Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Handb Clin Neurol ; 205: 145-154, 2024.
Article in English | MEDLINE | ID: mdl-39341651

ABSTRACT

Cell and gene therapies (CGTs) are intended to address many different diseases, including widespread diseases with millions of patients. The success of CGTs thus depends on the practicability of scaling cell manufacturing to population. It is obvious that process integration and automation are key for the reproducibility, quality, cost-effectiveness, and scalability of cell manufacturing. Still, different manufacturing concepts can be considered depending on the characteristics of cell therapies such as the degree of ex vivo manipulation of cells, the intended treatment scheme for the underlying medical indication, the prevalence of the indication, and the cell dose per final drug product. Here, we explain the characteristics of cellular products and their implications from the perspective of a manufacturer. We outline and exemplify with a list of devices' different strategies and scaling options for CGT manufacturing considering technical and regulatory aspects in the early and late clinical development of cellular products. Finally, we address the need for appropriate in-process and quality controls and the regulatory requirements and options for improvements of a cellular product at different manufacturing stages.


Subject(s)
Cell- and Tissue-Based Therapy , Genetic Therapy , Humans , Genetic Therapy/methods , Cell- and Tissue-Based Therapy/methods
2.
Nat Biotechnol ; 38(9): 1061-1072, 2020 09.
Article in English | MEDLINE | ID: mdl-32341565

ABSTRACT

Methods for differentiating human pluripotent stem cells to pancreatic and liver lineages in vitro have been limited by the inability to identify and isolate distinct endodermal subpopulations specific to these two organs. Here we report that pancreatic and hepatic progenitors can be isolated using the surface markers CD177/NB1 glycoprotein and inducible T-cell costimulatory ligand CD275/ICOSL, respectively, from seemingly homogeneous definitive endoderm derived from human pluripotent stem cells. Anterior definitive endoderm (ADE) subpopulations identified by CD177 and CD275 show inverse activation of canonical and noncanonical WNT signaling. CD177+ ADE expresses and synthesizes the secreted WNT, NODAL and BMP antagonist CERBERUS1 and is specified toward the pancreatic fate. CD275+ ADE receives canonical Wnt signaling and is specified toward the liver fate. Isolated CD177+ ADE differentiates more homogeneously into pancreatic progenitors and into more functionally mature and glucose-responsive ß-like cells in vitro compared with cells from unsorted differentiation cultures.


Subject(s)
Endoderm/cytology , Endoderm/metabolism , Insulin-Secreting Cells/cytology , Isoantigens/metabolism , Receptors, Cell Surface/metabolism , Adolescent , Adult , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cell Lineage , Cytokines/metabolism , Female , GPI-Linked Proteins/metabolism , Humans , Inducible T-Cell Co-Stimulator Ligand/metabolism , Insulin-Secreting Cells/metabolism , Liver/cytology , Liver/metabolism , Male , Middle Aged , Pancreas/cytology , Pancreas/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Receptors, CXCR4/metabolism , Wnt Signaling Pathway/physiology , Young Adult
3.
Sci Rep ; 8(1): 3907, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29500387

ABSTRACT

Mesenchymal stromal cells (MSCs) are a promising cell source to develop cell therapy for many diseases. Human platelet lysate (PLT) is increasingly used as an alternative to foetal calf serum (FCS) for clinical-scale MSC production. To date, the global surface protein expression of PLT-expended MSCs (MSC-PLT) is not known. To investigate this, paired MSC-PLT and MSC-FCS were analysed in parallel using high-throughput flow cytometry for the expression of 356 cell surface proteins. MSC-PLT showed differential surface protein expression compared to their MSC-FCS counterpart. Higher percentage of positive cells was observed in MSC-PLT for 48 surface proteins, of which 13 were significantly enriched on MSC-PLT. This finding was validated using multiparameter flow cytometry and further confirmed by quantitative staining intensity analysis. The enriched surface proteins are relevant to increased proliferation and migration capacity, as well as enhanced chondrogenic and osteogenic differentiation properties. In silico network analysis revealed that these enriched surface proteins are involved in three distinct networks that are associated with inflammatory responses, carbohydrate metabolism and cellular motility. This is the first study reporting differential cell surface protein expression between MSC-PLT and MSC-FSC. Further studies are required to uncover the impact of those enriched proteins on biological functions of MSC-PLT.


