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.
Mol Ther ; 29(5): 1903-1917, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33484963

ABSTRACT

Ornithine transcarbamylase deficiency (OTCD) is a monogenic disease of ammonia metabolism in hepatocytes. Severe disease is frequently treated by orthotopic liver transplantation. An attractive approach is the correction of a patient's own cells to regenerate the liver with gene-repaired hepatocytes. This study investigates the efficacy and safety of ex vivo correction of primary human hepatocytes. Hepatocytes isolated from an OTCD patient were genetically corrected ex vivo, through the deletion of a mutant intronic splicing site achieving editing efficiencies >60% and the restoration of the urea cycle in vitro. The corrected hepatocytes were transplanted into the liver of FRGN mice and repopulated to high levels (>80%). Animals transplanted and liver repopulated with genetically edited patient hepatocytes displayed normal ammonia, enhanced clearance of an ammonia challenge and OTC enzyme activity, as well as lower urinary orotic acid when compared to mice repopulated with unedited patient hepatocytes. Gene expression was shown to be similar between mice transplanted with unedited or edited patient hepatocytes. Finally, a genome-wide screening by performing CIRCLE-seq and deep sequencing of >70 potential off-targets revealed no unspecific editing. Overall analysis of disease phenotype, gene expression, and possible off-target editing indicated that the gene editing of a severe genetic liver disease was safe and effective.


Subject(s)
Gene Editing/methods , Hepatocytes/transplantation , Mutation , Ornithine Carbamoyltransferase Deficiency Disease/therapy , Ornithine Carbamoyltransferase/genetics , Adult , Aged , Ammonia/metabolism , Animals , Cells, Cultured , Child , Disease Models, Animal , Female , Gene Expression Regulation , Hepatocytes/chemistry , Hepatocytes/cytology , Humans , Introns , Male , Mice , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Orotic Acid/urine , RNA Splicing
2.
Cells ; 9(7)2020 07 15.
Article in English | MEDLINE | ID: mdl-32679793

ABSTRACT

Perinatal stem cells and epithelial cells isolated from full term amnion membrane, in particular, have attracted interest over the last decade, as a promising source of multipotent cells for cellular therapies. Human amnion epithelial cells (hAEC) have been used to treat monogenetic liver disease such as maple syrup urine disease or fibrosis of the liver in preclinical studies. In most studies xeno-transplants of hAEC were conducted without providing immunosuppression to recipients, reflecting the tolerogenic properties of hAEC. For many cell types, successful cryopreservation is critical for providing a readily available, off-the-shelf product. In this study, hAEC were isolated from full-term human placenta from 14 different donors, cryopreserved using a protocol and reagents commonly adopted for epithelial cell preservation. The cells were analyzed in terms of survival, recovery, and homogeneity, profiled for surface markers characteristic of epithelial, mesenchymal, endothelial, or hematopoietic cells. There were no significant differences observed in the percentage of cells with epithelial cell markers before and after cryopreservation. The relative proportion of stromal and hematopoietic cells was significantly reduced in hAEC preparations after cryopreservation. The expression of stem cell and immunomodulatory molecules were confirmed in the final product. Since multipotent cells are readily available from full-term placenta, this novel cell source might significantly increase the number of patients eligible to receive cellular therapies for liver and other diseases.


Subject(s)
Amnion/cytology , Cryopreservation , Epithelial Cells/cytology , Cell Survival , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Humans , Immunomodulation , Quality Control , Stem Cells/cytology , Stem Cells/metabolism
3.
Stem Cells Dev ; 28(14): 907-919, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31122128

ABSTRACT

Differentiation of stem cells to hepatocyte-like cells (HLCs) holds great promise for basic research, drug and toxicological investigations, and clinical applications. There are currently no protocols for the production of HLCs from stem cells, such as embryonic stem cells or induced pluripotent stem cells, that produce fully mature hepatocytes with a wide range of mature hepatic functions. This report describes a standard method to assess the maturation of stem cell-derived HLCs with a moderately high-throughput format, by analysing liver gene expression by quantitative RT-qPCR. This method also provides a robust data set of the expression of 62 genes expressed in normal liver, generated from 17 fetal and 25 mature human livers, so that investigators can quickly and easily compare the expression of these genes in their stem cell-derived HLCs with the values obtained in authentic fetal and mature human liver. The simple methods described in this study will provide a quick and accurate assessment of the efficacy of a differentiation protocol and will help guide the optimization of differentiation conditions.


