Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Stem Cells Transl Med ; 13(6): 522-531, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38619045

ABSTRACT

Placenta-derived human amniotic epithelial cells (hAEC) exhibit anti-inflammatory and anti-fibrotic effects in cirrhosis models. We conducted a first-in-human phase I clinical trial to assess the safety and tolerability of hAEC in adults with compensated cirrhosis. We examined increasing and repeated doses of hAEC in 9 patients in 3 cohorts. Cohort 1 patients received 0.5 × 106/kg hAEC in one IV infusion. Cohort 2 patients received 1 × 106/kg hAEC in one IV infusion. The patients in cohort 3 received 1 × 106/kg hAEC on days 0 and 28. Here, we report follow-up to post-infusion day 56 (D56), during which no serious adverse events occurred. Six patients experienced no study-related adverse events, while 3 patients reported mild (grade 1) headaches that were possibly infusion-related. A transient decrease in serum platelet count occurred in all patients, which returned to baseline screening values by day 5. FIB-4 values to assess fibrosis were significantly lower at D56. Although not statistically significant, serum AST levels and liver stiffness measurements at D56 were lower than those at baseline. The hepatic venous pressure gradient, a measure of portal hypertension, declined in 4 patients, did not change in 3 patients, and increased in 2 patients. In conclusion, intravenous infusion of allogeneic hAEC in patients with compensated cirrhosis at the doses used in this study was safe and well tolerated, with no difference observed between 1 and 2 doses. Decreased hepatic inflammation, liver stiffness, and portal hypertension support larger studies aimed at identifying patients who may benefit from this therapy. Clinical Trial registration: The trial was prospectively entered on the Australian Clinical Trials Registry (ANZCTR12616000437460).


Subject(s)
Amnion , Epithelial Cells , Liver Cirrhosis , Humans , Female , Amnion/transplantation , Liver Cirrhosis/therapy , Liver Cirrhosis/complications , Middle Aged , Male , Adult , Aged
2.
Stem Cell Res Ther ; 12(1): 429, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321089

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease is the most common liver disease globally and in its inflammatory form, non-alcoholic steatohepatitis (NASH), can progress to cirrhosis and hepatocellular carcinoma (HCC). Currently, patient education and lifestyle changes are the major tools to prevent the continued progression of NASH. Emerging therapies in NASH target known pathological processes involved in the progression of the disease including inflammation, fibrosis, oxidative stress and hepatocyte apoptosis. Human amniotic epithelial cells (hAECs) were previously shown to be beneficial in experimental models of chronic liver injury, reducing hepatic inflammation and fibrosis. Previous studies have shown that liver progenitor cells (LPCs) response plays a significant role in the development of fibrosis and HCC in mouse models of fatty liver disease. In this study, we examined the effect hAECs have on the LPC response and hepatic oxidative stress in an experimental model of NASH. METHODS: Experimental NASH was induced in C57BL/6 J male mice using a high-fat, high fructose diet for 42 weeks. Mice received either a single intraperitoneal injection of 2 × 106 hAECs at week 34 or an additional hAEC dose at week 38. Changes to the LPC response and oxidative stress regulators were measured. RESULTS: hAEC administration significantly reduced the expansion of LPCs and their mitogens, IL-6, IFNγ and TWEAK. hAEC administration also reduced neutrophil infiltration and myeloperoxidase production with a concurrent increase in heme oxygenase-1 production. These observations were accompanied by a significant increase in total levels of anti-fibrotic IFNß in mice treated with a single dose of hAECs, which appeared to be independent of c-GAS-STING activation. CONCLUSIONS: Expansion of liver progenitor cells, hepatic inflammation and oxidative stress associated with experimental NASH were attenuated by hAEC administration. Given that repeated doses did not significantly increase efficacy, future studies assessing the impact of dose escalation and/or timing of dose may provide insights into clinical translation.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Carcinoma, Hepatocellular/metabolism , Diet, High-Fat , Disease Models, Animal , Epithelial Cells , Liver/metabolism , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress , Stem Cells
3.
Cell Transplant ; 29: 963689720950221, 2020.
Article in English | MEDLINE | ID: mdl-32813573

ABSTRACT

Human amnion epithelial cells (hAECs) exert potent antifibrotic and anti-inflammatory effects when transplanted into preclinical models of tissue fibrosis. These effects are mediated in part via the secretion of soluble factors by hAECs which modulate signaling pathways and affect cell types involved in inflammation and fibrosis. Based on these reports, we hypothesized that these soluble factors may also support liver regeneration during chronic liver injury. To test this, we characterized the effect of both hAECs and hAEC-conditioned medium (CM) on liver repair in a mouse model of carbon tetrachloride (CCl4)-induced fibrosis. Liver repair was assessed by liver fibrosis, hepatocyte proliferation, and the liver progenitor cell (LPC) response. We found that the administration of hAECs or hAEC-CM reduced liver injury and fibrosis, sustained hepatocyte proliferation, and reduced LPC numbers during chronic liver injury. Additionally, we undertook in vitro studies to document both the cell-cell and paracrine-mediated effects of hAECs on LPCs by investigating the effects of co-culturing the LPCs and hAECs and hAEC-CM on LPCs. We found little change in LPCs co-cultured with hAECs. In contrast, hAEC-CM enhances LPC proliferation and differentiation. These findings suggest that paracrine factors secreted by hAECs enhance liver repair by reducing fibrosis while promoting regeneration during chronic liver injury.


