Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.730
Filtrer
1.
Methods Mol Biol ; 2835: 99-110, 2024.
Article de Anglais | MEDLINE | ID: mdl-39105909

RÉSUMÉ

Induced pluripotent stem cells (iPSCs) are generated through the reprogramming of somatic cells to an embryonic-like state by activating specific genes. They closely resemble embryonic stem cells (ESCs), in various aspects, including the expression of key stem cell genes, potency, and differentiation capabilities. iPSCs can be derived from various cell types such as fibroblasts, keratinocytes, and peripheral blood mononuclear cells (PBMCs). The ease of obtaining origin cells through non-invasive methods simplifies the generation of human iPSCs. Therefore, PBMCs are commonly preferred, with erythroid progenitor cells (EPCs) obtained through EPC enrichment being used as origin cells in this protocol. The EPC enrichment performed in this protocol not only reduces costs but also increases efficiency by enhancing the percentage of reprogrammable cells with progenitor characteristics. Human iPSCs are incredibly valuable for in vitro research, cell therapy, drug discovery, and tissue engineering. The outlined procedures below provide a general framework for inducing iPSCs from erythroid progenitor cells, pluripotency confirmation experiments, and cultivating them for downstream experiments.


Sujet(s)
Techniques de culture cellulaire , Différenciation cellulaire , Précurseurs érythroïdes , Cellules souches pluripotentes induites , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Précurseurs érythroïdes/cytologie , Précurseurs érythroïdes/métabolisme , Techniques de culture cellulaire/méthodes , Reprogrammation cellulaire/génétique , Cellules cultivées , Agranulocytes/cytologie , Agranulocytes/métabolisme , Fibroblastes/cytologie , Fibroblastes/métabolisme
2.
J Vis Exp ; (209)2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39141551

RÉSUMÉ

Peripheral blood mononuclear cells (PBMCs) are a heterogeneous population of monocytes and lymphocytes. Cryopreserved PBMCs have stable viability in long-term storage, making them an ideal cell type for many downstream research purposes, including flow cytometry, immunoassays, and genome sequencing. Typically, PBMCs are isolated via density gradient centrifugation, however, it is a low-throughput workflow that is difficult and costly to scale. This article presents a high-throughput workflow using a magnetic bead-based PBMC isolation method that is quick to implement. Total cell concentration, viability, and population distribution with PBMCs obtained using density gradient isolation were compared, and cell viability and proportion of cell types were comparable for both techniques. Isolated PBMCs demonstrated over 70% viability up to 9 days after blood collection, although yield decreased by half after 5 days compared to PBMCs processed within 24 h of collection. In summary, this article describes a PBMC protocol that utilizes a bead-based approach to adapt to a high throughput workflow and demonstrates that both manual and automated bead-based methods can increase processing capacity and provide flexibility for various budgets.


Sujet(s)
Séparation immunomagnétique , Agranulocytes , Humains , Agranulocytes/cytologie , Séparation immunomagnétique/méthodes , Buffy coat/cytologie
3.
Stem Cell Res ; 80: 103504, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39110999

RÉSUMÉ

We have successfully generated human induced pluripotent stem cells (hiPSC) from peripheral blood mononuclear cells (PBMCs) of a patient with COPA Syndrome. The patient, a 6 year old Caucasian male, has a spontaneous de novo missense mutation that replaced alanine with proline in the COPA gene. This paper confirms the differentiation potential of the hiPSC line, the presence of the p.Ala239Pro mutation, and the expression of typical pluripotency markers within the hiPSC line. The hiPSC line is ready for use as a cellular model of COPA Syndrome.


Sujet(s)
Cellules souches pluripotentes induites , Humains , Cellules souches pluripotentes induites/métabolisme , Mâle , Enfant , Lignée cellulaire , Hétérozygote , Différenciation cellulaire , Mutation , Agranulocytes/métabolisme , Agranulocytes/cytologie
4.
Cells ; 13(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38994943

