scholarly journals Single-cell RNA-seq highlights heterogeneity in human primary Wharton’s jelly mesenchymal stem/stromal cells cultured in vitro

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Changbin Sun ◽  
Lei Wang ◽  
Hailun Wang ◽  
Tingrun Huang ◽  
Wenwen Yao ◽  
...  
2019 ◽  
Author(s):  
Changbin Sun ◽  
Lei Wang ◽  
Hailun Wang ◽  
Tingrun Huang ◽  
Xi Zhang

SUMMARYMesenchymal Stem/Stromal cells (MSCs) are multipotent cells with promising application potential in regenerative medicine and immunomodulation. However, MSCs cultured in vitro exhibit functional heterogeneity. The underlying molecular mechanisms that define MSC heterogeneity remain unclear. Here, we investigated gene-expression heterogeneity of human primary Wharton’s Jelly-derived MSCs (WJMSCs) cultured in vitro via single-cell RNA-seq. At the single-cell level, highly variable genes (HVGs) are associated with functional characteristics of classic MSCs. Differentially expressed genes analysis revealed the existence of several distinct subpopulations exhibit different functional characteristics associated with proliferation, development, and inflammation response. By comparing our WJMSCs data with a public available adipose-derived MSCs (ADMSCs) single cell transcriptomic data, we found that HVGs from these two studies are largely overlapped and have similar functional enrichment. Taken together, these results suggested that these HVGs hold the potential to be used as candidate markers for further potency association studies.


2019 ◽  
Vol 15 (6) ◽  
pp. 900-918 ◽  
Author(s):  
Tiziana Corsello ◽  
Giandomenico Amico ◽  
Simona Corrao ◽  
Rita Anzalone ◽  
Francesca Timoneri ◽  
...  

2020 ◽  
Vol 2020 ◽  
pp. 1-8
Author(s):  
Hataiwan Kunkanjanawan ◽  
Tanut Kunkanjanawan ◽  
Veerapol Khemarangsan ◽  
Rungrueang Yodsheewan ◽  
Kasem Theerakittayakorn ◽  
...  

Coimplantation of endothelial cells (ECs) and mesenchymal stromal cells (MSCs) into the transplantation site could be a feasible option to achieve a sufficient level of graft-host vascularization. To find a suitable source of tissue that provides a large number of high-quality ECs and MSCs suited for future clinical application, we developed a simplified xeno-free strategy for isolation of human umbilical vein endothelial cells (HUVECs) and Wharton’s jelly-derived mesenchymal stromal cells (WJ-MSCs) from the same umbilical cord. We also assessed whether the coculture of HUVECs and WJ-MSCs derived from the same umbilical cord (autogenic cell source) or from different umbilical cords (allogenic cell sources) had an impact on in vitro angiogenic capacity. We found that HUVECs grown in 5 ng/ml epidermal growth factor (EGF) supplemented xeno-free condition showed higher proliferation potential compared to other conditions. HUVECs and WJ-MSCs obtained from this technic show an endothelial lineage (CD31 and von Willebrand factor) and MSC (CD73, CD90, and CD105) immunophenotype characteristic with high purity, respectively. It was also found that only the coculture of HUVEC/WJ-MSC, but not HUVEC or WJ-MSC mono-culture, provides a positive effect on vessel-like structure (VLS) formation, in vitro. Further investigations are needed to clarify the pros and cons of using autogenic or allogenic source of EC/MSC in tissue engineering applications. To the best of our knowledge, this study offers a simple, but reliable, xeno-free strategy to establish ECs and MSCs from the same umbilical cord, a new opportunity to facilitate the development of personal cell-based therapy.


2015 ◽  
Vol 102 (3) ◽  
pp. 368-378 ◽  
Author(s):  
Haiping He ◽  
Tokiko Nagamura-Inoue ◽  
Atsuko Takahashi ◽  
Yuka Mori ◽  
Yuki Yamamoto ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4531-4531
Author(s):  
Yahui Grace Chiu ◽  
Jacquelyn Lillis ◽  
Rakesh Singh ◽  
Jane L. Liesveld ◽  
Laura M. Calvi ◽  
...  

Abstract Introduction: Umbilical cord blood (UCB) is a salient source of primitive hematopoietic stem progenitor cells (HSPCs) for bone marrow (BM) reconstitution in patients with hematologic and non-hematologic malignancies. However, a relatively low number of HSPCs in UCB units and poor BM homing efficiency greatly hinders the clinical application of UCB CD34+ cells for transplantation. To overcome these hurdles, we developed two independent strategies that increase CD34+ cell numbers and improve BM homing efficiency of UCB HSPCs. First, we expanded UCB HSPCs by culturing them in decellularized Wharton's jelly matrix (DWJM), a biometric scaffold mimicking the 3-dimenstional (3D) microenvironment of BM. Second, we enhanced the in vitro transmigration and in vivo BM homing efficiency of UCB CD34+ cells by blocking EPO/EPOR signaling. Both approaches enhance UCB CD34+ cell migration toward stromal cell-derived factor 1 (SDF1). In this study, we employed RNA-Seq and RT-PCR approaches to analyze UCB HSPCs treated with EPO and co-cultured with DWJM, aiming to identify molecules that regulate UCB HSPC transmigration via EPO/EPOR signaling. Methods: CD34+ cells from highly enriched UCB units (>90% purity) were treated with EPO for 24 hours and separately co-cultured with DWJM for 1 week. UCB CD34+ cells were collected and subjected to RNA-Seq and real-time PCR (RT-PCR) analyses. In vitro transmigration toward SDF-1 was assessed by transwell assay. To assess the involvement of RasGRP3 in UCB CD34+ cell mobility, cells were treated with 100 nM ingenol-3-angelate (I3A), a diacylglycerol (DAG) analog that specifically targets RAS Guanyl Releasing-Protein 3 (RasGRP3), for 16 hours followed by transwell assay. Anti-EPOR antibody-treated or EPO-treated cells were used as controls. In addition, RasGRP3 gene expression was examined in CD34+ cells from peripheral blood (PB) and BM samples collected from the same donor, and compared to RasGRP3 expression in UCB CD34+ cells. Unpaired, 2-tailed t-test was used to analyze results. Results: RasGRP3 was identified by RNA-Seq from the two independent approaches, EPO treatment and DWJM co-culture. EPO downregulated and DWJM upregulated RasGRP3 gene expression in UCB CD34+ cells. RasGRP3 expression was confirmed by qPCR. UCB CD34+ cells that migrated to the bottom chamber of the transwell assays, a population that has a higher mobility, showed an elevated RasGRP3 gene expression and a decreased EPOR cell surface expression. Activation of RasGRP3 by DAG analog I3A induced a significant increase in RasGRP3 gene expression (control: I3A treatment = 1: 202 ± 58, p=0.00012) that was associated with an enhanced transmigration capability (control: I3A = 41%+/-5: 54%+/- 3, p=0.032). Knocking-down of RasGRP3 in K562 cells, a known EPOR expressing cell line, impaired the transmigration capability of K562. CD34+ cells in peripheral blood (PB) showed a higher level of RasGRP3 gene expression compared to CD34+ cells in BM samples from the same healthy donors. RasGRP3 expression in PB CD34+ cells was significantly higher than BM and UCB CD34+ cells (qPCR signals relative to BM, BM: PB: UCB = 1: 431±65: 21±8, p=0.0012, 0.0023, and <0.0001 for BM vs. PB, BM vs. UCB and PB vs. UCB, respectively). Conclusions: By employing transwell assays, flow cytometry and molecular analyses, we demonstrate for the first time that RasGRP3, a protein responsible for GDP/GTP exchange of Ras, regulates the transmigration ability of human CD34+ cells. In addition, our findings connect RasGRP3 expression to the EPOR-mediated signaling pathway in CD34+ cells. A significantly higher level of RasGRP3 expression in PB CD34+ cells than its counterparts in BM might provide an explanation for why PB HSPCs show relatively faster BM engraftment than BM HSPCs during transplantation. Ongoing follow-up studies will elucidate the molecular mechanism(s) underlying EPOR signaling, which holds clinical potential to improve the BM homing deficiency of UCB CD34+ cells via modulating EPOR and RasGRP3 expression (Figure 1). Disclosures Liesveld: Onconova: Other: DSMB; Abbvie: Honoraria. Aljitawi:Medpace: Consultancy; The University of Rochester Medical Center: Patents & Royalties: Pending patent related to decellularized Wharton's jelly matrix.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4959-4959 ◽  
Author(s):  
Dandan Li ◽  
Tara L. Lin ◽  
Richard Hopkins ◽  
Omar S. Aljitawi