Subject(s)
Amnion/metabolism , Epithelial Cells/metabolism , Liver Cirrhosis/physiopathology , Liver Cirrhosis/therapy , Liver Regeneration , Liver/injuries , Animals , Cell Count , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Gene Ontology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation/pathology , Liver/drug effects , Liver/pathology , Liver Regeneration/drug effects , Macrophages/drug effects , Macrophages/pathology , Male , Metabolic Networks and Pathways/drug effects , Mice, Inbred C57BL , Solubility , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology
4.
J Gastroenterol Hepatol ; 34(8): 1441-1449, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30821873

ABSTRACT

BACKGROUND AND AIM: Non-alcoholic steatohepatitis (NASH) can lead to cirrhosis and hepatocellular carcinoma. Currently, lifestyle modification is the only effective treatment. We have shown that human amnion epithelial cells (hAECs) reduce inflammation and fibrosis in toxin-induced liver injury models. We examined the effect of these cells and the soluble factors released by the cells into culture medium (hAEC conditioned medium [hAEC-CM]) in a diet-induced murine NASH model. METHODS: C57BL/6J male mice received a Western "fast food diet" for 42 weeks. Group 1 received an intraperitoneal injection of 2 × 106 hAECs at week 34, group 2 received an additional hAEC dose at week 38, and group 3 received thrice weekly hAEC-CM injections intraperitoneal for 8 weeks from week 34. Liver fibrosis area, inflammation, and fibrosis regulators were measured by immunohistochemistry, qPCR, and gelatin zymography. Metabolic parameters were also assessed. RESULTS: Fast food diet-fed mice demonstrated peri-cellular hepatic fibrosis, inflammation, and steatosis typical of NASH. Liver fibrosis area was reduced by 40% in hAEC-treated and hAEC-CM-treated mice. hAEC treatment significantly reduced pSMAD 2/3 signaling and the number of activated hepatic stellate cells and liver macrophages. Matrix metalloproteinase 2 and 9 gene and protein expression were variably affected. hAEC treatment did not alter the NASH activity score or metabolic parameters such as bodyweight, total cholesterol, or glucose tolerance. CONCLUSION: Human amnion epithelial cell and hAEC-CM significantly reduced hepatic inflammation and fibrosis in a diet-induced non-alcoholic fatty liver disease model. Although hAEC and hAEC-CM did not affect the metabolic components of NASH, their therapeutic potential is promising and warrants further investigation.


Subject(s)
Amnion/transplantation , Epithelial Cells/transplantation , Liver Cirrhosis, Experimental/prevention & control , Liver/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Amnion/cytology , Amnion/metabolism , Animals , Cells, Cultured , Diet, High-Fat , Epithelial Cells/metabolism , Female , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/pathology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Phosphorylation , Pregnancy , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism
5.
Front Pharmacol ; 8: 748, 2017.
Article in English | MEDLINE | ID: mdl-29114223

ABSTRACT

Background: Hepatic stellate cells (HSCs) are the primary collagen-secreting cells in the liver. While HSCs are the major cell type involved in the pathogenesis of liver fibrosis, hepatic macrophages also play an important role in mediating fibrogenesis and fibrosis resolution. Previously, we observed a reduction in HSC activation, proliferation, and collagen synthesis following exposure to human amnion epithelial cells (hAEC) and hAEC-conditioned media (hAEC-CM). This suggested that specific factors secreted by hAEC might be effective in ameliorating liver fibrosis. hAEC-derived extracellular vesicles (hAEC-EVs), which are nanosized (40-100 nm) membrane bound vesicles, may act as novel cell-cell communicators. Accordingly, we evaluated the efficacy of hAEC-EV in modulating liver fibrosis in a mouse model of chronic liver fibrosis and in human HSC. Methods: The hAEC-EVs were isolated and characterized. C57BL/6 mice with CCl4-induced liver fibrosis were administered hAEC-EV, hAEC-CM, or hAEC-EV depleted medium (hAEC-EVDM). LX2 cells, a human HSC line, and bone marrow-derived mouse macrophages were exposed to hAEC-EV, hAEC-CM, and hAEC-EVDM. Mass spectrometry was used to examine the proteome profile of each preparation. Results: The extent of liver fibrosis and number of activated HSCs were reduced significantly in CCl4-treated mice given hAEC-EVs, hAEC-CM, and hAEC EVDM compared to untreated controls. Hepatic macrophages were significantly decreased in all treatment groups, where a predominant M2 phenotype was observed. Human HSCs cultured with hAEC-EV and hAEC-CM displayed a significant reduction in collagen synthesis and hAEC-EV, hAEC-CM, and hAEC-EVDM altered macrophage polarization in bone marrow-derived mouse macrophages. Proteome analysis showed that 164 proteins were unique to hAEC-EV in comparison to hAEC-CM and hAEC-EVDM, and 51 proteins were co-identified components with the hAEC-EV fraction. Conclusion: This study provides novel data showing that hAEC-derived EVs significantly reduced liver fibrosis and macrophage infiltration to an extent similar to hAEC-EVDM and hAEC-CM. hAEC-EV-based therapy may be a potential therapeutic option for liver fibrosis.

6.
World J Gastroenterol ; 23(31): 5692-5699, 2017 Aug 21.
Article in English | MEDLINE | ID: mdl-28883694

ABSTRACT

AIM: To evaluate the role of tissue factor (TF) and protease activated receptor (PAR)-2 in liver fibrosis. METHODS: Using CCl4 administration for eight weeks, we induced hepatic fibrosis in wild-type C57BL/6 mice and in mice with deletion of the cytoplasmic signalling domain of TF (TF§CT/§CT), deletion of PAR-2 (PAR-2-/-) and combined deletion of TF signalling domain and PAR-2 (TF§CT/§CT/PAR-2-/-). Hepatic fibrosis area was assessed by quantitative imaging of picrosirius red staining. Hepatic collagen content was assessed by hydroxyproline levels. Hepatic stellate cells (αSMA positive) and hepatic macrophages (CD68 positive) were identified by immunohistochemistry. Hepatic gene expression was determined by PCR and liver TGFß1 content by ELISA. RESULTS: CCl4 treated mice with deletion of the PAR-2 gene (PAR-2-/-) and the cytoplasmic domain of TF (TF§CT/§CT) developed significantly less hepatic fibrosis, characterised by reduced liver fibrosis area and hydroxyproline content, compared to control wildtype mice treated with CCl4. The observed reduction in histological fibrosis was accompanied by a significant decrease in the hepatic content of TGFß, the prototypic fibrogenic cytokine, as well as fewer activated hepatic stellate cells and hepatic macrophages. Deletion of the TF cytoplasmic signalling domain reduced hepatic fibrosis to levels similar to those observed in mice lacking PAR-2 signalling but combined deletion provided no added protection against fibrosis indicating a lack of mutual modulating effects that have been observed in other contexts such as angiogenic responses. CONCLUSION: Tissue factor cytoplasmic domain is involved in TF-PAR-2 signalling initiating hepatic fibrosis and is a potential therapeutic target, as its deletion would not impact coagulation.


Subject(s)
Liver Cirrhosis, Experimental/pathology , Liver/pathology , Protein Domains/genetics , Receptor, PAR-2/metabolism , Thromboplastin/metabolism , Amino Acid Sequence/genetics , Animals , Blood Coagulation , Carbon Tetrachloride/toxicity , Collagen/analysis , Hepatic Stellate Cells/immunology , Humans , Hydroxyproline/analysis , Liver/cytology , Liver/immunology , Liver Cirrhosis, Experimental/blood , Liver Cirrhosis, Experimental/chemically induced , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, PAR-2/genetics , Sequence Deletion , Signal Transduction , Thromboplastin/genetics , Transforming Growth Factor beta/analysis
7.
PLoS One ; 10(10): e0129110, 2015.
Article in English | MEDLINE | ID: mdl-26479247

ABSTRACT

The placenta forms the interface between the maternal and fetal circulation and is critical for the establishment of a healthy pregnancy. Trophoblast cell proliferation, migration and invasion into the endometrium are fundamental events in the initiation of placentation. Leukemia inhibitory factor (LIF) has been shown to promote trophoblast invasion in vitro, however its precise role in trophoblast invasion in vivo is unknown. We hypothesized that LIF would be required for normal trophoblast invasion and spiral artery remodeling in mice. Both LIF and its receptor (LIFRα) co-localized with cytokeratin-positive invasive endovascular extravillous trophoblasts (EVT) in mouse implantation sites during mid-gestation. Temporally blocking LIF action during specific periods of placental development via administration of our unique LIFRα antagonist, PEGLA, resulted in abnormal trophoblast invasion and impaired spiral artery remodeling compared to PEG control. PEGLA-treated mouse decidual vessels were characterized by retention of α-smooth muscle actin (αSMA)-positive vascular smooth muscle cells (VSMCs), while PEG control decidual vessels were remodelled by cytokeratin-positive trophoblasts. LIF blockade did not alter F4/80-positive decidual macrophage numbers between treatment groups, but resulted in down-regulation of decidual transcript levels of monocyte chemoattractant protein-1 (MCP-1) and interleukin-10 (IL-10), which are important immune cell activation factors that promote spiral artery remodeling during pregnancy. Our data suggest that LIF plays an important role in trophoblast invasion in vivo and may facilitate trophoblast-decidual-immune cell cross talk to enable adequate spiral artery remodeling.