RÉSUMÉ

Gingival fibroblasts (GFs) can differentiate into osteoblast-like cells and induce osteoclast precursors to differentiate into osteoclasts. As it is unclear whether these two processes influence each other, we investigated how osteogenic differentiation of GFs affects their osteoclast-inducing capacity. To establish step-wise mineralization, GFs were cultured in four groups for 3 weeks, without or with osteogenic medium for the final 1, 2, or all 3 weeks. The mineralization was assessed by ALP activity, calcium concentration, scanning electron microscopy (SEM), Alizarin Red staining, and quantitative PCR (qPCR). To induce osteoclast differentiation, these cultures were then co-cultured for a further 3 weeks with peripheral blood mononuclear cells (PBMCs) containing osteoclast precursors. Osteoclast formation was assessed at different timepoints with qPCR, enzyme-linked immunosorbent assay (ELISA), TRAcP activity, and staining. ALP activity and calcium concentration increased significantly over time. As confirmed with the Alizarin Red staining, SEM images showed that the mineralization process occurred over time. Osteoclast numbers decreased in the GF cultures that had undergone osteogenesis. TNF-α secretion, a costimulatory molecule for osteoclast differentiation, was highest in the control group. GFs can differentiate into osteoblast-like cells and their degree of differentiation reduces their osteoclast-inducing capacity, indicating that, with appropriate stimulation, GFs could be used in regenerative periodontal treatments.


Sujet(s)
Différenciation cellulaire , Fibroblastes , Gencive , Ostéoclastes , Ostéogenèse , Humains , Ostéoclastes/métabolisme , Ostéoclastes/cytologie , Gencive/cytologie , Fibroblastes/métabolisme , Fibroblastes/cytologie , Cellules cultivées , Calcium/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Techniques de coculture , Agranulocytes/cytologie , Agranulocytes/métabolisme
5.
Cells ; 13(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38994999

RÉSUMÉ

Natural killer (NK) cells hold promise in cancer treatment due to their ability to spontaneously lyse cancer cells. For clinical use, high quantities of pure, functional NK cells are necessary. Combining adherence-based isolation with specialized media showed the unreliability of the isolation method, but demonstrated the superiority of the NK MACS® medium, particularly in suboptimal conditions. Neither human pooled serum, fetal calf serum (FCS), human platelet lysate, nor chemically defined serum replacement could substitute human AB serum. Interleukin (IL-)2, IL-15, IL-21, and combined CD2/NKp46 stimulation were assessed. IL-21 and CD2/NKp46 stimulation increased cytotoxicity, but reduced NK cell proliferation. IL-15 stimulation alone achieved the highest proliferation, but the more affordable IL-2 performed similarly. The RosetteSep™ human NK cell enrichment kit was effective for isolation, but the presence of peripheral blood mononuclear cells (PBMCs) in the culture enhanced NK cell proliferation, despite similar expression levels of CD16, NKp46, NKG2D, and ICAM-1. In line with this, purified NK cells cultured in NK MACS® medium with human AB serum and IL-2 demonstrated high cytotoxicity against primary glioblastoma stem cells.


Sujet(s)
Prolifération cellulaire , Milieux de culture , Cellules tueuses naturelles , Humains , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Techniques de culture cellulaire/méthodes , Interleukine-2/métabolisme , Cytotoxicité immunologique , Interleukine-15/métabolisme , Agranulocytes/métabolisme , Agranulocytes/cytologie , Cellules souches tumorales/métabolisme , Glioblastome/immunologie , Glioblastome/anatomopathologie , Séparation cellulaire/méthodes
6.
Stem Cell Res ; 79: 103501, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39029293

RÉSUMÉ

Apolipoprotein E (APOE)is the gene with greatest genetic risk for Alzheimer's disease (AD). We successfully established a human induced pluripotent stem cell(iPSC) line from a woman mutated by APOE gene. The cell line was isolated from this woman's peripheral blood mononuclear cells using a non-integrated Sendai virus, which retained the original genotype, showed a normal karyotype, highly expressed pluripotent markers and could differentiate into three germ layers.


