Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Stem Cell Res Ther ; 15(1): 71, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38475825

ABSTRACT

BACKGROUND: Liver transplantation remains the only curative treatment for end-stage liver diseases. Unfortunately, there is a drastic organ donor shortage. Hepatocyte transplantation emerged as a viable alternative to liver transplantation. Considering their unique expansion capabilities and their potency to be driven toward a chosen cell fate, pluripotent stem cells are extensively studied as an unlimited cell source of hepatocytes for cell therapy. It has been previously shown that freshly prepared hepatocyte-like cells can cure mice from acute and chronic liver failure and restore liver function. METHODS: Human PSC-derived immature hepatic progenitors (GStemHep) were generated using a new protocol with current good manufacturing practice compliant conditions from PSC amplification and hepatic differentiation to cell cryopreservation. The therapeutic potential of these cryopreserved cells was assessed in two clinically relevant models of acute liver failure, and the mode of action was studied by several analytical methods, including unbiased proteomic analyses. RESULTS: GStemHep cells present an immature hepatic phenotype (alpha-fetoprotein positive, albumin negative), secrete hepatocyte growth factor and do not express major histocompatibility complex. A single dose of thawed GStemHep rescue mice from sudden death caused by acetaminophen and thioacetamide-induced acute liver failure, both in immunodeficient and immunocompetent animals in the absence of immunosuppression. Therapeutic biological effects were observed as soon as 3 h post-cell transplantation with a reduction in serum transaminases and in liver necrosis. The swiftness of the therapeutic effect suggests a paracrine mechanism of action of GStemHep leading to a rapid reduction of inflammation as well as a rapid cytoprotective effect with as a result a proteome reprograming of the host hepatocytes. The mode of action of GStemHep relie on the alleviation of inhibitory factors of liver regeneration, an increase in proliferation-promoting factors and a decrease in liver inflammation. CONCLUSIONS: We generated cryopreserved and current good manufacturing practice-compliant human pluripotent stem cell-derived immature hepatic progenitors that were highly effective in treating acute liver failure through rapid paracrine effects reprogramming endogenous hepatocytes. This is also the first report highlighting that human allogeneic cells could be used as cryopreserved cells and in the absence of immunosuppression for human PSC-based regenerative medicine for acute liver failure.


Subject(s)
Liver Failure, Acute , Pluripotent Stem Cells , Humans , Animals , Mice , Proteomics , Liver/metabolism , Hepatocytes/metabolism , Liver Failure, Acute/therapy , Cell Differentiation , Inflammation/metabolism
2.
Xenotransplantation ; 27(1): e12544, 2020 01.
Article in English | MEDLINE | ID: mdl-31342573

ABSTRACT

Pluripotent stem cells have been investigated as a renewable source of therapeutic hepatic cells, in order to overcome the lack of transplantable donor hepatocytes. Whereas different studies were able to correct hepatic defects in animal models, they focused on the most mature phenotype of hepatocyte-like cells (HLCs) derived from pluripotent stem cells and needed freshly prepared cells, which limits clinical applications of HLCs. Here, we report the production of hepatic stem cells (pHSCs) from human-induced pluripotent stem cells (hiPSCs) in xeno-free, feeder-free, and chemically defined conditions using as extracellular matrix a recombinant laminin instead of Matrigel, an undefined animal-derived matrix. Freshly prepared and frozen pHSCs were transplanted via splenic injection in Gunn rats, the animal model for Crigler-Najjar syndrome. Following cell transplantation and daily immunosuppression treatment, bilirubinemia was significantly decreased (around 30% decrease, P < .05) and remained stable throughout the 6-month study. The transplanted pHSCs underwent maturation in vivo to restore the deficient metabolic hepatic function (bilirubin glucuronidation by UGT1A1). In conclusion, we demonstrate for the first time the differentiation of hiPSCs into pHSCs that (a) are produced using a differentiation protocol compatible with Good Manufacturing Practices, (b) can be frozen, and (c) are sufficient to demonstrate in vivo therapeutic efficacy to significantly lower hyperbilirubinemia in a model of inherited liver disease, despite their immature phenotype. Thus, our approach provides major advances toward future clinical applications and would facilitate cell therapy manufacturing from human pluripotent stem cells.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Crigler-Najjar Syndrome/therapy , Hepatocytes/physiology , Hyperbilirubinemia/therapy , Induced Pluripotent Stem Cells/physiology , Liver/physiology , Stem Cell Transplantation/methods , Animals , Cell Differentiation , Cells, Cultured , Cryopreservation , Disease Models, Animal , Humans , Liver/surgery , Rats , Rats, Gunn , Regenerative Medicine/methods , Transplantation, Heterologous
3.
Stem Cell Res Ther ; 10(1): 221, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358055