Subject(s)
Arteries/drug effects , Arteries/physiology , Leukemia Inhibitory Factor/antagonists & inhibitors , Trophoblasts/cytology , Trophoblasts/drug effects , Vascular Remodeling/drug effects , Animals , Chemokine CCL2/genetics , Cytokine Receptor gp130/genetics , Decidua/blood supply , Decidua/drug effects , Embryo Implantation/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Interleukin-10/genetics , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Placentation/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Pregnancy , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/metabolism
8.
Sci Rep ; 5: 13237, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26272398

ABSTRACT

The placenta forms the interface between the maternal and fetal circulation and is critical for the establishment of a healthy pregnancy. Specialized trophoblast cells derived from the embryonic trophectoderm play a pivotal role in the establishment of the placenta. Leukemia inhibitory factor (LIF) is one of the predominant cytokines present in the placenta during early pregnancy. LIF has been shown to regulate trophoblast adhesion and invasion in vitro, however its precise role in vivo is unknown. We hypothesized that LIF would be required for normal placental development in mice. LIF and LIFRα were immunolocalized to placental trophoblasts and fetal vessels in mouse implantation sites during mid-gestation. Temporally blocking LIF action during specific periods of placental development via intraperitoneal administration of our specific LIFRα antagonist, PEGLA, resulted in abnormal placental trophoblast and vascular morphology and reduced activated STAT3 but not ERK. Numerous genes regulating angiogenesis and oxidative stress were altered in the placenta in response to LIF inhibition. Pregnancy viability was also significantly compromised in PEGLA treated mice. Our data suggest that LIF plays an important role in placentation in vivo and the maintenance of healthy pregnancy.


Subject(s)
Abortion, Spontaneous/physiopathology , Leukemia Inhibitory Factor/metabolism , Placenta Diseases/physiopathology , Placentation , Animals , Female , Leukemia Inhibitory Factor/antagonists & inhibitors , Leukemia Inhibitory Factor/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyethylene Glycols/pharmacology , Pregnancy , Receptors, OSM-LIF/antagonists & inhibitors
9.
Endocrinology ; 155(7): 2602-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24708242

ABSTRACT

Emerging evidence suggest sex-specific regulation of dopamine neurons may underlie susceptibility of males to disorders such as Parkinson's disease (PD). In healthy male dopamine neurons, the Y-chromosome gene product, the sex-determining region on the Y chromosome (SRY) modulates dopamine biosynthesis and motor function. We investigated the regulation and function of SRY in a model of dopamine cell injury. Treatment with the dopaminergic toxin, 6-hydroxydopamine (6-OHDA), significantly elevated SRY mRNA expression (9-fold) in human male dopamine M17 cells. SRY up-regulation occurred via the p-quinone pathway, associated with a 3.5-fold increase in expression of GADD45γ, a DNA damage inducible factor gene and known SRY regulator. In turn, a signaling cascade involving GADD45γ/p38-MAPK/GATA activated the SRY promoter. Knockdown of SRY mRNA in 6-OHDA-treated M17 cells was deleterious, increasing levels of reactive oxygen species (ROS), pro-apoptotic marker PUMA mRNA, and cell injury (+25%, +32% and +34%, respectively). Conversely, ectopic over-expression of SRY in 6-OHDA-treated female SH-SY5Y cells was protective, decreasing ROS, PUMA, and cell injury (-40%, -46%, and -30%, respectively). However, the 6-OHDA-induced increase in SRY expression was diminished with higher concentrations of toxins or with chronic exposure to 6-OHDA. We conclude that SRY upregulation after dopamine cell injury is initially a protective response in males, but diminishes with gradual loss in dopamine cells. We speculate that dysregulation of SRY may contribute the susceptibility of males to PD.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Sex-Determining Region Y Protein/genetics , Up-Regulation/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Benzoquinones/metabolism , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Dopaminergic Neurons/drug effects , GATA Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen Peroxide/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Oxidopamine/pharmacology , PC12 Cells , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Rats , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sex-Determining Region Y Protein/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , GADD45 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...