Sujet(s)
Apolipoprotéines E , Cellules souches pluripotentes induites , Mutation , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Femelle , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Lignée cellulaire , Différenciation cellulaire , Caryotype , Agranulocytes/métabolisme , Agranulocytes/cytologie
7.
Stem Cell Res Ther ; 15(1): 231, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075608

RÉSUMÉ

BACKGROUND: Hematopoietic stem and progenitor cells (HSPCs) mobilize from bone marrow to peripheral blood in response to stress. The impact of alloresponse-induced stress on HSPCs mobilization in human liver transplantation (LTx) recipients remains under-investigated. METHODS: Peripheral blood mononuclear cell (PBMC) samples were longitudinally collected from pre- to post-LTx for one year from 36 recipients with acute rejection (AR), 74 recipients without rejection (NR), and 5 recipients with graft-versus-host disease (GVHD). 28 PBMC samples from age-matched healthy donors were collected as healthy control (HC). Multi-color flow cytometry (MCFC) was used to immunophenotype HSPCs and their subpopulations. Donor recipient-distinguishable major histocompatibility complex (MHC) antibodies determined cell origin. RESULTS: Before LTx, patients who developed AR after transplant contained more HSPCs in PBMC samples than HC, while the NR group patients contained fewer HSPCs than HC. After LTx, the HSPC ratio in the AR group sharply decreased and became less than HC within six months, and dropped to a comparable NR level afterward. During the one-year follow-up period, myeloid progenitors (MPs) biased differentiation was observed in all LTx recipients who were under tacrolimus-based immunosuppressive treatment. During both AR and GVHD episodes, the recipient-derived and donor-derived HSPCs mobilized into the recipient's blood-circulation and migrated to the target tissue, respectively. The HSPCs percentage in blood reduced after the disease was cured. CONCLUSIONS: A preoperative high HSPC ratio in blood characterizes recipients who developed AR after LTx. Recipients exhibited a decline in blood-circulating HSPCs after transplant, the cells mobilized into the blood and migrated to target tissue during alloresponse.


Sujet(s)
Maladie du greffon contre l'hôte , Mobilisation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques , Transplantation hépatique , Humains , Mâle , Femelle , Mobilisation de cellules souches hématopoïétiques/méthodes , Adulte , Adulte d'âge moyen , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/cytologie , Rejet du greffon/immunologie , Donneurs de tissus , Agranulocytes/métabolisme , Agranulocytes/cytologie , Transplantation de cellules souches hématopoïétiques/méthodes
8.
Eur J Cell Biol ; 103(3): 151440, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38954934

RÉSUMÉ

One of the deficits of knowledge on bone remodelling, is to what extent cells that are driven towards osteogenic differentiation can contribute to osteoclast formation. The periodontal ligament fibroblast (PdLFs) is an ideal model to study this, since they play a role in osteogenesis, and can also orchestrate osteoclastogenesis.when co-cultured with a source of osteoclast-precursor such as peripheral blood mononuclear cells (PBMCs). Here, the osteogenic differentiation of PdLFs and the effects of this process on the formation of osteoclasts were investigated. PdLFs were obtained from extracted teeth and exposed to osteogenic medium for 0, 7, 14, or 21 out of 21 days. After this 21-day culturing period, the cells were co-cultured with peripheral blood mononuclear cells (PBMCs) for an additional 21 days to study osteoclast formation. Alkaline phosphatase (ALP) activity, calcium concentration, and gene expression of osteogenic markers were assessed at day 21 to evaluate the different stages of osteogenic differentiation. Alizarin red staining and scanning electron microscopy were used to visualise mineralisation. Tartrate-resistant acid phosphatase (TRAcP) activity, TRAcP staining, multinuclearity, the expression of osteoclastogenesis-related genes, and TNF-α and IL-1ß protein levels were assessed to evaluate osteoclastogenesis. The osteogenesis assays revealed that PdLFs became more differentiated as they were exposed to osteogenic medium for a longer period of time. Mineralisation by these osteogenic cells increased with the progression of differentiation. Culturing PdLFs in osteogenic medium before co-culturing them with PMBCs led to a significant decrease in osteoclast formation. qPCR revealed significantly lower DCSTAMP expression in cultures that had been supplemented with osteogenic medium. Protein levels of osteoclastogenesis stimulator TNF-α were also lower in these cultures. The present study shows that the osteogenic differentiation of PdLFs reduces the osteoclastogenic potential of these cells. Immature cells of the osteoblastic lineage may facilitate osteoclastogenesis, whereas mature mineralising cells may suppress the formation of osteoclasts. Therefore, mature and immature osteogenic cells may have different roles in maintaining bone homeostasis.


