Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 30(12): 1610-1623.e7, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38065068

ABSTRACT

Hematopoietic stem cells (HSCs) are the rare cells responsible for the lifelong curative effects of hematopoietic cell (HC) transplantation. The demand for clinical-grade HSCs has increased significantly in recent decades, leading to major difficulties in treating patients. A promising but not yet achieved goal is the generation of HSCs from pluripotent stem cells. Here, we have obtained vector- and stroma-free transplantable HSCs by differentiating human induced pluripotent stem cells (hiPSCs) using an original one-step culture system. After injection into immunocompromised mice, cells derived from hiPSCs settle in the bone marrow and form a robust multilineage hematopoietic population that can be serially transplanted. Single-cell RNA sequencing shows that this repopulating activity is due to a hematopoietic population that is transcriptionally similar to human embryonic aorta-derived HSCs. Overall, our results demonstrate the generation of HSCs from hiPSCs and will help identify key regulators of HSC production during human ontogeny.


Subject(s)
Hematopoietic Stem Cell Transplantation , Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Humans , Mice , Animals , Cell Differentiation , Hematopoietic Stem Cells , Bone Marrow
2.
Stem Cells Transl Med ; 8(8): 822-832, 2019 08.
Article in English | MEDLINE | ID: mdl-31037857

ABSTRACT

We previously demonstrated that intracardiac delivery of autologous peripheral blood-derived CD34+ stem cells (SCs), mobilized by granulocyte-colony stimulating factor (G-CSF) and collected by leukapheresis after myocardial infarction, structurally and functionally repaired the damaged myocardial area. When used for cardiac indication, CD34+ cells are now considered as Advanced Therapy Medicinal Products (ATMPs). We have industrialized their production by developing an automated device for ex vivo CD34+ -SC expansion, starting from a whole blood (WB) sample. Blood samples were collected from healthy donors after G-CSF mobilization. Manufacturing procedures included: (a) isolation of total nuclear cells, (b) CD34+ immunoselection, (c) expansion and cell culture recovery in the device, and (d) expanded CD34+ cell immunoselection and formulation. The assessment of CD34+ cell counts, viability, and immunophenotype and sterility tests were performed as quality tests. We established graft acceptance criteria and performed validation processes in three cell therapy centers. 59.4 × 106 ± 36.8 × 106 viable CD34+ cells were reproducibly generated as the final product from 220 ml WB containing 17.1 × 106 ± 8.1 × 106 viable CD34+ cells. CD34+ identity, genetic stability, and telomere length were consistent with those of basal CD34+ cells. Gram staining and mycoplasma and endotoxin analyses were negative in all cases. We confirmed the therapeutic efficacy of both CD34+ -cell categories in experimental acute myocardial infarct (AMI) in immunodeficient rats during preclinical studies. This reproducible, automated, and standardized expansion process produces high numbers of CD34+ cells corresponding to the approved ATMP and paves the way for a phase I/IIb study in AMI, which is currently recruiting patients. Stem Cells Translational Medicine 2019;8:822&832.


Subject(s)
Antigens, CD34/genetics , Automation, Laboratory/methods , Flow Cytometry/methods , Myocardial Infarction/therapy , Peripheral Blood Stem Cell Transplantation/methods , Peripheral Blood Stem Cells/cytology , Adult , Animals , Antigens, CD34/metabolism , Cells, Cultured , Clinical Trials as Topic , Humans , Immunophenotyping/methods , Male , Middle Aged , Peripheral Blood Stem Cells/metabolism , Primary Cell Culture/methods , Rats
3.
PLoS One ; 8(5): e62303, 2013.
Article in English | MEDLINE | ID: mdl-23658722

ABSTRACT

A role for caspase activation in erythroid differentiation has been established, yet its precise mode of action remains elusive. A drawback of all previous investigations on caspase activation in ex vivo erythroid differentiation is the lack of an in vitro model producing full enucleation of erythroid cells. Using a culture system which renders nearly 100% enucleated red cells from human CD34(+) cells, we investigated the role of active caspase-3 in erythropoiesis. Profound effects of caspase-3 inhibition were found on erythroid cell growth and differentiation when inhibitors were added to CD34(+) cells at the start of the culture and showed dose-response to the concentration of inhibitor employed. Enucleation was only reduced as a function of the reduced maturity of the culture and the increased cell death of mature cells while the majority of cells retained their ability to extrude their nuclei. Cell cycle analysis after caspase-3 inhibition showed caspase-3 to play a critical role in cell proliferation and highlighted a novel function of this protease in erythroid differentiation, i.e. its contribution to cell cycle regulation at the mitotic phase. While the effect of caspase-3 inhibitor treatment on CD34(+) derived cells was not specific to the erythroid lineage, showing a similar reduction of cell expansion in myeloid cultures, the mechanism of action in both lineages appeared to be distinct with a strong induction of apoptosis causing the decreased yield of myeloid cells. Using a series of colony-forming assays we were able to pinpoint the stage at which cells were most sensitive to caspase-3 inhibition and found activated caspase-3 to play a signalling role in erythroid differentiation by targeting mature BFU-E and CFU-E but not early BFU-E.


Subject(s)
Caspase 3/metabolism , Cell Differentiation , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/enzymology , Signal Transduction , Apoptosis/drug effects , Caspase Inhibitors/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Erythroid Precursor Cells/drug effects , G2 Phase/drug effects , Humans , Signal Transduction/drug effects
4.
Blood ; 118(19): 5071-9, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21885599

ABSTRACT

In vitro RBC production from stem cells could represent an alternative to classic transfusion products. Until now the clinical feasibility of this concept has not been demonstrated. We addressed the question of the capacity of cultured RBCs (cRBCs) to survive in humans. By using a culture protocol permitting erythroid differentiation from peripheral CD34(+) HSC, we generated a homogeneous population of cRBC functional in terms of their deformability, enzyme content, capacity of their hemoglobin to fix/release oxygen, and expression of blood group antigens. We then demonstrated in the nonobese diabetes/severe combined immunodeficiency mouse that cRBC encountered in vivo the conditions necessary for their complete maturation. These data provided the rationale for injecting into one human a homogeneous sample of 10(10) cRBCs generated under good manufacturing practice conditions and labeled with (51)Cr. The level of these cells in the circulation 26 days after injection was between 41% and 63%, which compares favorably with the reported half-life of 28 ± 2 days for native RBCs. Their survival in vivo testifies globally to their quality and functionality. These data establish the proof of principle for transfusion of in vitro-generated RBCs and path the way toward new developments in transfusion medicine. This study is registered at http://www.clinicaltrials.gov as NCT0929266.


Subject(s)
Erythrocyte Transfusion/methods , Animals , Antigens, CD34/blood , Blood Group Antigens/blood , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Erythrocyte Aging , Erythrocyte Deformability , Erythrocytes/cytology , Erythrocytes/immunology , Erythrocytes/metabolism , Erythropoiesis , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hemoglobins/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...