Subject(s)
Blood Platelets/metabolism , Bone Marrow/growth & development , Chondrogenesis , Mesenchymal Stem Cells/cytology , Osteogenesis , Receptors, Cell Surface/metabolism , Bone Marrow/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , Mesenchymal Stem Cells/metabolism , Phenotype
4.
Stem Cell Reports ; 9(4): 1207-1220, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28943253

ABSTRACT

Human pluripotent stem cell (hPSC)-derived mesencephalic dopaminergic (mesDA) neurons can relieve motor deficits in animal models of Parkinson's disease (PD). Clinical translation of differentiation protocols requires standardization of production procedures, and surface-marker-based cell sorting is considered instrumental for reproducible generation of defined cell products. Here, we demonstrate that integrin-associated protein (IAP) is a cell surface marker suitable for enrichment of hPSC-derived mesDA progenitor cells. Immunomagnetically sorted IAP+ mesDA progenitors showed increased expression of ventral midbrain floor plate markers, lacked expression of pluripotency markers, and differentiated into mature dopaminergic (DA) neurons in vitro. Intrastriatal transplantation of IAP+ cells sorted at day 16 of differentiation in a rat model of PD resulted in functional recovery. Grafts from sorted IAP+ mesDA progenitors were more homogeneous in size and DA neuron density. Thus, we suggest IAP-based sorting for reproducible prospective enrichment of mesDA progenitor cells in clinical cell replacement strategies.


Subject(s)
CD47 Antigen/metabolism , Dopamine/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Stem Cell Transplantation , Animals , Biomarkers , Cell Differentiation , Cell Survival , Cell- and Tissue-Based Therapy , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Graft Survival , Humans , Immunomagnetic Separation , Immunophenotyping , Mesencephalon/metabolism , Rats , Regeneration
5.
Nat Methods ; 13(5): 446-52, 2016 05.
Article in English | MEDLINE | ID: mdl-27088312

ABSTRACT

We report that the efficiency of reprogramming human somatic cells to induced pluripotent stem cells (hiPSCs) can be dramatically improved in a microfluidic environment. Microliter-volume confinement resulted in a 50-fold increase in efficiency over traditional reprogramming by delivery of synthetic mRNAs encoding transcription factors. In these small volumes, extracellular components of the TGF-ß and other signaling pathways exhibited temporal regulation that appears critical to acquisition of pluripotency. The high quality and purity of the resulting hiPSCs (µ-hiPSCs) allowed direct differentiation into functional hepatocyte- and cardiomyocyte-like cells in the same platform without additional expansion.


Subject(s)
Cellular Reprogramming Techniques/methods , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Microfluidics/methods , Cells, Cultured , Fibroblasts/cytology , Humans , RNA, Messenger/genetics
6.
Breast Cancer Res ; 17(1): 146, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26607327

ABSTRACT

INTRODUCTION: Chemotherapy resistance resulting in incomplete pathologic response is associated with high risk of metastasis and early relapse in breast cancer. The aim of this study was to identify and evaluate biomarkers of treatment-resistant tumor cells. METHODS: We performed a cell surface marker screen in triple-negative breast cancer patient-derived xenograft models treated with standard care genotoxic chemotherapy. Global expression profiling was used to further characterize the identified treatment-resistant subpopulations. RESULTS: High expression of sialyl-glycolipid stage-specific embryonic antigen 4 (SSEA4) was found in residual tumor cells surviving chemotherapy and in samples from metastatic patients who relapsed after neoadjuvant chemotherapy. Gene and microRNA (miRNA) expression profiling linked SSEA4 positivity with a mesenchymal phenotype and a deregulation of drug resistance pathways. Functional assays demonstrated a direct link between epithelial-mesenchymal transition (EMT) and SSEA4 expression. Interestingly, SSEA4 expression, EMT, and drug resistance seemed to be regulated posttranscriptionally. Finally, high expression of CMP-N-acetylneuraminate-ß-galactosamide-α-2,3-sialyltransferase 2 (ST3GAL2), the rate-limiting enzyme of SSEA4 synthesis, was found to be associated with poor clinical outcome in breast and ovarian cancer patients treated with chemotherapy. CONCLUSIONS: In this study, we identified SSEA4 as highly expressed in a subpopulation of tumor cells resistant to multiple commonly used chemotherapy drugs, as well as ST3GAL2, the rate-limiting enzyme of SSEA4 synthesis, as a predictive marker of poor outcome for breast and ovarian cancer patients undergoing chemotherapy. Both biomarkers and additionally identified regulatory miRNAs may be used to further understand chemoresistance, to stratify patient groups in order to avoid ineffective and painful therapies, and to develop alternative treatment regimens for breast cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Stage-Specific Embryonic Antigens/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Neoplasm Transplantation
7.
PLoS One ; 9(7): e102171, 2014.
Article in English | MEDLINE | ID: mdl-25029550