Sujet(s)
Différenciation cellulaire , Fibroblastes , Ostéoclastes , Ostéogenèse , Desmodonte , Desmodonte/cytologie , Desmodonte/métabolisme , Ostéoclastes/métabolisme , Ostéoclastes/cytologie , Humains , Fibroblastes/métabolisme , Fibroblastes/cytologie , Cellules cultivées , Techniques de coculture , Agranulocytes/métabolisme , Agranulocytes/cytologie
9.
Stem Cell Res ; 80: 103514, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39083857

RÉSUMÉ

The human leukocyte antigen (HLA) system comprises cell-surface proteins responsible for the presentation of peptide antigens. HLAs play an essential role in the regulation of the human immune system, and their studies have been crucial to its understanding. To create a sustainable model for the investigation of HLAs, we successfully generated the human iPSC line MURAi003-A derived from the peripheral blood mononuclear cells of a donor with homozygous Class I and Class II HLAs (A*11:01, B*46:01; C*01:02; DRB1*09:01; DQB1*03:03) using non-integrative reprogramming episomes. MURAi003-A exhibited pluripotent stem cell characteristics with consistent demonstrations of pluripotency and expression of stem cell-associated markers.


Sujet(s)
Cellules souches pluripotentes induites , Agranulocytes , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Agranulocytes/métabolisme , Agranulocytes/cytologie , Lignée cellulaire , Homozygote , Différenciation cellulaire , Antigènes d'histocompatibilité de classe II/métabolisme , Antigènes d'histocompatibilité de classe II/génétique , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/génétique
10.
FEBS Lett ; 598(15): 1909-1918, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38955545

RÉSUMÉ

The poliovirus (PV) enters the central nervous system (CNS) via the bloodstream, suggesting the existence of a mechanism to cross the blood-brain barrier. Here, we report that PV capsid proteins (VP1 and VP3) can penetrate cells, with VP3 being more invasive. Two independent parts of VP3 are responsible for this function. Both peptides can penetrate human umbilical cord vascular endothelial cells, and one peptide of VP3 could also penetrate peripheral blood mononuclear cells. In an in vitro blood-brain barrier model using rat-derived astrocytes, pericytes, and endothelial cells, both peptides were observed to traverse from the blood side to the brain side at 6 h after administration. These results provide insights into the molecular mechanisms underlying PV invasion into the CNS.


Sujet(s)
Barrière hémato-encéphalique , Protéines de capside , Poliovirus , Protéines de capside/métabolisme , Protéines de capside/génétique , Humains , Poliovirus/génétique , Poliovirus/métabolisme , Poliovirus/physiologie , Animaux , Rats , Barrière hémato-encéphalique/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Agranulocytes/métabolisme , Agranulocytes/cytologie , Astrocytes/métabolisme
11.
Cells ; 13(11)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38891103

RÉSUMÉ

Patients with chronic hypoxia show a higher tumor incidence; however, no primary common cause has been recognized. Given the similarities between cellular reprogramming and oncogenic transformation, we directly compared these processes in human cells subjected to hypoxia. Mouse embryonic fibroblasts were employed as controls to compare transfection and reprogramming efficiency; human adipose-derived mesenchymal stem cells were employed as controls in human cells. Easily obtainable human peripheral blood mononuclear cells (PBMCs) were chosen to establish a standard protocol to compare cell reprogramming (into induced pluripotent stem cells (iPSCs)) and oncogenic focus formation efficiency. Cell reprogramming was achieved for all three cell types, generating actual pluripotent cells capable for differentiating into the three germ layers. The efficiencies of the cell reprogramming and oncogenic transformation were similar. Hypoxia slightly increased the reprogramming efficiency in all the cell types but with no statistical significance for PBMCs. Various PBMC types can respond to hypoxia differently; lymphocytes and monocytes were, therefore, reprogrammed separately, finding a significant difference between normoxia and hypoxia in monocytes in vitro. These differences were then searched for in vivo. The iPSCs and oncogenic foci were generated from healthy volunteers and patients with chronic obstructive pulmonary disease (COPD). Although higher iPSC generation efficiency in the patients with COPD was found for lymphocytes, this increase was not statistically significant for oncogenic foci. Remarkably, a higher statistically significant efficiency in COPD monocytes was demonstrated for both processes, suggesting that physiological hypoxia exerts an effect on cell reprogramming and oncogenic transformation in vivo in at least some cell types.


