cell engraftment
Recently Published Documents


TOTAL DOCUMENTS

625
(FIVE YEARS 72)

H-INDEX

60
(FIVE YEARS 5)

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Silvia Preciado ◽  
Mª Salomé Sirerol-Piquer ◽  
Sandra Muntión ◽  
Lika Osugui ◽  
Gerardo J. Martí-Chillón ◽  
...  

Abstract Background Poor graft function or graft failure after allogeneic stem cell transplantation is an unmet medical need, in which mesenchymal stromal cells (MSC) constitute an attractive potential therapeutic approach. Hypoxia-inducible factor-1α (HIF-1α) overexpression in MSC (HIF-MSC) potentiates the angiogenic and immunomodulatory properties of these cells, so we hypothesized that co-transplantation of MSC-HIF with CD34+ human cord blood cells would also enhance hematopoietic stem cell engraftment and function both in vitro and in vivo. Methods Human MSC were obtained from dental pulp. Lentiviral overexpression of HIF-1α was performed transducing cells with pWPI-green fluorescent protein (GFP) (MSC WT) or pWPI-HIF-1α-GFP (HIF-MSC) expression vectors. Human cord blood CD34+ cells were co-cultured with MSC WT or HIF-MSC (4:1) for 72 h. Then, viability (Annexin V and 7-AAD), cell cycle, ROS expression and immunophenotyping of key molecules involved in engraftment (CXCR4, CD34, ITGA4, c-KIT) were evaluated by flow cytometry in CD34+ cells. In addition, CD34+ cells clonal expansion was analyzed by clonogenic assays. Finally, in vivo engraftment was measured by flow cytometry 4-weeks after CD34+ cell transplantation with or without intrabone MSC WT or HIF-MSC in NOD/SCID mice. Results We did not observe significant differences in viability, cell cycle and ROS expression between CD34+ cells co-cultured with MSC WT or HIF-MSC. Nevertheless, a significant increase in CD34, CXCR4 and ITGA4 expression (p = 0.009; p = 0.001; p = 0.013, respectively) was observed in CD34+ cells co-cultured with HIF-MSC compared to MSC WT. In addition, CD34+ cells cultured with HIF-MSC displayed a higher CFU-GM clonogenic potential than those cultured with MSC WT (p = 0.048). We also observed a significant increase in CD34+ cells engraftment ability when they were co-transplanted with HIF-MSC compared to CD34+ co-transplanted with MSC WT (p = 0.016) or alone (p = 0.015) in both the injected and contralateral femurs (p = 0.024, p = 0.008 respectively). Conclusions Co-transplantation of human CD34+ cells with HIF-MSC enhances cell engraftment in vivo. This is probably due to the ability of HIF-MSC to increase clonogenic capacity of hematopoietic cells and to induce the expression of adhesion molecules involved in graft survival in the hematopoietic niche.


2021 ◽  
Vol 22 (22) ◽  
pp. 12563
Author(s):  
Olivier Schussler ◽  
Pierre E. Falcoz ◽  
Juan C. Chachques ◽  
Marco Alifano ◽  
Yves Lecarpentier

Currently, the clinical impact of cell therapy after a myocardial infarction (MI) is limited by low cell engraftment due to low cell retention, cell death in inflammatory and poor angiogenic infarcted areas, secondary migration. Cells interact with their microenvironment through integrin mechanoreceptors that control their survival/apoptosis/differentiation/migration and proliferation. The association of cells with a three-dimensional material may be a way to improve interactions with their integrins, and thus outcomes, especially if preparations are epicardially applied. In this review, we will focus on the rationale for using collagen as a polymer backbone for tissue engineering of a contractile tissue. Contractilities are reported for natural but not synthetic polymers and for naturals only for: collagen/gelatin/decellularized-tissue/fibrin/Matrigel™ and for different material states: hydrogels/gels/solids. To achieve a thick/long-term contractile tissue and for cell transfer, solid porous compliant scaffolds are superior to hydrogels or gels. Classical methods to produce solid scaffolds: electrospinning/freeze-drying/3D-printing/solvent-casting and methods to reinforce and/or maintain scaffold properties by reticulations are reported. We also highlight the possibility of improving integrin interaction between cells and their associated collagen by its functionalizing with the RGD-peptide. Using a contractile patch that can be applied epicardially may be a way of improving ventricular remodeling and limiting secondary cell migration.