Subject(s)
Cell Differentiation , Gene Expression Profiling , Gene Expression Regulation , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Liver/metabolism , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Liver/cytology
4.
J Inherit Metab Dis ; 42(6): 1054-1063, 2019 11.
Article in English | MEDLINE | ID: mdl-30843237

ABSTRACT

A liver-humanized mouse model for CPS1-deficiency was generated by the high-level repopulation of the mouse liver with CPS1-deficient human hepatocytes. When compared with mice that are highly repopulated with CPS1-proficient human hepatocytes, mice that are repopulated with CPS1-deficient human hepatocytes exhibited characteristic symptoms of human CPS1 deficiency including an 80% reduction in CPS1 metabolic activity, delayed clearance of an ammonium chloride infusion, elevated glutamine and glutamate levels, and impaired metabolism of [15 N]ammonium chloride into urea, with no other obvious phenotypic differences. Because most metabolic liver diseases result from mutations that alter critical pathways in hepatocytes, a model that incorporates actual disease-affected, mutant human hepatocytes is useful for the investigation of the molecular, biochemical, and phenotypic differences induced by that mutation. The model is also expected to be useful for investigations of modified RNA, gene, and cellular and small molecule therapies for CPS1-deficiency. Liver-humanized models for this and other monogenic liver diseases afford the ability to assess the therapy on actual disease-affected human hepatocytes, in vivo, for long periods of time and will provide data that are highly relevant for investigations of the safety and efficacy of gene-editing technologies directed to human hepatocytes and the translation of gene-editing technology to the clinic.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , Carbamoyl-Phosphate Synthase I Deficiency Disease/pathology , Hepatocytes/transplantation , Hydrolases/genetics , Liver/metabolism , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Cells, Cultured , Child , Disease Models, Animal , Female , Hepatocytes/metabolism , Humans , Hydrolases/metabolism , Infant , Infant, Newborn , Liver/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Organ Specificity/genetics
5.
Cytotherapy ; 21(1): 113-124, 2019 01.
Article in English | MEDLINE | ID: mdl-30409699

ABSTRACT

Placenta is a non-controversial and promising source of cells for the treatment of several liver diseases. We previously reported that transplanted human amnion epithelial cells (hAECs) differentiate into hepatocyte-like cells, resulting in correction of mouse models of metabolic liver disease or acute hepatic failure. As part of preclinical safety studies, we investigated the distribution of hAECs using two routes of administration to efficiently deliver hAECs to the liver. Optical imaging is commonly used because it can provide fast, high-throughput, whole-body imaging, thus DiR-labeled hAECs were injected into immunodeficient mice, via the spleen or the tail vein. The cell distribution was monitored using an in vivo imaging system over the next 24 h. After splenic injection, the DiR signal was detected in liver and spleen at 1, 3 and 24 h post-transplant. The distribution was confirmed by analysis of human DNA content at 24 h post-transplant and human-specific cytokeratin 8/18 staining. Tail vein infusion resulted in cell engraftment mainly in the lungs, with minimal detection in the liver. Delivery of cells to the portal vein, via the spleen, resulted in efficient delivery of hAECs to the liver, with minimal, off-target distribution to lungs or other organs.


Subject(s)
Amnion/cytology , Cell Transplantation/methods , Epithelial Cells/transplantation , Liver Diseases/therapy , Placenta/cytology , Animals , Cells, Cultured , Female , Humans , Liver/metabolism , Lung , Mice , Models, Animal , Portal Vein , Pregnancy , Spleen , Splenic Vein
6.
J Immunol ; 202(3): 724-735, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30587530

ABSTRACT

This study investigates the mechanism(s) underlying the immunoregulatory activities of placenta-derived human amnion epithelial cells (hAEC). The working hypothesis is that NAD+ and ATP, along with ectoenzymes involved in their metabolism, play a significant role in hAEC-mediated immune regulation. Proof of principle of the hypothesis was obtained by analyzing the interactions between hAEC and the main human leukocyte populations. The results obtained indicate that hAEC constitutively express a unique combination of functional ectoenzymes, driving the production of adenosine (ADO) via canonical (CD39, CD73) and alternative (CD38, CD203a/PC-1, CD73) pathways. Further, the picture is completed by the observation that hAEC express A1, A2a, and A2b ADO receptors as well as ADO deaminase, the enzyme involved in ADO catabolism. The contribution of the purinergic mediator to immunomodulation was confirmed by exposing in vitro different immune effector cells to the action of primary hAECs. B cells showed an enhanced proliferation and diminished spontaneous apoptosis when in contact with hAEC. T cell proliferation was partially inhibited by hAEC through ADO production, as confirmed by using specific ectoenzyme inhibitors. Further, hAEC induced an expansion of both T and B regulatory cells. Last, hAEC inhibited NK cell proliferation. However, the involvement of ADO-producing ectoenzymes is less apparent in this context. In conclusion, hAEC exert different in vitro immunoregulatory effects, per se, as a result of interactions with different populations of immune effector cells. These results support the view that hAEC are instrumental for regenerative medicine as well as in therapeutic applications for immune-related diseases.