Sujet(s)
Transformation cellulaire néoplasique , Reprogrammation cellulaire , Cellules souches pluripotentes induites , Humains , Reprogrammation cellulaire/génétique , Cellules souches pluripotentes induites/métabolisme , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Animaux , Souris , Hypoxie cellulaire , Agranulocytes/métabolisme , Agranulocytes/cytologie , Mâle , Femelle , Adulte d'âge moyen , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Différenciation cellulaire/génétique , Sujet âgé
12.
J Vis Exp ; (207)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38856231

RÉSUMÉ

Peripheral mononuclear cells (PBMCs) exhibit robust changes in mitochondrial respiratory capacity in response to health and disease. While these changes do not always reflect what occurs in other tissues, such as skeletal muscle, these cells are an accessible and valuable source of viable mitochondria from human subjects. PBMCs are exposed to systemic signals that impact their bioenergetic state. Thus, expanding our tools to interrogate mitochondrial metabolism in this population will elucidate mechanisms related to disease progression. Functional assays of mitochondria are often limited to using respiratory outputs following maximal substrate, inhibitor, and uncoupler concentrations to determine the full range of respiratory capacity, which may not be achievable in vivo. The conversion of adenosine diphosphate (ADP) to adenosine triphosphate (ATP) by ATP-synthase results in a decrease in mitochondrial membrane potential (mMP) and an increase in oxygen consumption. To provide a more integrated analysis of mitochondrial dynamics, this article describes the use of high-resolution fluorespirometry to measure the simultaneous response of oxygen consumption and mitochondrial membrane potential (mMP) to physiologically relevant concentrations of ADP. This technique uses tetramethylrhodamine methylester (TMRM) to measure mMP polarization in response to ADP titrations following maximal hyperpolarization with complex I and II substrates. This technique can be used to quantify how changes in health status, such as aging and metabolic disease, affect the sensitivity of mitochondrial response to energy demand in PBMCs, T-cells, and monocytes from human subjects.


Sujet(s)
Agranulocytes , Potentiel de membrane mitochondriale , Humains , Potentiel de membrane mitochondriale/physiologie , Agranulocytes/métabolisme , Agranulocytes/cytologie , Rhodamines/composition chimique , ADP/métabolisme , ADP/pharmacologie , Consommation d'oxygène/physiologie , Mitochondries/métabolisme , Colorants fluorescents/composition chimique
13.
Stem Cell Res ; 79: 103465, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38880015

RÉSUMÉ

Peripheral blood mononuclear cells (PBMCs) were obtained from a patient diagnosed with Familial Hemiplegic Migraine Type 3, who carried a heterozygous A > C mutation in the SCN1A gene and reprogrammed using CytoTuneTM-iPS 2.0 Sendai Reprogramming Kit. The iPSC line maintained the mutation while expressing markers of pluripotency. Additionally, it exhibited a normal karyotype and demonstrated potential for in vitro differentiation into cells representing all three embryonic germ layers.


Sujet(s)
Cellules souches pluripotentes induites , Agranulocytes , Humains , Cellules souches pluripotentes induites/métabolisme , Agranulocytes/métabolisme , Agranulocytes/cytologie , Différenciation cellulaire , Lignée cellulaire , Migraine avec aura/génétique , Canal sodique voltage-dépendant NAV1.1/génétique , Mâle , Femelle
14.
Nat Methods ; 21(7): 1166-1170, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38877315

RÉSUMÉ

The growth of omic data presents evolving challenges in data manipulation, analysis and integration. Addressing these challenges, Bioconductor provides an extensive community-driven biological data analysis platform. Meanwhile, tidy R programming offers a revolutionary data organization and manipulation standard. Here we present the tidyomics software ecosystem, bridging Bioconductor to the tidy R paradigm. This ecosystem aims to streamline omic analysis, ease learning and encourage cross-disciplinary collaborations. We demonstrate the effectiveness of tidyomics by analyzing 7.5 million peripheral blood mononuclear cells from the Human Cell Atlas, spanning six data frameworks and ten analysis tools.