ABSTRACT

BACKGROUND: Familial hypercholesterolemia type IIA (FH) is due to mutations in the low-density lipoprotein receptor (LDLR) resulting in elevated levels of low-density lipoprotein cholesterol (LDL-c) in plasma and in premature cardiovascular diseases. As hepatocytes are the only cells capable of metabolizing cholesterol, they are therefore the target cells for cell/gene therapy approaches in the treatment of lipid metabolism disorders. Furthermore, the LDLR has been reported to be involved in hepatitis C virus (HCV) entry into hepatocytes; however, its role in the virus infection cycle is still disputed. METHODS: We generated induced pluripotent stem cells (iPSCs) from a homozygous LDLR-null FH-patient (FH-iPSCs). We constructed a correction cassette bearing LDLR cDNA under the control of human hepatic apolipoprotein A2 promoter that targets the adeno-associated virus integration site AAVS1. We differentiated both FH-iPSCs and corrected FH-iPSCs (corr-FH-iPSCs) into hepatocytes to study statin-mediated regulation of genes involved in cholesterol metabolism. Upon HCV particle inoculation, viral replication and production were quantified in these cells. RESULTS: We showed that FH-iPSCs displayed the disease phenotype. Using homologous recombination mediated by the CRISPR/Cas9 system, FH-iPSCs were genetically corrected by the targeted integration of a correction cassette at the AAVS1 locus. Both FH-iPSCs and corr-FH-iPSCs were then differentiated into functional polarized hepatocytes using a stepwise differentiation approach (FH-iHeps and corr-FH-iHeps). The correct insertion and expression of the correction cassette resulted in restoration of LDLR expression and function (LDL-c uptake) in corr-FH-iHeps. We next demonstrated that pravastatin treatment increased the expression of genes involved in cholesterol metabolism in both cell models. Moreover, LDLR expression and function were also enhanced in corr-FH-iHeps after pravastatin treatment. Finally, we demonstrated that both FH-iHeps and corr-FH-iHeps were as permissive to viral infection as primary human hepatocytes but that virus production in FH-iHeps was significantly decreased compared to corr-FH-iHeps, suggesting a role of the LDLR in HCV morphogenesis. CONCLUSIONS: Our work provides the first LDLR-null FH cell model and its corrected counterpart to study the regulation of cholesterol metabolism and host determinants of HCV life cycle, and a platform to screen drugs for treating dyslipidemia and HCV infection.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Hepatitis C/pathology , Hyperlipoproteinemia Type II/pathology , Receptors, LDL/genetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Apolipoprotein A-II/genetics , Cell Differentiation , Cholesterol/metabolism , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatitis C/drug therapy , Hepatitis C/virology , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hyperlipoproteinemia Type II/metabolism , Induced Pluripotent Stem Cells/cytology , Phenotype , Plasmids/genetics , Plasmids/metabolism , Promoter Regions, Genetic , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Receptors, LDL/metabolism , Sofosbuvir/pharmacology , Sofosbuvir/therapeutic use , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
4.
Sci Rep ; 8(1): 8222, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844473

ABSTRACT

Despite decades of investigation on the proliferation of adult human primary hepatocytes, their expansion in vitro still remains challenging. To later be able to consider hepatocytes as a cell therapy alternative or bridge to liver transplantation, dramatically impeded by a shortage in liver donors, the first step is having an almost unlimited source of these cells. The banking of transplantable hepatocytes also implies a protocol for their expansion that can be compatible with large-scale production. We show that adult human primary hepatocytes when grown in 3D organoids are easily amplified, providing a substantial source of functional hepatocytes ready for transplantation. Following their plating, differentiated human hepatocytes are amplified during a transient and reversible step as liver progenitors, and can subsequently be converted back to mature differentiated hepatocytes. The protocol we propose is not only compatible with automated and high-throughput cell culture systems, thanks to the expansion of hepatocytes in suspension, but also guarantees the generation of a high number of functional cells from the same patient sample, with a relatively easy set up.