Abstract Background and objective: Acute myeloid leukemia (AML) develops from leukemia stem cells (LSCs), a small subset of leukemia cells possessing both self-renewal and multilineage differentiation potential similar to normal hematopoietic stem cells. The stem cell niche of the bone marrow microenvironment protects LSCs from chemotherapy, resulting in subsequent leukemia relapse. The study of AML LSCs in vitro is limited because of the lack of an ideal culture system mimicking the protective bone marrow microenvironment. The bone marrow stem cell niche is mainly composed of stromal cells, soluble cytokines and growth factors, as well as extracellular matrix (ECM). We therefore developed a 3-dimensional ECM model using decellularized Wharton's jelly from human umbilical cords to better characterize AML LSCs in vitro. Previously we have shown that leukemia cells grown in DWJM changed morphology to become spindle shaped and maintained viability but had decreased proliferation as measured by Alamar blue assay. Herein, we further characterize leukemia cells cultured in DWJM. Methods: Wharton's jelly decellularization process included multiple osmotic shock cycles using hypertonic and hypotonic solutions, a non-ionic detergent Triton-x, an anionic detergent sodium lauroyl succinate, and an enzyme digestion with recombinant endonuclease Benzonase™. We examined three human leukemia cell lines: HL60, Kasumi I and MV411. We characterized leukemia cell proliferation by CellTrace proliferation assay and phenotype by flow cytometry for stem cell markers. Serial colony forming unit (CFU) assays were used to test the self-renewal of leukemia cells. Results: CellTrace proliferation assay showed that, compared to cells in suspension, cells cultured in DWJM divided less frequently. To assess for LSCs properties, we measured the ALDH+ population by Aldefluor assay and found that the ALDH+ cells from Kasumi I and HL60 increased significantly in DWJM compared to suspension (p<0.05). Lastly, serial CFU showed significantly increased colony forming units in both primary and secondary plating in MV411 and HL60 cells cultured in DWJM versus suspension (P<0.05). CFU also increased in Kasumi-I cells cultured in DWJM versus suspension, though the increase was not statistically significant (P=0.08). Taken together, our findings suggest that leukemia cells cultured in DWJM demonstrated an increased self-renewal ability due to the enrichment of LSCs rather than progenitors. Conclusion: Decellularized Wharton's jelly matrix may serve as a practical in vitro ECM model to enrich for LSCs and study the ECM-LSC interactions. Disclosures No relevant conflicts of interest to declare.


Placenta ◽  
2016 ◽  
pp. 91-128 ◽  
Author(s):  
Marta Magatti ◽  
Mohamed H. Abumaree ◽  
Antonietta R. Silini ◽  
Rita Anzalone ◽  
Salvatore Saieva ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1212-1212 ◽  
Author(s):  
Aristea Batsali ◽  
Charalampos Pontikoglou ◽  
Elisavet Kouvidi ◽  
Athina Damianaki ◽  
Aikaterini Stratigi ◽  
...  

Abstract Bone narrow (BM)- derived mesenchymal stem/stromal cells (MSCs) represent the most extensively studied population of adult MSCs and are considered as the gold-standard for MSC-based clinical applications. Yet, it is now becoming increasingly clear that BM may not represent the most suitable source for MSC collection. Indeed, Umbilical cord (UC) has emerged as a more abundant and easily attainable source of MSCs and several reports have shown that MSCs can be efficiently isolated from the connective tissue that surrounds UC vessels, namely the Wharton's jelly (WJ). According to the existing literature, WJ-MSCs display typical MSC characteristics, however a head-to-head comparison with BM-MSCs is still lacking. Provided that ex vivo MSC expansion is a prerequisite for clinical MSC-applications, in the present study we seek to comparatively investigate the characteristics of WJ- and BM-MSCs, cultured under identical conditions. MSCs were isolated and expanded from consenting healthy donors’ BM aspirates (n=5) and from the WJ of full-term neonates (n=10) after written informed consent of the family. MSCs were in vitro expanded and re-seeded for a total of 10 passages (P) and phenotypically characterized by flow cytometry (FC). MSCs were induced to differentiate in vitro to adipocytes and osteoblasts. Differentiation was assessed by cytochemical stains and by the expression of adipocyte- and osteocyte-specific genes. Relative gene expression was calculated by the ΔCt method. MSC growth characteristics were assessed by evaluating the population doubling time (DT) and by a methyl-triazolyl-tetrazolium (MTT)-assay throughout passages. Cell-cycle analysis was performed using propidium iodide (PI) staining. MSC survival was evaluated by FC with 7-Aminoactinomycin D (7-AAD) and senescence was estimated by the percentage of SA-b-gal+ cells in cultures. Moreover, MSC karyotypic stability was assessed with classic G-banding. Finally the expression of genes related to Wnt-mediated signal transduction was also investigated, using a PCR array. Total RNA was thus isolated from 6 representative BM- and 6 WJ-MSC cultures at P2. The fold change (FC) for each gene between the group of WJ- and the group of BM-MSCs was calculated with the ΔΔCt method (FC=2-ΔΔCt). WJ-MSCs displayed a spindle-shape morphology, similar to BM-MSCs. Furthermore, WJ- and BM-MSCs displayed identical immunophenotype, as evidenced by the expression of CD90,CD105,CD44,CD29,CD73 and the lack of expression of CD45,CD14,CD34,CD31. WJ-MSCs displayed superior proliferative potential compared to BM-MSCs throughout passages (p<0.05). Moreover, the proportion of proliferating (S/G2/M) WJ-MSCs was higher compared to BM-MSCs at P4 (p<0.001), while there was no significant difference between two MSC populations in the proportion of 7-AADbright/dim –cells at P4. Regarding senescence, significantly fewer SA-b-gal+ cells were observed in WJ-MSC cultures, as compared to BM-MSCs at P10 (p<0.05). Compared to their bone marrow counterparts, WJ-MSCs displayed inferior capacity to differentiate into adipocytes and osteoblasts as evidenced by Oil Red O and Alizarin Red staining, respectively, and also by the weaker expression of adipocyte- (PPAR-g, p<0.0002; CEBP-a, p<0.0001) and osteocyte-specific markers (RUNX2, p<0.0006; DLX5, p<0.0001; ALP, p<0.0042). No chromosomal abnormalities were observed in either WJ- or BM-MSCs during in vitro expansion. Regarding the Wnt-pathway signaling molecules, the Wnt antagonist sFRP4, which induces adipogenesis, as well the Wnt/b-catenin target gene Wisp-1, a regulator of osteogenesis were significantly down-regulated in WJ-MSCs (FC=22.3825, p<0,05; FC=20.18, p<0.0001, respectively). On the other hand, the expression of Wnt/b-catenin target gene Cyclin D1, which induces MSC proliferation and represses adipogenesis, was up-regulated in WJ-MSCs (FC=2.8, p<0.05). Taken together WJ-MSCs display decreased cellular senescence after extended in vitro culture, increased proliferative capacity and reduced potential to differentiate in vitro to adipocytes and osteocytes, as compared to BM-MSCs. The last two observations can be explained, at least partly, by the aberrant expression of Wnt-signaling molecules in WJ-MSCs. The emerging role of Wnt-signaling pathway in WJ-MSC biology is currently under investigation. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document