Sujet(s)
Logiciel , Humains , Biologie informatique/méthodes , Agranulocytes/métabolisme , Agranulocytes/cytologie , Génomique/méthodes , Analyse de données
15.
Biomolecules ; 14(6)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38927093

RÉSUMÉ

Special attention is given to cow's milk and its variants, with ongoing discussions about health-related impacts primarily focusing on the A1 variant in contrast to the A2 variant. The difference between these variants lies in a single amino acid alteration at position 67 of ß-casein. This alteration is presumed to make the A1 variant more susceptible to enzymatic breakdown during milk digestion, leading to an increased release of the peptide ß-casomorphin-7 (BCM-7). BCM-7 is hypothesized to interact with µ-opioid receptors on immune cells in humans. Although BCM-7 has demonstrated both immunosuppressive and inflammatory effects, its direct impact on the immune system remains unclear. Thus, we examined the influence of A1 and A2 milk on Concanavalin A (ConA)-stimulated human peripheral blood mononuclear cells (PBMCs), as well as the effect of experimentally digested A1 and A2 milk, containing different amounts of free BCM-7 from ß-casein cleavage. Additionally, we evaluated the effects of pure BCM-7 on the proliferation of ConA-stimulated PBMCs and purified CD4+ T cells. Milk fundamentally inhibited PBMC proliferation, independent of the ß-casein variant. In contrast, experimentally digested milk of both variants and pure BCM-7 showed no influence on the proliferation of PBMCs or isolated CD4+ T cells. Our results indicate that milk exerts an anti-inflammatory effect on PBMCs, regardless of the A1 or A2 ß-casein variant, which is nullified after in vitro digestion. Consequently, we deem BCM-7 unsuitable as a biomarker for food-induced inflammation.


Sujet(s)
Caséines , Prolifération cellulaire , Endorphines , Agranulocytes , Lait , Fragments peptidiques , Humains , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Agranulocytes/cytologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lait/composition chimique , Endorphines/pharmacologie , Endorphines/métabolisme , Animaux , Caséines/pharmacologie , Caséines/métabolisme , Fragments peptidiques/pharmacologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD4+/cytologie , Concanavaline A/pharmacologie , Bovins
16.
Stem Cell Res ; 78: 103466, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38852425

RÉSUMÉ

The Rh-negative type O blood group (O Rh-) is considered a universal donor for emergency blood transfusions. Due to the constant shortage of this rare blood group, the production of blood cells from iPSCs derived from the O Rh- donor could potentially serve as a limitless blood source for transfusions. In this report, we establish a MUSIi017-A iPSC line from peripheral blood mononuclear cells of a healthy donor with the O Rh- blood group. The established iPSC line exhibited a normal karyotype, showed identical STR compared to donor peripheral blood mononuclear cells, and could differentiate to all three germ layers.


Sujet(s)
Cellules souches pluripotentes induites , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Lignée cellulaire , Système ABO de groupes sanguins , Agranulocytes/cytologie , Agranulocytes/métabolisme , Différenciation cellulaire , Donneurs de sang
17.
Cell Transplant ; 33: 9636897241251619, 2024.
Article de Anglais | MEDLINE | ID: mdl-38761062

RÉSUMÉ

Pressure injuries, or pressure ulcers, are a common problem that may lead to infections and major complications, besides being a social and economic burden due to the costs of treatment and hospitalization. While surgery is sometimes necessary, this also has complications such as recurrence or wound dehiscence. Among the newer methods of pressure injury treatment, advanced therapies are an interesting option. This study examines the healing properties of bone marrow mononuclear cells (BM-MNCs) embedded in a plasma-based scaffold in a mouse model. Pressure ulcers were created on the backs of mice (2 per mouse) using magnets and assigned to a group of ulcers that were left untreated (Control, n = 15), treated with plasma scaffold (Plasma, n = 15), or treated with plasma scaffold containing BM-MNC (Plasma + BM-MNC, n = 15). Each group was examined at three time points (3, 7, and 14 days) after the onset of treatment. At each time point, animals were subjected to biometric assessment, bioluminescence imaging, and tomography. Once treatment had finished, skin biopsies were processed for histological and wound healing reverse transcription polymerase chain reaction (RT-PCR) array studies. While wound closure percentages were higher in the Plasma and Plasma + BM-MNC groups, differences were not significant, and thus descriptive data are provided. In all individuals, the presence of donor cells was revealed by immunohistochemistry on posttreatment onset Days 3, 7, and 14. In the Plasma + BM-MNC group, less inflammation was observed by positron emission tomography-computed tomography (PET/CT) imaging of the mice at 7 days, and a complete morphometabolic response was produced at 14 days, in accordance with histological results. A much more pronounced inflammatory process was observed in controls than in the other two groups, and this persisted until Day 14 after treatment onset. RT-PCR array gene expression patterns were also found to vary significantly, with the greatest difference noted between both treatments at 14 days when 11 genes were differentially expressed.