2021 ◽  
Vol 8 (11) ◽  
pp. 153
Author(s):  
Dongmin Yu ◽  
Xiaowei Wang ◽  
Lei Ye

Poor cell engraftment rate is one of the primary factors limiting the effectiveness of cell transfer therapy for cardiac repair. Recent studies have shown that the combination of cell-based therapy and tissue engineering technology can improve stem cell engraftment and promote the therapeutic effects of the treatment for myocardial infarction. This mini-review summarizes the recent progress in cardiac tissue engineering of cardiovascular cells from differentiated human pluripotent stem cells (PSCs), highlights their therapeutic applications for the treatment of myocardial infarction, and discusses the present challenges of cardiac tissue engineering and possible future directions from a clinical perspective.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2936-2936
Author(s):  
Daisuke Araki ◽  
Christi Salisbury-Ruf ◽  
Waleed Hakami ◽  
Keyvan Keyvanfar ◽  
Richard H. Smith ◽  
...  

Abstract Transplantation of genetically modified autologous hematopoietic stem and progenitor cells (HSPCs) holds a curative potential for subjects with inherited blood disorders. In recent years, transfer of a therapeutic gene to HSPCs has been successfully achieved using replication-incompetent integrating lentiviral vectors. More recently, advances have emerged to more precisely edit cellular genomes by specific correction of mutations or targeted gene addition at endogenous genomic loci. However, cellular processes triggered in HSPCs by the programmable nucleases utilized in these gene editing approaches may negatively impact their ability to reconstitute and maintain hematopoiesis long-term in recipient hosts. Granulocyte colony-stimulating factor (G-CSF) use after autologous HSPC transplantation is generally recommended to shorten the duration of severe neutropenia. However, little is known about the safety and efficacy of G-CSF use after transplantation of genetically modified autologous HSPCs. G-CSF is the principal cytokine regulating granulopoiesis, but also plays an important role in regulating hematopoietic stem cell (HSC) function (Schuttpelz, Leukemia 2014). Studies have suggested that G-CSF can exacerbate HSC damage caused by chemotherapeutic agents and irradiation by promoting differentiation at the expense of self-renewal and by inducing cellular senescence (van Os, Stem Cells 2000; Li, Cell Biosci 2015). Here, we asked whether G-CSF use after transplantation of gene edited HSPCs may negatively affect their long-term repopulating (LTR) and self-renewal capacities. To assess the effect of G-CSF use post-transplant on HSPC repopulating function after gene editing, mobilized human CD34+ cells were stimulated for 2 days, electroporated with AAVS1-specific sgRNA/Cas9 ribonucleoprotein complexes, and subsequently transplanted into NSG mice following busulfan conditioning. We subcutaneously injected G-CSF (125 mcg/kg/day) or PBS from post-transplant day 1 to 14 and compared hematopoietic reconstitution between both groups. The use of G-CSF initially increased human CD45+ cells in peripheral blood (PB) at 2 weeks post-transplant by enhancing CD13+ myeloid cell proliferation from committed progenitors (Fig. A). However, starting at 10 weeks post-transplant when hematopoiesis begins to emerge from the most primitive HSPCs, administration of G-CSF resulted in a 3 to 4-fold reduction in PB human cell engraftment compared to untreated mice (Fig. A). Similarly, G-CSF treated mice had significantly lower bone marrow (BM) and splenic engraftment at the endpoint (22 weeks) analysis, with comparable editing efficiency and lineage composition detected within human CD45+ cells (Fig. B, C). Importantly, percentages of immunophenotypic HSCs were 2-fold lower within the BM of G-CSF treated mice relative to the untreated group (Fig. D). To determine whether the negative effect of G-CSF post-transplant is specific to CRISPR-Cas9 gene editing, similar experiments were conducted using unmanipulated CD34+ cells or CD34+ cells transduced with a lentivirus vector expressing GFP. Interestingly, we found no differences in engraftment levels or immunophenotypic HSC frequencies between G-CSF treated and untreated mice. To assess the self-renewal capacity and quantify the frequency of gene edited LTR-HSCs, human CD45+ cells obtained from the BM of primary mice were serially transplanted into secondary recipient (NBSGW) mice at limiting dilution and BM engraftment was analyzed at 20 weeks post-transplant (total period of engraftment was 42 weeks). Notably, the secondary mice in the untreated group showed significantly superior human CD45+ cell engraftment compared with those in G-CSF treated group at the highest dose tested (Fig. E). The extreme limiting dilution analysis indicated that the frequency of LTR-HSCs was 5.1-fold higher (p = 0.011) in the untreated group compared with G-CSF treated group (Fig. F, G). Considering total engraftment in primary mice and the frequency of edited LTR-HSCs in secondary mice, we estimated the frequency of edited LTR-HSCs was reduced by 10-fold with G-CSF administration post-transplant. Collectively, our data suggest that G-CSF use post-transplant significantly reduces LTR and self-renewal capacities of CRISPR-Cas9 gene edited HSPCs. This understanding could have important clinical implications in HSPC gene therapy protocol. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1707-1707
Author(s):  
Wesley Wilson ◽  
Fei Miao ◽  
Alfred L Garfall ◽  
Dexiu Bu ◽  
Elena J Orlando ◽  
...  

Abstract Background: Autologous chimeric antigen receptor T (CAR-T) cell therapies show significant clinical activity against hematologic malignancies including multiple myeloma (MM). Numerous factors influence the effectiveness of this therapeutic approach. The fitness of a CAR-T cell product is an important factor affecting T cell engraftment and persistence, a pre-requisite for effective CAR-T cell therapy. In an effort to deconstruct the cellular heterogeneity of CAR-T cell products and explore relationships between T cell states within the cellular product, gene expression and CAR-T cell engraftment following adoptive transfer, we performed single cell RNA-sequencing (scRNA-seq) on CAR-T cell products from a phase I trial of BCMA-specific CAR-T cells in relapsed/refractory MM patients that included cohorts receiving a similar CAR-T cell dose with and without preconditioning with cyclophosphamide (NCT02546167, Cohen et al, J Clin Invest 2019). Method: scRNA-seq was performed on 25 unique products with an average of 8823 cells captured per product at an average depth of 40779 reads per cell. Batch effects were controlled by integration using established metrics for quality control. The linked inference of genomic experimental relationships (LIGER) method exhibited the most effective integration of 228,752 single cells across the 25 products. Annotation was completed by comparing transcriptomes of cells to the Blueprint/ENCODE references of pure immune cells. Differential gene expression (DGE) analysis was conducted by both Wilcoxon rank sum test testing and negative binomial distribution. Gene enrichment analysis was performed by comparing DGE with the Molecular Signatures Database v7.4. Cell to cell communication utilized the KEGG signaling pathway maps and curated lists of interactions from the literature. The intercellular communication probability was estimated on the DE genes before statistically significant intercellular communications were calculated by a permutation test, with dominant senders, receivers, mediators, and influencers identified using graph theory. Patients were grouped based upon "good" or "poor" engraftment using a peak blood vector copies per cell cutoff of 10,000. Association of transcriptional variation with neurotoxicity was explored for all patients. Results: The CD4:CD8 ratio of 2.51 single cells in the data was comparable to the observed average CD4:CD8 ratio of total T cells in the product based upon flow cytometric analysis of the products at harvest. Cell annotation showed significant heterogeneity of CD4+ and CD8+ T cells with cells exhibiting a CD4 Tcm transcriptional profile comprising the largest subset of T cells within the product. DGE analysis found 205 genes that were up or down regulated between the "good" vs "poor" engraftment phenotypes with 75 genes being detected using both mathematical approaches. CAR-BCMA expression was increased in "good" engraftment groups which has not previously been shown in CAR sorted cells. Within both CD4+ and CD8+ T cell subsets, transcriptional pathways associated with the RXF5/RFXAP/RFXANK transcriptional activator complex and the IL-2/STAT5 signaling were identified as upregulated in the "good" engrafted products. Cell to cell communication analysis for both secreted signaling and cell to cell contact revealed similar ligand-receptor interaction differences between the engraftment groups. Within the neurotoxicity groups CAR-BCMA expression was not associated with either neurotoxicity or with high and low neurotoxicity effects in the product. High vs low neurotoxicity showed a shift with interferon gamma and cytokine-cytokine interactions. We also detected IL-6/JAK/STAT3 signaling activation increases in this group. Conclusion: The RFX5/RFXAP/RFXANK pathway associated with MHC class II expression and IL-2/STAT5 signaling show a significant association with engraftment of BCMA-specific CAR-T cells. Although IL-2 signaling is well known to be critical to T cell survival and a potential key driver for long-term persistence, the role of the RFX5 transcriptional activator complex in T cells, outside of its important role in regulating MHC expression, is largely unknown. Both pathways deserve further investigation. Cell to cell communication between engraftment groups suggests CD4/CD8 interactions that might be beneficial to engraftment at the product manufacturing stage. Figure 1 Figure 1. Disclosures Garfall: Amgen: Honoraria; CRISPR Therapeutics: Research Funding; GlaxoSmithKline: Honoraria; Janssen: Honoraria, Research Funding; Novartis: Research Funding; Tmunity: Research Funding. Bu: Novartis: Current Employment, Patents & Royalties: Co-inventor on patent applications. Orlando: Novartis: Current Employment. Brogdon: Novartis Institutes for Biomedical Research: Current Employment. Pruteanu-Malinici: Novartis: Current Employment. Cohen: Janssen: Consultancy; Oncopeptides: Consultancy; Genentech/Roche: Consultancy; BMS/Celgene: Consultancy; AstraZeneca: Consultancy; Novartis: Research Funding; Takeda: Consultancy; GlaxoSmithKline: Consultancy, Research Funding.


Author(s):  
Tsung-Ming LEE ◽  
Cheng-Che LEE ◽  
Horng-Jyh HARN ◽  
Tzyy-Wen Chiou ◽  
Ming-Hsi CHUANG ◽  
...  

Abstract Cognitive impairment is a serious side effect of post-myocardial infarction (MI) course. We have recently demonstrated that human adipose-derived stem cells (hADSCs) ameliorated myocardial injury after MI by attenuating reactive oxygen species (ROS) levels. Here, we studied whether the beneficial effects of intramyocardial hADSC transplantation can extend to the brain and how they may attenuate cognitive dysfunction via modulating ROS after MI. After coronary ligation, male Wistar rats were randomized via an intramyocardial route to receive either vehicle, hADSC transplantation (1 × 106 cells), or the combination of hADSCs and 3-Morpholinosydnonimine (SIN-1, a peroxynitrite donor). Whether hADSCs migrated into the hippocampus was assessed by using human-specific primers in qPCR reactions. Passive avoidance test was used to assess cognitive performance. Postinfarction was associated with increased oxidative stress in the myocardium, circulation, and hippocampus. This was coupled with decreased numbers of dendritic spines as well as a significant downregulation of synaptic plasticity consisting of synaptophysin and PSD95. Step-through latency during passive avoidance test was impaired in vehicle-treated rats after MI. Intramyocardial hADSC injection exerted therapeutic benefits in improving cardiac function and cognitive impairment. None of hADSCs was detected in rat’s hippocampus at the 3rd day after intramyocardial injection. The beneficial effects of hADSCs on MI-induced histological and cognitive changes were abolished after adding SIN-1. MI-induced ROS attacked the hippocampus to induce neurodegeneration, resulting in cognitive deficit. The remotely intramyocardial administration of hADSCs has the capacity of improved synaptic neuroplasticity in the hippocampus mediated by ROS, not the cell engraftment, after MI. Key messages Human adipose-derived stem cells (hADSCs) ameliorated injury after myocardial infarction by attenuating reactive oxygen species (ROS) levels. Intramyocardial administration of hADSCs remotely exerted therapeutic benefits in improving cognitive impairment after myocardial infarction. The improved synaptic neuroplasticity in the hippocampus was mediated by hADSC-inhibiting ROS, not by the stem cell engraftment.


2021 ◽  
Vol 22 (17) ◽  
pp. 9126
Author(s):  
Gloria Abizanda ◽  
Leyre López-Muneta ◽  
Javier Linares ◽  
Luis I. Ramos ◽  
Arantxa Baraibar-Churio ◽  
...  

The success of cell therapy for the treatment of myocardial infarction depends on finding novel approaches that can substantially implement the engraftment of the transplanted cells. In order to enhance cell engraftment, most studies have focused on the pretreatment of transplantable cells. Here we have considered an alternative approach that involves the preconditioning of infarcted heart tissue to reduce endogenous cell activity and thus provide an advantage to our exogenous cells. This treatment is routinely used in other tissues such as bone marrow and skeletal muscle to improve cell engraftment, but it has never been taken in cardiac tissue. To avoid long-term cardiotoxicity induced by full heart irradiation we developed a rat model of a catheter-based heart irradiation system to locally impact a delimited region of the infarcted cardiac tissue. As proof of concept, we transferred ZsGreen+ iPSCs in the infarcted heart, due to their ease of use and detection. We found a very significant increase in cell engraftment in preirradiated rats. In this study, we demonstrate for the first time that preconditioning the infarcted cardiac tissue with local irradiation can substantially enhance cell engraftment.


Sign in / Sign up

Export Citation Format

Share Document