ABSTRACT

Cellular reprogramming of somatic cells into induced pluripotent stem cells (iPSC) opens up new avenues for basic research and regenerative medicine. However, the low efficiency of the procedure remains a major limitation. To identify iPSC, many studies to date relied on the activation of pluripotency-associated transcription factors. Such strategies are either retrospective or depend on genetically modified reporter cells. We aimed at identifying naturally occurring surface proteins in a systematic approach, focusing on antibody-targeted markers to enable live-cell identification and selective isolation. We tested 170 antibodies for differential expression between mouse embryonic fibroblasts (MEF) and mouse pluripotent stem cells (PSC). Differentially expressed markers were evaluated for their ability to identify and isolate iPSC in reprogramming cultures. Epithelial cell adhesion molecule (EPCAM) and stage-specific embryonic antigen 1 (SSEA1) were upregulated early during reprogramming and enabled enrichment of OCT4 expressing cells by magnetic cell sorting. Downregulation of somatic marker FAS was equally suitable to enrich OCT4 expressing cells, which has not been described so far. Furthermore, FAS downregulation correlated with viral transgene silencing. Finally, using the marker SSEA-1 we exemplified that magnetic separation enables the establishment of bona fide iPSC and propose strategies to enrich iPSC from a variety of human source tissues.


Subject(s)
Cell Separation/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , fas Receptor/metabolism , Animals , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cell Line , Cellular Reprogramming , Epithelial Cell Adhesion Molecule , Gene Expression Regulation , Humans , Lewis X Antigen/metabolism , Magnetics , Mice
8.
Glia ; 60(6): 894-907, 2012 May.
Article in English | MEDLINE | ID: mdl-22374709

ABSTRACT

Astrocytes show large morphological and functional heterogeneity and are involved in many aspects of neural function. Progress in defining astrocyte subpopulations has been hampered by the lack of a suitable antibody for their direct detection and isolation. Here, we describe a new monoclonal antibody, ACSA-1, which was generated by immunization of GLAST1 knockout mice. The antibody specifically detects an extracellular epitope of the astrocyte-specific L-glutamate/L-aspartate transporter GLAST (EAAT1, Slc1a3). As shown by immunohistochemistry, immunocytochemistry, and flow cytometry, ACSA-1 was cross-reactive for mouse, human, and rat. It labeled virtually all astrocytes positive for GFAP, GS, BLBP, RC2, and Nestin, including protoplastic, fibrous, and reactive astrocytes as well as Bergmann glia, Müller glia, and radial glia. Oligodendrocytes, microglia, neurons, and neuronal progenitors were negative for ACSA-1. Using an immunomagnetic approach, we established a method for the isolation of GLAST-positive cells with high purity. Binding of the antibody to GLAST and subsequent sorting of GLAST-positive cells neither interfered with cellular glutamate transport nor compromised astrocyte viability in vitro. The ACSA-1 antibody is not only a valuable tool to identify and track astrocytes by immunostaining, but also provides the possibility of separation and further analysis of pure astrocytes.


Subject(s)
Antibodies, Monoclonal/metabolism , Astrocytes/metabolism , Brain/cytology , Excitatory Amino Acid Transporter 1/immunology , Excitatory Amino Acid Transporter 1/metabolism , Animals , Animals, Newborn , Ascorbic Acid , Aspartic Acid/metabolism , Brain/metabolism , CD11b Antigen/metabolism , Cells, Cultured , Electroporation/methods , Excitatory Amino Acid Transporter 1/deficiency , Excitatory Amino Acid Transporter 1/pharmacology , Female , Flow Cytometry , Gangliosides/metabolism , Glutamate-Ammonia Ligase/metabolism , Humans , Magnesium , Mice , Mice, Knockout , Myelin Proteins/metabolism , Myelin-Oligodendrocyte Glycoprotein , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurons/metabolism , Rats , Sialic Acids/metabolism , Tritium/metabolism , Vitamin B 6
9.
Biochim Biophys Acta ; 1783(1): 34-48, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17950478

ABSTRACT

Latent transforming growth factor-beta binding proteins are a family of extracellular matrix proteins comprising four isoforms (LTBP-1, -2, -3, -4) with different structures, tissue expression patterns and affinity for TGF-beta. So far, respective knockout models have highlighted some essential functions for LTBP-2, LTBP-3 and LTBP-4, while the physiological significance of LTBP-1 is only superficially known. Here we report for the first time the generation and characterization of a mouse model lacking both the long and short LTBP-1 isoform. Surprisingly, respective mice are viable and fertile. However, detailed X-ray analysis of the skull revealed a modified facial profile. In addition, the gene disruption induces a reduced biological activity of TGF-beta that became evident in an experimental model of hepatic fibrogenesis in which the LTBP-1 knockout animals were less prone to hepatic fibrogenesis. Furthermore, comparative cDNA microarray gene expression profiling of cultured hepatic stellate cells confirmed that respective nulls were less receptive to cellular activation and transdifferentiation into myofibroblasts. Therefore, we conclude that LTBP-1 has essential functions in the control of TGF-beta activation.


Subject(s)
Face/anatomy & histology , Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Down-Regulation , Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Messenger/genetics , Up-Regulation/genetics
10.
Mol Biochem Parasitol ; 137(1): 65-74, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15279952

ABSTRACT

Eukaryotic translation initiation factor (eIF-5A) is a highly conserved and essential protein that contains the unique amino acid hypusine. The first step in the post-translational biosynthesis of hypusine, the transfer of an aminobutyl moiety from the polyamine substrate spermidine to the -amino group of a specific lysine residue in the eIF-5A precursor, is catalyzed by the enzyme deoxyhypusine synthase. A cDNA encoding a protein homologous to eIF-5A was isolated by plaque hybridization from a cDNA library of Plasmodium falciparum. The cloned cDNA contains an open reading frame encoding a protein of 161 amino acids, which shares a high sequence identity with other eukaryotic eIF-5A sequences. A phylogenetic tree constructed with eIF-5A from P. falciparum and 16 other eIF-5A sequences of eukaryotic and archaeal origin reveals that plasmodial eIF-5A together with other apicomplexan eIF-5A show a higher degree of homology to plant proteins than to animal and fungal sequences. The plasmodial eIF-5A gene was expressed as a six-histidine tagged fusion protein in Escherichia coli. Radioactive incorporation studies with [1,8-3H] spermidine indicated that this protein can serve as a substrate for human deoxyhypusine synthase. Results of quantitative real-time PCR studies with synchronized erythrocytic stages of P. falciparum revealed no significant induction or downregulation but only some variation in the expression level of plasmodial eIF-5A in ring, trophozoite and schizont stage.


Subject(s)
Peptide Initiation Factors/chemistry , Peptide Initiation Factors/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Conserved Sequence , DNA, Protozoan/analysis , DNA, Protozoan/chemistry , Erythrocytes/parasitology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Molecular Sequence Data , Open Reading Frames , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Initiation Factors/biosynthesis , Peptide Initiation Factors/isolation & purification , Phylogeny , Plasmodium falciparum/growth & development , RNA, Messenger/analysis , RNA, Protozoan/analysis , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology , Spermidine/metabolism , Eukaryotic Translation Initiation Factor 5A
SELECTION OF CITATIONS
SEARCH DETAIL