Sujet(s)
Cellules de la moelle osseuse , Modèles animaux de maladie humaine , Escarre , Cicatrisation de plaie , Animaux , Escarre/thérapie , Escarre/anatomopathologie , Souris , Cellules de la moelle osseuse/cytologie , Mâle , Structures d'échafaudage tissulaires/composition chimique , Souris de lignée C57BL , Transplantation de moelle osseuse/méthodes , Agranulocytes/cytologie , Agranulocytes/métabolisme , Agranulocytes/transplantation
18.
Cell Transplant ; 33: 9636897241256462, 2024.
Article de Anglais | MEDLINE | ID: mdl-38808671

RÉSUMÉ

Regulatory cell therapies have shown promise in tolerance-induction protocols in living donor organ transplantation. These protocols should be pursued in deceased donor transplantation. Donor peripheral mononuclear cells (PBMCs) are an optimal source of donor antigens for the induction of donor-specific regulatory cells. During the development of a regulatory cell tolerance-induction protocol with organs from deceased donors, we compared 3 methods of obtaining PBMCs from deceased donors focusing on cell yield, viability, and contamination of unwanted cell types. PBMC procurement methods: 1. During organ procurement at the time of cold perfusion, blood was collected from the vena cava and placed into a 10-liter blood collection bag, and thereafter transported to Karolinska University Hospital, where leukapheresis was performed (BCL). 2. Blood was collected via the vena cava into blood donation bags before cold perfusion. The bags underwent buffy coat separation and thereafter automated leukocyte isolation system (BCS). 3. To collect PBMCs, leukapheresis was performed via a central dialysis catheter on deceased donors in the intensive care unit (ICU) prior to the organ procurement procedure (LEU).All 3 methods to obtain PBMC from deceased donors were safe and did not affect the procurement of organs. BCL contained around 50% of NK cells in lymphocytes population. LEU had a highest yield of donor PBMC among 3 groups. LEU had the lower amount of granulocyte contamination, compared to BCS and BCL. Based on these results, we choose LEU as the preferred method to obtain donor PBMC in the development of our tolerance-induction protocol.


Sujet(s)
Leucaphérèse , Agranulocytes , Donneurs de tissus , Humains , Agranulocytes/cytologie , Agranulocytes/métabolisme , Adulte , Adulte d'âge moyen , Mâle , Femelle , Leucaphérèse/méthodes , Sujet âgé , Tolérance immunitaire
19.
Stem Cell Res ; 77: 103434, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38703667

RÉSUMÉ

The Jra antigen, the only antigen within the JR blood group system, is a high-prevalence red blood cell (RBC) antigen found in over 99 % of the global population. An induced pluripotent stem cell line (YUCMi020-A) was generated from peripheral blood drawn from a Jr(a-) phenotype individual, who was homozygous for a null mutation of ABCG2*01N.01 (rs72552713, c.376C>T; p.Gln126*). The generated line exhibited pluripotent characteristics and no chromosomal aberrations. This cell line will serve as a cell source, enabling us to produce RBCs with the Jr(a-) phenotype in vitro, which can be used for transfusing individuals with anti-Jra antibodies.


Sujet(s)
Cellules souches pluripotentes induites , Agranulocytes , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Femelle , Agranulocytes/cytologie , Agranulocytes/métabolisme , Antigènes de groupe sanguin/métabolisme , Lignée cellulaire , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Différenciation cellulaire
20.
Stem Cell Res ; 77: 103433, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38718593

RÉSUMÉ

An induced pluripotent stem cell (iPSC) line (SDUCHi001-A) was established using peripheral blood mononuclear cells (PBMCs) from a healthy 6 years old boy. Reprogramming of the PBMCs was achieved through non-integrating delivery of OCT4, SOX2, KFL4, BCL-XL, and c-MYC. The iPSC line expressed pluripotency markers, had a normal karyotype and trilineage differentiation potential.


Sujet(s)
Cellules souches pluripotentes induites , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Humains , Mâle , Enfant , Différenciation cellulaire , Agranulocytes/métabolisme , Agranulocytes/cytologie , Lignée cellulaire , Reprogrammation cellulaire , Caryotype
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE