Network


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

Hotspot


Dive into the research topics where Ewa Ellis is active.

Publication


Featured researches published by Ewa Ellis.


Nature Biotechnology | 2007

Robust expansion of human hepatocytes in Fah −/− / Rag2 −/− / Il2rg −/− mice

Hisaya Azuma; Nicole K. Paulk; Aarati Ranade; Craig Dorrell; Muhsen Al-Dhalimy; Ewa Ellis; Stephen C. Strom; Mark A. Kay; Milton J. Finegold; Markus Grompe

Mice that could be highly repopulated with human hepatocytes would have many potential uses in drug development and research applications. The best available model of liver humanization, the uroplasminogen-activator transgenic model, has major practical limitations. To provide a broadly useful hepatic xenorepopulation system, we generated severely immunodeficient, fumarylacetoacetate hydrolase (Fah)-deficient mice. After pretreatment with a urokinase-expressing adenovirus, these animals could be highly engrafted (up to 90%) with human hepatocytes from multiple sources, including liver biopsies. Furthermore, human cells could be serially transplanted from primary donors and repopulate the liver for at least four sequential rounds. The expanded cells displayed typical human drug metabolism. This system provides a robust platform to produce high-quality human hepatocytes for tissue culture. It may also be useful for testing the toxicity of drug metabolites and for evaluating pathogens dependent on human liver cells for replication.


Gastroenterology | 2009

Differentiation and Transplantation of Human Embryonic Stem Cell–Derived Hepatocytes

Hesham Basma; Alejandro Soto-Gutierrez; Govardhana Rao Yannam; Liping Liu; Ryotaro Ito; Toshiyuki Yamamoto; Ewa Ellis; Steven D. Carson; Shintaro Sato; Yong Chen; David Muirhead; Nalu Navarro-Alvarez; Ronald J. Wong; Jayanta Roy-Chowdhury; Jeffrey L. Platt; David F. Mercer; John D. Miller; Stephen C. Strom; Naoya Kobayashi; Ira J. Fox

BACKGROUND & AIMS The ability to obtain unlimited numbers of human hepatocytes would improve the development of cell-based therapies for liver diseases, facilitate the study of liver biology, and improve the early stages of drug discovery. Embryonic stem cells are pluripotent, potentially can differentiate into any cell type, and therefore could be developed as a source of human hepatocytes. METHODS To generate human hepatocytes, human embryonic stem cells were differentiated by sequential culture in fibroblast growth factor 2 and human activin-A, hepatocyte growth factor, and dexamethasone. Functional hepatocytes were isolated by sorting for surface asialoglycoprotein-receptor expression. Characterization was performed by real-time polymerase chain reaction, immunohistochemistry, immunoblot, functional assays, and transplantation. RESULTS Embryonic stem cell-derived hepatocytes expressed liver-specific genes, but not genes representing other lineages, secreted functional human liver-specific proteins similar to those of primary human hepatocytes, and showed human hepatocyte cytochrome P450 metabolic activity. Serum from rodents given injections of embryonic stem cell-derived hepatocytes contained significant amounts of human albumin and alpha1-antitrypsin. Colonies of cytokeratin-18 and human albumin-expressing cells were present in the livers of recipient animals. CONCLUSIONS Human embryonic stem cells can be differentiated into cells with many characteristics of primary human hepatocytes. Hepatocyte-like cells can be enriched and recovered based on asialoglycoprotein-receptor expression and potentially could be used in drug discovery research and developed as therapeutics.


Cell | 2015

Long-term culture of genome-stable bipotent stem cells from adult human liver

Meritxell Huch; Helmuth Gehart; Ruben van Boxtel; Karien Hamer; Francis Blokzijl; Monique M.A. Verstegen; Ewa Ellis; Martien van Wenum; Sabine A. Fuchs; Joep de Ligt; Marc van de Wetering; Nobuo Sasaki; Susanne J. Boers; Hans Kemperman; Jeroen de Jonge; Jan N. M. IJzermans; Edward E. S. Nieuwenhuis; Ruurdtje Hoekstra; Stephen C. Strom; Robert G. Vries; Luc J. W. van der Laan; Edwin Cuppen; Hans Clevers

