physical biomarker., Conclusion: By understanding progressive and resolving liver fibrosis, a series of therapeutic targets have been identified. At the same time, key mediators of fibrosis may have an integral role to play as soluble or physical biomarkers., (© 2017 S. Karger AG, Basel.)
- Published
- 2017
- Full Text
- View/download PDF
42. BREXIT and science, where do we go from here.
- Author
-
Iredale JP
- Subjects
- Biomedical Research organization & administration, Biomedical Research trends, Research Support as Topic trends, United Kingdom, Universities organization & administration, Universities trends, European Union, Science trends
- Published
- 2016
- Full Text
- View/download PDF
43. Increased levels of 3-hydroxykynurenine parallel disease severity in human acute pancreatitis.
- Author
-
Skouras C, Zheng X, Binnie M, Homer NZ, Murray TB, Robertson D, Briody L, Paterson F, Spence H, Derr L, Hayes AJ, Tsoumanis A, Lyster D, Parks RW, Garden OJ, Iredale JP, Uings IJ, Liddle J, Wright WL, Dukes G, Webster SP, and Mole DJ
- Abstract
Inhibition of kynurenine 3-monooxygenase (KMO) protects against multiple organ dysfunction (MODS) in experimental acute pancreatitis (AP). We aimed to precisely define the kynurenine pathway activation in relation to AP and AP-MODS in humans, by carrying out a prospective observational study of all persons presenting with a potential diagnosis of AP for 90 days. We sampled peripheral venous blood at 0, 3, 6, 12, 24, 48, 72 and 168 hours post-recruitment. We measured tryptophan metabolite concentrations and analysed these in the context of clinical data and disease severity indices, cytokine profiles and C-reactive protein (CRP) concentrations. 79 individuals were recruited (median age: 59.6 years; 47 males, 59.5%). 57 met the revised Atlanta definition of AP: 25 had mild, 23 moderate, and 9 severe AP. Plasma 3-hydroxykynurenine concentrations correlated with contemporaneous APACHE II scores (R
2 = 0.273; Spearman rho = 0.581; P < 0.001) and CRP (R2 = 0.132; Spearman rho = 0.455, P < 0.001). Temporal profiling showed early tryptophan depletion and contemporaneous 3-hydroxykynurenine elevation. Furthermore, plasma concentrations of 3-hydroxykynurenine paralleled systemic inflammation and AP severity. These findings support the rationale for investigating early intervention with a KMO inhibitor, with the aim of reducing the incidence and severity of AP-associated organ dysfunction.- Published
- 2016
- Full Text
- View/download PDF
44. Overexpression of human kynurenine-3-monooxygenase protects against 3-hydroxykynurenine-mediated apoptosis through bidirectional nonlinear feedback.
- Author
-
Wilson K, Auer M, Binnie M, Zheng X, Pham NT, Iredale JP, Webster SP, and Mole DJ
- Subjects
- Amino Acid Chloromethyl Ketones pharmacology, Enzyme Inhibitors pharmacology, HEK293 Cells, Humans, Kynurenine toxicity, Kynurenine 3-Monooxygenase antagonists & inhibitors, Kynurenine 3-Monooxygenase genetics, Microscopy, Confocal, Mitochondria metabolism, Nitric Oxide Synthase Type II metabolism, Pentosyltransferases antagonists & inhibitors, Pentosyltransferases genetics, Pentosyltransferases metabolism, Plasmids genetics, Plasmids metabolism, RNA Interference, RNA, Small Interfering metabolism, Real-Time Polymerase Chain Reaction, Time-Lapse Imaging, Transfection, Apoptosis drug effects, Kynurenine analogs & derivatives, Kynurenine 3-Monooxygenase metabolism
- Abstract
Kynurenine 3-monooxygenase (KMO) is a critical regulator of inflammation. The preferred KMO substrate, kynurenine, is converted to 3-hydroxykynurenine (3HK), and this product exhibits cytotoxicity through mechanisms that culminate in apoptosis. Here, we report that overexpression of human KMO with orthotopic localisation to mitochondria creates a metabolic environment during which the cell exhibits increased tolerance for exogenous 3HK-mediated cellular injury. Using the selective KMO inhibitor Ro61-8048, we show that KMO enzyme function is essential for cellular protection. Pan-caspase inhibition with Z-VAD-FMK confirmed apoptosis as the mode of cell death. By defining expression of pathway components upstream and downstream of KMO, we observed alterations in other key kynurenine pathway components, particularly tryptophan-2,3-dioxygenase upregulation, through bidirectional nonlinear feedback. KMO overexpression also increased expression of inducible nitric oxide synthase (iNOS). These changes in gene expression are functionally relevant, because siRNA knockdown of the pathway components kynureninase and quinolinate phosphoribosyl transferase caused cells to revert to a state of susceptibility to 3HK-mediated apoptosis. In summary, KMO overexpression, and importantly KMO activity, have metabolic repercussions that fundamentally affect resistance to cell stress.
- Published
- 2016
- Full Text
- View/download PDF
45. Prostaglandin E₂ constrains systemic inflammation through an innate lymphoid cell-IL-22 axis.
- Author
-
Duffin R, O'Connor RA, Crittenden S, Forster T, Yu C, Zheng X, Smyth D, Robb CT, Rossi F, Skouras C, Tang S, Richards J, Pellicoro A, Weller RB, Breyer RM, Mole DJ, Iredale JP, Anderton SM, Narumiya S, Maizels RM, Ghazal P, Howie SE, Rossi AG, and Yao C
- Subjects
- Animals, Bacterial Infections genetics, Bacterial Infections immunology, Gene Expression, Humans, Immunity, Innate, Inflammation drug therapy, Inflammation microbiology, Intestines microbiology, Mice, Receptors, Prostaglandin E, EP4 Subtype antagonists & inhibitors, Receptors, Prostaglandin E, EP4 Subtype genetics, Signal Transduction, Interleukin-22, Dinoprostone immunology, Inflammation immunology, Interleukins immunology, Intestines immunology, Lymphocytes immunology, Receptors, Prostaglandin E, EP4 Subtype immunology
- Abstract
Systemic inflammation, which results from the massive release of proinflammatory molecules into the circulatory system, is a major risk factor for severe illness, but the precise mechanisms underlying its control are not fully understood. We observed that prostaglandin E2 (PGE2), through its receptor EP4, is down-regulated in human systemic inflammatory disease. Mice with reduced PGE2 synthesis develop systemic inflammation, associated with translocation of gut bacteria, which can be prevented by treatment with EP4 agonists. Mechanistically, we demonstrate that PGE2-EP4 signaling acts directly on type 3 innate lymphoid cells (ILCs), promoting their homeostasis and driving them to produce interleukin-22 (IL-22). Disruption of the ILC-IL-22 axis impairs PGE2-mediated inhibition of systemic inflammation. Hence, the ILC-IL-22 axis is essential in protecting against gut barrier dysfunction, enabling PGE2-EP4 signaling to impede systemic inflammation., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
46. Kynurenine-3-monooxygenase inhibition prevents multiple organ failure in rodent models of acute pancreatitis.
- Author
-
Mole DJ, Webster SP, Uings I, Zheng X, Binnie M, Wilson K, Hutchinson JP, Mirguet O, Walker A, Beaufils B, Ancellin N, Trottet L, Bénéton V, Mowat CG, Wilkinson M, Rowland P, Haslam C, McBride A, Homer NZ, Baily JE, Sharp MG, Garden OJ, Hughes J, Howie SE, Holmes DS, Liddle J, and Iredale JP
- Subjects
- Acute Disease, Animals, Chromatography, Liquid, Crystallography, X-Ray, Disease Models, Animal, HEK293 Cells, Hepatocytes metabolism, Humans, In Vitro Techniques, Kidney metabolism, Kidney pathology, Kynurenine 3-Monooxygenase genetics, Lung metabolism, Lung pathology, Mice, Mice, Knockout, Multiple Organ Failure etiology, Multiple Organ Failure pathology, Pancreas metabolism, Pancreas pathology, Pancreatitis complications, Pancreatitis pathology, Rats, Tandem Mass Spectrometry, Tryptophan metabolism, Benzoxazoles pharmacology, Kynurenine 3-Monooxygenase antagonists & inhibitors, Multiple Organ Failure genetics, Oxazolidinones pharmacology, Pancreatitis genetics, Propionates pharmacology, RNA, Messenger metabolism
- Abstract
Acute pancreatitis (AP) is a common and devastating inflammatory condition of the pancreas that is considered to be a paradigm of sterile inflammation leading to systemic multiple organ dysfunction syndrome (MODS) and death. Acute mortality from AP-MODS exceeds 20% (ref. 3), and the lifespans of those who survive the initial episode are typically shorter than those of the general population. There are no specific therapies available to protect individuals from AP-MODS. Here we show that kynurenine-3-monooxygenase (KMO), a key enzyme of tryptophan metabolism, is central to the pathogenesis of AP-MODS. We created a mouse strain that is deficient for Kmo (encoding KMO) and that has a robust biochemical phenotype that protects against extrapancreatic tissue injury to the lung, kidney and liver in experimental AP-MODS. A medicinal chemistry strategy based on modifications of the kynurenine substrate led to the discovery of the oxazolidinone GSK180 as a potent and specific inhibitor of KMO. The binding mode of the inhibitor in the active site was confirmed by X-ray co-crystallography at 3.2 Å resolution. Treatment with GSK180 resulted in rapid changes in the levels of kynurenine pathway metabolites in vivo, and it afforded therapeutic protection against MODS in a rat model of AP. Our findings establish KMO inhibition as a novel therapeutic strategy in the treatment of AP-MODS, and they open up a new area for drug discovery in critical illness.
- Published
- 2016
- Full Text
- View/download PDF
47. Polymer Supported Directed Differentiation Reveals a Unique Gene Signature Predicting Stable Hepatocyte Performance.
- Author
-
Villarin BL, Cameron K, Szkolnicka D, Rashidi H, Bates N, Kimber SJ, Flint O, Forbes SJ, Iredale JP, Bradley M, and Hay DC
- Subjects
- Cells, Cultured, Embryonic Stem Cells cytology, Humans, Liver cytology, Pluripotent Stem Cells cytology, Pluripotent Stem Cells metabolism, Quality Control, Transfection, Cell Differentiation, Hepatocytes metabolism, Polymers chemistry
- Abstract
In theory, pluripotent stem cells can give rise to all somatic cell types found in the human body. The ability to generate renewable sources of human cells has enormous potential to improve human health and wealth. One major obstacle to the routine deployment of stem cell-derived cells is their instability in culture. To tackle this issue a synthetic polymer surface is used., (© 2015 The Authors. Published by WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
48. Polyurethane: Stable Cell Phenotype Requires Plasticity: Polymer Supported Directed Differentiation Reveals a Unique Gene Signature Predicting Stable Hepatocyte Performance (Adv. Healthcare Mater. 12/2015).
- Author
-
Villarin BL, Cameron K, Szkolnicka D, Rashidi H, Bates N, Kimber SJ, Flint O, Forbes SJ, Iredale JP, Bradley M, and Hay DC
- Abstract
One major obstacle to the routine deployment of stem cell-derived cells is their instability in culture. On page 1820 David C. Hay and co-workers describe the use of a synthetic polymer surface. The image shows stem cell-derived hepatocytes replated on this polyurethane surface. Importantly the cells express Zonal Occludin (green stain) at the cell surface, which indicates that the cells display elements of polarization. The blue stain is DAPI, which demarks the nucleus., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
49. Hepatic progenitor cells of biliary origin with liver repopulation capacity.
- Author
-
Lu WY, Bird TG, Boulter L, Tsuchiya A, Cole AM, Hay T, Guest RV, Wojtacha D, Man TY, Mackinnon A, Ridgway RA, Kendall T, Williams MJ, Jamieson T, Raven A, Hay DC, Iredale JP, Clarke AR, Sansom OJ, and Forbes SJ
- Subjects
- Animals, Apoptosis, Bile Ducts metabolism, Bile Ducts pathology, Biomarkers metabolism, Cell Separation, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Epithelial Cells metabolism, Epithelial Cells pathology, Female, Genotype, Hepatocytes metabolism, Hepatocytes pathology, Male, Mice, Inbred C57BL, Mice, Knockout, Necrosis, Phenotype, Proto-Oncogene Proteins c-mdm2 deficiency, Proto-Oncogene Proteins c-mdm2 genetics, Time Factors, Bile Ducts transplantation, Cell Lineage, Cell Proliferation, Epithelial Cells transplantation, Hepatocytes transplantation, Liver metabolism, Liver pathology, Liver Regeneration, Stem Cell Transplantation, Stem Cells metabolism, Stem Cells pathology
- Abstract
Hepatocytes and cholangiocytes self-renew following liver injury. Following severe injury hepatocytes are increasingly senescent, but whether hepatic progenitor cells (HPCs) then contribute to liver regeneration is unclear. Here, we describe a mouse model where the E3 ubiquitin ligase Mdm2 is inducibly deleted in more than 98% of hepatocytes, causing apoptosis, necrosis and senescence with nearly all hepatocytes expressing p21. This results in florid HPC activation, which is necessary for survival, followed by complete, functional liver reconstitution. HPCs isolated from genetically normal mice, using cell surface markers, were highly expandable and phenotypically stable in vitro. These HPCs were transplanted into adult mouse livers where hepatocyte Mdm2 was repeatedly deleted, creating a non-competitive repopulation assay. Transplanted HPCs contributed significantly to restoration of liver parenchyma, regenerating hepatocytes and biliary epithelia, highlighting their in vivo lineage potency. HPCs are therefore a potential future alternative to hepatocyte or liver transplantation for liver disease.
- Published
- 2015
- Full Text
- View/download PDF
50. Liver fibrosis: Therapeutic armory 40 years on.
- Author
-
Iredale JP, Sci FM, and Pellicoro A
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.