Subject(s)
Adenosine Deaminase/metabolism , Adenosine/biosynthesis , Amnion/cytology , Cell Proliferation , Epithelial Cells/enzymology , Adenosine Deaminase/genetics , B-Lymphocytes/cytology , Cells, Cultured , Epithelial Cells/immunology , Humans , Killer Cells, Natural/cytology , Lymphocyte Activation , Metabolic Networks and Pathways , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , T-Lymphocytes/cytology
7.
Curr Protoc Stem Cell Biol ; 37: 1E.10.1-1E.10.13, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27171794

ABSTRACT

Different cell types can be isolated from human placental tissues, and some have been reported to retain phenotypic plasticity and characteristics that make them a promising source of cells for regenerative medicine. Among these are human amnion epithelial cells (hAECs). Adoption of current good manufacturing practices (cGMP) and enhanced quality control is essential when isolating hAECs in order to deliver a safe and effective cellular product for clinical purposes. This unit describes a detailed protocol for selective isolation of hAECs from human term placenta with little to no contamination by other cell types. A method for characterizing the heterogeneity of the hAEC suspension is also provided. The resulting cell product will be useful for clinical as well as basic research applications. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Amnion/cytology , Cell Separation/methods , Cell Separation/standards , Epithelial Cells/cytology , Female , Flow Cytometry , Humans , Placenta/cytology , Pregnancy
8.
Biol Open ; 4(12): 1782-91, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26621830

ABSTRACT

Epithelial invagination is a morphological process in which flat cell sheets transform into three-dimensional structures through bending of the tissue. It is accompanied by apical constriction, in which the apical cell surface is reduced in relation to the basal cell surface. Although much is known about the intra-cellular molecular machinery driving apical constriction and epithelial invagination, information of how extra-cellular signals affect these processes remains insufficient. In this study we have established several in vivo assays of placodal invagination to explore whether the external signal BMP regulates processes connected to epithelial invagination. By inhibiting BMP activity in prospective cranial placodes, we provide evidence that BMP signals are required for RhoA and F-actin rearrangements, apical constriction, cell elongation and epithelial invagination. The failure of placode invagination after BMP inhibition appears to be a direct consequence of disrupted apical accumulation of RhoA and F-actin, rather than changes in cell death or proliferation. In addition, our results show that epithelial invagination and acquisition of placode-specific identities are two distinct and separable developmental processes. In summary, our results provide evidence that BMP signals promote epithelial invagination by acting upstream of the intracellular molecular machinery that drives apical constriction and cell elongation.

9.
Eur Surg Res ; 54(3-4): 162-77, 2015.
Article in English | MEDLINE | ID: mdl-25633583

ABSTRACT

Since the first human hepatocyte transplants (HTx) in 1992, clinical studies have clearly established proof of principle for this therapy as a treatment for patients with acquired or inherited liver disease. Although major accomplishments have been made, there are still some specific limitations to this technology, which, if overcome, could greatly enhance the efficacy and implementation of this therapy. Here, we describe what in our view are the most significant obstacles to the clinical application of HTx and review the solutions currently proposed. The obstacles of significance include the limited number and quality of liver tissues as a cell source, the lack of clinical grade reagents, quality control evaluation of hepatocytes prior to transplantation, hypothermic storage of cells prior to transplantation, preconditioning treatments to enhance engraftment and proliferation of donor cells, tracking or monitoring cells after transplantation, and the optimal immunosuppression protocols for transplant recipients.


Subject(s)
Hepatocytes/transplantation , Liver Diseases/therapy , Cell Separation , Humans , Immunosuppression Therapy , Liver/cytology , Liver/physiology , Liver/surgery , Preservation, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...