Summary Despite the enormous replication potential of the human liver, there are currently no culture systems available that sustain hepatocyte replication and/or function in vitro. We have shown previously that single mouse Lgr5+ liver stem cells can be expanded as epithelial organoids in vitro and can be differentiated into functional hepatocytes in vitro and in vivo. We now describe conditions allowing long-term expansion of adult bile duct-derived bipotent progenitor cells from human liver. The expanded cells are highly stable at the chromosome and structural level, while single base changes occur at very low rates. The cells can readily be converted into functional hepatocytes in vitro and upon transplantation in vivo. Organoids from α1-antitrypsin deficiency and Alagille syndrome patients mirror the in vivo pathology. Clonal long-term expansion of primary adult liver stem cells opens up experimental avenues for disease modeling, toxicology studies, regenerative medicine, and gene therapy.


Cell Transplantation | 2012

Improving the techniques for human hepatocyte transplantation: Report from a consensus meeting in London

Juliana Puppi; Stephen C. Strom; Robin D. Hughes; Sanjay Bansal; José V. Castell; Ibrahim Dagher; Ewa Ellis; Greg Nowak; Bo Göran Ericzon; Ira J. Fox; José M. Gómez-Lechón; Chandan Guha; Sanjeev Gupta; Ragai R. Mitry; Kazuo Ohashi; Michael Ott; Lola M. Reid; Jayanta Roy-Chowdhury; Etienne Sokal; Anne Weber; Anil Dhawan

On September 6 and 7, 2009 a meeting was held in London to identify and discuss what are perceived to be current roadblocks to effective hepatocyte transplantation as it is currently practiced in the clinics and, where possible, to offer suggestions to overcome the blocks and improve the outcomes for this cellular therapy. Present were representatives of most of the active clinical hepatocyte transplant programs along with other scientists who have contributed substantial basic research to this field. Over the 2-day sessions based on the experience of the participants, numerous roadblocks or challenges were identified, including the source of cells for the transplants and problems with tracking cells following transplantation. Much of the discussion was focused on methods to improve engraftment and proliferation of donor cells posttransplantation. The group concluded that, for now, parenchymal hepatocytes isolated from donor livers remain the best cell source for transplantation. It was reported that investigations with other cell sources, including stem cells, were at the preclinical and early clinical stages. Numerous methods to modulate the immune reaction and vascular changes that accompany hepatocyte transplantation were proposed. It was agreed that, to obtain sufficient levels of repopulation of liver with donor cells in patients with metabolic liver disease, some form of liver preconditioning would likely be required to enhance the engraftment and/or proliferation of donor cells. It was reported that clinical protocols for preconditioning by hepatic irradiation, portal vein embolization, and surgical resection had been developed and that clinical studies using these protocols would be initiated in the near future. Participants concluded that sharing information between the groups, including standard information concerning the quality and function of the transplanted cells prior to transplantation, clinical information on outcomes, and standard preconditioning protocols, would help move the field forward and was encouraged.


Genes & Development | 2010

GPS2-dependent corepressor/SUMO pathways govern anti-inflammatory actions of LRH-1 and LXRβ in the hepatic acute phase response

Nicolas Venteclef; Tomas Jakobsson; Anna Ehrlund; Anastasios E. Damdimopoulos; Laura Mikkonen; Ewa Ellis; Lisa M. Nilsson; Paolo Parini; Olli A. Jänne; Jan Åke Gustafsson; Knut R. Steffensen; Eckardt Treuter

The orphan receptor LRH-1 and the oxysterol receptors LXRalpha and LXRbeta are established transcriptional regulators of lipid metabolism that appear to control inflammatory processes. Here, we investigate the anti-inflammatory actions of these nuclear receptors in the hepatic acute phase response (APR). We report that selective synthetic agonists induce SUMOylation-dependent recruitment of either LRH-1 or LXR to hepatic APR promoters and prevent the clearance of the N-CoR corepressor complex upon cytokine stimulation. Investigations of the APR in vivo, using LXR knockout mice, indicate that the anti-inflammatory actions of LXR agonists are triggered selectively by the LXRbeta subtype. We further find that hepatic APR responses in small ubiquitin-like modifier-1 (SUMO-1) knockout mice are increased, which is due in part to diminished LRH-1 action at APR promoters. Finally, we provide evidence that the metabolically important coregulator GPS2 functions as a hitherto unrecognized transrepression mediator of interactions between SUMOylated nuclear receptors and the N-CoR corepressor complex. Our study extends the knowledge of anti-inflammatory mechanisms and pathways directed by metabolic nuclear receptor-corepressor networks to the control of the hepatic APR, and implies alternative pharmacological strategies for the treatment of human metabolic diseases associated with inflammation.


