Publications

2022

  1. Single cell RNA sequencing and lineage tracing confirm mesenchyme to epithelial transformation (MET) contributes to repair of the endometrium at menstruation
    P. M. Kirkwood, D. A. Gibson, I. Shaw, R. Dobie, O. Kelepouri, N. C. Henderson, and P. T. K. Saunders
    Elife Dec 2022

    The human endometrium experiences repetitive cycles of tissue wounding characterised by piecemeal shedding of the surface epithelium and rapid restoration of tissue homeostasis. In this study we used a mouse model of endometrial repair and three transgenic lines of mice to investigate whether epithelial cells that become incorporated into the newly formed luminal epithelium have their origins in one or more of the mesenchymal cell types present in the stromal compartment of the endometrium. Using scRNAseq we identified a novel population of PDGFRb+ mesenchymal stromal cells that developed a unique transcriptomic signature in response to endometrial breakdown/repair. These cells expressed genes usually considered specific to epithelial cells and in silico trajectory analysis suggested they were stromal fibroblasts in transition to becoming epithelial cells. To confirm our hypothesis we used a lineage tracing strategy to compare the fate of stromal fibroblasts (PDGFRa+) and stromal perivascular cells (NG2/CSPG4+). We demonstrated that stromal fibroblasts can undergo a mesenchyme to epithelial transformation and become incorporated into the re-epithelialised luminal surface of the repaired tissue. This study is the first to discover a novel population of wound-responsive, plastic endometrial stromal fibroblasts that contribute to the rapid restoration of an intact luminal epithelium during endometrial repair. These findings form a platform for comparisons both to endometrial pathologies which involve a fibrotic response (Asherman’s syndrome, endometriosis) as well as other mucosal tissues which have a variable response to wounding.

    @article{pmid36524724,
      author = {Kirkwood, P. M. and Gibson, D. A. and Shaw, I. and Dobie, R. and Kelepouri, O. and Henderson, N. C. and Saunders, P. T. K.},
      title = {{{S}ingle cell {R}{N}{A} sequencing and lineage tracing confirm mesenchyme to epithelial transformation ({M}{E}{T}) contributes to repair of the endometrium at menstruation}},
      journal = {Elife},
      year = {2022},
      volume = {11},
      month = dec
    }
  2. Single-cell analysis of senescent epithelia reveals targetable mechanisms promoting fibrosis
    E. D. O’Sullivan, K. J. Mylonas, R. Bell, C. Carvalho, D. P. Baird, C. Cairns, K. M. Gallagher, R. Campbell, M. Docherty, A. Laird, N. C. Henderson, T. Chandra, K. Kirschner, B. Conway, G. H. Dihazi, M. Zeisberg, J. Hughes, L. Denby, H. Dihazi, and D. A. Ferenbach
    JCI Insight Nov 2022

    -mediated fibroblast activation. Inhibition of PDIA3 in vivo significantly reduced kidney fibrosis during ongoing renal injury and as such represented a new potential therapeutic pathway. Analysis of the signaling pathways of senescent epithelia connected senescence to organ fibrosis, permitting rational design of antifibrotic therapies.

    @article{pmid36509292,
      author = {O'Sullivan, E. D. and Mylonas, K. J. and Bell, R. and Carvalho, C. and Baird, D. P. and Cairns, C. and Gallagher, K. M. and Campbell, R. and Docherty, M. and Laird, A. and Henderson, N. C. and Chandra, T. and Kirschner, K. and Conway, B. and Dihazi, G. H. and Zeisberg, M. and Hughes, J. and Denby, L. and Dihazi, H. and Ferenbach, D. A.},
      title = {{{S}ingle-cell analysis of senescent epithelia reveals targetable mechanisms promoting fibrosis}},
      journal = {JCI Insight},
      year = {2022},
      volume = {7},
      number = {22},
      month = nov
    }
  3. Opposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis
    A. Filliol, Y. Saito, A. Nair, D. H. Dapito, L. X. Yu, A. Ravichandra, S. Bhattacharjee, S. Affo, N. Fujiwara, H. Su, Q. Sun, T. M. Savage, J. R. Wilson-Kanamori, J. M. Caviglia, L. Chin, D. Chen, X. Wang, S. Caruso, J. K. Kang, A. D. Amin, S. Wallace, R. Dobie, D. Yin, O. M. Rodriguez-Fiallos, C. Yin, A. Mehal, B. Izar, R. A. Friedman, R. G. Wells, U. B. Pajvani, Y. Hoshida, H. E. Remotti, N. Arpaia, J. Zucman-Rossi, M. Karin, N. C. Henderson, I. Tabas, and R. F. Schwabe
    Nature Oct 2022

    1 in established tumours. An increased HSC imbalance between cytokine-producing HSCs and myofibroblastic HSCs during liver disease progression was associated with increased HCC risk in patients. In summary, the dynamic shift in HSC subpopulations and their mediators during chronic liver disease is associated with a switch from HCC protection to HCC promotion.

    @article{pmid36198802,
      author = {Filliol, A. and Saito, Y. and Nair, A. and Dapito, D. H. and Yu, L. X. and Ravichandra, A. and Bhattacharjee, S. and Affo, S. and Fujiwara, N. and Su, H. and Sun, Q. and Savage, T. M. and Wilson-Kanamori, J. R. and Caviglia, J. M. and Chin, L. and Chen, D. and Wang, X. and Caruso, S. and Kang, J. K. and Amin, A. D. and Wallace, S. and Dobie, R. and Yin, D. and Rodriguez-Fiallos, O. M. and Yin, C. and Mehal, A. and Izar, B. and Friedman, R. A. and Wells, R. G. and Pajvani, U. B. and Hoshida, Y. and Remotti, H. E. and Arpaia, N. and Zucman-Rossi, J. and Karin, M. and Henderson, N. C. and Tabas, I. and Schwabe, R. F.},
      title = {{{O}pposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis}},
      journal = {Nature},
      year = {2022},
      volume = {610},
      number = {7931},
      pages = {356--365},
      month = oct
    }
  4. R. S. Taylor, R. Ruiz Daniels, R. Dobie, S. Naseer, T. C. Clark, N. C. Henderson, P. Boudinot, S. A. M. Martin, and D. J. Macqueen
    Front Immunol Oct 2022

    T cells, and candidate populations of monocytes/macrophages and dendritic cells. A dominant hepatocyte population radically remodeled its transcriptome following infection to activate the acute phase response and other defense functions, while repressing routine functions such as metabolism. These defense-specialized hepatocytes showed strong activation of genes controlling protein synthesis and secretion, presumably to support the release of acute phase proteins into circulation. The infection response further involved up-regulation of numerous genes in an immune-cell specific manner, reflecting functions in pathogen recognition and killing, antigen presentation, phagocytosis, regulation of inflammation, B cell differentiation and T cell activation. Overall, this study greatly enhances our understanding of the multifaceted role played by liver immune and non-immune cells in host defense and metabolic remodeling following infection and provides many novel cell-specific marker genes to empower future studies of this organ in fishes.

    @article{pmid36091005,
      author = {Taylor, R. S. and Ruiz Daniels, R. and Dobie, R. and Naseer, S. and Clark, T. C. and Henderson, N. C. and Boudinot, P. and Martin, S. A. M. and Macqueen, D. J.},
      title = {{}},
      journal = {Front Immunol},
      year = {2022},
      volume = {13},
      pages = {984799}
    }
  5. Pax6 limits the competence of developing cerebral cortical cells to respond to inductive intercellular signals
    M. Manuel, K. B. Tan, Z. Kozic, M. Molinek, T. S. Marcos, M. F. A. Razak, D. Dobolyi, R. Dobie, B. E. P. Henderson, N. C. Henderson, W. K. Chan, M. I. Daw, J. O. Mason, and D. J. Price
    PLoS Biol Sep 2022

    The development of stable specialized cell types in multicellular organisms relies on mechanisms controlling inductive intercellular signals and the competence of cells to respond to such signals. In developing cerebral cortex, progenitors generate only glutamatergic excitatory neurons despite being exposed to signals with the potential to initiate the production of other neuronal types, suggesting that their competence is limited. Here, we tested the hypothesis that this limitation is due to their expression of transcription factor Pax6. We used bulk and single-cell RNAseq to show that conditional cortex-specific Pax6 deletion from the onset of cortical neurogenesis allowed some progenitors to generate abnormal lineages resembling those normally found outside the cortex. Analysis of selected gene expression showed that the changes occurred in specific spatiotemporal patterns. We then compared the responses of control and Pax6-deleted cortical cells to in vivo and in vitro manipulations of extracellular signals. We found that Pax6 loss increased cortical progenitors’ competence to generate inappropriate lineages in response to extracellular factors normally present in developing cortex, including the morphogens Shh and Bmp4. Regional variation in the levels of these factors could explain spatiotemporal patterns of fate change following Pax6 deletion in vivo. We propose that Pax6’s main role in developing cortical cells is to minimize the risk of their development being derailed by the potential side effects of morphogens engaged contemporaneously in other essential functions.

    @article{pmid36067211,
      author = {Manuel, M. and Tan, K. B. and Kozic, Z. and Molinek, M. and Marcos, T. S. and Razak, M. F. A. and Dobolyi, D. and Dobie, R. and Henderson, B. E. P. and Henderson, N. C. and Chan, W. K. and Daw, M. I. and Mason, J. O. and Price, D. J.},
      title = {{{P}ax6 limits the competence of developing cerebral cortical cells to respond to inductive intercellular signals}},
      journal = {PLoS Biol},
      year = {2022},
      volume = {20},
      number = {9},
      pages = {e3001563},
      month = sep
    }
  6. Cells play a dual role as hematopoietic precursors and niche cells during mouse ontogeny
    D. Bandeira, A. M. Kilpatrick, M. Marques, M. Gomez-Salazar, T. Ventura, Z. N. Gonzalez, D. Stefancova, F. Rossi, M. Vermeren, C. S. Vink, M. Beltran, N. C. Henderson, B. Jung, R. Linden, H. J. G. Werken, W. F. J. Ijcken, C. Betsholtz, S. J. Forbes, H. Cuervo, and M. Crisan
    Cell Rep Jul 2022

    Embryonic precursors give rise to a subset of HSPCs that persist into adulthood. These findings provide crucial information for the controlled production of HSPCs in vitro.

    @article{pmid35858557,
      author = {Sá da Bandeira, D. and Kilpatrick, A. M. and Marques, M. and Gomez-Salazar, M. and Ventura, T. and Gonzalez, Z. N. and Stefancova, D. and Rossi, F. and Vermeren, M. and Vink, C. S. and Beltran, M. and Henderson, N. C. and Jung, B. and van der Linden, R. and van de Werken, H. J. G. and van Ijcken, W. F. J. and Betsholtz, C. and Forbes, S. J. and Cuervo, H. and Crisan, M.},
      title = {{Cells play a dual role as hematopoietic precursors and niche cells during mouse ontogeny}},
      journal = {Cell Rep},
      year = {2022},
      volume = {40},
      number = {3},
      pages = {111114},
      month = jul
    }
  7. Understanding the cellular interactome of non-alcoholic fatty liver disease
    S. J. Wallace, F. Tacke, R. F. Schwabe, and N. C. Henderson
    JHEP Rep Aug 2022

    Non-alcoholic fatty liver disease (NAFLD) is reaching epidemic proportions, with a global prevalence of 25% in the adult population. Non-alcoholic steatohepatitis (NASH), which can lead to cirrhosis, has become the leading indication for liver transplantation in both Europe and the USA. Liver fibrosis is the consequence of sustained, iterative liver injury, and the main determinant of outcomes in NASH. The liver possesses remarkable inherent plasticity, and liver fibrosis can regress when the injurious agent is removed, thus providing opportunities to alter long-term outcomes through therapeutic interventions. Although hepatocyte injury is a key driver of NASH, multiple other cell lineages within the hepatic fibrotic niche play major roles in the perpetuation of inflammation, mesenchymal cell activation, extracellular matrix accumulation as well as fibrosis resolution. The constituents of this cellular interactome, and how the various subpopulations within the fibrotic niche interact to drive fibrogenesis is an area of active research. Important cellular components of the fibrotic niche include endothelial cells, macrophages, passaging immune cell populations and myofibroblasts. In this review, we will describe how rapidly evolving technologies such as single-cell genomics, spatial transcriptomics and single-cell ligand-receptor analyses are transforming our understanding of the cellular interactome in NAFLD/NASH, and how this new, high-resolution information is being leveraged to develop rational new therapies for patients with NASH.

    @article{pmid35845296,
      author = {Wallace, S. J. and Tacke, F. and Schwabe, R. F. and Henderson, N. C.},
      title = {{{U}nderstanding the cellular interactome of non-alcoholic fatty liver disease}},
      journal = {JHEP Rep},
      year = {2022},
      volume = {4},
      number = {8},
      pages = {100524},
      month = aug
    }
  8. The purinergic P2Y14 receptor links hepatocyte death to hepatic stellate cell activation and fibrogenesis in the liver
    I. Mederacke, A. Filliol, S. Affo, A. Nair, C. Hernandez, Q. Sun, F. Hamberger, F. Brundu, Y. Chen, A. Ravichandra, P. Huebener, H. Anke, H. Shi, R. A. Torre, J. R. Smith, N. C. Henderson, F. W. R. Vondran, C. V. Rothlin, H. Baehre, I. Tabas, P. Sancho-Bru, and R. F. Schwabe
    Sci Transl Med Apr 2022

    Fibrosis contributes to  45% of deaths in western countries. In chronic liver disease, fibrosis is a major factor determining outcomes, but efficient antifibrotic therapies are lacking. Although platelet-derived growth factor and transforming growth factor-β constitute key fibrogenic mediators, they do not account for the well-established link between cell death and fibrosis in the liver. Here, we hypothesized that damage-associated molecular patterns (DAMPs) may link epithelial cell death to fibrogenesis in the injured liver. DAMP receptor screening identified purinergic receptor P2Y14 among several candidates as highly enriched in hepatic stellate cells (HSCs), the main fibrogenic cell type of the liver. Conversely, P2Y14 ligands uridine 5’-diphosphate (UDP)-glucose and UDP-galactose were enriched in hepatocytes and were released upon different modes of cell death. Accordingly, ligand-receptor interaction analysis that combined proteomic and single-cell RNA sequencing data revealed P2Y14 ligands and P2Y14 receptor as a link between dying cells and HSCs, respectively. Treatment with P2Y14 ligands or coculture with dying hepatocytes promoted HSC activation in a P2Y14-dependent manner. P2Y14 ligands activated extracellular signal-regulated kinase (ERK) and Yes-associated protein (YAP) signaling in HSCs, resulting in ERK-dependent HSC activation. Global and HSC-selective P2Y14 deficiency attenuated liver fibrosis in multiple mouse models of liver injury. Functional expression of P2Y14 was confirmed in healthy and diseased human liver and human HSCs. In conclusion, P2Y14 ligands and their receptor constitute a profibrogenic DAMP pathway that directly links cell death to fibrogenesis.

    @article{pmid35385339,
      author = {Mederacke, I. and Filliol, A. and Affo, S. and Nair, A. and Hernandez, C. and Sun, Q. and Hamberger, F. and Brundu, F. and Chen, Y. and Ravichandra, A. and Huebener, P. and Anke, H. and Shi, H. and Martínez García de la Torre, R. A. and Smith, J. R. and Henderson, N. C. and Vondran, F. W. R. and Rothlin, C. V. and Baehre, H. and Tabas, I. and Sancho-Bru, P. and Schwabe, R. F.},
      title = {{{T}he purinergic {P}2{Y}14 receptor links hepatocyte death to hepatic stellate cell activation and fibrogenesis in the liver}},
      journal = {Sci Transl Med},
      year = {2022},
      volume = {14},
      number = {639},
      pages = {eabe5795},
      month = apr
    }
  9. Mapping the developing human cardiac endothelium at single cell resolution identifies MECOM as a regulator of arteriovenous gene expression
    I. R. McCracken, R. Dobie, M. Bennett, R. Passi, A. Beqqali, N. C. Henderson, J. C. Mountford, P. R. Riley, C. P. Ponting, N. Smart, M. Brittan, and A. H. Baker
    Cardiovasc Res Feb 2022

    Coronary vasculature formation is a critical event during cardiac development, essential for heart function throughout perinatal and adult life. However, current understanding of coronary vascular development has largely been derived from transgenic mouse models. The aim of this study was to characterise the transcriptome of the human fetal cardiac endothelium using single-cell RNA sequencing (scRNA-seq) to provide critical new insights into the cellular heterogeneity and transcriptional dynamics that underpin endothelial specification within the vasculature of the developing heart.\{We acquired scRNA-seq data of over 10,000 fetal cardiac endothelial cells (EC), revealing divergent EC subtypes including endocardial, capillary, venous, arterial, and lymphatic populations. Gene regulatory network analyses predicted roles for SMAD1 and MECOM in determining the identity of capillary and arterial populations, respectively. Trajectory inference analysis suggested an endocardial contribution to the coronary vasculature and subsequent arterialisation of capillary endothelium accompanied by increasing MECOM expression. Comparative analysis of equivalent data from murine cardiac development demonstrated that transcriptional signatures defining endothelial subpopulations are largely conserved between human and mouse. Comprehensive characterisation of the transcriptional response to MECOM knockdown in human embryonic stem cell-derived EC (hESC-EC) demonstrated an increase in the expression of non-arterial markers, including those enriched in venous EC.\{scRNA-seq of the human fetal cardiac endothelium identified distinct EC populations. A predicted endocardial contribution to the developing coronary vasculature was identified, as well as subsequent arterial specification of capillary EC. Loss of MECOM in hESC-EC increased expression of non-arterial markers, suggesting a role in maintaining arterial EC identity.\{Endogenous blood vessel formation in the adult heart following myocardial infarction is insufficient to support adequate survival of the remaining myocardium, often ultimately leading to heart failure. Improved understanding of the mechanisms regulating human coronary vessel formation is required to inform therapeutic strategies to reactivate developmental pathways promoting therapeutic angiogenesis in patients. We applied scRNA-seq to map the transcriptome of the endothelium of the developing human heart. We identified novel transcriptional signatures underlying the cellular heterogeneity and dynamic changes occurring within the developing cardiac endothelium. This included identifying and validating MECOM as a novel regulator of arterial EC identity which may serve as a target for therapeutic neovascularization.

    @article{pmid35212715,
      author = {McCracken, I. R. and Dobie, R. and Bennett, M. and Passi, R. and Beqqali, A. and Henderson, N. C. and Mountford, J. C. and Riley, P. R. and Ponting, C. P. and Smart, N. and Brittan, M. and Baker, A. H.},
      title = {{{M}apping the developing human cardiac endothelium at single cell resolution identifies {M}{E}{C}{O}{M} as a regulator of arteriovenous gene expression}},
      journal = {Cardiovasc Res},
      year = {2022},
      month = feb
    }
  10. Liver zonation, revisited
    J. Paris, and N. C. Henderson
    Hepatology Feb 2022

    The concept of hepatocyte functional zonation is well established, with differences in metabolism and xenobiotic processing determined by multiple factors including oxygen and nutrient levels across the hepatic lobule. However, recent advances in single-cell genomics technologies, including single-cell and nuclei RNA sequencing, and the rapidly evolving fields of spatial transcriptomic and proteomic profiling have greatly increased our understanding of liver zonation. Here we discuss how these transformative experimental strategies are being leveraged to dissect liver zonation at unprecedented resolution and how this new information should facilitate the emergence of novel precision medicine-based therapies for patients with liver disease.

    @article{pmid35175659,
      author = {Paris, J. and Henderson, N. C.},
      title = {{{L}iver zonation, revisited}},
      journal = {Hepatology},
      year = {2022},
      month = feb
    }
  11. Genome-wide analysis identifies gallstone-susceptibility loci including genes regulating gastrointestinal motility
    C. J. Fairfield, T. M. Drake, R. Pius, A. D. Bretherick, A. Campbell, D. W. Clark, J. A. Fallowfield, C. Hayward, N. C. Henderson, A. Iakovliev, P. K. Joshi, N. L. Mills, D. J. Porteous, P. Ramachandran, R. K. Semple, C. A. Shaw, C. L. W. Sudlow, P. R. H. J. Timmers, J. F. Wilson, S. J. Wigmore, A. Spiliopoulou, and E. M. Harrison
    Hepatology May 2022

    Genome-wide association studies (GWAS) have identified several risk loci for gallstone disease. As with most polygenic traits, it is likely that many genetic determinants are undiscovered. The aim of this study was to identify genetic variants that represent new targets for gallstone research and treatment.\ ), of which 46 were new. Pathway enrichment revealed associations with lipid homeostasis, glucuronidation, phospholipid metabolism, and gastrointestinal motility. Anoctamin 1 (ANO1) and transmembrane Protein 147 (TMEM147), both in novel, replicated loci, are expressed in the gallbladder and gastrointestinal tract. Both regulate gastrointestinal motility. The gallstone risk allele rs7599-A leads to suppression of hepatic TMEM147 expression, suggesting that the protein protects against gallstone formation. The highest decile of the PRS demonstrated a 6-fold increased odds of gallstones compared with the lowest decile. The PRS was strongly associated with increased body mass index, serum liver enzymes, and C-reactive protein concentrations, and decreased lipoprotein cholesterol concentrations.\{This GWAS demonstrates the polygenic nature of gallstone risk and identifies 46 novel susceptibility loci. We implicate genes influencing gastrointestinal motility in the pathogenesis of gallstones.

    @article{pmid34651315,
      author = {Fairfield, C. J. and Drake, T. M. and Pius, R. and Bretherick, A. D. and Campbell, A. and Clark, D. W. and Fallowfield, J. A. and Hayward, C. and Henderson, N. C. and Iakovliev, A. and Joshi, P. K. and Mills, N. L. and Porteous, D. J. and Ramachandran, P. and Semple, R. K. and Shaw, C. A. and Sudlow, C. L. W. and Timmers, P. R. H. J. and Wilson, J. F. and Wigmore, S. J. and Spiliopoulou, A. and Harrison, E. M.},
      title = {{{G}enome-wide analysis identifies gallstone-susceptibility loci including genes regulating gastrointestinal motility}},
      journal = {Hepatology},
      year = {2022},
      volume = {75},
      number = {5},
      pages = {1081--1094},
      month = may
    }
  12. Differential abundance testing on single-cell data using k-nearest neighbor graphs
    E. Dann, N. C. Henderson, S. A. Teichmann, M. D. Morgan, and J. C. Marioni
    Nat Biotechnol Feb 2022

    Current computational workflows for comparative analyses of single-cell datasets typically use discrete clusters as input when testing for differential abundance among experimental conditions. However, clusters do not always provide the appropriate resolution and cannot capture continuous trajectories. Here we present Milo, a scalable statistical framework that performs differential abundance testing by assigning cells to partially overlapping neighborhoods on a k-nearest neighbor graph. Using simulations and single-cell RNA sequencing (scRNA-seq) data, we show that Milo can identify perturbations that are obscured by discretizing cells into clusters, that it maintains false discovery rate control across batch effects and that it outperforms alternative differential abundance testing strategies. Milo identifies the decline of a fate-biased epithelial precursor in the aging mouse thymus and identifies perturbations to multiple lineages in human cirrhotic liver. As Milo is based on a cell-cell similarity structure, it might also be applicable to single-cell data other than scRNA-seq. Milo is provided as an open-source R software package at https://github.com/MarioniLab/miloR .

    @article{pmid34594043,
      author = {Dann, E. and Henderson, N. C. and Teichmann, S. A. and Morgan, M. D. and Marioni, J. C.},
      title = {{{D}ifferential abundance testing on single-cell data using k-nearest neighbor graphs}},
      journal = {Nat Biotechnol},
      year = {2022},
      volume = {40},
      number = {2},
      pages = {245--253},
      month = feb
    }
  13. Genome-Wide Association Study of NAFLD Using Electronic Health Records
    C. J. Fairfield, T. M. Drake, R. Pius, A. D. Bretherick, A. Campbell, D. W. Clark, J. A. Fallowfield, C. Hayward, N. C. Henderson, P. K. Joshi, N. L. Mills, D. J. Porteous, P. Ramachandran, R. K. Semple, C. A. Shaw, C. L. M. Sudlow, P. R. H. J. Timmers, J. F. Wilson, S. J. Wigmore, E. M. Harrison, and A. Spiliopoulou
    Hepatol Commun Feb 2022

    ) is a missense variant within the APOE gene determining ϵ4 versus ϵ2/ϵ3 alleles. The ϵ4 allele of APOE offered protection against NAFLD (odds ratio for heterozygotes 0.84 [95% confidence interval 0.78-0.90] and homozygotes 0.64 [0.50-0.79]). Conclusion: This GWAS replicates six known NAFLD-susceptibility loci and confirms that the ϵ4 allele of APOE is associated with protection against NAFLD. The results are consistent with published GWAS using histological and radiological measures of NAFLD, confirming that NAFLD identified through diagnostic codes from consensus guidelines is a valid alternative to more invasive and costly approaches.

    @article{pmid34535985,
      author = {Fairfield, C. J. and Drake, T. M. and Pius, R. and Bretherick, A. D. and Campbell, A. and Clark, D. W. and Fallowfield, J. A. and Hayward, C. and Henderson, N. C. and Joshi, P. K. and Mills, N. L. and Porteous, D. J. and Ramachandran, P. and Semple, R. K. and Shaw, C. A. and Sudlow, C. L. M. and Timmers, P. R. H. J. and Wilson, J. F. and Wigmore, S. J. and Harrison, E. M. and Spiliopoulou, A.},
      title = {{{G}enome-{W}ide {A}ssociation {S}tudy of {N}{A}{F}{L}{D} {U}sing {E}lectronic {H}ealth {R}ecords}},
      journal = {Hepatol Commun},
      year = {2022},
      volume = {6},
      number = {2},
      pages = {297--308},
      month = feb
    }
  14. Single-cell RNA sequencing profiling of mouse endothelial cells in response to pulmonary arterial hypertension
    J. Rodor, S. H. Chen, J. P. Scanlon, J. P. Monteiro, A. Caudrillier, S. Sweta, K. R. Stewart, A. Shmakova, R. Dobie, B. E. P. Henderson, K. Stewart, P. W. F. Hadoke, M. Southwood, S. D. Moore, P. D. Upton, N. W. Morrell, Z. Li, S. Y. Chan, A. Handen, R. Lafyatis, L. P. M. H. Rooij, N. C. Henderson, P. Carmeliet, A. M. Spiroski, M. Brittan, and A. H. Baker
    Cardiovasc Res Aug 2022

    Endothelial cell (EC) dysfunction drives the initiation and pathogenesis of pulmonary arterial hypertension (PAH). We aimed to characterize EC dynamics in PAH at single-cell resolution.\{We carried out single-cell RNA sequencing (scRNA-seq) of lung ECs isolated from an EC lineage-tracing mouse model in Control and SU5416/hypoxia-induced PAH conditions. EC populations corresponding to distinct lung vessel types, including two discrete capillary populations, were identified in both Control and PAH mice. Differential gene expression analysis revealed global PAH-induced EC changes that were confirmed by bulk RNA-seq. This included upregulation of the major histocompatibility complex class II pathway, supporting a role for ECs in the inflammatory response in PAH. We also identified a PAH response specific to the second capillary EC population including upregulation of genes involved in cell death, cell motility, and angiogenesis. Interestingly, four genes with genetic variants associated with PAH were dysregulated in mouse ECs in PAH. To compare relevance across PAH models and species, we performed a detailed analysis of EC heterogeneity and response to PAH in rats and humans through whole-lung PAH scRNA-seq datasets, revealing that 51% of up-regulated mouse genes were also up-regulated in rat or human PAH. We identified promising new candidates to target endothelial dysfunction including CD74, the knockdown of which regulates EC proliferation and barrier integrity in vitro. Finally, with an in silico cell ordering approach, we identified zonation-dependent changes across the arteriovenous axis in mouse PAH and showed upregulation of the Serine/threonine-protein kinase Sgk1 at the junction between the macro- and microvasculature.\{This study uncovers PAH-induced EC transcriptomic changes at a high resolution, revealing novel targets for potential therapeutic candidate development.

    @article{pmid34528097,
      author = {Rodor, J. and Chen, S. H. and Scanlon, J. P. and Monteiro, J. P. and Caudrillier, A. and Sweta, S. and Stewart, K. R. and Shmakova, A. and Dobie, R. and Henderson, B. E. P. and Stewart, K. and Hadoke, P. W. F. and Southwood, M. and Moore, S. D. and Upton, P. D. and Morrell, N. W. and Li, Z. and Chan, S. Y. and Handen, A. and Lafyatis, R. and de Rooij, L. P. M. H. and Henderson, N. C. and Carmeliet, P. and Spiroski, A. M. and Brittan, M. and Baker, A. H.},
      title = {{{S}ingle-cell {R}{N}{A} sequencing profiling of mouse endothelial cells in response to pulmonary arterial hypertension}},
      journal = {Cardiovasc Res},
      year = {2022},
      volume = {118},
      number = {11},
      pages = {2519--2534},
      month = aug
    }
  15. Deciphering Mesenchymal Drivers of Human Dupuytren’s Disease at Single-Cell Level
    R. Dobie, C. C. West, B. E. P. Henderson, J. R. Wilson-Kanamori, D. Markose, L. J. Kitto, J. R. Portman, M. Beltran, S. Sohrabi, A. R. Akram, P. Ramachandran, L. Y. Yong, D. Davidson, and N. C. Henderson
    J Invest Dermatol Jan 2022

    mesenchymal cells, demonstrating a functional role for these genes in DD. The power of single-cell RNA sequencing is utilized to identify the major pathogenic mesenchymal subpopulations driving DD and the key molecular pathways regulating the DD-specific myofibroblast phenotype. Using this precision medicine approach, inhibition of TNFRSF12A has shown potential clinical utility in the treatment of DD.

    @article{pmid34274346,
      author = {Dobie, R. and West, C. C. and Henderson, B. E. P. and Wilson-Kanamori, J. R. and Markose, D. and Kitto, L. J. and Portman, J. R. and Beltran, M. and Sohrabi, S. and Akram, A. R. and Ramachandran, P. and Yong, L. Y. and Davidson, D. and Henderson, N. C.},
      title = {{{D}eciphering {M}esenchymal {D}rivers of {H}uman {D}upuytren's {D}isease at {S}ingle-{C}ell {L}evel}},
      journal = {J Invest Dermatol},
      year = {2022},
      volume = {142},
      number = {1},
      pages = {114--123},
      month = jan
    }

2021

  1. Selective Myeloid Depletion of Galectin-3 Offers Protection Against Acute and Chronic Lung Injury
    D. C. Humphries, R. Mills, R. Dobie, N. C. Henderson, T. Sethi, and A. C. Mackinnon
    Front Pharmacol Jan 2021

    Myeloid cell derived Galectin-3 drives acute and chronic lung inflammation and supports direct targeting of galectin-3 as an attractive new therapy for lung inflammation.

    @article{pmid34526900,
      author = {Humphries, D. C. and Mills, R. and Dobie, R. and Henderson, N. C. and Sethi, T. and Mackinnon, A. C.},
      title = {{{S}elective {M}yeloid {D}epletion of {G}alectin-3 {O}ffers {P}rotection {A}gainst {A}cute and {C}hronic {L}ung {I}njury}},
      journal = {Front Pharmacol},
      year = {2021},
      volume = {12},
      pages = {715986}
    }
  2. Dynamic cell contacts between periportal mesenchyme and ductal epithelium act as a rheostat for liver cell proliferation
    L. Cordero-Espinoza, A. M. Dowbaj, T. N. Kohler, B. Strauss, O. Sarlidou, G. Belenguer, C. Pacini, N. P. Martins, R. Dobie, J. R. Wilson-Kanamori, R. Butler, N. Prior, P. Serup, F. Jug, N. C. Henderson, F. Hollfelder, and M. Huch
    Cell Stem Cell Nov 2021

    In the liver, ductal cells rarely proliferate during homeostasis but do so transiently after tissue injury. These cells can be expanded as organoids that recapitulate several of the cell-autonomous mechanisms of regeneration but lack the stromal interactions of the native tissue. Here, using organoid co-cultures that recapitulate the ductal-to-mesenchymal cell architecture of the portal tract, we demonstrate that a subpopulation of mouse periportal mesenchymal cells exerts dual control on proliferation of the epithelium. Ductal cell proliferation is either induced and sustained or, conversely, completely abolished, depending on the number of direct mesenchymal cell contacts, through a mechanism mediated, at least in part, by Notch signaling. Our findings expand the concept of the cellular niche in epithelial tissues, whereby not only soluble factors but also cell-cell contacts are the key regulatory cues involved in the control of cellular behaviors, suggesting a critical role for cell-cell contacts during regeneration.

    @article{pmid34343491,
      author = {Cordero-Espinoza, L. and Dowbaj, A. M. and Kohler, T. N. and Strauss, B. and Sarlidou, O. and Belenguer, G. and Pacini, C. and Martins, N. P. and Dobie, R. and Wilson-Kanamori, J. R. and Butler, R. and Prior, N. and Serup, P. and Jug, F. and Henderson, N. C. and Hollfelder, F. and Huch, M.},
      title = {{{D}ynamic cell contacts between periportal mesenchyme and ductal epithelium act as a rheostat for liver cell proliferation}},
      journal = {Cell Stem Cell},
      year = {2021},
      volume = {28},
      number = {11},
      pages = {1907--1921},
      month = nov
    }
  3. Loss of Adam10 Disrupts Ion Transport in Immortalized Kidney Collecting Duct Cells
    A. Assmus, L. Mullins, M. Ward, R. Dobie, R. Hunter, N. C. Henderson, and J. J. Mullins
    Function (Oxf) Nov 2021

    (11βHSD2), but no significant changes in transcription of components of the Notch pathway were observed. Immunostaining in the knockout clone confirmed the decrease in expression of γ-ENaC and importantly, showed an altered, diffuse distribution of PC and IC markers, suggesting altered trafficking in the Adam10 knockout clone as an explanation for the loss of polarization.

    @article{pmid34131651,
      author = {Assmus, A. and Mullins, L. and Ward, M. and Dobie, R. and Hunter, R. and Henderson, N. C. and Mullins, J. J.},
      title = {{{L}oss of {A}dam10 {D}isrupts {I}on {T}ransport in {I}mmortalized {K}idney {C}ollecting {D}uct {C}ells}},
      journal = {Function (Oxf)},
      year = {2021},
      volume = {2},
      number = {4},
      pages = {zqab024}
    }
  4. A unique macrophage subpopulation signals directly to progenitor cells to promote regenerative neurogenesis in the zebrafish spinal cord
    L. Cavone, T. McCann, L. K. Drake, E. A. Aguzzi, A. M. Oprişoreanu, E. Pedersen, S. Sandi, J. Selvarajah, T. M. Tsarouchas, D. Wehner, M. Keatinge, K. S. Mysiak, B. E. P. Henderson, R. Dobie, N. C. Henderson, T. Becker, and C. G. Becker
    Dev Cell Jun 2021

    Central nervous system injury re-initiates neurogenesis in anamniotes (amphibians and fishes), but not in mammals. Activation of the innate immune system promotes regenerative neurogenesis, but it is fundamentally unknown whether this is indirect through the activation of known developmental signaling pathways or whether immune cells directly signal to progenitor cells using mechanisms that are unique to regeneration. Using single-cell RNA-seq of progenitor cells and macrophages, as well as cell-type-specific manipulations, we provide evidence for a direct signaling axis from specific lesion-activated macrophages to spinal progenitor cells to promote regenerative neurogenesis in zebrafish. Mechanistically, TNFa from pro-regenerative macrophages induces Tnfrsf1a-mediated AP-1 activity in progenitors to increase regeneration-promoting expression of hdac1 and neurogenesis. This establishes the principle that macrophages directly communicate to spinal progenitor cells via non-developmental signals after injury, providing potential targets for future interventions in the regeneration-deficient spinal cord of mammals.

    @article{pmid34033756,
      author = {Cavone, L. and McCann, T. and Drake, L. K. and Aguzzi, E. A. and Oprişoreanu, A. M. and Pedersen, E. and Sandi, S. and Selvarajah, J. and Tsarouchas, T. M. and Wehner, D. and Keatinge, M. and Mysiak, K. S. and Henderson, B. E. P. and Dobie, R. and Henderson, N. C. and Becker, T. and Becker, C. G.},
      title = {{{A} unique macrophage subpopulation signals directly to progenitor cells to promote regenerative neurogenesis in the zebrafish spinal cord}},
      journal = {Dev Cell},
      year = {2021},
      volume = {56},
      number = {11},
      pages = {1617--1630},
      month = jun
    }
  5. scRNA Transcription Profile of Adult Zebrafish Podocytes Using a Novel Reporter Strain
    C. Brown, L. J. Mullins, K. Wesencraft, G. McConnell, M. Beltran, N. C. Henderson, B. Conway, S. Hoffmann, S. Rider, and J. J. Mullins
    Cell Physiol Biochem May 2021

    The role of podocytes is well conserved across species from drosophila to teleosts, and mammals. Identifying the molecular markers that actively maintain the integrity of the podocyte will enable a greater understanding of the changes that lead to damage.\{We generated transgenic zebrafish, expressing fluorescent reporters driven by the podocin promoter, for the visualization and isolation of podocytes. We have conducted single cell RNA sequencing (scRNA-seq) on isolated podocytes from a zebrafish reporter line.\{We demonstrated that the LifeAct-TagRFP-T fluorescent reporter faithfully replicated podocin expression in vivo. We were also able to show spontaneous GCaMP6s fluorescence using light sheet (single plane illumination) microscopy. We identified many podocyte transcripts, encoding proteins related to calcium-binding and actin filament assembly, in common with those expressed in human and mouse mature podocytes.\{We describe the establishment of novel transgenic zebrafish and their use to identify and isolate podocyte cells for the preparation of a scRNA-seq library from normal podocytes. The scRNA-seq data identifies distinct populations of cells and potential gene switching between clusters. These data provide a foundation for future comparative studies and for exploiting the zebrafish as a model for kidney development, disease, injury and repair.

    @article{pmid33945241,
      author = {Brown, C. and Mullins, L. J. and Wesencraft, K. and McConnell, G. and Beltran, M. and Henderson, N. C. and Conway, B. and Hoffmann, S. and Rider, S. and Mullins, J. J.},
      title = {{sc{R}{N}{A} {T}ranscription {P}rofile of {A}dult {Z}ebrafish {P}odocytes {U}sing a {N}ovel {R}eporter {S}train}},
      journal = {Cell Physiol Biochem},
      year = {2021},
      volume = {55},
      number = {S4},
      pages = {35--47},
      month = may
    }
  6. Single-cell RNA sequencing redefines the mesenchymal cell landscape of mouse endometrium
    P. M. Kirkwood, D. A. Gibson, J. R. Smith, J. R. Wilson-Kanamori, O. Kelepouri, A. Esnal-Zufiaurre, R. Dobie, N. C. Henderson, and P. T. K. Saunders
    FASEB J Apr 2021

    The endometrium is a dynamic tissue that exhibits remarkable resilience to repeated episodes of differentiation, breakdown, regeneration, and remodeling. Endometrial physiology relies on a complex interplay between the stromal and epithelial compartments with the former containing a mixture of fibroblasts, vascular, and immune cells. There is evidence for rare populations of putative mesenchymal progenitor cells located in the perivascular niche of human endometrium, but the existence of an equivalent cell population in mouse is unclear. We used the Pdgfrb-BAC-eGFP transgenic reporter mouse in combination with bulk and single-cell RNA sequencing to redefine the endometrial mesenchyme. In contrast to previous reports we show that CD146 is expressed in both PDGFRβ + perivascular cells and CD31 + endothelial cells. Bulk RNAseq revealed cells in the perivascular niche which express the high levels of Pdgfrb as well as genes previously identified in pericytes and/or vascular smooth muscle cells (Acta2, Myh11, Olfr78, Cspg4, Rgs4, Rgs5, Kcnj8, and Abcc9). scRNA-seq identified five subpopulations of cells including closely related pericytes/vascular smooth muscle cells and three subpopulations of fibroblasts. All three fibroblast populations were PDGFRα+/CD34 + but were distinct in their expression of Ngfr/Spon2/Angptl7 (F1), Cxcl14/Smoc2/Rgs2 (F2), and Clec3b/Col14a1/Mmp3 (F3), with potential functions in the regulation of immune responses, response to wounding, and organization of extracellular matrix, respectively. Immunohistochemistry was used to investigate the spatial distribution of these populations revealing F1/NGFR + cells in most abundance beside epithelial cells. We provide the first definitive analysis of mesenchymal cells in the adult mouse endometrium identifying five subpopulations providing a platform for comparisons between mesenchymal cells in endometrium and other adult tissues which are prone to fibrosis.

    @article{pmid33710643,
      author = {Kirkwood, P. M. and Gibson, D. A. and Smith, J. R. and Wilson-Kanamori, J. R. and Kelepouri, O. and Esnal-Zufiaurre, A. and Dobie, R. and Henderson, N. C. and Saunders, P. T. K.},
      title = {{{S}ingle-cell {R}{N}{A} sequencing redefines the mesenchymal cell landscape of mouse endometrium}},
      journal = {FASEB J},
      year = {2021},
      volume = {35},
      number = {4},
      pages = {e21285},
      month = apr
    }
  7. MIR503HG Loss Promotes Endothelial-to-Mesenchymal Transition in Vascular Disease
    J. P. Monteiro, J. Rodor, A. Caudrillier, J. P. Scanlon, A. M. Spiroski, T. Dudnakova, B. Pflüger-Müller, A. Shmakova, A. Kriegsheim, L. Deng, R. S. Taylor, J. R. Wilson-Kanamori, S. H. Chen, K. Stewart, A. Thomson, T. Mitić, J. D. McClure, J. Iynikkel, P. W. F. Hadoke, L. Denby, A. C. Bradshaw, P. Caruso, N. W. Morrell, J. C. Kovacic, I. Ulitsky, N. C. Henderson, A. Caporali, M. S. Leisegang, R. P. Brandes, and A. H. Baker
    Circ Res Apr 2021

    [Figure: see text].

    @article{pmid33703914,
      author = {Monteiro, J. P. and Rodor, J. and Caudrillier, A. and Scanlon, J. P. and Spiroski, A. M. and Dudnakova, T. and Pflüger-Müller, B. and Shmakova, A. and von Kriegsheim, A. and Deng, L. and Taylor, R. S. and Wilson-Kanamori, J. R. and Chen, S. H. and Stewart, K. and Thomson, A. and Mitić, T. and McClure, J. D. and Iynikkel, J. and Hadoke, P. W. F. and Denby, L. and Bradshaw, A. C. and Caruso, P. and Morrell, N. W. and Kovacic, J. C. and Ulitsky, I. and Henderson, N. C. and Caporali, A. and Leisegang, M. S. and Brandes, R. P. and Baker, A. H.},
      title = {{{M}{I}{R}503{H}{G} {L}oss {P}romotes {E}ndothelial-to-{M}esenchymal {T}ransition in {V}ascular {D}isease}},
      journal = {Circ Res},
      year = {2021},
      volume = {128},
      number = {8},
      pages = {1173--1190},
      month = apr
    }
  8. Hepatic Stellate Cell Regulation of Liver Regeneration and Repair
    L. J. Kitto, and N. C. Henderson
    Hepatol Commun Mar 2021

    The hepatic mesenchyme has been studied extensively in the context of liver fibrosis; however, much less is known regarding the role of mesenchymal cells during liver regeneration. As our knowledge of the cellular and molecular mechanisms driving hepatic regeneration deepens, the key role of the mesenchymal compartment during the regenerative response has been increasingly appreciated. Single-cell genomics approaches have recently uncovered both spatial and functional zonation of the hepatic mesenchyme in homeostasis and following liver injury. Here we discuss how the use of preclinical models, from in vivo mouse models to organoid-based systems, are helping to shape our understanding of the role of the mesenchyme during liver regeneration, and how these approaches should facilitate the precise identification of highly targeted, pro-regenerative therapies for patients with liver disease.

    @article{pmid33681672,
      author = {Kitto, L. J. and Henderson, N. C.},
      title = {{{H}epatic {S}tellate {C}ell {R}egulation of {L}iver {R}egeneration and {R}epair}},
      journal = {Hepatol Commun},
      year = {2021},
      volume = {5},
      number = {3},
      pages = {358--370},
      month = mar
    }
  9. SOX9 is required for kidney fibrosis and activates NAV3 to drive renal myofibroblast function
    S. Raza, E. Jokl, J. Pritchett, K. Martin, K. Su, K. Simpson, L. Birchall, A. F. Mullan, V. S. Athwal, D. T. Doherty, L. Zeef, N. C. Henderson, P. A. Kalra, N. A. Hanley, and K. Piper Hanley
    Sci Signal Mar 2021

    Renal fibrosis is a common end point for kidney injury and many chronic kidney diseases. Fibrogenesis depends on the sustained activation of myofibroblasts, which deposit the extracellular matrix that causes progressive scarring and organ failure. Here, we showed that the transcription factor SOX9 was associated with kidney fibrosis in humans and required for experimentally induced kidney fibrosis in mice. From genome-wide analysis, we identified Neuron navigator 3 (NAV3) as acting downstream of SOX9 in kidney fibrosis. NAV3 increased in abundance and colocalized with SOX9 after renal injury in mice, and both SOX9 and NAV3 were present in diseased human kidneys. In an in vitro model of renal pericyte transdifferentiation into myofibroblasts, we demonstrated that NAV3 was required for multiple aspects of fibrogenesis, including actin polymerization linked to cell migration and sustained activation of the mechanosensitive transcription factor YAP1. In summary, our work identifies a SOX9-NAV3-YAP1 axis involved in the progression of kidney fibrosis and points to NAV3 as a potential target for pharmacological intervention.

    @article{pmid33653921,
      author = {Raza, S. and Jokl, E. and Pritchett, J. and Martin, K. and Su, K. and Simpson, K. and Birchall, L. and Mullan, A. F. and Athwal, V. S. and Doherty, D. T. and Zeef, L. and Henderson, N. C. and Kalra, P. A. and Hanley, N. A. and Piper Hanley, K.},
      title = {{{S}{O}{X}9 is required for kidney fibrosis and activates {N}{A}{V}3 to drive renal myofibroblast function}},
      journal = {Sci Signal},
      year = {2021},
      volume = {14},
      number = {672},
      month = mar
    }
  10. Decoding myofibroblast origins in human kidney fibrosis
    C. Kuppe, M. M. Ibrahim, J. Kranz, X. Zhang, S. Ziegler, J. Perales-Patón, J. Jansen, K. C. Reimer, J. R. Smith, R. Dobie, J. R. Wilson-Kanamori, M. Halder, Y. Xu, N. Kabgani, N. Kaesler, M. Klaus, L. Gernhold, V. G. Puelles, T. B. Huber, P. Boor, S. Menzel, R. M. Hoogenboezem, E. M. J. Bindels, J. Steffens, J. Floege, R. K. Schneider, J. Saez-Rodriguez, N. C. Henderson, and R. Kramann
    Nature Jan 2021

    . Here, using single-cell RNA sequencing, we profiled the transcriptomes of cells from the proximal and non-proximal tubules of healthy and fibrotic human kidneys to map the entire human kidney. This analysis enabled us to map all matrix-producing cells at high resolution, and to identify distinct subpopulations of pericytes and fibroblasts as the main cellular sources of scar-forming myofibroblasts during human kidney fibrosis. We used genetic fate-tracing, time-course single-cell RNA sequencing and ATAC-seq (assay for transposase-accessible chromatin using sequencing) experiments in mice, and spatial transcriptomics in human kidney fibrosis, to shed light on the cellular origins and differentiation of human kidney myofibroblasts and their precursors at high resolution. Finally, we used this strategy to detect potential therapeutic targets, and identified NKD2 as a myofibroblast-specific target in human kidney fibrosis.

    @article{pmid33176333,
      author = {Kuppe, C. and Ibrahim, M. M. and Kranz, J. and Zhang, X. and Ziegler, S. and Perales-Patón, J. and Jansen, J. and Reimer, K. C. and Smith, J. R. and Dobie, R. and Wilson-Kanamori, J. R. and Halder, M. and Xu, Y. and Kabgani, N. and Kaesler, N. and Klaus, M. and Gernhold, L. and Puelles, V. G. and Huber, T. B. and Boor, P. and Menzel, S. and Hoogenboezem, R. M. and Bindels, E. M. J. and Steffens, J. and Floege, J. and Schneider, R. K. and Saez-Rodriguez, J. and Henderson, N. C. and Kramann, R.},
      title = {{{D}ecoding myofibroblast origins in human kidney fibrosis}},
      journal = {Nature},
      year = {2021},
      volume = {589},
      number = {7841},
      pages = {281--286},
      month = jan
    }

2020

  1. Monocytes as key regulators of human monocyte transcriptional response to Toxoplasma
    A. Patir, A. Gossner, P. Ramachandran, J. Alves, T. C. Freeman, N. C. Henderson, M. Watson, and M. A. Hassan
    Sci Rep Dec 2020

    Functional cytokine assays on sorted monocyte populations show that the infection-distinguishing monocytes secrete high levels of chemokines, such as CCL2 and CXCL5. These findings uncover the Toxoplasma-induced monocyte transcriptional heterogeneity and shed new light on the cell populations that largely define cytokine and chemokine secretion in human monocytes exposed to Toxoplasma.

    @article{pmid33273621,
      author = {Patir, A. and Gossner, A. and Ramachandran, P. and Alves, J. and Freeman, T. C. and Henderson, N. C. and Watson, M. and Hassan, M. A.},
      title = {{Monocytes as key regulators of human monocyte transcriptional response to {T}oxoplasma}},
      journal = {Sci Rep},
      year = {2020},
      volume = {10},
      number = {1},
      pages = {21047},
      month = dec
    }
  2. Transfer of hepatocellular microRNA regulates cytochrome P450 2E1 in renal tubular cells
    O. Matthews, E. E. Morrison, J. D. Tranter, P. Starkey Lewis, I. S. Toor, A. Srivastava, R. Sargeant, H. Rollison, K. P. Matchett, T. J. Kendall, G. A. Gray, C. Goldring, K. Park, L. Denby, N. Dhaun, M. A. Bailey, N. C. Henderson, D. Williams, and J. W. Dear
    EBioMedicine Dec 2020

    Extracellular microRNAs enter kidney cells and modify gene expression. We used a Dicer-hepatocyte-specific microRNA conditional-knock-out (Dicer-CKO) mouse to investigate microRNA transfer from liver to kidney.\{mice were treated with a Cre recombinase-expressing adenovirus (AAV8) to selectively inhibit hepatocyte microRNA production (Dicer-CKO). Organ microRNA expression was measured in health and following paracetamol toxicity. The functional consequence of hepatic microRNA transfer was determined by measuring the expression and activity of cytochrome P450 2E1 (target of the hepatocellular miR-122), and by measuring the effect of serum extracellular vesicles (ECVs) on proximal tubular cell injury. In humans with liver injury we measured microRNA expression in urinary ECVs. A murine model of myocardial infarction was used as a non-hepatic model of microRNA release.\{Dicer-CKO mice demonstrated a decrease in kidney miR-122 in the absence of other microRNA changes. During hepatotoxicity, miR-122 increased in kidney tubular cells; this was abolished in Dicer-CKO mice. Depletion of hepatocyte microRNA increased kidney cytochrome P450 2E1 expression and activity. Serum ECVs from mice with hepatotoxicity increased proximal tubular cell miR-122 and prevented cisplatin toxicity. miR-122 increased in urinary ECVs during human hepatotoxicity. Transfer of microRNA was not restricted to liver injury -miR-499 was released following cardiac injury and correlated with an increase in the kidney.\{Physiological transfer of functional microRNA to the kidney is increased by liver injury and this signalling represents a new paradigm for understanding the relationship between liver injury and renal function.\{Kidney Research UK, Medical Research Scotland, Medical Research Council.

    @article{pmid33232872,
      author = {Matthews, O. and Morrison, E. E. and Tranter, J. D. and Starkey Lewis, P. and Toor, I. S. and Srivastava, A. and Sargeant, R. and Rollison, H. and Matchett, K. P. and Kendall, T. J. and Gray, G. A. and Goldring, C. and Park, K. and Denby, L. and Dhaun, N. and Bailey, M. A. and Henderson, N. C. and Williams, D. and Dear, J. W.},
      title = {{{T}ransfer of hepatocellular micro{R}{N}{A} regulates cytochrome {P}450 2{E}1 in renal tubular cells}},
      journal = {EBioMedicine},
      year = {2020},
      volume = {62},
      pages = {103092},
      month = dec
    }
  3. Kidney Single-Cell Atlas Reveals Myeloid Heterogeneity in Progression and Regression of Kidney Disease
    B. R. Conway, E. D. O’Sullivan, C. Cairns, J. O’Sullivan, D. J. Simpson, A. Salzano, K. Connor, P. Ding, D. Humphries, K. Stewart, O. Teenan, R. Pius, N. C. Henderson, C. Bénézech, P. Ramachandran, D. Ferenbach, J. Hughes, T. Chandra, and L. Denby
    J Am Soc Nephrol Dec 2020

    Little is known about the roles of myeloid cell subsets in kidney injury and in the limited ability of the organ to repair itself. Characterizing these cells based only on surface markers using flow cytometry might not provide a full phenotypic picture. Defining these cells at the single-cell, transcriptomic level could reveal myeloid heterogeneity in the progression and regression of kidney disease.\{Integrated droplet- and plate-based single-cell RNA sequencing were used in the murine, reversible, unilateral ureteric obstruction model to dissect the transcriptomic landscape at the single-cell level during renal injury and the resolution of fibrosis. Paired blood exchange tracked the fate of monocytes recruited to the injured kidney.\{macrophage subset acts during repair.\{Complementary technologies identified novel myeloid subtypes, based on transcriptomics in single cells, that represent therapeutic targets to inhibit progression or promote regression of kidney disease.

    @article{pmid32978267,
      author = {Conway, B. R. and O'Sullivan, E. D. and Cairns, C. and O'Sullivan, J. and Simpson, D. J. and Salzano, A. and Connor, K. and Ding, P. and Humphries, D. and Stewart, K. and Teenan, O. and Pius, R. and Henderson, N. C. and Bénézech, C. and Ramachandran, P. and Ferenbach, D. and Hughes, J. and Chandra, T. and Denby, L.},
      title = {{{K}idney {S}ingle-{C}ell {A}tlas {R}eveals {M}yeloid {H}eterogeneity in {P}rogression and {R}egression of {K}idney {D}isease}},
      journal = {J Am Soc Nephrol},
      year = {2020},
      volume = {31},
      number = {12},
      pages = {2833--2854},
      month = dec
    }
  4. Single-cell analyses and machine learning define hematopoietic progenitor and HSC-like cells derived from human PSCs
    A. Fidanza, P. S. Stumpf, P. Ramachandran, S. Tamagno, A. Babtie, M. Lopez-Yrigoyen, A. H. Taylor, J. Easterbrook, B. E. P. Henderson, R. Axton, N. C. Henderson, A. Medvinsky, K. Ottersbach, N. Romanò, and L. M. Forrester
    Blood Dec 2020

    Hematopoietic stem and progenitor cells (HSPCs) develop in distinct waves at various anatomical sites during embryonic development. The in vitro differentiation of human pluripotent stem cells (hPSCs) recapitulates some of these processes; however, it has proven difficult to generate functional hematopoietic stem cells (HSCs). To define the dynamics and heterogeneity of HSPCs that can be generated in vitro from hPSCs, we explored single-cell RNA sequencing (scRNAseq) in combination with single-cell protein expression analysis. Bioinformatics analyses and functional validation defined the transcriptomes of naïve progenitors and erythroid-, megakaryocyte-, and leukocyte-committed progenitors, and we identified CD44, CD326, ICAM2/CD9, and CD18, respectively, as markers of these progenitors. Using an artificial neural network that we trained on scRNAseq derived from human fetal liver, we identified a wide range of hPSC-derived HSPCs phenotypes, including a small group classified as HSCs. This transient HSC-like population decreased as differentiation proceeded, and was completely missing in the data set that had been generated using cells selected on the basis of CD43 expression. By comparing the single-cell transcriptome of in vitro-generated HSC-like cells with those generated within the fetal liver, we identified transcription factors and molecular pathways that can be explored in the future to improve the in vitro production of HSCs.

    @article{pmid32614947,
      author = {Fidanza, A. and Stumpf, P. S. and Ramachandran, P. and Tamagno, S. and Babtie, A. and Lopez-Yrigoyen, M. and Taylor, A. H. and Easterbrook, J. and Henderson, B. E. P. and Axton, R. and Henderson, N. C. and Medvinsky, A. and Ottersbach, K. and Romanò, N. and Forrester, L. M.},
      title = {{{S}ingle-cell analyses and machine learning define hematopoietic progenitor and {H}{S}{C}-like cells derived from human {P}{S}{C}s}},
      journal = {Blood},
      year = {2020},
      volume = {136},
      number = {25},
      pages = {2893--2904},
      month = dec
    }
  5. Single-cell genomics and spatial transcriptomics: Discovery of novel cell states and cellular interactions in liver physiology and disease biology
    A. Saviano, N. C. Henderson, and T. F. Baumert
    J Hepatol Nov 2020

    Transcriptome analysis enables the study of gene expression in human tissues and is a valuable tool to characterise liver function and gene expression dynamics during liver disease, as well as to identify prognostic markers or signatures, and to facilitate discovery of new therapeutic targets. In contrast to whole tissue RNA sequencing analysis, single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics enables the study of transcriptional activity at the single cell or spatial level. ScRNA-seq has paved the way for the discovery of previously unknown cell types and subtypes in normal and diseased liver, facilitating the study of rare cells (such as liver progenitor cells) and the functional roles of non-parenchymal cells in chronic liver disease and cancer. By adding spatial information to scRNA-seq data, spatial transcriptomics has transformed our understanding of tissue functional organisation and cell-to-cell interactions in situ. These approaches have recently been applied to investigate liver regeneration, organisation and function of hepatocytes and non-parenchymal cells, and to profile the single cell landscape of chronic liver diseases and cancer. Herein, we review the principles and technologies behind scRNA-seq and spatial transcriptomic approaches, highlighting the recent discoveries and novel insights these methodologies have yielded in both liver physiology and disease biology.

    @article{pmid32534107,
      author = {Saviano, A. and Henderson, N. C. and Baumert, T. F.},
      title = {{{S}ingle-cell genomics and spatial transcriptomics: {D}iscovery of novel cell states and cellular interactions in liver physiology and disease biology}},
      journal = {J Hepatol},
      year = {2020},
      volume = {73},
      number = {5},
      pages = {1219--1230},
      month = nov
    }
  6. Pericyte FAK negatively regulates Gas6/Axl signalling to suppress tumour angiogenesis and tumour growth
    T. Lechertier, L. E. Reynolds, H. Kim, A. R. Pedrosa, J. Gómez-Escudero, J. M. Muñoz-Félix, S. Batista, M. Dukinfield, F. Demircioglu, P. P. Wong, K. P. Matchett, N. C. Henderson, G. D’Amico, M. Parsons, C. Harwood, P. Meier, and K. M. Hodivala-Dilke
    Nat Commun Jun 2020

    The overexpression of the protein tyrosine kinase, Focal adhesion kinase (FAK), in endothelial cells has implicated its requirement in angiogenesis and tumour growth, but how pericyte FAK regulates tumour angiogenesis is unknown. We show that pericyte FAK regulates tumour growth and angiogenesis in multiple mouse models of melanoma, lung carcinoma and pancreatic B-cell insulinoma and provide evidence that loss of pericyte FAK enhances Gas6-stimulated phosphorylation of the receptor tyrosine kinase, Axl with an upregulation of Cyr61, driving enhanced tumour growth. We further show that pericyte derived Cyr61 instructs tumour cells to elevate expression of the proangiogenic/protumourigenic transmembrane receptor Tissue Factor. Finally, in human melanoma we show that when 50% or more tumour blood vessels are pericyte-FAK negative, melanoma patients are stratified into those with increased tumour size, enhanced blood vessel density and metastasis. Overall our data uncover a previously unknown mechanism of tumour growth by pericytes that is controlled by pericyte FAK.

    @article{pmid32499572,
      author = {Lechertier, T. and Reynolds, L. E. and Kim, H. and Pedrosa, A. R. and Gómez-Escudero, J. and Muñoz-Félix, J. M. and Batista, S. and Dukinfield, M. and Demircioglu, F. and Wong, P. P. and Matchett, K. P. and Henderson, N. C. and D'Amico, G. and Parsons, M. and Harwood, C. and Meier, P. and Hodivala-Dilke, K. M.},
      title = {{{P}ericyte {F}{A}{K} negatively regulates {G}as6/{A}xl signalling to suppress tumour angiogenesis and tumour growth}},
      journal = {Nat Commun},
      year = {2020},
      volume = {11},
      number = {1},
      pages = {2810},
      month = jun
    }
  7. Single-cell technologies in hepatology: new insights into liver biology and disease pathogenesis
    P. Ramachandran, K. P. Matchett, R. Dobie, J. R. Wilson-Kanamori, and N. C. Henderson
    Nat Rev Gastroenterol Hepatol Aug 2020

    Liver disease is a major global health-care problem, affecting an estimated 844 million people worldwide. Despite this substantial burden, therapeutic options for liver disease remain limited, in part owing to a paucity of detailed analyses defining the cellular and molecular mechanisms that drive these conditions in humans. Single-cell transcriptomic technologies are transforming our understanding of cellular diversity and function in health and disease. In this Review, we discuss how these technologies have been applied in hepatology, advancing our understanding of cellular heterogeneity and providing novel insights into fundamental liver biology such as the metabolic zonation of hepatocytes, endothelial cells and hepatic stellate cells, and the cellular mechanisms underpinning liver regeneration. Application of these methodologies is also uncovering critical pathophysiological changes driving disease states such as hepatic fibrosis, where distinct populations of macrophages, endothelial cells and mesenchymal cells reside within a spatially distinct fibrotic niche and interact to promote scar formation. In addition, single-cell approaches are starting to dissect key cellular and molecular functions in liver cancer. In the near future, new techniques such as spatial transcriptomics and multiomic approaches will further deepen our understanding of disease pathogenesis, enabling the identification of novel therapeutic targets for patients across the spectrum of liver diseases.

    @article{pmid32483353,
      author = {Ramachandran, P. and Matchett, K. P. and Dobie, R. and Wilson-Kanamori, J. R. and Henderson, N. C.},
      title = {{{S}ingle-cell technologies in hepatology: new insights into liver biology and disease pathogenesis}},
      journal = {Nat Rev Gastroenterol Hepatol},
      year = {2020},
      volume = {17},
      number = {8},
      pages = {457--472},
      month = aug
    }
  8. Cancer Burden Is Controlled by Mural Cell-β3-Integrin Regulated Crosstalk with Tumor Cells
    P. P. Wong, J. M. Muñoz-Félix, M. Hijazi, H. Kim, S. D. Robinson, B. De Luxán-Delgado, I. Rodríguez-Hernández, O. Maiques, Y. M. Meng, Q. Meng, N. Bodrug, M. S. Dukinfield, L. E. Reynolds, G. Elia, A. Clear, C. Harwood, Y. Wang, J. J. Campbell, R. Singh, P. Zhang, T. J. Schall, K. P. Matchett, N. C. Henderson, P. W. Szlosarek, S. A. Dreger, S. Smith, J. L. Jones, J. G. Gribben, P. R. Cutillas, P. Meier, V. Sanz-Moreno, and K. M. Hodivala-Dilke
    Cell Jun 2020

    Enhanced blood vessel (BV) formation is thought to drive tumor growth through elevated nutrient delivery. However, this observation has overlooked potential roles for mural cells in directly affecting tumor growth independent of BV function. Here we provide clinical data correlating high percentages of mural-β3-integrin-negative tumor BVs with increased tumor sizes but no effect on BV numbers. Mural-β3-integrin loss also enhances tumor growth in implanted and autochthonous mouse tumor models with no detectable effects on BV numbers or function. At a molecular level, mural-cell β3-integrin loss enhances signaling via FAK-p-HGFR-p-Akt-p-p65, driving CXCL1, CCL2, and TIMP-1 production. In particular, mural-cell-derived CCL2 stimulates tumor cell MEK1-ERK1/2-ROCK2-dependent signaling and enhances tumor cell survival and tumor growth. Overall, our data indicate that mural cells can control tumor growth via paracrine signals regulated by β3-integrin, providing a previously unrecognized mechanism of cancer growth control.

    @article{pmid32473126,
      author = {Wong, P. P. and Muñoz-Félix, J. M. and Hijazi, M. and Kim, H. and Robinson, S. D. and De Luxán-Delgado, B. and Rodríguez-Hernández, I. and Maiques, O. and Meng, Y. M. and Meng, Q. and Bodrug, N. and Dukinfield, M. S. and Reynolds, L. E. and Elia, G. and Clear, A. and Harwood, C. and Wang, Y. and Campbell, J. J. and Singh, R. and Zhang, P. and Schall, T. J. and Matchett, K. P. and Henderson, N. C. and Szlosarek, P. W. and Dreger, S. A. and Smith, S. and Jones, J. L. and Gribben, J. G. and Cutillas, P. R. and Meier, P. and Sanz-Moreno, V. and Hodivala-Dilke, K. M.},
      title = {{{C}ancer {B}urden {I}s {C}ontrolled by {M}ural {C}ell-β3-{I}ntegrin {R}egulated {C}rosstalk with {T}umor {C}ells}},
      journal = {Cell},
      year = {2020},
      volume = {181},
      number = {6},
      pages = {1346--1363},
      month = jun
    }
  9. Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis
    T. Tsukui, K. H. Sun, J. B. Wetter, J. R. Wilson-Kanamori, L. A. Hazelwood, N. C. Henderson, T. S. Adams, J. C. Schupp, S. D. Poli, I. O. Rosas, N. Kaminski, M. A. Matthay, P. J. Wolters, and D. Sheppard
    Nat Commun Apr 2020

    Collagen-producing cells maintain the complex architecture of the lung and drive pathologic scarring in pulmonary fibrosis. Here we perform single-cell RNA-sequencing to identify all collagen-producing cells in normal and fibrotic lungs. We characterize multiple collagen-producing subpopulations with distinct anatomical localizations in different compartments of murine lungs. One subpopulation, characterized by expression of Cthrc1 (collagen triple helix repeat containing 1), emerges in fibrotic lungs and expresses the highest levels of collagens. Single-cell RNA-sequencing of human lungs, including those from idiopathic pulmonary fibrosis and scleroderma patients, demonstrate similar heterogeneity and CTHRC1-expressing fibroblasts present uniquely in fibrotic lungs. Immunostaining and in situ hybridization show that these cells are concentrated within fibroblastic foci. We purify collagen-producing subpopulations and find disease-relevant phenotypes of Cthrc1-expressing fibroblasts in in vitro and adoptive transfer experiments. Our atlas of collagen-producing cells provides a roadmap for studying the roles of these unique populations in homeostasis and pathologic fibrosis.

    @article{pmid32317643,
      author = {Tsukui, T. and Sun, K. H. and Wetter, J. B. and Wilson-Kanamori, J. R. and Hazelwood, L. A. and Henderson, N. C. and Adams, T. S. and Schupp, J. C. and Poli, S. D. and Rosas, I. O. and Kaminski, N. and Matthay, M. A. and Wolters, P. J. and Sheppard, D.},
      title = {{{C}ollagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis}},
      journal = {Nat Commun},
      year = {2020},
      volume = {11},
      number = {1},
      pages = {1920},
      month = apr
    }
  10. Stromal Cells Covering Omental Fat-Associated Lymphoid Clusters Trigger Formation of Neutrophil Aggregates to Capture Peritoneal Contaminants
    L. H. Jackson-Jones, P. Smith, J. R. Portman, M. S. Magalhaes, K. J. Mylonas, M. M. Vermeren, M. Nixon, B. E. P. Henderson, R. Dobie, S. Vermeren, L. Denby, N. C. Henderson, D. J. Mole, and C. Bénézech
    Immunity Apr 2020

    mesothelial cells, which we termed FALC cover cells. Blockade of CXCL1 inhibited the recruitment and aggregation of neutrophils at FALCs during zymosan-induced peritonitis. Inhibition of protein arginine deiminase 4, an enzyme important for the release of neutrophil extracellular traps, abolished neutrophil aggregation and the capture of peritoneal contaminants by omental FALCs. Analysis of omental samples from patients with acute appendicitis confirmed neutrophil recruitment and bacterial capture at FALCs. Thus, specialized omental mesothelial cells coordinate the recruitment and aggregation of neutrophils to capture peritoneal contaminants.

    @article{pmid32294409,
      author = {Jackson-Jones, L. H. and Smith, P. and Portman, J. R. and Magalhaes, M. S. and Mylonas, K. J. and Vermeren, M. M. and Nixon, M. and Henderson, B. E. P. and Dobie, R. and Vermeren, S. and Denby, L. and Henderson, N. C. and Mole, D. J. and Bénézech, C.},
      title = {{{S}tromal {C}ells {C}overing {O}mental {F}at-{A}ssociated {L}ymphoid {C}lusters {T}rigger {F}ormation of {N}eutrophil {A}ggregates to {C}apture {P}eritoneal {C}ontaminants}},
      journal = {Immunity},
      year = {2020},
      volume = {52},
      number = {4},
      pages = {700--715},
      month = apr
    }
  11. The circadian clock protein REVERBα inhibits pulmonary fibrosis development
    P. S. Cunningham, P. Meijer, A. Nazgiewicz, S. G. Anderson, L. A. Borthwick, J. Bagnall, G. B. Kitchen, M. Lodyga, N. Begley, R. V. Venkateswaran, R. Shah, P. F. Mercer, H. J. Durrington, N. C. Henderson, K. Piper-Hanley, A. J. Fisher, R. C. Chambers, D. A. Bechtold, J. E. Gibbs, A. S. Loudon, M. K. Rutter, B. Hinz, D. W. Ray, and J. F. Blaikley
    Proc Natl Acad Sci U S A Jan 2020

    Pulmonary inflammatory responses lie under circadian control; however, the importance of circadian mechanisms in the underlying fibrotic phenotype is not understood. Here, we identify a striking change to these mechanisms resulting in a gain of amplitude and lack of synchrony within pulmonary fibrotic tissue. These changes result from an infiltration of mesenchymal cells, an important cell type in the pathogenesis of pulmonary fibrosis. Mutation of the core clock protein REVERBα in these cells exacerbated the development of bleomycin-induced fibrosis, whereas mutation of REVERBα in club or myeloid cells had no effect on the bleomycin phenotype. Knockdown of REVERBα revealed regulation of the little-understood transcription factor TBPL1. Both REVERBα and TBPL1 altered integrinβ1 focal-adhesion formation, resulting in increased myofibroblast activation. The translational importance of our findings was established through analysis of 2 human cohorts. In the UK Biobank, circadian strain markers (sleep length, chronotype, and shift work) are associated with pulmonary fibrosis, making them risk factors. In a separate cohort, REVERBα expression was increased in human idiopathic pulmonary fibrosis (IPF) lung tissue. Pharmacological targeting of REVERBα inhibited myofibroblast activation in IPF fibroblasts and collagen secretion in organotypic cultures from IPF patients, thus suggesting that targeting of REVERBα could be a viable therapeutic approach.

    @article{pmid31879343,
      author = {Cunningham, P. S. and Meijer, P. and Nazgiewicz, A. and Anderson, S. G. and Borthwick, L. A. and Bagnall, J. and Kitchen, G. B. and Lodyga, M. and Begley, N. and Venkateswaran, R. V. and Shah, R. and Mercer, P. F. and Durrington, H. J. and Henderson, N. C. and Piper-Hanley, K. and Fisher, A. J. and Chambers, R. C. and Bechtold, D. A. and Gibbs, J. E. and Loudon, A. S. and Rutter, M. K. and Hinz, B. and Ray, D. W. and Blaikley, J. F.},
      title = {{{T}he circadian clock protein {R}{E}{V}{E}{R}{B}α inhibits pulmonary fibrosis development}},
      journal = {Proc Natl Acad Sci U S A},
      year = {2020},
      volume = {117},
      number = {2},
      pages = {1139--1147},
      month = jan
    }
  12. Transcriptional dynamics of pluripotent stem cell-derived endothelial cell differentiation revealed by single-cell RNA sequencing
    I. R. McCracken, R. S. Taylor, F. O. Kok, F. Cuesta, R. Dobie, B. E. P. Henderson, J. C. Mountford, A. Caudrillier, N. C. Henderson, C. P. Ponting, and A. H. Baker
    Eur Heart J Mar 2020

    Pluripotent stem cell-derived endothelial cell products possess therapeutic potential in ischaemic vascular disease. However, the factors that drive endothelial differentiation from pluripotency and cellular specification are largely unknown. The aims of this study were to use single-cell RNA sequencing (scRNA-seq) to map the transcriptional landscape and cellular dynamics of directed differentiation of human embryonic stem cell-derived endothelial cells (hESC-EC) and to compare these cells to mature endothelial cells from diverse vascular beds.\{A highly efficient directed 8-day differentiation protocol was used to generate a hESC-derived endothelial cell product (hESC-ECP), in which 66% of cells co-expressed CD31 and CD144. We observed largely homogeneous hESC and mesodermal populations at Days 0 and 4, respectively, followed by a rapid emergence of distinct endothelial and mesenchymal populations. Pseudotime trajectory identified transcriptional signatures of endothelial commitment and maturation during the differentiation process. Concordance in transcriptional signatures was verified by scRNA-seq analysis using both a second hESC line RC11, and an alternative hESC-EC differentiation protocol. In total, 105 727 cells were subjected to scRNA-seq analysis. Global transcriptional comparison revealed a transcriptional architecture of hESC-EC that differs from freshly isolated and cultured human endothelial cells and from organ-specific endothelial cells.\{A transcriptional bifurcation into endothelial and mesenchymal lineages was identified, as well as novel transcriptional signatures underpinning commitment and maturation. The transcriptional architecture of hESC-ECP was distinct from mature and foetal human EC.

    @article{pmid31242503,
      author = {McCracken, I. R. and Taylor, R. S. and Kok, F. O. and de la Cuesta, F. and Dobie, R. and Henderson, B. E. P. and Mountford, J. C. and Caudrillier, A. and Henderson, N. C. and Ponting, C. P. and Baker, A. H.},
      title = {{{T}ranscriptional dynamics of pluripotent stem cell-derived endothelial cell differentiation revealed by single-cell {R}{N}{A} sequencing}},
      journal = {Eur Heart J},
      year = {2020},
      volume = {41},
      number = {9},
      pages = {1024--1036},
      month = mar
    }
  13. Nature
    Fibrosis: from mechanisms to medicines
    Neil C Henderson, Florian Rieder, and Thomas A Wynn
    Nature Mar 2020

    Fibrosis can affect any organ and is responsible for up to 45% of all deaths in the industrialized world. It has long been thought to be relentlessly progressive and irreversible, but both preclinical models and clinical trials in various organ systems have shown that fibrosis is a highly dynamic process. This has clear implications for therapeutic interventions that are designed to capitalize on this inherent plasticity. However, despite substantial progress in our understanding of the pathobiology of fibrosis, a translational gap remains between the identification of putative antifibrotic targets and conversion of this knowledge into effective treatments in humans. Here we discuss the transformative experimental strategies that are being leveraged to dissect the key cellular and molecular mechanisms that regulate fibrosis, and the translational approaches that are enabling the emergence of precision medicine-based therapies for patients with fibrosis.

    @article{henderson2020fibrosis,
      abbr = {Nature},
      url = {https://doi.org/10.1038/s41586-020-2938-9},
      title = {Fibrosis: from mechanisms to medicines},
      author = {Henderson, Neil C and Rieder, Florian and Wynn, Thomas A},
      journal = {Nature},
      volume = {587},
      number = {7835},
      pages = {555--566},
      year = {2020},
      publisher = {Nature Publishing Group},
      selected = {true}
    }

2019

  1. Unravelling fibrosis using single-cell transcriptomics
    R. Dobie, and N. C. Henderson
    Curr Opin Pharmacol Dec 2019

    Fibrosis, the excessive accumulation of extracellular matrix, is a major global healthcare burden. Despite major advances in our understanding of the mechanisms regulating fibrosis, treatment options for patients with fibrosis remain very limited. However, recent developments in the rapidly evolving field of single-cell transcriptomics are enabling the interrogation of individual pathogenic cell populations in the context of fibrosis at unprecedented resolution. In this review, we will discuss how single-cell transcriptomics is driving this step change in our understanding of fibrotic disease pathogenesis, and how these cutting-edge approaches should accelerate the precise identification of novel, relevant and potentially druggable therapeutic targets to treat patients with fibrosis.

    @article{pmid31670054,
      author = {Dobie, R. and Henderson, N. C.},
      title = {{{U}nravelling fibrosis using single-cell transcriptomics}},
      journal = {Curr Opin Pharmacol},
      year = {2019},
      volume = {49},
      pages = {71--75},
      month = dec
    }
  2. Mice depleted for Exchange Proteins Directly Activated by cAMP (Epac) exhibit irregular liver regeneration in response to partial hepatectomy
    K. Sivertsen Åsrud, L. Pedersen, R. Aesoy, H. Muwonge, E. Aasebø, I. K. Nitschke Pettersen, L. Herfindal, R. Dobie, S. Jenkins, R. K. Berge, N. C. Henderson, F. Selheim, S. O. Døskeland, and M. Bakke
    Sci Rep Sep 2019

    The exchange proteins directly activated by cAMP 1 and 2 (Epac1 and Epac2) are expressed in a cell specific manner in the liver, but their biological functions in this tissue are poorly understood. The current study was undertaken to begin to determine the potential roles of Epac1 and Epac2 in liver physiology and disease. Male C57BL/6J mice in which expression of Epac1 and/or Epac2 are deleted, were subjected to partial hepatectomy and the regenerating liver was analyzed with regard to lipid accumulation, cell replication and protein expression. In response to partial hepatectomy, deletion of Epac1 and/or Epac2 led to increased hepatocyte proliferation 36 h post surgery, and the transient steatosis observed in wild type mice was virtually absent in mice lacking both Epac1 and Epac2. The expression of the protein cytochrome P4504a14, which is implicated in hepatic steatosis and fibrosis, was substantially reduced upon deletion of Epac1/2, while a number of factors involved in lipid metabolism were significantly decreased. Moreover, the number of Küpffer cells was affected, and Epac2 expression was increased in the liver of wild type mice in response to partial hepatectomy, further supporting a role for these proteins in liver function. This study establishes hepatic phenotypic abnormalities in mice deleted for Epac1/2 for the first time, and introduces Epac1/2 as regulators of hepatocyte proliferation and lipid accumulation in the regenerative process.

    @article{pmid31551444,
      author = {Sivertsen Åsrud, K. and Pedersen, L. and Aesoy, R. and Muwonge, H. and Aasebø, E. and Nitschke Pettersen, I. K. and Herfindal, L. and Dobie, R. and Jenkins, S. and Berge, R. K. and Henderson, N. C. and Selheim, F. and Døskeland, S. O. and Bakke, M.},
      title = {{{M}ice depleted for {E}xchange {P}roteins {D}irectly {A}ctivated by c{A}{M}{P} ({E}pac) exhibit irregular liver regeneration in response to partial hepatectomy}},
      journal = {Sci Rep},
      year = {2019},
      volume = {9},
      number = {1},
      pages = {13789},
      month = sep
    }
  3. Development of mouse models of angiosarcoma driven by p53
    D. M. Salter, M. Griffin, M. Muir, K. Teo, J. Culley, J. R. Smith, L. Gomez-Cuadrado, K. Matchett, A. H. Sims, L. Hayward, N. C. Henderson, and V. G. Brunton
    Dis Model Mech Jul 2019

    mice provided a more consistent and rapid model of angiosarcoma than the two spontaneous models. The ability to passage tumour fragments through the mouse provides a novel model which is amenable to preclinical studies and will help the development of potential new therapies for angiosarcoma.

    @article{pmid31221668,
      author = {Salter, D. M. and Griffin, M. and Muir, M. and Teo, K. and Culley, J. and Smith, J. R. and Gomez-Cuadrado, L. and Matchett, K. and Sims, A. H. and Hayward, L. and Henderson, N. C. and Brunton, V. G.},
      title = {{{D}evelopment of mouse models of angiosarcoma driven by p53}},
      journal = {Dis Model Mech},
      year = {2019},
      volume = {12},
      number = {7},
      month = jul
    }
  4. Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction
    Z. Li, E. G. Solomonidis, M. Meloni, R. S. Taylor, R. Duffin, R. Dobie, M. S. Magalhaes, B. E. P. Henderson, P. A. Louwe, G. D’Amico, K. M. Hodivala-Dilke, A. M. Shah, N. L. Mills, B. D. Simons, G. A. Gray, N. C. Henderson, A. H. Baker, and M. Brittan
    Eur Heart J Aug 2019

    A better understanding of the pathways that regulate regeneration of the coronary vasculature is of fundamental importance for the advancement of strategies to treat patients with heart disease. Here, we aimed to investigate the origin and clonal dynamics of endothelial cells (ECs) associated with neovascularization in the adult mouse heart following myocardial infarction (MI). Furthermore, we sought to define murine cardiac endothelial heterogeneity and to characterize the transcriptional profiles of pro-angiogenic resident ECs in the adult mouse heart, at single-cell resolution.\{An EC-specific multispectral lineage-tracing mouse (Pdgfb-iCreERT2-R26R-Brainbow2.1) was used to demonstrate that structural integrity of adult cardiac endothelium following MI was maintained through clonal proliferation by resident ECs in the infarct border region, without significant contributions from bone marrow cells or endothelial-to-mesenchymal transition. Ten transcriptionally discrete heterogeneous EC states, as well as the pathways through which each endothelial state is likely to enhance neovasculogenesis and tissue regeneration following ischaemic injury were defined. Plasmalemma vesicle-associated protein (Plvap) was selected for further study, which showed an endothelial-specific and increased expression in both the ischaemic mouse and human heart, and played a direct role in regulating human endothelial proliferation in vitro.\{We present a single-cell gene expression atlas of cardiac specific resident ECs, and the transcriptional hierarchy underpinning endogenous vascular repair following MI. These data provide a rich resource that could assist in the development of new therapeutic interventions to augment endogenous myocardial perfusion and enhance regeneration in the injured heart.

    @article{pmid31162546,
      author = {Li, Z. and Solomonidis, E. G. and Meloni, M. and Taylor, R. S. and Duffin, R. and Dobie, R. and Magalhaes, M. S. and Henderson, B. E. P. and Louwe, P. A. and D'Amico, G. and Hodivala-Dilke, K. M. and Shah, A. M. and Mills, N. L. and Simons, B. D. and Gray, G. A. and Henderson, N. C. and Baker, A. H. and Brittan, M.},
      title = {{{S}ingle-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction}},
      journal = {Eur Heart J},
      year = {2019},
      volume = {40},
      number = {30},
      pages = {2507--2520},
      month = aug
    }
  5. A Macrophage-Pericyte Axis Directs Tissue Restoration via Amphiregulin-Induced Transforming Growth Factor Beta Activation
    C. M. Minutti, R. V. Modak, F. Macdonald, F. Li, D. J. Smyth, D. A. Dorward, N. Blair, C. Husovsky, A. Muir, E. Giampazolias, R. Dobie, R. M. Maizels, T. J. Kendall, D. W. Griggs, M. Kopf, N. C. Henderson, and D. M. Zaiss
    Immunity Mar 2019

    activation. Using acute injury models in two different tissues, we found that by inducing TGF-β activation on mesenchymal stromal cells (pericytes), Amphiregulin induced their differentiation into myofibroblasts, thereby selectively contributing to the restoration of vascular barrier function within injured tissue. Furthermore, we identified macrophages as a critical source of Amphiregulin, revealing a direct effector mechanism by which these cells contribute to tissue restoration after acute injury. Combined, these observations expose a so far under-appreciated mechanism of how cells of the immune system selectively control the differentiation of tissue progenitor cells during tissue repair and inflammation.

    @article{pmid30770250,
      author = {Minutti, C. M. and Modak, R. V. and Macdonald, F. and Li, F. and Smyth, D. J. and Dorward, D. A. and Blair, N. and Husovsky, C. and Muir, A. and Giampazolias, E. and Dobie, R. and Maizels, R. M. and Kendall, T. J. and Griggs, D. W. and Kopf, M. and Henderson, N. C. and Zaiss, D. M.},
      title = {{{A} {M}acrophage-{P}ericyte {A}xis {D}irects {T}issue {R}estoration via {A}mphiregulin-{I}nduced {T}ransforming {G}rowth {F}actor {B}eta {A}ctivation}},
      journal = {Immunity},
      year = {2019},
      volume = {50},
      number = {3},
      pages = {645--654},
      month = mar
    }
  6. An Orally Active Galectin-3 Antagonist Inhibits Lung Adenocarcinoma Growth and Augments Response to PD-L1 Blockade
    L. Vuong, E. Kouverianou, C. M. Rooney, B. J. McHugh, S. E. M. Howie, C. D. Gregory, S. J. Forbes, N. C. Henderson, F. R. Zetterberg, U. J. Nilsson, H. Leffler, P. Ford, A. Pedersen, L. Gravelle, S. Tantawi, H. Schambye, T. Sethi, and A. C. MacKinnon
    Cancer Res Apr 2019

    http://cancerres.aacrjournals.org/content/canres/79/7/1480/F1.large.jpg.

    @article{pmid30674531,
      author = {Vuong, L. and Kouverianou, E. and Rooney, C. M. and McHugh, B. J. and Howie, S. E. M. and Gregory, C. D. and Forbes, S. J. and Henderson, N. C. and Zetterberg, F. R. and Nilsson, U. J. and Leffler, H. and Ford, P. and Pedersen, A. and Gravelle, L. and Tantawi, S. and Schambye, H. and Sethi, T. and MacKinnon, A. C.},
      title = {{{A}n {O}rally {A}ctive {G}alectin-3 {A}ntagonist {I}nhibits {L}ung {A}denocarcinoma {G}rowth and {A}ugments {R}esponse to {P}{D}-{L}1 {B}lockade}},
      journal = {Cancer Res},
      year = {2019},
      volume = {79},
      number = {7},
      pages = {1480--1492},
      month = apr
    }
  7. Fibroblast-specific integrin-alpha V differentially regulates type 17 and type 2 driven inflammation and fibrosis
    J. C. Sciurba, R. L. Gieseck, N. Jiwrajka, S. D. White, E. P. Karmele, J. Redes, K. M. Vannella, N. C. Henderson, T. A. Wynn, and K. M. Hart
    J Pathol May 2019

    Sons, Ltd.

    @article{pmid30536905,
      author = {Sciurba, J. C. and Gieseck, R. L. and Jiwrajka, N. and White, S. D. and Karmele, E. P. and Redes, J. and Vannella, K. M. and Henderson, N. C. and Wynn, T. A. and Hart, K. M.},
      title = {{{F}ibroblast-specific integrin-alpha {V} differentially regulates type 17 and type 2 driven inflammation and fibrosis}},
      journal = {J Pathol},
      year = {2019},
      volume = {248},
      number = {1},
      pages = {16--29},
      month = may
    }
  8. Loss of Integrin αvβ8 in Murine Hepatocytes Accelerates Liver Regeneration
    S. N. Greenhalgh, K. P. Matchett, R. S. Taylor, K. Huang, J. T. Li, K. Saeteurn, M. C. Donnelly, E. E. M. Simpson, J. L. Pollack, A. Atakilit, K. J. Simpson, J. J. Maher, J. P. Iredale, D. Sheppard, and N. C. Henderson
    Am J Pathol Feb 2019

    ;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.

    @article{pmid30448409,
      author = {Greenhalgh, S. N. and Matchett, K. P. and Taylor, R. S. and Huang, K. and Li, J. T. and Saeteurn, K. and Donnelly, M. C. and Simpson, E. E. M. and Pollack, J. L. and Atakilit, A. and Simpson, K. J. and Maher, J. J. and Iredale, J. P. and Sheppard, D. and Henderson, N. C.},
      title = {{{L}oss of {I}ntegrin αvβ8 in {M}urine {H}epatocytes {A}ccelerates {L}iver {R}egeneration}},
      journal = {Am J Pathol},
      year = {2019},
      volume = {189},
      number = {2},
      pages = {258--271},
      month = feb
    }
  9. Nature
    Resolving the fibrotic niche of human liver cirrhosis at single-cell level
    Prakash Ramachandran, Ross Dobie, John R Wilson-Kanamori, EF Dora, BEP Henderson, NT Luu, Jordan R Portman, Kylie P Matchett, Madara Brice, JA Marwick, RS Taylor, M Efremova, R Vento-Tormo, NO Carragher, TJ Kendall, JA Fallowfield, EM Harrison, DJ Mole, SJ Wigmore, PN Newsome, J Weston, JP Iredale, F Tacke, JW Pollard, Chris Ponting, John C Marioni, Sarah A Teichmann, and Neil C Henderson
    Nature Feb 2019

    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.

    @article{ramachandran2019resolving,
      abbr = {Nature},
      url = {https://doi.org/10.1038/s41586-019-1631-3},
      title = {Resolving the fibrotic niche of human liver cirrhosis at single-cell level},
      author = {Ramachandran, Prakash and Dobie, Ross and Wilson-Kanamori, John R and Dora, EF and Henderson, BEP and Luu, NT and Portman, Jordan R and Matchett, Kylie P and Brice, Madara and Marwick, JA and Taylor, RS and Efremova, M and Vento-Tormo, R and Carragher, NO and Kendall, TJ and Fallowfield, JA and Harrison, EM and Mole, DJ and Wigmore, SJ and Newsome, PN and Weston, J and Iredale, JP and Tacke, F and Pollard, JW and Ponting, Chris and Marioni, John C and Teichmann, Sarah A and Henderson, Neil C},
      journal = {Nature},
      volume = {575},
      number = {7783},
      pages = {512--518},
      year = {2019},
      publisher = {Nature Publishing Group},
      selected = {true}
    }
  10. CellRep
    Single-cell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis
    Ross Dobie, John R Wilson-Kanamori, Beth EP Henderson, James R Smith, Kylie P Matchett, Jordan R Portman, Karolina Wallenborg, Simone Picelli, Anna Zagorska, Swetha V Pendem, TE Hudson, MM Wu, GR Budas, DG Breckenridge, EM Harrison, DJ Mole, SJ Wigmore, Prakash Ramachandran, Chris P Ponting, Sarah A Teichmann, John C Marioni, and Neil C Henderson
    Cell reports Feb 2019

    Iterative liver injury results in progressive fibrosis disrupting hepatic architecture, regeneration potential, and liver function. Hepatic stellate cells (HSCs) are a major source of pathological matrix during fibrosis and are thought to be a functionally homogeneous population. Here, we use single-cell RNA sequencing to deconvolve the hepatic mesenchyme in healthy and fibrotic mouse liver, revealing spatial zonation of HSCs across the hepatic lobule. Furthermore, we show that HSCs partition into topographically diametric lobule regions, designated portal vein-associated HSCs (PaHSCs) and central vein-associated HSCs (CaHSCs). Importantly we uncover functional zonation, identifying CaHSCs as the dominant pathogenic collagen-producing cells in a mouse model of centrilobular fibrosis. Finally, we identify LPAR1 as a therapeutic target on collagen-producing CaHSCs, demonstrating that blockade of LPAR1 inhibits liver fibrosis in a rodent NASH model. Taken together, our work illustrates the power of single-cell transcriptomics to resolve the key collagen-producing cells driving liver fibrosis with high precision.

    @article{dobie2019single,
      abbr = {CellRep},
      title = {Single-cell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis},
      author = {Dobie, Ross and Wilson-Kanamori, John R and Henderson, Beth EP and Smith, James R and Matchett, Kylie P and Portman, Jordan R and Wallenborg, Karolina and Picelli, Simone and Zagorska, Anna and Pendem, Swetha V and Hudson, TE and Wu, MM and Budas, GR and Breckenridge, DG and Harrison, EM and Mole, DJ and Wigmore, SJ and Ramachandran, Prakash and Ponting, Chris P and Teichmann, Sarah A and Marioni, John C and Henderson, Neil C},
      journal = {Cell reports},
      volume = {29},
      number = {7},
      pages = {1832--1847},
      year = {2019},
      publisher = {Elsevier},
      selected = {true}
    }

2018

  1. Unravelling atherosclerotic heterogeneity by single cell RNA sequencing
    I. R. McCracken, R. S. Taylor, N. C. Henderson, J. C. Sluimer, and A. H. Baker
    Curr Opin Lipidol Dec 2018
    @article{pmid30379738,
      author = {McCracken, I. R. and Taylor, R. S. and Henderson, N. C. and Sluimer, J. C. and Baker, A. H.},
      title = {{{U}nravelling atherosclerotic heterogeneity by single cell {R}{N}{A} sequencing}},
      journal = {Curr Opin Lipidol},
      year = {2018},
      volume = {29},
      number = {6},
      pages = {488--489},
      month = dec
    }
  2. The STAT3-IL-10-IL-6 Pathway Is a Novel Regulator of Macrophage Efferocytosis and Phenotypic Conversion in Sterile Liver Injury
    L. Campana, P. J. Starkey Lewis, A. Pellicoro, R. L. Aucott, J. Man, E. O’Duibhir, S. E. Mok, S. Ferreira-Gonzalez, E. Livingstone, S. N. Greenhalgh, K. L. Hull, T. J. Kendall, D. Vernimmen, N. C. Henderson, L. Boulter, C. D. Gregory, Y. Feng, S. M. Anderton, S. J. Forbes, and J. P. Iredale
    J Immunol Feb 2018

    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.

    @article{pmid29263216,
      author = {Campana, L. and Starkey Lewis, P. J. and Pellicoro, A. and Aucott, R. L. and Man, J. and O'Duibhir, E. and Mok, S. E. and Ferreira-Gonzalez, S. and Livingstone, E. and Greenhalgh, S. N. and Hull, K. L. and Kendall, T. J. and Vernimmen, D. and Henderson, N. C. and Boulter, L. and Gregory, C. D. and Feng, Y. and Anderton, S. M. and Forbes, S. J. and Iredale, J. P.},
      title = {{{T}he {S}{T}{A}{T}3-{I}{L}-10-{I}{L}-6 {P}athway {I}s a {N}ovel {R}egulator of {M}acrophage {E}fferocytosis and {P}henotypic {C}onversion in {S}terile {L}iver {I}njury}},
      journal = {J Immunol},
      year = {2018},
      volume = {200},
      number = {3},
      pages = {1169--1187},
      month = feb
    }
  3. Beta-hydroxysteroid dehydrogenase-1 deficiency or inhibition enhances hepatic myofibroblast activation in murine liver fibrosis
    X. Zou, P. Ramachandran, T. J. Kendall, A. Pellicoro, E. Dora, R. L. Aucott, K. Manwani, T. Y. Man, K. E. Chapman, N. C. Henderson, S. J. Forbes, S. P. Webster, J. P. Iredale, B. R. Walker, and Z. Michailidou
    Hepatology Jun 2018

    mice, including greater fibrosis.\ 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).

    @article{pmid29251794,
      author = {Zou, X. and Ramachandran, P. and Kendall, T. J. and Pellicoro, A. and Dora, E. and Aucott, R. L. and Manwani, K. and Man, T. Y. and Chapman, K. E. and Henderson, N. C. and Forbes, S. J. and Webster, S. P. and Iredale, J. P. and Walker, B. R. and Michailidou, Z.},
      title = {{{B}eta-hydroxysteroid dehydrogenase-1 deficiency or inhibition enhances hepatic myofibroblast activation in murine liver fibrosis}},
      journal = {Hepatology},
      year = {2018},
      volume = {67},
      number = {6},
      pages = {2167--2181},
      month = jun
    }

2017

  1. Sphingosine-1-Phosphate Prevents Egress of Hematopoietic Stem Cells From Liver to Reduce Fibrosis
    A. King, D. D. Houlihan, D. Kavanagh, D. Haldar, N. Luu, A. Owen, S. Suresh, N. N. Than, G. Reynolds, J. Penny, H. Sumption, P. Ramachandran, N. C. Henderson, N. Kalia, J. Frampton, D. H. Adams, and P. N. Newsome
    Gastroenterology Jul 2017

    There is growing interest in the use of bone marrow cells to treat liver fibrosis, however, little is known about their antifibrotic efficacy or the identity of their effector cell(s). Sphingosine-1-phosphate (S1P) mediates egress of immune cells from the lymphoid organs into the lymphatic vessels; we investigated its role in the response of hematopoietic stem cells (HSCs) to liver fibrosis in mice.\ ) or placement on a methionine-choline-deficient diet. Some mice were irradiated and given transplants of bone marrow cells from C57BL6 mice, with or without the S1P antagonist FTY720; we then studied HSC mobilization and localization. Migration of HSC lines was quantified in Transwell assays. Levels of S1P in liver, bone marrow, and lymph fluid were measured using an enzyme-linked immunosorbent assay. Liver tissues were collected and analyzed by immunohistochemical quantitative polymerase chain reaction and sphingosine kinase activity assays. We performed quantitative polymerase chain reaction analyses of the expression of sphingosine kinase 1 and 2, sphingosine-1-phosphate lyase 1, and sphingosine-1-phosphate phosphatase 1 in normal human liver and cirrhotic liver from patients with alcohol-related liver disease (n = 6).\ .05), and further reductions in fibrosis.\{In studies of mice with chronic liver injury, we showed the antifibrotic effects of repeated infusions of purified HSCs. We found that HSCs promote recruitment of endogenous macrophages and neutrophils. Strategies to reduce SIP signaling and increase retention of HSCs in the liver could increase their antifibrotic activities and be developed for treatment of patients with liver fibrosis.

    @article{pmid28363640,
      author = {King, A. and Houlihan, D. D. and Kavanagh, D. and Haldar, D. and Luu, N. and Owen, A. and Suresh, S. and Than, N. N. and Reynolds, G. and Penny, J. and Sumption, H. and Ramachandran, P. and Henderson, N. C. and Kalia, N. and Frampton, J. and Adams, D. H. and Newsome, P. N.},
      title = {{{S}phingosine-1-{P}hosphate {P}revents {E}gress of {H}ematopoietic {S}tem {C}ells {F}rom {L}iver to {R}educe {F}ibrosis}},
      journal = {Gastroenterology},
      year = {2017},
      volume = {153},
      number = {1},
      pages = {233--248},
      month = jul
    }
  2. Galectin-3, histone deacetylases, and Hedgehog signaling: Possible convergent targets in schistosomiasis-induced liver fibrosis
    F. L. Oliveira, K. Carneiro, J. M. Brito, M. Cabanel, J. X. Pereira, L. A. Paiva, W. Syn, N. C. Henderson, and M. C. El-Cheikh
    PLoS Negl Trop Dis Feb 2017

    Schistosomiasis affects approximately 240 million people in the world. Schistosoma mansoni eggs in the liver induce periportal fibrosis and hepatic failure driven by monocyte recruitment and macrophage activation, resulting in robust Th2 response. Here, we suggested a possible involvement of Galectin-3 (Gal-3), histone deacetylases (HDACs), and Hedgehog (Hh) signaling with macrophage activation during Th1/Th2 immune responses, fibrogranuloma reaction, and tissue repair during schistosomiasis. Gal-3 is highly expressed by liver macrophages (Kupffer cells) around Schistosoma eggs. HDACs and Hh regulate macrophage polarization and hepatic stellate cell activation during schistosomiasis-associated fibrogenesis. Previously, we demonstrated an abnormal extracellular matrix distribution in the liver that correlated with atypical monocyte-macrophage differentiation in S. mansoni-infected, Gal-3-deficient (Lgals3-/-) mice. New findings explored in this review focus on the chronic phase, when wild-type (Lgals3+/+) and Lgals3-/- mice were analyzed 90 days after cercariae infection. In Lgals3-/- infected mice, there was significant inflammatory infiltration with myeloid cells associated with egg destruction (hematoxylin and eosin staining), phagocytes (specifically Kupffer cells), numerically reduced and diffuse matrix extracellular deposition in fibrotic areas (Gomori trichrome staining), and severe disorganization of collagen fibers surrounding the S. mansoni eggs (reticulin staining). Granuloma-derived stromal cells (GR cells) of Lgals3-/- infected mice expressed lower levels of alpha smooth muscle actin (α-SMA) and eotaxin and higher levels of IL-4 than Lgals3+/+ mice (real-time PCR). The relevant participation of macrophages in these events led us to suggest distinct mechanisms of activation that culminate in defective fibrosis in the liver of Lgals3-/- infected mice. These aspects were discussed in this review, as well as the possible interference between Gal-3, HDACs, and Hh signaling during progressive liver fibrosis in S. mansoni-infected mice. Further studies focused on macrophage roles could elucidate these questions and clear the potential utility of these molecules as antifibrotic targets.

    @article{pmid28231240,
      author = {de Oliveira, F. L. and Carneiro, K. and Brito, J. M. and Cabanel, M. and Pereira, J. X. and Paiva, L. A. and Syn, W. and Henderson, N. C. and El-Cheikh, M. C.},
      title = {{{G}alectin-3, histone deacetylases, and {H}edgehog signaling: {P}ossible convergent targets in schistosomiasis-induced liver fibrosis}},
      journal = {PLoS Negl Trop Dis},
      year = {2017},
      volume = {11},
      number = {2},
      pages = {e0005137},
      month = feb
    }
  3. Longitudinal in vivo bioimaging of hepatocyte transcription factor activity following cholestatic liver injury in mice
    J. M. Delhove, S. M. Buckley, D. P. Perocheau, R. Karda, P. Arbuthnot, N. C. Henderson, S. N. Waddington, and T. R. McKay
    Sci Rep Feb 2017

    Molecular mechanisms regulating liver repair following cholestatic injury remain largely unknown. We have combined a mouse model of acute cholestatic liver injury, partial bile duct ligation (pBDL), with a novel longitudinal bioimaging methodology to quantify transcription factor activity during hepatic injury and repair. We administered lentiviral transcription factor activated luciferase/eGFP reporter (TFAR) cassettes to neonatal mice enabling longitudinal TFAR profiling by continued bioimaging throughout the lives of the animals and following pBDL in adulthood. Neonatal intravascular injection of VSV-G pseudotyped lentivirus resulted in almost exclusive transduction of hepatocytes allowing analysis of hepatocyte-specific transcription factor activity. We recorded acute but transient responses with NF-κB and Smad2/3 TFAR whilst our Notch reporter was repressed over the 40 days of evaluation post-pBDL. The bipotent hepatic progenitor cell line, HepaRG, can be directed to differentiate into hepatocytes and biliary epithelia. We found that forced expression of the Notch inhibitor NUMB in HepaRG resulted in enhanced hepatocyte differentiation and proliferation whereas over-expressing the Notch agonist JAG1 resulted in biliary epithelial differentiation. In conclusion, our data demonstrates that hepatocytes rapidly upregulate NF-κB and Smad2/3 activity, whilst repressing Notch signalling. This transcriptional response to cholestatic liver injury likely promotes partial de-differentiation to allow pro-regenerative proliferation of hepatocytes.

    @article{pmid28157201,
      author = {Delhove, J. M. and Buckley, S. M. and Perocheau, D. P. and Karda, R. and Arbuthnot, P. and Henderson, N. C. and Waddington, S. N. and McKay, T. R.},
      title = {{{L}ongitudinal in vivo bioimaging of hepatocyte transcription factor activity following cholestatic liver injury in mice}},
      journal = {Sci Rep},
      year = {2017},
      volume = {7},
      pages = {41874},
      month = feb
    }
  4. Low-dose acetaminophen induces early disruption of cell-cell tight junctions in human hepatic cells and mouse liver
    W. Gamal, P. Treskes, K. Samuel, G. J. Sullivan, R. Siller, V. Srsen, K. Morgan, A. Bryans, A. Kozlowska, A. Koulovasilopoulos, I. Underwood, S. Smith, J. Del-Pozo, S. Moss, A. I. Thompson, N. C. Henderson, P. C. Hayes, J. N. Plevris, P. O. Bagnaninchi, and L. J. Nelson
    Sci Rep Jan 2017

    Dysfunction of cell-cell tight junction (TJ) adhesions is a major feature in the pathogenesis of various diseases. Liver TJs preserve cellular polarity by delimiting functional bile-canalicular structures, forming the blood-biliary barrier. In acetaminophen-hepatotoxicity, the mechanism by which tissue cohesion and polarity are affected remains unclear. Here, we demonstrate that acetaminophen, even at low-dose, disrupts the integrity of TJ and cell-matrix adhesions, with indicators of cellular stress with liver injury in the human hepatic HepaRG cell line, and primary hepatocytes. In mouse liver, at human-equivalence (therapeutic) doses, dose-dependent loss of intercellular hepatic TJ-associated ZO-1 protein expression was evident with progressive clinical signs of liver injury. Temporal, dose-dependent and specific disruption of the TJ-associated ZO-1 and cytoskeletal-F-actin proteins, correlated with modulation of hepatic ultrastructure. Real-time impedance biosensing verified in vitro early, dose-dependent quantitative decreases in TJ and cell-substrate adhesions. Whereas treatment with NAPQI, the reactive metabolite of acetaminophen, or the PKCα-activator and TJ-disruptor phorbol-12-myristate-13-acetate, similarly reduced TJ integrity, which may implicate oxidative stress and the PKC pathway in TJ destabilization. These findings are relevant to the clinical presentation of acetaminophen-hepatotoxicity and may inform future mechanistic studies to identify specific molecular targets and pathways that may be altered in acetaminophen-induced hepatic depolarization.

    @article{pmid28134251,
      author = {Gamal, W. and Treskes, P. and Samuel, K. and Sullivan, G. J. and Siller, R. and Srsen, V. and Morgan, K. and Bryans, A. and Kozlowska, A. and Koulovasilopoulos, A. and Underwood, I. and Smith, S. and Del-Pozo, J. and Moss, S. and Thompson, A. I. and Henderson, N. C. and Hayes, P. C. and Plevris, J. N. and Bagnaninchi, P. O. and Nelson, L. J.},
      title = {{{L}ow-dose acetaminophen induces early disruption of cell-cell tight junctions in human hepatic cells and mouse liver}},
      journal = {Sci Rep},
      year = {2017},
      volume = {7},
      pages = {37541},
      month = jan
    }

2016

  1. Antifibrotics in chronic liver disease: tractable targets and translational challenges
    P. Ramachandran, and N. C. Henderson
    Lancet Gastroenterol Hepatol Dec 2016

    Chronic liver disease prevalence is increasing globally. Iterative liver damage, secondary to any cause of liver injury, results in progressive fibrosis, disrupted hepatic architecture, and aberrant regeneration, which are defining characteristics of liver cirrhosis. Liver transplantation is an effective treatment for end-stage liver disease; however, demand greatly outweighs donor organ supply, and in many parts of the world liver transplantation is unavailable. Hence, effective antifibrotic therapies are urgently required. In the past decade, rapid progress has been made in our understanding of the pathophysiology of liver fibrosis and a large number of potential cellular and molecular antifibrotic targets have been identified. This has led to numerous clinical trials of antifibrotic agents in patients with chronic liver disease. However, none of these have resulted in a robust and reproducible effect on fibrosis. It is therefore imperative that the ongoing translational challenges are addressed, to convert scientific discoveries into potent antifibrotics and enable bridging of the translational gap between putative therapeutic targets and effective treatments for patients with chronic liver disease.

    @article{pmid28404203,
      author = {Ramachandran, P. and Henderson, N. C.},
      title = {{{A}ntifibrotics in chronic liver disease: tractable targets and translational challenges}},
      journal = {Lancet Gastroenterol Hepatol},
      year = {2016},
      volume = {1},
      number = {4},
      pages = {328--340},
      month = dec
    }
  2. Mesenchymal stromal cells and liver fibrosis: a complicated relationship
    D. Haldar, N. C. Henderson, G. Hirschfield, and P. N. Newsome
    FASEB J Dec 2016

    Mesenchymal stromal cell (MSC) therapy demands the attention of clinicians and scientists because of its potential in clinical fields that are bereft of medical options, but also because of the controversies that underlie its mode of action. MSCs are potent immune modulators, yet their biologic activity may not be innate, requiring licensing by their microenvironment. This property has prompted researchers to explore unique ways in which MSCs may be able to exert distinct biologic effects in different pathologic settings. More than 400 clinical trials have investigated the therapeutic capacity of MSCs in different pathologies, including liver disease. Along with their anti-inflammatory action, there are data to suggest that MSCs may exert direct antifibrotic effects, although enthusiasm for their use in patients has been tempered by concerns of a possible profibrotic role of endogenous MSCs in response to injury. There is a significant need for antifibrotic therapy to combat the increasing burden of patients with cirrhosis, and a concerted effort is required to determine the mechanisms by which MSCs modulate the liver’s response to injury, both endogenously and after adoptive transfer. This review critically appraises the preclinical published data with regard to the capacity of MSCs to influence fibrotic response to liver injury and will explore the potential mechanisms that underpin the reported beneficial effects of MSC therapy in the context of liver injury and fibrosis.-Haldar, D., Henderson, N. C., Hirschfield, G., Newsome, P. N. Mesenchymal stromal cells and liver fibrosis: a complicated relationship.

    @article{pmid27601441,
      author = {Haldar, D. and Henderson, N. C. and Hirschfield, G. and Newsome, P. N.},
      title = {{{M}esenchymal stromal cells and liver fibrosis: a complicated relationship}},
      journal = {FASEB J},
      year = {2016},
      volume = {30},
      number = {12},
      pages = {3905--3928},
      month = dec
    }
  3. PAK proteins and YAP-1 signalling downstream of integrin beta-1 in myofibroblasts promote liver fibrosis
    K. Martin, J. Pritchett, J. Llewellyn, A. F. Mullan, V. S. Athwal, R. Dobie, E. Harvey, L. Zeef, S. Farrow, C. Streuli, N. C. Henderson, S. L. Friedman, N. A. Hanley, and K. Piper Hanley
    Nat Commun Aug 2016

    Fibrosis due to extracellular matrix (ECM) secretion from myofibroblasts complicates many chronic liver diseases causing scarring and organ failure. Integrin-dependent interaction with scar ECM promotes pro-fibrotic features. However, the pathological intracellular mechanism in liver myofibroblasts is not completely understood, and further insight could enable therapeutic efforts to reverse fibrosis. Here, we show that integrin beta-1, capable of binding integrin alpha-11, regulates the pro-fibrotic phenotype of myofibroblasts. Integrin beta-1 expression is upregulated in pro-fibrotic myofibroblasts in vivo and is required in vitro for production of fibrotic ECM components, myofibroblast proliferation, migration and contraction. Serine/threonine-protein kinase proteins, also known as P21-activated kinase (PAK), and the mechanosensitive factor, Yes-associated protein 1 (YAP-1) are core mediators of pro-fibrotic integrin beta-1 signalling, with YAP-1 capable of perpetuating integrin beta-1 expression. Pharmacological inhibition of either pathway in vivo attenuates liver fibrosis. PAK protein inhibition, in particular, markedly inactivates the pro-fibrotic myofibroblast phenotype, limits scarring from different hepatic insults and represents a new tractable therapeutic target for treating liver fibrosis.

    @article{pmid27535340,
      author = {Martin, K. and Pritchett, J. and Llewellyn, J. and Mullan, A. F. and Athwal, V. S. and Dobie, R. and Harvey, E. and Zeef, L. and Farrow, S. and Streuli, C. and Henderson, N. C. and Friedman, S. L. and Hanley, N. A. and Piper Hanley, K.},
      title = {{{P}{A}{K} proteins and {Y}{A}{P}-1 signalling downstream of integrin beta-1 in myofibroblasts promote liver fibrosis}},
      journal = {Nat Commun},
      year = {2016},
      volume = {7},
      pages = {12502},
      month = aug
    }
  4. Homing in on the hepatic scar: recent advances in cell-specific targeting of liver fibrosis
    R. Dobie, and N. C. Henderson
    F1000Res Aug 2016

    Despite the high prevalence of liver disease globally, there are currently no approved anti-fibrotic therapies to treat patients with liver fibrosis. A major goal in anti-fibrotic therapy is the development of drug delivery systems that allow direct targeting of the major pro-scarring cell populations within the liver (hepatic myofibroblasts) whilst not perturbing the homeostatic functions of other mesenchymal cell types present within both the liver and other organ systems. In this review we will outline some of the recent advances in our understanding of myofibroblast biology, discussing both the origin of myofibroblasts and possible myofibroblast fates during hepatic fibrosis progression and resolution. We will then discuss the various strategies currently being employed to increase the precision with which we deliver potential anti-fibrotic therapies to patients with liver fibrosis.

    @article{pmid27508067,
      author = {Dobie, R. and Henderson, N. C.},
      title = {{{H}oming in on the hepatic scar: recent advances in cell-specific targeting of liver fibrosis}},
      journal = {F1000Res},
      year = {2016},
      volume = {5}
    }
  5. Recent progress on targeting the αvβ1 integrin for the treatment of tissue fibrosis
    J. R. Smith, and N. C. Henderson
    Expert Opin Drug Discov Aug 2016
    @article{pmid27216763,
      author = {Smith, J. R. and Henderson, N. C.},
      title = {{{R}ecent progress on targeting the αvβ1 integrin for the treatment of tissue fibrosis}},
      journal = {Expert Opin Drug Discov},
      year = {2016},
      volume = {11},
      number = {8},
      pages = {749--751},
      month = aug
    }
  6. αv integrins: key regulators of tissue fibrosis
    K. P. Conroy, L. J. Kitto, and N. C. Henderson
    Cell Tissue Res Sep 2016

    Chronic tissue injury with fibrosis results in the disruption of tissue architecture, organ dysfunction and eventual organ failure. Therefore, the development of effective anti-fibrotic therapies is urgently required. During fibrogenesis, complex interplay occurs between cellular and extracellular matrix components of the wound healing response. Integrins, a family of transmembrane cell adhesion molecules, play a key role in mediating intercellular and cell-matrix interactions. Thus, integrins provide a major node of communication between the extracellular matrix, inflammatory cells, fibroblasts and parenchymal cells and, as such, are intimately involved in the initiation, maintenance and resolution of tissue fibrosis. Modulation of members of the αv integrin family has exhibited profound effects on fibrosis in multiple organs and disease states. In this review, we discuss the current knowledge of the mechanisms of αv-integrin-mediated regulation of fibrogenesis and show that the therapeutic targeting of specific αv integrins represents a promising avenue to treat patients with a broad range of fibrotic diseases.

    @article{pmid27139180,
      author = {Conroy, K. P. and Kitto, L. J. and Henderson, N. C.},
      title = {{αv integrins: key regulators of tissue fibrosis}},
      journal = {Cell Tissue Res},
      year = {2016},
      volume = {365},
      number = {3},
      pages = {511--519},
      month = sep
    }
  7. CD248/endosialin critically regulates hepatic stellate cell proliferation during chronic liver injury via a PDGF-regulated mechanism
    A. Wilhelm, V. Aldridge, D. Haldar, A. J. Naylor, C. J. Weston, D. Hedegaard, A. Garg, J. Fear, G. M. Reynolds, A. P. Croft, N. C. Henderson, C. D. Buckley, and P. N. Newsome
    Gut Jul 2016

    CD248 (endosialin) is a stromal cell marker expressed on fibroblasts and pericytes. During liver injury, myofibroblasts are the main source of fibrotic matrix.\{To determine the role of CD248 in the development of liver fibrosis in the rodent and human setting.\ CD248 expression was studied by immunostaining and quantitative PCR in both normal and diseased human and murine liver tissue and isolated hepatic stellate cells (HSCs). Hepatic fibrosis was induced in CD248(-/-) and wild-type controls with carbon tetrachloride (CCl4) treatment.\{Expression of CD248 was seen in normal liver of humans and mice but was significantly increased in liver injury using both immunostaining and gene expression assays. CD248 was co-expressed with a range of fibroblast/HSC markers including desmin, vimentin and α-smooth muscle actin (α-SMA) in murine and human liver sections. CD248 expression was restricted to isolated primary murine and human HSC. Collagen deposition and α-SMA expression, but not inflammation and neoangiogenesis, was reduced in CD248(-/-) mice compared with wild-type mice after CCl4 treatment. Isolated HSC from wild-type and CD248(-/-) mice expressed platelet-derived growth factor receptor α (PDGFR-α) and PDGFR-β at similar levels. As expected, PDGF-BB stimulation induced proliferation of wild-type HSC, whereas CD248(-/-) HSC did not demonstrate a proliferative response to PDGF-BB. Abrogated PDGF signalling in CD248(-/-) HSC was confirmed by significantly reduced c-fos expression in CD248(-/-) HSC compared with wild-type HSC.\{Our data show that deletion of CD248 reduces susceptibility to liver fibrosis via an effect on PDGF signalling, making it an attractive clinical target for the treatment of liver injury.

    @article{pmid26078290,
      author = {Wilhelm, A. and Aldridge, V. and Haldar, D. and Naylor, A. J. and Weston, C. J. and Hedegaard, D. and Garg, A. and Fear, J. and Reynolds, G. M. and Croft, A. P. and Henderson, N. C. and Buckley, C. D. and Newsome, P. N.},
      title = {{{C}{D}248/endosialin critically regulates hepatic stellate cell proliferation during chronic liver injury via a {P}{D}{G}{F}-regulated mechanism}},
      journal = {Gut},
      year = {2016},
      volume = {65},
      number = {7},
      pages = {1175--1185},
      month = jul
    }

2015

  1. Acute Liver Injury Is Independent of B Cells or Immunoglobulin M
    J. A. Richards, M. Bucsaiova, E. E. Hesketh, C. Ventre, N. C. Henderson, K. Simpson, C. O. Bellamy, S. E. Howie, S. M. Anderton, J. Hughes, and S. J. Wigmore
    PLoS One Jul 2015

    Acute liver injury is a clinically important pathology and results in the release of Danger Associated Molecular Patterns, which initiate an immune response. Withdrawal of the injurious agent and curtailing any pathogenic secondary immune response may allow spontaneous resolution of injury. The role B cells and Immunoglobulin M (IgM) play in acute liver injury is largely unknown and it was proposed that B cells and/or IgM would play a significant role in its pathogenesis.\{Tissue from 3 models of experimental liver injury (ischemia-reperfusion injury, concanavalin A hepatitis and paracetamol-induced liver injury) and patients transplanted following paracetamol overdose were stained for evidence of IgM deposition. Mice deficient in B cells (and IgM) were used to dissect out the role B cells and/or IgM played in the development or resolution of injury. Serum transfer into mice lacking IgM was used to establish the role IgM plays in injury.\ 0.001), but not B cell deficient (μMT) mice (p = 0.93), were significantly protected from injury. Further interrogation with T cell deficient (CD3εKO) mice confirmed that the T cell component is a key mediator of sterile liver injury. Mice deficient in B cells and IgM mice did not have a significant delay in resolution following acute liver injury.\{IgM deposition appears to be common feature of both human and murine sterile liver injury. However, neither IgM nor B cells, play a significant role in the development of or resolution from acute liver injury. T cells appear to be key mediators of injury. In conclusion, the therapeutic targeting of IgM or B cells (e.g. with Rituximab) would have limited benefit in protecting patients from acute liver injury.

    @article{pmid26406765,
      author = {Richards, J. A. and Bucsaiova, M. and Hesketh, E. E. and Ventre, C. and Henderson, N. C. and Simpson, K. and Bellamy, C. O. and Howie, S. E. and Anderton, S. M. and Hughes, J. and Wigmore, S. J.},
      title = {{{A}cute {L}iver {I}njury {I}s {I}ndependent of {B} {C}ells or {I}mmunoglobulin {M}}},
      journal = {PLoS One},
      year = {2015},
      volume = {10},
      number = {9},
      pages = {e0138688}
    }
  2. Hepatic stellate cells: central modulators of hepatic carcinogenesis
    A. I. Thompson, K. P. Conroy, and N. C. Henderson
    BMC Gastroenterol May 2015

    Hepatocellular carcinoma (HCC) represents the second most common cause of cancer-related death worldwide, and is increasing in incidence. Currently, our therapeutic repertoire for the treatment of HCC is severely limited, and therefore effective new therapies are urgently required. Recently, there has been increasing interest focusing on the cellular and molecular interactions between cancer cells and their microenvironment. HCC represents a unique opportunity to study the relationship between a diseased stroma and promotion of carcinogenesis, as 90% of HCCs arise in a cirrhotic liver. Hepatic stellate cells (HSC) are the major source of extracellular proteins during fibrogenesis, and may directly, or via secreted products, contribute to tumour initiation and progression. In this review we explore the complex cellular and molecular interplay between HSC biology and hepatocarcinogenesis. We focus on the molecular mechanisms by which HSC modulate HCC growth, immune cell evasion and angiogenesis. This is followed by a discussion of recent progress in the field in understanding the mechanistic crosstalk between HSC and HCC, and the pathways that are potentially amenable to therapeutic intervention. Furthermore, we summarise the exciting recent developments in strategies to target HSC specifically, and novel techniques to deliver pharmaceutical agents directly to HSC, potentially allowing tailored, cell-specific therapy for HCC.

    @article{pmid26013123,
      author = {Thompson, A. I. and Conroy, K. P. and Henderson, N. C.},
      title = {{{H}epatic stellate cells: central modulators of hepatic carcinogenesis}},
      journal = {BMC Gastroenterol},
      year = {2015},
      volume = {15},
      pages = {63},
      month = may
    }
  3. Cre-ativity in the liver: transgenic approaches to targeting hepatic nonparenchymal cells
    S. N. Greenhalgh, K. P. Conroy, and N. C. Henderson
    Hepatology Jun 2015

    Rapid evolution in transgenic (Tg) mouse technology now permits cell-specific and temporal control of fluorescent cell-labeling and gene inactivation. Here, we discuss the principal strategies that have been utilized to target, label, and manipulate hepatic nonparenchymal cells, with emphasis on the utility of constitutive and inducible Cre-lox systems. We summarize key findings of studies employing Tg technology to target hepatic stellate cells, myofibroblasts, liver sinusoidal endothelial cells, and macrophages to illustrate the power of these approaches in identifying cell-specific molecular mechanisms critical to the pathophysiology of liver disease. Increasing adoption of Tg techniques will help to answer fundamental questions regarding the pathogenesis of hepatic diseases and provide the mechanistic rationale to allow identification of novel drug targets, ultimately translating into effective therapies for patients with liver disease.

    @article{pmid25412828,
      author = {Greenhalgh, S. N. and Conroy, K. P. and Henderson, N. C.},
      title = {{{C}re-ativity in the liver: transgenic approaches to targeting hepatic nonparenchymal cells}},
      journal = {Hepatology},
      year = {2015},
      volume = {61},
      number = {6},
      pages = {2091--2099},
      month = jun
    }
  4. Galectin-3 regulates hepatic progenitor cell expansion during liver injury
    W. C. Hsieh, A. C. Mackinnon, W. Y. Lu, J. Jung, L. Boulter, N. C. Henderson, K. J. Simpson, B. Schotanus, D. Wojtacha, T. G. Bird, C. N. Medine, D. C. Hay, T. Sethi, J. P. Iredale, and S. J. Forbes
    Gut Feb 2015

    Following chronic liver injury or when hepatocyte proliferation is impaired, ductular reactions containing hepatic progenitor cells (HPCs) appear in the periportal regions and can regenerate the liver parenchyma. HPCs exist in a niche composed of myofibroblasts, macrophages and laminin matrix. Galectin-3 (Gal-3) is a β-galactoside-binding lectin that binds to laminin and is expressed in injured liver in mice and humans.\{We examined the role of Gal-3 in HPC activation. HPC activation was studied following dietary induced hepatocellular (choline-deficient ethionine-supplemented diet) and biliary (3,5-diethoxycarbonyl-1,4-dihydrocollidine supplemented diet) injury in wild type and Gal-3(-/-) mice.\{HPC proliferation was significantly reduced in Gal-3(-/-) mice. Gal-3(-/-) mice failed to form a HPC niche, with reduced laminin formation. HPCs isolated from wild type mice secrete Gal-3 which enhanced adhesion and proliferation of HPCs on laminin in an undifferentiated form. These effects were attenuated in Gal3(-/-) HPCs and in wild type HPCs treated with the Gal-3 inhibitor lactose. Gal-3(-/-) HPCs in vitro showed increased hepatocyte function and prematurely upregulated both biliary and hepatocyte differentiation markers and regulated cell cycle genes leading to arrest in G0/G1.\{We conclude that Gal-3 is required for the undifferentiated expansion of HPCs in their niche in injured liver.

    @article{pmid24837171,
      author = {Hsieh, W. C. and Mackinnon, A. C. and Lu, W. Y. and Jung, J. and Boulter, L. and Henderson, N. C. and Simpson, K. J. and Schotanus, B. and Wojtacha, D. and Bird, T. G. and Medine, C. N. and Hay, D. C. and Sethi, T. and Iredale, J. P. and Forbes, S. J.},
      title = {{{G}alectin-3 regulates hepatic progenitor cell expansion during liver injury}},
      journal = {Gut},
      year = {2015},
      volume = {64},
      number = {2},
      pages = {312--321},
      month = feb
    }

2013

  1. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs
    N. C. Henderson, T. D. Arnold, Y. Katamura, M. M. Giacomini, J. D. Rodriguez, J. H. McCarty, A. Pellicoro, E. Raschperger, C. Betsholtz, P. G. Ruminski, D. W. Griggs, M. J. Prinsen, J. J. Maher, J. P. Iredale, A. Lacy-Hulbert, R. H. Adams, and D. Sheppard
    Nat Med Dec 2013

    Myofibroblasts are the major source of extracellular matrix components that accumulate during tissue fibrosis, and hepatic stellate cells (HSCs) are believed to be the major source of myofibroblasts in the liver. To date, robust systems to genetically manipulate these cells have not been developed. We report that Cre under control of the promoter of Pdgfrb (Pdgfrb-Cre) inactivates loxP-flanked genes in mouse HSCs with high efficiency. We used this system to delete the gene encoding α(v) integrin subunit because various α(v)-containing integrins have been suggested as central mediators of fibrosis in multiple organs. Such depletion protected mice from carbon tetrachloride-induced hepatic fibrosis, whereas global loss of β₃, β₅ or β₆ integrins or conditional loss of β₈ integrins in HSCs did not. We also found that Pdgfrb-Cre effectively targeted myofibroblasts in multiple organs, and depletion of the α(v) integrin subunit using this system was protective in other models of organ fibrosis, including pulmonary and renal fibrosis. Pharmacological blockade of α(v)-containing integrins by a small molecule (CWHM 12) attenuated both liver and lung fibrosis, including in a therapeutic manner. These data identify a core pathway that regulates fibrosis and suggest that pharmacological targeting of all α(v) integrins may have clinical utility in the treatment of patients with a broad range of fibrotic diseases.

    @article{pmid24216753,
      author = {Henderson, N. C. and Arnold, T. D. and Katamura, Y. and Giacomini, M. M. and Rodriguez, J. D. and McCarty, J. H. and Pellicoro, A. and Raschperger, E. and Betsholtz, C. and Ruminski, P. G. and Griggs, D. W. and Prinsen, M. J. and Maher, J. J. and Iredale, J. P. and Lacy-Hulbert, A. and Adams, R. H. and Sheppard, D.},
      title = {{{T}argeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs}},
      journal = {Nat Med},
      year = {2013},
      volume = {19},
      number = {12},
      pages = {1617--1624},
      month = dec
    }
  2. Origins of fibrosis: pericytes take centre stage
    S. N. Greenhalgh, J. P. Iredale, and N. C. Henderson
    F1000Prime Rep Sep 2013

    Pericytes are ubiquitous perivascular cells that have recently attracted interest as potential myofibroblast precursors. In turn, myofibroblasts are the major source of extracellular matrix components that accumulate during tissue fibrosis. Given the worldwide burden of fibrotic disease and paucity of therapeutic options available to halt its progression, elucidating the origins of myofibroblasts is of prime importance. The advent of genetic strategies that permit fate-mapping of specific cell populations through permanent and heritable expression of reporter proteins has begun to shed light on the source of the fibrogenic myofibroblast. Here we discuss recent studies in multiple organs that highlight the central role of pericytes in the origins of fibrosis.

    @article{pmid24049641,
      author = {Greenhalgh, S. N. and Iredale, J. P. and Henderson, N. C.},
      title = {{{O}rigins of fibrosis: pericytes take centre stage}},
      journal = {F1000Prime Rep},
      year = {2013},
      volume = {5},
      pages = {37},
      month = sep
    }
  3. Eosinophils secrete IL-4 to facilitate liver regeneration
    Y. P. Goh, N. C. Henderson, J. E. Heredia, A. Red Eagle, J. I. Odegaard, N. Lehwald, K. D. Nguyen, D. Sheppard, L. Mukundan, R. M. Locksley, and A. Chawla
    Proc Natl Acad Sci U S A Jun 2013

    The liver is a central organ for the synthesis and storage of nutrients, production of serum proteins and hormones, and breakdown of toxins and metabolites. Because the liver is susceptible to toxin- or pathogen-mediated injury, it maintains a remarkable capacity to regenerate by compensatory growth. Specifically, in response to injury, quiescent hepatocytes enter the cell cycle and undergo DNA replication to promote liver regrowth. Despite the elucidation of a number of regenerative factors, the mechanisms by which liver injury triggers hepatocyte proliferation are incompletely understood. We demonstrate here that eosinophils stimulate liver regeneration after partial hepatectomy and toxin-mediated injury. Liver injury results in rapid recruitment of eosinophils, which secrete IL-4 to promote the proliferation of quiescent hepatocytes. Surprisingly, signaling via the IL-4Rα in macrophages, which have been implicated in tissue repair, is dispensable for hepatocyte proliferation and liver regrowth after injury. Instead, IL-4 exerts its proliferative actions via IL-4Rα in hepatocytes. Our findings thus provide a unique mechanism by which eosinophil-derived IL-4 stimulates hepatocyte proliferation in regenerating liver.

    @article{pmid23716700,
      author = {Goh, Y. P. and Henderson, N. C. and Heredia, J. E. and Red Eagle, A. and Odegaard, J. I. and Lehwald, N. and Nguyen, K. D. and Sheppard, D. and Mukundan, L. and Locksley, R. M. and Chawla, A.},
      title = {{{E}osinophils secrete {I}{L}-4 to facilitate liver regeneration}},
      journal = {Proc Natl Acad Sci U S A},
      year = {2013},
      volume = {110},
      number = {24},
      pages = {9914--9919},
      month = jun
    }
  4. Extracellular matrix degradation in liver fibrosis: Biochemistry and regulation
    J. P. Iredale, A. Thompson, and N. C. Henderson
    Biochim Biophys Acta Jul 2013

    Fibrosis is a highly conserved wound healing response and represents the final common pathway of virtually all chronic inflammatory injuries. Over the past 3 decades detailed analysis of hepatic extracellular matrix synthesis and degradation using approaches incorporating human disease, experimental animal models and cell culture have highlighted the extraordinarily dynamic nature of tissue repair and remodelling in this solid organ. Furthermore emerging studies of fibrosis in other organs demonstrate that basic common mechanisms exist, suggesting that bidirectionality of the fibrotic process may not solely be the preserve of the liver. In this review we will examine the cellular and molecular mechanisms that govern extracellular matrix degradation and fibrosis resolution, and highlight how manipulation of these processes may result in the development of effective anti-fibrotic therapies. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.

    @article{pmid23149387,
      author = {Iredale, J. P. and Thompson, A. and Henderson, N. C.},
      title = {{{E}xtracellular matrix degradation in liver fibrosis: {B}iochemistry and regulation}},
      journal = {Biochim Biophys Acta},
      year = {2013},
      volume = {1832},
      number = {7},
      pages = {876--883},
      month = jul
    }
  5. Integrin-mediated regulation of TGFβ in fibrosis
    N. C. Henderson, and D. Sheppard
    Biochim Biophys Acta Jul 2013

    Fibrosis is a major cause of morbidity and mortality worldwide. Currently, therapeutic options for tissue fibrosis are severely limited, and organ transplantation is the only effective treatment for end-stage fibrotic disease. However, demand for donor organs greatly outstrips supply, and so effective anti-fibrotic treatments are urgently required. In recent years, the integrin family of cell adhesion receptors has gained prominence as key regulators of chronic inflammation and fibrosis. Fibrosis models in multiple organs have demonstrated that integrins have profound effects on the fibrotic process. There is now abundant in vivo data demonstrating critical regulatory roles for integrins expressed on different cell types during tissue fibrogenesis. In this review, we will examine the ways in which integrins regulate these processes and discuss how the manipulation of integrins using function blocking antibodies and small molecule inhibitors may have clinical utility in the treatment of patients with a broad range of fibrotic diseases. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.

    @article{pmid23046811,
      author = {Henderson, N. C. and Sheppard, D.},
      title = {{{I}ntegrin-mediated regulation of {T}{G}{F}β in fibrosis}},
      journal = {Biochim Biophys Acta},
      year = {2013},
      volume = {1832},
      number = {7},
      pages = {891--896},
      month = jul
    }

2012

  1. Standing down the guard: stellate cells leave quietly
    N. C. Henderson, and J. P. Iredale
    Gastroenterology Oct 2012
    @article{pmid22917861,
      author = {Henderson, N. C. and Iredale, J. P.},
      title = {{{S}tanding down the guard: stellate cells leave quietly}},
      journal = {Gastroenterology},
      year = {2012},
      volume = {143},
      number = {4},
      pages = {890--892},
      month = oct
    }

2011

  1. Impaired gluconeogenesis in a porcine model of paracetamol induced acute liver failure
    K. J. Dabos, H. R. Whalen, P. N. Newsome, J. A. Parkinson, N. C. Henderson, I. H. Sadler, P. C. Hayes, and J. N. Plevris
    World J Gastroenterol Mar 2011

    To investigate glucose homeostasis and in particular gluconeogenesis in a large animal model of acute liver failure (ALF).\{Six pigs with paracetamol induced ALF under general anaesthesia were studied over 25 h. Plasma samples were withdrawn every five hours from a central vein. Three animals were used as controls and were maintained under anaesthesia only. Using (1)H NMR spectroscopy we identified most gluconeogenic amino acids along with lactate and pyruvate in the animal plasma samples.\ 0.05).\{Our pig model of ALF is characterized by an altered gluconeogenetic capacity, an impaired tricarboxylic acid (TCA) cycle and a glycolytic state.

    @article{pmid21472104,
      author = {Dabos, K. J. and Whalen, H. R. and Newsome, P. N. and Parkinson, J. A. and Henderson, N. C. and Sadler, I. H. and Hayes, P. C. and Plevris, J. N.},
      title = {{{I}mpaired gluconeogenesis in a porcine model of paracetamol induced acute liver failure}},
      journal = {World J Gastroenterol},
      year = {2011},
      volume = {17},
      number = {11},
      pages = {1457--1461},
      month = mar
    }

2010

  1. Development of an invasively monitored porcine model of acetaminophen-induced acute liver failure
    P. N. Newsome, N. C. Henderson, L. J. Nelson, C. Dabos, C. Filippi, C. Bellamy, F. Howie, R. E. Clutton, T. King, A. Lee, P. C. Hayes, and J. N. Plevris
    BMC Gastroenterol Mar 2010

    The development of effective therapies for acute liver failure (ALF) is limited by our knowledge of the pathophysiology of this condition, and the lack of suitable large animal models of acetaminophen toxicity. Our aim was to develop a reproducible invasively-monitored porcine model of acetaminophen-induced ALF.\ 35kg pigs were maintained under general anaesthesia and invasively monitored. Control pigs received a saline infusion, whereas ALF pigs received acetaminophen intravenously for 12 hours to maintain blood concentrations between 200-300 mg/l. Animals surviving 28 hours were euthanased.\ 5%. Intracranial hypertension was not seen (ICP monitoring), but there was biochemical evidence of encephalopathy by the reduction of Fischer’s ratio from 5.6 +/- 1.1 to 0.45 +/- 0.06.\{We have developed a reproducible large animal model of acetaminophen-induced liver failure, which allows in-depth investigation of the pathophysiological basis of this condition. Furthermore, this represents an important large animal model for testing artificial liver support systems.

    @article{pmid20353598,
      author = {Newsome, P. N. and Henderson, N. C. and Nelson, L. J. and Dabos, C. and Filippi, C. and Bellamy, C. and Howie, F. and Clutton, R. E. and King, T. and Lee, A. and Hayes, P. C. and Plevris, J. N.},
      title = {{{D}evelopment of an invasively monitored porcine model of acetaminophen-induced acute liver failure}},
      journal = {BMC Gastroenterol},
      year = {2010},
      volume = {10},
      pages = {34},
      month = mar
    }

2009

  1. c-Jun terminal kinase-2 gene deleted mice overexpress hemeoxygenase-1 and are protected from hepatic ischemia reperfusion injury
    L. Devey, E. Mohr, C. Bellamy, K. Simpson, N. Henderson, E. M. Harrison, J. A. Ross, and S. J. Wigmore
    Transplantation Aug 2009

    Targeted deletion of c-jun amino terminal kinase-2 (jnk-2) upregulates the activator protein-1 transcription factor system. We hypothesized that this would lead to induction of hemeoxygenase-1 (HO-1) and confer protection from hepatic ischemia reperfusion injury.\{Wild-type and jnk-2 -/- animals were subjected to hepatic ischemia reperfusion insults in two models: a total hepatic ischemia model involving timed Pringle maneuver, and a partial hepatic ischemia model involving selective occlusion of the portal pedicle supplying the left hepatic lobe. Optimal durations of injury were calibrated for each model. After 24 hr, animals were killed, and blood and tissues were collected for alanine aminotransferase, histologic injury scoring, and other analyses. Before total or partial hepatic ischemia reperfusion insults, some animals were subject to HO-1 inhibition with chromium mesoporphyrin IX or Kupffer cell depletion with liposomal clodronate. Bone marrow-derived monocytes were grown from hemopoietic progenitors taken from wild-type and jnk-2 -/- mice before stimulation with lipopolysaccharide and measurement of tumour necrosis factor-alpha production.\{Jnk-2 -/- animals were protected from hepatic ischemia reperfusion injury. HO-1 expression and activity was elevated in jnk-2 -/- animals (2.2-fold; P=0.006). Most HO-1 was expressed in Kupffer cells. Inhibition of HO-1 in jnk-2 -/- animals led to the loss of protection from ischemia. Depletion of Kupffer cells using liposomal clodronate led to loss of hepatic HO-1 expression and much more severe injury in wild-type and jnk-2 -/- animals. In vitro studies of cultured macrophages demonstrated reduced tumour necrosis factor-alpha secretion after lipopolysaccharide stimulus, an effect lost after HO-1 inhibition.

    @article{pmid19667931,
      author = {Devey, L. and Mohr, E. and Bellamy, C. and Simpson, K. and Henderson, N. and Harrison, E. M. and Ross, J. A. and Wigmore, S. J.},
      title = {{c-{J}un terminal kinase-2 gene deleted mice overexpress hemeoxygenase-1 and are protected from hepatic ischemia reperfusion injury}},
      journal = {Transplantation},
      year = {2009},
      volume = {88},
      number = {3},
      pages = {308--316},
      month = aug
    }
  2. The regulation of inflammation by galectin-3
    N. C. Henderson, and T. Sethi
    Immunol Rev Jul 2009

    Galectin-3 is a beta-galactoside-binding animal lectin of approximately 30 kDa and is evolutionarily highly conserved. Galectin-3 is promiscuous, its localization within the tissue micro-environment may be extracellular, cytoplasmic, or nuclear, and it has a concentration-dependent ability to be monomeric or form oligomers. These properties impart great flexibility on galectin-3 as a specific regulator of many biological systems including inflammation. For example, in acute tissue damage galectin-3 is a key component in the host defense against microbes such as Streptococcus pneumoniae. However, if tissue injury becomes repetitive galectin-3 also appears to be intimately involved in the transition to chronic inflammation, facilitating the walling off of tissue injury with fibrogenesis and organ scarring. Therefore galectin-3 can be viewed as a regulatory molecule acting at various stages along the continuum from acute inflammation to chronic inflammation and tissue fibrogenesis. In this review, we examine the role of galectin-3 in inflammation, and discuss the manipulation of galectin-3 expression as a potentially novel therapeutic strategy in the treatment of a broad range of inflammatory diseases.

    @article{pmid19594635,
      author = {Henderson, N. C. and Sethi, T.},
      title = {{{T}he regulation of inflammation by galectin-3}},
      journal = {Immunol Rev},
      year = {2009},
      volume = {230},
      number = {1},
      pages = {160--171},
      month = jul
    }
  3. The utilization of liver transplantation in the management of acute liver failure: comparison between acetaminophen and non-acetaminophen etiologies
    K. J. Simpson, C. M. Bates, N. C. Henderson, S. J. Wigmore, O. J. Garden, A. Lee, A. Pollok, G. Masterton, and P. C. Hayes
    Liver Transpl Jun 2009

    0.01). This was due to an increase in the number of patients with psychiatric contraindications to transplantation (predominantly resistant and severe alcohol dependence). In conclusion, at all decision steps between admission and emergency LT, LT is favored in non-acetaminophen patients, and nonoperative management is favored in acetaminophen ALF patients.

    @article{pmid19479803,
      author = {Simpson, K. J. and Bates, C. M. and Henderson, N. C. and Wigmore, S. J. and Garden, O. J. and Lee, A. and Pollok, A. and Masterton, G. and Hayes, P. C.},
      title = {{{T}he utilization of liver transplantation in the management of acute liver failure: comparison between acetaminophen and non-acetaminophen etiologies}},
      journal = {Liver Transpl},
      year = {2009},
      volume = {15},
      number = {6},
      pages = {600--609},
      month = jun
    }
  4. Gut homing receptors on CD8 T cells are retinoic acid dependent and not maintained by liver dendritic or stellate cells
    B. Eksteen, J. R. Mora, E. L. Haughton, N. C. Henderson, L. Lee-Turner, E. J. Villablanca, S. M. Curbishley, A. I. Aspinall, U. H. Andrian, and D. H. Adams
    Gastroenterology Jul 2009

    Lymphocytes primed by intestinal dendritic cells (DC) express the gut-homing receptors CCR9 and alpha4beta7, which recognize CCL25 and mucosal addressin cell-adhesion molecule-1 in the intestine promoting the development of regional immunity. In mice, imprinting of CCR9 and alpha4beta7 is dependent on retinoic acid during T-cell activation. Tissue specificity is lost in primary sclerosing cholangitis (PSC), an extraintestinal manifestation of inflammatory bowel disease, when ectopic expression of mucosal addressin cell-adhesion molecule-1 and CCL25 in the liver promotes recruitment of CCR9+alpha4beta7+ T cells to the liver. We investigated the processes that control enterohepatic T-cell migration and whether the ability to imprint CCR9 and alpha4beta7 is restricted to intestinal DCs or can under some circumstances be acquired by hepatic DCs in diseases such as PSC.\{Human and murine DCs from gut, liver, or portal lymph nodes and hepatic stellate cells were used to activate CD8 T cells. Imprinting of CCR9 and alpha4beta7 and functional migration responses were determined. Crossover activation protocols assessed plasticity of gut homing.\{Activation by gut DCs imprinted high levels of functional CCR9 and alpha4beta7 on naïve CD8 T cells, whereas hepatic DCs and stellate cells proved inferior. Imprinting was RA dependent and demonstrated plasticity.\{Imprinting and plasticity of gut-homing human CD8 T cells requires primary activation or reactivation by gut DCs and is retinoic acid dependent. The inability of liver DCs to imprint gut tropism implies that alpha4beta7+CCR9+ T cell that infiltrate the liver in PSC are primed in the gut.

    @article{pmid19233184,
      author = {Eksteen, B. and Mora, J. R. and Haughton, E. L. and Henderson, N. C. and Lee-Turner, L. and Villablanca, E. J. and Curbishley, S. M. and Aspinall, A. I. and von Andrian, U. H. and Adams, D. H.},
      title = {{{G}ut homing receptors on {C}{D}8 {T} cells are retinoic acid dependent and not maintained by liver dendritic or stellate cells}},
      journal = {Gastroenterology},
      year = {2009},
      volume = {137},
      number = {1},
      pages = {320--329},
      month = jul
    }

2008

  1. Hepatic fibrogenesis: from within and outwith
    N. C. Henderson, and S. J. Forbes
    Toxicology Dec 2008

    Liver disease is now the fifth commonest cause of death in the United Kingdom and the incidence is increasing. Chronic injury to the liver typically due to toxic insult, viral infection, immunological or metabolic diseases usually results in a stereotypical response with both parenchymal regeneration and wound healing. Chronic hepatic injury results in liver fibrosis with eventual progression to cirrhosis and end stage liver disease. At this point the majority of the clinical complications arise such as portal hypertension and the development of liver cancer. If the causative disease can be effectively treated the liver can regenerate and at the least partial resolution of liver fibrosis may occur. Unfortunately, unless the primary disease can be eradicated there are no specific anti-fibrotic treatments in routine clinical use. This highlights the urgent need to both increase our understanding of the mechanisms of hepatic fibrogenesis and to develop novel therapies to arrest or reverse the fibrotic process. This article initially outlines the main cellular pathway of fibrogenesis within the liver-the activation of the quiescent hepatic stellate cell into an activated myofibroblast phenotype, resulting in the production of fibrillar collagen. We will then discuss newly emerging sources of scar forming cells during hepatic injury together with the role of hepatic macrophages which have a regulatory function in both the formation and resolution of liver fibrosis.

    @article{pmid18824072,
      author = {Henderson, N. C. and Forbes, S. J.},
      title = {{{H}epatic fibrogenesis: from within and outwith}},
      journal = {Toxicology},
      year = {2008},
      volume = {254},
      number = {3},
      pages = {130--135},
      month = dec
    }
  2. Regulation of alternative macrophage activation by galectin-3
    A. C. MacKinnon, S. L. Farnworth, P. S. Hodkinson, N. C. Henderson, K. M. Atkinson, H. Leffler, U. J. Nilsson, C. Haslett, S. J. Forbes, and T. Sethi
    J Immunol Feb 2008

    Alternative macrophage activation is implicated in diverse disease pathologies such as asthma, organ fibrosis, and granulomatous diseases, but the mechanisms underlying macrophage programming are not fully understood. Galectin-3 is a carbohydrate-binding lectin present on macrophages. We show that disruption of the galectin-3 gene in 129sv mice specifically restrains IL-4/IL-13-induced alternative macrophage activation in bone marrow-derived macrophages in vitro and in resident lung and recruited peritoneal macrophages in vivo without affecting IFN-gamma/LPS-induced classical activation or IL-10-induced deactivation. IL-4-mediated alternative macrophage activation is inhibited by siRNA-targeted deletion of galectin-3 or its membrane receptor CD98 and by inhibition of PI3K. Increased galectin-3 expression and secretion is a feature of alternative macrophage activation. IL-4 stimulates galectin-3 expression and release in parallel with other phenotypic markers of alternative macrophage activation. By contrast, classical macrophage activation with LPS inhibits galectin-3 expression and release. Galectin-3 binds to CD98, and exogenous galectin-3 or cross-linking CD98 with the mAb 4F2 stimulates PI3K activation and alternative activation. IL-4-induced alternative activation is blocked by bis-(3-deoxy-3-(3-methoxybenzamido)-beta-D-galactopyranosyl) sulfane, a specific inhibitor of extracellular galectin-3 carbohydrate binding. These results demonstrate that a galectin-3 feedback loop drives alternative macrophage activation. Pharmacological modulation of galectin-3 function represents a novel therapeutic strategy in pathologies associated with alternatively activated macrophages.

    @article{pmid18250477,
      author = {MacKinnon, A. C. and Farnworth, S. L. and Hodkinson, P. S. and Henderson, N. C. and Atkinson, K. M. and Leffler, H. and Nilsson, U. J. and Haslett, C. and Forbes, S. J. and Sethi, T.},
      title = {{{R}egulation of alternative macrophage activation by galectin-3}},
      journal = {J Immunol},
      year = {2008},
      volume = {180},
      number = {4},
      pages = {2650--2658},
      month = feb
    }
  3. Galectin-3 reduces the severity of pneumococcal pneumonia by augmenting neutrophil function
    S. L. Farnworth, N. C. Henderson, A. C. Mackinnon, K. M. Atkinson, T. Wilkinson, K. Dhaliwal, K. Hayashi, A. J. Simpson, A. G. Rossi, C. Haslett, and T. Sethi
    Am J Pathol Feb 2008

    The Gram-positive Streptococcus pneumoniae is the leading cause of community-acquired pneumonia worldwide, resulting in high mortality. Our in vivo studies show that galectin-3(-/-) mice develop more severe pneumonia after infection with S. pneumoniae, as demonstrated by increased bacteremia and lung damage compared to wild-type mice and that galectin-3 reduces the severity of pneumococcal pneumonia in part by augmenting neutrophil function. Specifically, we show that 1) galectin-3 directly acts as a neutrophil-activating agent and potentiates the effect of fMLP, 2) exogenous galectin-3 augments neutrophil phagocytosis of bacteria and delays neutrophil apoptosis, 3) phagocytosis of apoptotic neutrophils by galectin-3(-/-) macrophages is less efficient compared to wild type, and 4) galectin-3 demonstrates bacteriostatic properties against S. pneumoniae in vitro. Furthermore, ad-back of recombinant galectin-3 in vivo protects galectin-3-deficient mice from developing severe pneumonia. Together, these results demonstrate that galectin-3 is a key molecule in the host defense against pneumococcal infection. Therapeutic strategies designed to augment galectin-3 activity may both enhance inflammatory cell function (by directly affecting neutrophil responsiveness and prolonging neutrophil longevity) and have direct bacteriostatic activity, improving clinical outcomes after severe pneumococcal infection.

    @article{pmid18202191,
      author = {Farnworth, S. L. and Henderson, N. C. and Mackinnon, A. C. and Atkinson, K. M. and Wilkinson, T. and Dhaliwal, K. and Hayashi, K. and Simpson, A. J. and Rossi, A. G. and Haslett, C. and Sethi, T.},
      title = {{{G}alectin-3 reduces the severity of pneumococcal pneumonia by augmenting neutrophil function}},
      journal = {Am J Pathol},
      year = {2008},
      volume = {172},
      number = {2},
      pages = {395--405},
      month = feb
    }
  4. Galectin-3 expression and secretion links macrophages to the promotion of renal fibrosis
    N. C. Henderson, A. C. Mackinnon, S. L. Farnworth, T. Kipari, C. Haslett, J. P. Iredale, F. T. Liu, J. Hughes, and T. Sethi
    Am J Pathol Feb 2008

    Macrophages have been proposed as a key cell type in the pathogenesis of renal fibrosis; however, the mechanism by which macrophages drive fibrosis is still unclear. We show that expression of galectin-3, a beta-galactoside-binding lectin, is up-regulated in a mouse model of progressive renal fibrosis (unilateral ureteric obstruction, UUO), and absence of galectin-3 protects against renal myofibroblast accumulation/activation and fibrosis. Furthermore, specific depletion of macrophages using CD11b-DTR mice reduces fibrosis severity after UUO demonstrating that macrophages are key cells in the pathogenesis of renal fibrosis. Disruption of the galectin-3 gene does not affect macrophage recruitment after UUO, or macrophage proinflammatory cytokine profiles in response to interferon-gamma/lipopolysaccharide. In addition, absence of galectin-3 does not affect transforming growth factor-beta expression or Smad 2/3 phosphorylation in obstructed kidneys. Adoptive transfer of wild-type but not galectin-3(-/-) macrophages did, however, restore the fibrotic phenotype in galectin-3(-/-) mice. Cross-over experiments using wild-type and galectin-3(-/-) macrophage supernatants and renal fibroblasts confirmed that secretion of galectin-3 by macrophages is critical in the activation of renal fibroblasts to a profibrotic phenotype. Therefore, we demonstrate for the first time that galectin-3 expression and secretion by macrophages is a major mechanism linking macrophages to the promotion of renal fibrosis.

    @article{pmid18202187,
      author = {Henderson, N. C. and Mackinnon, A. C. and Farnworth, S. L. and Kipari, T. and Haslett, C. and Iredale, J. P. and Liu, F. T. and Hughes, J. and Sethi, T.},
      title = {{{G}alectin-3 expression and secretion links macrophages to the promotion of renal fibrosis}},
      journal = {Am J Pathol},
      year = {2008},
      volume = {172},
      number = {2},
      pages = {288--298},
      month = feb
    }

2007

  1. Liver fibrosis: cellular mechanisms of progression and resolution
    N. C. Henderson, and J. P. Iredale
    Clin Sci (Lond) Mar 2007

    Liver fibrosis represents a major worldwide health care burden. The last 15 years have seen a rapid growth in our understanding of the pathogenesis of this clinically relevant model of inflammation and repair. This work is likely to inform the design of effective antifibrotic therapies in the near future. In this review, we examine how the innate and adaptive immune response interacts with other key cell types in the liver, such as the myofibroblast, regulating the process of hepatic fibrosis and, where relevant, resolution of fibrosis with remodelling. Emphasis is placed on the increasing knowledge that has been generated by the use of transgenic animals and animals in which specific cell lines have been deleted. Additionally, we review the increasing evidence that, although significant numbers of wound-healing myofibroblasts are derived from the hepatic stellate cell, significant contributions may occur from other cell lineages, including those from distant sites such as bone marrow stem cells.

    @article{pmid17261089,
      author = {Henderson, N. C. and Iredale, J. P.},
      title = {{{L}iver fibrosis: cellular mechanisms of progression and resolution}},
      journal = {Clin Sci (Lond)},
      year = {2007},
      volume = {112},
      number = {5},
      pages = {265--280},
      month = mar
    }
  2. Collagen I and thrombin activate MMP-2 by MMP-14-dependent and -independent pathways: implications for airway smooth muscle migration
    N. Henderson, L. J. Markwick, S. R. Elshaw, A. M. Freyer, A. J. Knox, and S. R. Johnson
    Am J Physiol Lung Cell Mol Physiol Apr 2007

    Increased proinflammatory mediators and ECM deposition are key features of the airways in asthma. Matrix metalloproteinases (MMPs) are produced by airway smooth muscle (ASM) cells and have multiple roles in inflammation and tissue remodeling. We hypothesized that components of the asthmatic airway would stimulate MMP production and activation by ASM and contribute to airway remodeling. We measured human ASM-derived MMP mRNA, protein, and activity by real-time RT-PCR, zymography, Western blotting, and MMP activity assay. Collagen I and thrombin caused a synergistic increase in MMP-2 protein and total MMP activity but paradoxically decreased MMP-2 mRNA. Additionally, collagen I activated MMP-2 in culture supernatants independent of the cell surface. Together, collagen I and thrombin strongly enhanced MMP-14 mRNA and protein but had no effect individually, suggesting increased MMP-14, the activating protease for MMP-2, may be partially responsible for MMP-2 activation. Furthermore, collagen I reduced tissue inhibitor of metalloproteinase-2 protein (TIMP-2). We examined the role of MMPs in functions of ASM related to airway remodeling and found migration and proliferation were MMP dependent, whereas adhesion and apoptosis were not. Ilomastat inhibited migration by 25%, which was also inhibited by TIMPs 1-4 and increased by the MMP-2 activator thrombin. These in vitro findings suggest that the environment within the airways of patients with asthma enhances MMP-2 and -14 protein and activity by a complex interaction of transcriptional and posttranscriptional mechanisms, which may contribute to ASM migration.

    @article{pmid17189319,
      author = {Henderson, N. and Markwick, L. J. and Elshaw, S. R. and Freyer, A. M. and Knox, A. J. and Johnson, S. R.},
      title = {{{C}ollagen {I} and thrombin activate {M}{M}{P}-2 by {M}{M}{P}-14-dependent and -independent pathways: implications for airway smooth muscle migration}},
      journal = {Am J Physiol Lung Cell Mol Physiol},
      year = {2007},
      volume = {292},
      number = {4},
      pages = {L1030--1038},
      month = apr
    }
  3. Critical role of c-jun (NH2) terminal kinase in paracetamol- induced acute liver failure
    N. C. Henderson, K. J. Pollock, J. Frew, A. C. Mackinnon, R. A. Flavell, R. J. Davis, T. Sethi, and K. J. Simpson
    Gut Jul 2007

    Acute hepatic failure secondary to paracetamol poisoning is associated with high mortality. C-jun (NH2) terminal kinase (JNK) is a member of the mitogen-activated protein kinase family and is a key intracellular signalling molecule involved in controlling the fate of cells.\{To examine the role of JNK in paracetamol-induced acute liver failure (ALF).\{A previously developed mouse model of paracetamol poisoning was used to examine the role of JNK in paracetamol-induced ALF.\{Paracetamol-induced hepatic JNK activation both in human and murine paracetamol hepatotoxicity and in our murine model preceded the onset of hepatocyte death. JNK inhibition in vivo (using two JNK inhibitors with different mechanisms of action) markedly reduced mortality in murine paracetamol hepatotoxicity, with a significant reduction in hepatic necrosis and apoptosis. In addition, delayed administration of the JNK inhibitor was more effective than N-acetylcysteine after paracetamol poisoning in mice. JNK inhibition was not protective in acute carbon tetrachloride-mediated or anti-Fas antibody-mediated hepatic injury, suggesting specificity for the role of JNK in paracetamol hepatotoxicity. Furthermore, disruption of the JNK1 or JNK2 genes did not protect against paracetamol-induced hepatic damage. Pharmacological JNK inhibition had no effect on paracetamol metabolism, but markedly inhibited hepatic tumour necrosis foctor alpha (TNF alpha) production after paracetamol poisoning.\{These data demonstrated a central role for JNK in the pathogenesis of paracetamol-induced liver failure, thereby identifying JNK as an important therapeutic target in the treatment of paracetamol hepatotoxicity.

    @article{pmid17185352,
      author = {Henderson, N. C. and Pollock, K. J. and Frew, J. and Mackinnon, A. C. and Flavell, R. A. and Davis, R. J. and Sethi, T. and Simpson, K. J.},
      title = {{{C}ritical role of c-jun ({N}{H}2) terminal kinase in paracetamol- induced acute liver failure}},
      journal = {Gut},
      year = {2007},
      volume = {56},
      number = {7},
      pages = {982--990},
      month = jul
    }

2006

  1. Galectin-3 regulates myofibroblast activation and hepatic fibrosis
    N. C. Henderson, A. C. Mackinnon, S. L. Farnworth, F. Poirier, F. P. Russo, J. P. Iredale, C. Haslett, K. J. Simpson, and T. Sethi
    Proc Natl Acad Sci U S A Mar 2006

    Central to fibrogenesis and the scarring of organs is the activation of fibroblasts into matrix-secreting myofibroblasts. We demonstrate that Galectin-3 expression is up-regulated in established human fibrotic liver disease and is temporally and spatially related to the induction and resolution of experimental hepatic fibrosis. Disruption of the Galectin-3 gene blocks myofibroblast activation and procollagen (I) expression in vitro and in vivo, markedly attenuating liver fibrosis. Addition of exogenous recombinant Galectin-3 in vitro reversed this abnormality. The reduction in hepatic fibrosis observed in the Galectin-3(-/-) mouse occurred despite equivalent liver injury and inflammation, and similar tissue expression of TGF-beta. TGF-beta failed to transactivate Galectin-3(-/-) hepatic stellate cells, in contrast with WT hepatic stellate cells; however, TGF-beta-stimulated Smad-2 and -3 activation was equivalent. These data suggest that Galectin-3 is required for TGF-beta mediated myofibroblast activation and matrix production. Finally, in vivo siRNA knockdown of Galectin-3 inhibited myofibroblast activation after hepatic injury and may therefore provide an alternative therapeutic approach to the prevention and treatment of liver fibrosis.

    @article{pmid16549783,
      author = {Henderson, N. C. and Mackinnon, A. C. and Farnworth, S. L. and Poirier, F. and Russo, F. P. and Iredale, J. P. and Haslett, C. and Simpson, K. J. and Sethi, T.},
      title = {{{G}alectin-3 regulates myofibroblast activation and hepatic fibrosis}},
      journal = {Proc Natl Acad Sci U S A},
      year = {2006},
      volume = {103},
      number = {13},
      pages = {5060--5065},
      month = mar
    }

2005

  1. Modeling liver fibrosis in rodents
    C. Constandinou, N. Henderson, and J. P. Iredale
    Methods Mol Med Mar 2005

    Animal models of hepatic fibrosis provide a means to study the cell and molecular mediators of fibrosis in a serial manner during both progression and recovery. Several approaches to induction of fibrosis have been described. Of these, CCl(4) intoxication in rats and mice is probably the most widely studied. In addition, the CCl(4) model is the best characterized with respect to histological, biochemical, cell, and molecular changes associated with the development of fibrosis. CCl(4) can be given intraperitoneally or by oral gavage; it induces zone III necrosis and hepatocyte apoptosis with associated hepatic stellate cell activation and tissue fibrosis. With repetitive dosing CCl(4) can be used to induce bridging hepatic fibrosis (4 wk of twice-weekly dosing), cirrhosis (8 wk of twice-weekly dosing) and advanced micronodular cirrhosis (12 wk of twice-weekly dosing). In addition, for each of these models spontaneous recovery from fibrosis can be studied after cessation of dosing. Mechanistic studies using gene knockout and transgenic animals can also be established using CCl(4). Together these models have provided unparalleled insights into the mechanisms underlying hepatic fibrosis.

    @article{pmid16118456,
      author = {Constandinou, C. and Henderson, N. and Iredale, J. P.},
      title = {{{M}odeling liver fibrosis in rodents}},
      journal = {Methods Mol Med},
      year = {2005},
      volume = {117},
      pages = {237--250}
    }

2004

  1. CD98hc (SLC3A2) interaction with beta 1 integrins is required for transformation
    N. C. Henderson, E. A. Collis, A. C. Mackinnon, K. J. Simpson, C. Haslett, R. Zent, M. Ginsberg, and T. Sethi
    J Biol Chem Dec 2004

    CD98hc (SLC3A2) constitutively and specifically associates with beta(1) integrins and is highly expressed on the surface of human tumor cells irrespective of the tissue of origin. We have found here that expression of CD98hc promotes both anchorage- and serum-independent growth. This oncogenic activity is dependent on beta(1) integrin-mediated phosphoinositol 3-hydroxykinase stimulation and the level of surface expression of CD98hc. Using chimeras of CD98hc and the type II membrane protein CD69, we show that the transmembrane domain of CD98hc is necessary and sufficient for integrin association in cells. Furthermore, CD98hc/beta(1) integrin association is required for focal adhesion kinase-dependent phosphoinositol 3-hydroxykinase activation and cellular transformation. Amino acids 82-87 in the putative cytoplasmic/transmembrane region appear to be critical for the oncogenic potential of CD98hc and provide a novel mechanism for tumor promotion by integrins. These results explain how high expression of CD98hc in human cancers contributes to transformation; furthermore, the transmembrane association of CD98hc and beta(1) integrins may provide a new target for cancer therapy.

    @article{pmid15485886,
      author = {Henderson, N. C. and Collis, E. A. and Mackinnon, A. C. and Simpson, K. J. and Haslett, C. and Zent, R. and Ginsberg, M. and Sethi, T.},
      title = {{{C}{D}98hc ({S}{L}{C}3{A}2) interaction with beta 1 integrins is required for transformation}},
      journal = {J Biol Chem},
      year = {2004},
      volume = {279},
      number = {52},
      pages = {54731--54741},
      month = dec
    }
  2. Matrix metalloproteinase expression and activity in human airway smooth muscle cells
    S. R. Elshaw, N. Henderson, A. J. Knox, S. A. Watson, D. J. Buttle, and S. R. Johnson
    Br J Pharmacol Aug 2004

    Airway remodelling is a feature of chronic asthma comprising smooth muscle hypertrophy and deposition of extracellular matrix (ECM) proteins. Matrix metalloproteinases (MMPs) breakdown ECM, are involved in tissue remodelling and have been implicated in airway remodelling. Although mesenchymal cells are an important source of MMPs, little data are available on airway smooth muscle (ASM) derived MMPs. We therefore investigated MMP and tissue inhibitor of metalloproteinase (TIMP) production and activity in human ASM cells. MMPs and TIMPs were examined using quantitative real-time RT-PCR, Western blotting, zymography and a quench fluorescence (QF) assay of total MMP activity. The most abundant MMPs were pro-MMP-2, pro- MMP-3, active MMP-3 and MT1-MMP. TIMP-1 and TIMP-2 expression was low in cell lysates but high in conditioned medium. High TIMP secretion was confirmed by the ability of ASM-conditioned medium to inhibit recombinant MMP-2 in a QF assay. Thrombin increased MMP activity by activation of pro-MMP-2 independent of the conventional smooth muscle thrombin receptors PAR 1 and 4. In conclusion, ASM cells express pro-MMP-2, pro and active MMP-3, MMP-9 and MT1-MMP. Unstimulated cells secrete excess TIMP 1 and 2, preventing proteolytic activity. MMP-2 can be activated by thrombin which may contribute to airway remodelling.

    @article{pmid15265805,
      author = {Elshaw, S. R. and Henderson, N. and Knox, A. J. and Watson, S. A. and Buttle, D. J. and Johnson, S. R.},
      title = {{{M}atrix metalloproteinase expression and activity in human airway smooth muscle cells}},
      journal = {Br J Pharmacol},
      year = {2004},
      volume = {142},
      number = {8},
      pages = {1318--1324},
      month = aug
    }

2003

  1. Chemokines in the pathogenesis of liver disease: so many players with poorly defined roles
    K. J. Simpson, N. C. Henderson, C. L. Bone-Larson, N. W. Lukacs, C. M. Hogaboam, and S. L. Kunkel
    Clin Sci (Lond) Jan 2003

    Many new chemokines have been described in recent years, resulting in a new classification of these chemoattractant proteins. The characterization of the biological functions of most chemokines relates to their ability to induce chemotaxis in circulating inflammatory cells. However, it is now clear that chemokines have a much wider biological role, including angiogenesis, carcinogenesis and involvement in the pathogenesis of HIV infection. Our understanding of the role of chemokines in the pathogenesis of disorders of the lungs and brain outstrips that with regard to disorders of the liver. An increased understanding of the role of chemokines in the pathogenesis of liver disease may lead to the development of novel therapies for hepatic disease.

    @article{pmid12519087,
      author = {Simpson, K. J. and Henderson, N. C. and Bone-Larson, C. L. and Lukacs, N. W. and Hogaboam, C. M. and Kunkel, S. L.},
      title = {{{C}hemokines in the pathogenesis of liver disease: so many players with poorly defined roles}},
      journal = {Clin Sci (Lond)},
      year = {2003},
      volume = {104},
      number = {1},
      pages = {47--63},
      month = jan
    }