Subject(s)
Hepatocytes/cytology , Organoids/cytology , Stem Cells/cytology , Adult , Aged , Cell Differentiation , Cells, Cultured , Collagen , Drug Combinations , Female , Humans , Laminin , Male , Proteoglycans , Tissue Engineering
5.
PLoS One ; 9(9): e107324, 2014.
Article in English | MEDLINE | ID: mdl-25211016

ABSTRACT

With the goal of identifying splicing alterations in myotonic dystrophy 1 (DM1) tissues that may yield insights into targets or mechanisms, we have surveyed mis-splicing events in three systems using a RT-PCR screening and validation platform. First, a transgenic mouse model expressing CUG-repeats identified splicing alterations shared with other mouse models of DM1. Second, using cell cultures from human embryonic muscle, we noted that DM1-associated splicing alterations were significantly enriched in cytoskeleton (e.g. SORBS1, TACC2, TTN, ACTN1 and DMD) and channel (e.g. KCND3 and TRPM4) genes. Third, of the splicing alterations occurring in adult DM1 tissues, one produced a dominant negative variant of the splicing regulator RBFOX1. Notably, half of the splicing events controlled by MBNL1 were co-regulated by RBFOX1, and several events in this category were mis-spliced in DM1 tissues. Our results suggest that reduced RBFOX1 activity in DM1 tissues may amplify several of the splicing alterations caused by the deficiency in MBNL1.


Subject(s)
Muscle, Skeletal/metabolism , Myotonic Dystrophy/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology , Adult , Alternative Splicing , Animals , Cell Line , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Myoblasts/metabolism , Myotonic Dystrophy/genetics , RNA Splicing Factors , RNA-Binding Proteins/metabolism
6.
Anaerobe ; 17(1): 23-31, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20152920

ABSTRACT

Strain R1333, isolated from commercially available smoked salmon, was identified as Lactobacillus sakei based on biochemical tests, sugar fermentation reactions (API 50 CHL), PCR with species-specific primers and sequencing of the 16S rRNA gene. Strain R1333 produces a 3811 kDa class IIa bacteriocin, active against Streptococcus caprinus, Streptococcus macedonicus, Streptococcus spp., L. sakei, Lactococcus lactis subsp. lactis, Listeria innocua, Listeria ivanovii subsp. ivanovii and Listeria monocytogenes. The mode of activity against L. innocua 2030C and L. ivanovii subsp. ivanovii ATCC 19119 was bactericidal, resulting in cell lysis and enzyme- and DNA-leakage. The highest level of activity (1600 AU/mL) was recorded when cells were grown at 30°C in MRS broth (initial pH 6.5). Only 800 AU/mL was recorded when strain R1333 was grown in MRS without Tween 80. Lower levels of bacteriocin production were recorded when strain R1333 was grown in MRS at 20°C. Peptide R1333 adsorbs at low levels (200 AU/mL) to producer cells. Purification of bacteriocin R1333 was performed by 60% ammonium sulfate precipitation, followed by separation on a SepPak C(18) column and reverse-phase HPLC on a Nucleosil C(18) column with a linear gradient from 0.1% TFA to 90% acetonitryl. A molecular mass of 3811 kDa was determined by mass spectrometry. Based on mass spectrometry and sequencing of the PCR amplified fragment targeting the sakG gene, L. sakei R1333 is a potential producer of sakacin G. This is the first report of the identification of sakacin G produced by L. sakei isolated from smoked salmon.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacteriocins/metabolism , Food Microbiology , Lactobacillus/metabolism , Salmon/microbiology , Animals , Anti-Bacterial Agents/isolation & purification , Bacterial Typing Techniques , Bacteriocins/chemistry , Bacteriocins/isolation & purification , Chemical Fractionation , Chromatography, High Pressure Liquid , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Lactobacillus/classification , Lactobacillus/drug effects , Lactobacillus/isolation & purification , Lactococcus/drug effects , Listeria/drug effects , Mass Spectrometry , Molecular Weight , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Streptococcus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...