Scientific Reports | 2016

Characterization of primary human hepatocyte spheroids as a model system for drug-induced liver injury, liver function and disease

Catherine C. Bell; Delilah F. G. Hendriks; Sabrina M.L. Moro; Ewa Ellis; Joanne Walsh; Anna Renblom; Lisa Fredriksson Puigvert; Anita C. A. Dankers; Frank Jacobs; Jan Snoeys; Rowena Sison-Young; Rosalind E. Jenkins; Åsa Nordling; Souren Mkrtchian; B. Kevin Park; Neil R. Kitteringham; Christopher E. Goldring; Volker M. Lauschke; Magnus Ingelman-Sundberg

Liver biology and function, drug-induced liver injury (DILI) and liver diseases are difficult to study using current in vitro models such as primary human hepatocyte (PHH) monolayer cultures, as their rapid de-differentiation restricts their usefulness substantially. Thus, we have developed and extensively characterized an easily scalable 3D PHH spheroid system in chemically-defined, serum-free conditions. Using whole proteome analyses, we found that PHH spheroids cultured this way were similar to the liver in vivo and even retained their inter-individual variability. Furthermore, PHH spheroids remained phenotypically stable and retained morphology, viability, and hepatocyte-specific functions for culture periods of at least 5 weeks. We show that under chronic exposure, the sensitivity of the hepatocytes drastically increased and toxicity of a set of hepatotoxins was detected at clinically relevant concentrations. An interesting example was the chronic toxicity of fialuridine for which hepatotoxicity was mimicked after repeated-dosing in the PHH spheroid model, not possible to detect using previous in vitro systems. Additionally, we provide proof-of-principle that PHH spheroids can reflect liver pathologies such as cholestasis, steatosis and viral hepatitis. Combined, our results demonstrate that the PHH spheroid system presented here constitutes a versatile and promising in vitro system to study liver function, liver diseases, drug targets and long-term DILI.


Hepatology | 2011

Overexpression of Cholesterol 7α-hydroxylase promotes hepatic bile acid synthesis and secretion and maintains cholesterol homeostasis

Tiangang Li; Michelle Matozel; Shannon Boehme; Bo Kong; Lisa-Mari Nilsson; Grace L. Guo; Ewa Ellis; John Y. L. Chiang

We reported previously that mice overexpressing cytochrome P450 7a1 (Cyp7a1; Cyp7a1‐tg mice) are protected against high fat diet–induced hypercholesterolemia, obesity, and insulin resistance. Here, we investigated the underlying mechanism of bile acid signaling in maintaining cholesterol homeostasis in Cyp7a1‐tg mice. Cyp7a1‐tg mice had two‐fold higher Cyp7a1 activity and bile acid pool than did wild‐type mice. Gallbladder bile acid composition changed from predominantly cholic acid (57%) in wild‐type to chenodeoxycholic acid (54%) in Cyp7a1‐tg mice. Cyp7a1‐tg mice had higher biliary and fecal cholesterol and bile acid secretion rates than did wild‐type mice. Surprisingly, hepatic de novo cholesterol synthesis was markedly induced in Cyp7a1‐tg mice but intestine fractional cholesterol absorption in Cyp7a1‐tg mice remained the same as wild‐type mice despite the presence of increased intestine bile acids. Interestingly, hepatic but not intestinal expression of several cholesterol (adenosine triphosphate–binding cassette G5/G8 [ABCG5/G8], scavenger receptor class B, member 1) and bile acid (ABCB11) transporters were significantly induced in Cyp7a1‐tg mice. Treatment of mouse or human hepatocytes with a farnesoid X receptor (FXR) agonist GW4064 or bile acids induced hepatic Abcg5/g8 expression. A functional FXR binding site was identified in the Abcg5 gene promoter. Study of tissue‐specific Fxr knockout mice demonstrated that loss of the Fxr gene in the liver attenuated bile acid induction of hepatic Abcg5/g8 and gallbladder cholesterol content, suggesting a role of FXR in the regulation of cholesterol transport. Conclusion: This study revealed a new mechanism by which increased Cyp7a1 activity expands the hydrophobic bile acid pool, stimulating hepatic cholesterol synthesis and biliary cholesterol secretion without increasing intestinal cholesterol absorption. This study demonstrated that Cyp7a1 plays a critical role in maintaining cholesterol homeostasis and underscores the importance of bile acid signaling in regulating overall cholesterol homeostasis. (HEPATOLOGY 2011)


Current protocols in stem cell biology | 2010

Isolation of Amniotic Epithelial Stem Cells

Toshio Miki; Fabio Marongiu; Kenneth Dorko; Ewa Ellis; Stephen C. Strom

Many of the cell types that can be isolated from placental tissues retain phenotypic plasticity that makes them an interesting source of cells for regenerative medicine. Several procedures for the isolation of stem cells from different parts of the placenta have been reported. This unit describes a detailed and simple protocol for the selective isolation of amniotic epithelial cells from human term placenta without disturbing the mesenchymal layer. We also introduce a simple density separation technique for the enrichment of the population for SSEA-4 positive cells.


Genes & Development | 2009

Bile acid signaling pathways increase stability of Small Heterodimer Partner (SHP) by inhibiting ubiquitin–proteasomal degradation

Ji Miao; Zhen Xiao; Deepthi Kanamaluru; Gyesik Min; Peter M. Yau; Timothy D. Veenstra; Ewa Ellis; Steve Strom; Kelly Suino-Powell; H. Eric Xu; Jongsook Kim Kemper

Small Heterodimer Partner (SHP) inhibits activities of numerous transcription factors involved in diverse biological pathways. As an important metabolic regulator, SHP plays a key role in maintaining cholesterol and bile acid homeostasis by inhibiting cholesterol conversion to bile acids. While SHP gene induction by increased bile acids is well established, whether SHP activity is also modulated remains unknown. Here, we report surprising findings that SHP is a rapidly degraded protein via the ubiquitin-proteasomal pathway and that bile acids or bile acid-induced intestinal fibroblast growth factor 19 (FGF19) increases stability of hepatic SHP by inhibiting proteasomal degradation in an extracellular signal-regulated kinase (ERK)-dependent manner. SHP was ubiquitinated at Lys122 and Lys123, and mutation of these sites altered its stability and repression activity. Tandem mass spectrometry revealed that upon bile acid treatment, SHP was phosphorylated at Ser26, within an ERK motif in SHP, and mutation of this site dramatically abolished SHP stability. Surprisingly, SHP stability was abnormally elevated in ob/ob mice and diet-induced obese mice. These results demonstrate an important role for regulation of SHP stability in bile acid signaling in normal conditions, and that abnormal stabilization of SHP may be associated with metabolic disorders, including obesity and diabetes.


Journal of Immunology | 2015

Cutting Edge: Identification and Characterization of Human Intrahepatic CD49a+ NK Cells

Nicole Marquardt; Vivien Béziat; Sanna Nyström; Julia Hengst; Martin A. Ivarsson; Eliisa Kekäläinen; Helene Johansson; Jenny Mjösberg; Magnus Westgren; Tim O. Lankisch; Heiner Wedemeyer; Ewa Ellis; Hans-Gustaf Ljunggren; Jakob Michaëlsson; Niklas K. Björkström

Although NK cells are considered innate, recent studies in mice revealed the existence of a unique lineage of hepatic CD49a+DX5− NK cells with adaptive-like features. Development of this NK cell lineage is, in contrast to conventional NK cells, dependent on T-bet but not Eomes. In this study, we describe the identification of a T-bet+Eomes−CD49a+ NK cell subset readily detectable in the human liver, but not in afferent or efferent hepatic venous or peripheral blood. Human intrahepatic CD49a+ NK cells express killer cell Ig-like receptor and NKG2C, indicative of having undergone clonal-like expansion, are CD56bright, and express low levels of CD16, CD57, and perforin. After stimulation, CD49a+ NK cells express high levels of inflammatory cytokines but degranulate poorly. CD49a+ NK cells retain their phenotype after expansion in long-term in vitro cultures. These results demonstrate the presence of a likely human counterpart of mouse intrahepatic NK cells with adaptive-like features.

Collaboration


Dive into the Ewa Ellis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo-Göran Ericzon

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Carl Jorns

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Greg Nowak

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helene Johansson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Curt Einarsson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Kenneth Dorko

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Ingemar Björkhem

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Ira J. Fox

University of Nebraska Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge