1
|
Kim JY, Hong N, Ham SW, Park S, Seo S, Kim H. Cancer-wide in silico analyses using differentially expressed genes demonstrate the functions and clinical relevance of JAG, DLL, and NOTCH. PLoS One 2024; 19:e0307943. [PMID: 39074091 DOI: 10.1371/journal.pone.0307943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 07/16/2024] [Indexed: 07/31/2024] Open
Abstract
Notch ligands [jagged (JAG) and, delta-like (DLL) families] and receptors [NOTCH family] are key regulators of Notch signaling. NOTCH signaling contributes to vascular development, tissue homeostasis, angiogenesis, and cancer progression. To elucidate the universal functions of the JAG, DLL, and NOTCH families and their connections with various biological functions, we examined 15 types of cancer using The Cancer Genome Atlas clinical database. We selected the differentially expressed genes (DEGs), which were positively correlated to the JAG, DLL, and NOTCH families in each cancer. We selected positive and negative hallmark signatures across cancer types. These indicated biological features associated with angiogenesis, hypoxia, KRAS signaling, cell cycle, and MYC targets by gene ontology and gene set enrichment analyses using DEGs. Furthermore, we analyzed single-cell RNA sequencing data to examine the expression of JAG, DLL, and NOTCH families and enrichment of hallmark signatures. Positive signatures identified using DEGs, such as KRAS signaling and hypoxia, were enriched in clusters with high expression of JAG, DLL, and NOTCH families. We subsequently validated the correlation between the JAG, DLL, and NOTCH families and clinical stages, including treatment response, metastasis, and recurrence. In addition, we performed survival analysis to identify hallmark signatures that critically affect patient survival when combining the expression of JAG, DLL, and NOTCH families. By combining the DEG enrichment and hallmark signature enrichment in survival analysis, we suggested unexplored regulatory functions and synergistic effects causing synthetic lethality. Taken together, our observations demonstrate the functions of JAG, DLL, and NOTCH families in cancer malignancy and provide insights into their molecular regulatory mechanisms.
Collapse
Affiliation(s)
- Jung Yun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Nayoung Hong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Seok Won Ham
- MEDIFIC Inc., Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Sehyeon Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Sunyoung Seo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Fereydani NM, Galehdari H, Hoveizi E, Alghasi A, Ajami M. Ex vivo expansion of hematopoietic stem cells in two/ three-dimensional co-cultures with various source of stromal cells. Tissue Cell 2024; 87:102331. [PMID: 38430847 DOI: 10.1016/j.tice.2024.102331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/19/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024]
Abstract
The ex vivo expansion of hematopoietic stem cells, with both high quantities and quality, is considered a paramount issue in cell and gene therapy for hematological diseases. Complex interactions between the bone marrow microenvironment and hematopoietic stem cells reveal the importance of using 2D and 3D coculture as a physiological system simulator in the proliferation, differentiation, and homeostasis of HSCs. Herein, the capacity of mesenchymal stem cells derived from different sources to support the expansion and maintenance of HSPC was compared with each other. We evaluated the fold increase of HSPC, CD34 marker expression, cytokine secretion profile of different MSCs, and the frequency of hematopoietic colony-forming unit parameters. Our results show that there was no significant difference between adipose tissue-MSC, Wharton jelly-MSC, and Endometrial-MSCs in HSPC expansion (fold increase: 34.74±4.38 in Wj-MSC, 32.22±5.07 in AD-MSC, 25.9±1.27 in En-MSCs); However, there were significantly more than the expansion media alone (4.4±0.69). The results obtained from the cytokine secretion analysis also confirm these results. Also, there were significant differences in the clonogenicity of Wj-MSC, En-MSCs, and expansion media (CFU-GEMM: 7±1.73, 2.3±1.15, and 2.3±1.52), which indicated that Wj-MSC could significantly maintain the primitive state. As a result, using Wj-mesenchymal stem cells on a 3D coculture system effectively increases the HSPC expansion and maintains the colonization potential of hematopoietic stem cells.
Collapse
Affiliation(s)
- Nasim Mayeli Fereydani
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Hamid Galehdari
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Elham Hoveizi
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Arash Alghasi
- Thalassemia & Hemoglobinopathy Research center, Health research institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Monireh Ajami
- Department of Hematology, School of Paramedical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
3
|
Wang H, Bi X, Zhang R, Yuan H, Xu J, Zhang K, Qi S, Zhang X, Jiang M. Adipose-Derived Mesenchymal Stem Cell Facilitate Hematopoietic Stem Cell Proliferation via the Jagged-1/Notch-1/Hes Signaling Pathway. Stem Cells Int 2023; 2023:1068405. [PMID: 38020206 PMCID: PMC10653966 DOI: 10.1155/2023/1068405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/29/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Poor graft function (PGF) is a life-threatening complication following hematopoietic stem cell transplantation (HSCT). Current therapies, such as CD34+ cell infusion, have shown limited effectiveness. Conversely, mesenchymal stem cells (MSCs) show potential in addressing PGF. Adipose-derived mesenchymal stem cells (ADSCs) effectively support long-term hematopoietic stem cell proliferation. Therefore, this study aimed to investigate the mechanisms underlying the long-term hematopoietic support provided by ADSCs. Methods ADSCs were isolated from mice and subsequently identified. In vitro experiments involved coculturing ADSCs as feeders with Lin-Sca-1+c-kit+ (LSK) cells from mice for 2 and 5 weeks. The number of LSK cells was quantified after coculture. Scanning electron microscopy was utilized to observe the interaction between ADSCs and LSK cells. Hes-1 expression was assessed using western blot and real-time quantitative PCR. An γ-secretase inhibitor (GSI) was used to confirm the involvement of the Jagged-1/Notch-1/Hes-1 pathway in LSK cell expansion. Additionally, Jagged-1 was knocked down in ADSCs to demonstrate its significance in ADSC-mediated hematopoietic support. In vivo experiments were conducted to study the hematopoietic support provided by ADSCs through the infusion of LSK, LSK + fibroblasts, and LSK + ADSCs, respectively. Mouse survival, platelet count, leukocyte count, and hemoglobin levels were monitored. Results ADSCs showed high-Jagged-1 expression and promoted LSK cell proliferation. There was a direct interaction between ADSCs and LSK cells. After coculture, Hes-1 expression increased in LSK cells. Moreover, GSI-reduced LSK cell proliferation and Hes-1 expression. Knockdown of Jagged-1 attenuated ADSCs-mediated promotion of LSK cell proliferation. Furthermore, ADSCs facilitated hematopoietic recovery and promoted the survival of NOD/SCID mice. Conclusion The hematopoietic support provided by ADSCs both in vivo and in vitro may be mediated, at least in part, through the Jagged-1/Notch-1 signaling pathway. These findings provide valuable insights into the mechanisms underlying ADSCs-mediated hematopoietic support and may have implications for improving the treatment of PGF following HSCT.
Collapse
Affiliation(s)
- Hongbo Wang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Xiaojuan Bi
- The State Key Laboratory of Pathogenesis and Prevention of Central Asian High Incidence Diseases, Institute of Clinical Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Rongyao Zhang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Hailong Yuan
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Jianli Xu
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Kaile Zhang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Songqing Qi
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Xue Zhang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Ming Jiang
- Hematology Center, The First Affiliated Hospital of Xinjiang Medical University (Xinjiang Uygur Autonomous Region Institute of Hematology), Urumqi 830054, China
- Stem Cell Research Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| |
Collapse
|
4
|
Cho HY, Lee S, Park JH, Kwak YH, Kweon H, Kang D. Competitive Hybridization of a Microarray Identifies CMKLR1 as an Up-Regulated Gene in Human Bone Marrow-Derived Mesenchymal Stem Cells Compared to Human Embryonic Fibroblasts. Curr Issues Mol Biol 2022; 44:1497-1512. [PMID: 35723360 PMCID: PMC9164045 DOI: 10.3390/cimb44040102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely applied to the regeneration of damaged tissue and the modulation of immune response. The purity of MSC preparation and the delivery of MSCs to a target region are critical factors for success in therapeutic application. In order to define the molecular identity of an MSC, the gene expression pattern of a human bone marrow-derived mesenchymal stem cell (hBMSC) was compared with that of a human embryonic fibroblast (hEF) by competitive hybridization of a microarray. A total of 270 and 173 genes were two-fold up- and down-regulated with FDR < 0.05 in the hBMSC compared to the hEF, respectively. The overexpressed genes in the hBMSC over the hEF, including transcription factors, were enriched for biological processes such as axial pattern formation, face morphogenesis and skeletal system development, which could be expected from the differentiation potential of MSCs. CD70 and CD339 were identified as additional CD markers that were up-regulated in the hBMSC over the hEF. The differential expression of CD70 and CD339 might be exploited to distinguish hEF and hBMSC. CMKLR1, a chemokine receptor, was up-regulated in the hBMSC compared to the hEF. RARRES2, a CMKLR1 ligand, stimulated specific migration of the hBMSC, but not of the hEF. RARRES2 manifested as ~two-fold less effective than SDF-1α in the directional migration of the hBMSC. The expression of CMKLR1 was decreased upon the osteoblastic differentiation of the hBMSC. However, the RARRES2-loaded 10% HA-silk scaffold did not recruit endogenous cells to the scaffold in vivo. The RARRES2−CMKLR1 axis could be employed in recruiting systemically delivered or endogenous MSCs to a specific target lesion.
Collapse
Affiliation(s)
- Hee-Yeon Cho
- Ilsong Institute of Life Science, Hallym University, Beodeunaru-ro 55, Seoul 07247, Korea; (H.-Y.C.); (S.L.); (J.-H.P.)
- Department of Biomedical Gerontology, Hallym University Graduate School, Chuncheon 24252, Korea
| | - Sooho Lee
- Ilsong Institute of Life Science, Hallym University, Beodeunaru-ro 55, Seoul 07247, Korea; (H.-Y.C.); (S.L.); (J.-H.P.)
| | - Ji-Hong Park
- Ilsong Institute of Life Science, Hallym University, Beodeunaru-ro 55, Seoul 07247, Korea; (H.-Y.C.); (S.L.); (J.-H.P.)
- Department of Biomedical Gerontology, Hallym University Graduate School, Chuncheon 24252, Korea
| | - Yoon Hae Kwak
- Department of Orthopaedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul 05505, Korea;
| | - HaeYong Kweon
- Industrial Insect and Sericulture Division, National Institute of Agricultural Sciences, RDA, Wanju-gun 55365, Korea;
| | - Dongchul Kang
- Ilsong Institute of Life Science, Hallym University, Beodeunaru-ro 55, Seoul 07247, Korea; (H.-Y.C.); (S.L.); (J.-H.P.)
- Department of Biomedical Gerontology, Hallym University Graduate School, Chuncheon 24252, Korea
- Correspondence: ; Tel.: +82-2-6923-8230
| |
Collapse
|
5
|
Takam Kamga P, Bazzoni R, Dal Collo G, Cassaro A, Tanasi I, Russignan A, Tecchio C, Krampera M. The Role of Notch and Wnt Signaling in MSC Communication in Normal and Leukemic Bone Marrow Niche. Front Cell Dev Biol 2021; 8:599276. [PMID: 33490067 PMCID: PMC7820188 DOI: 10.3389/fcell.2020.599276] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
Notch and Wnt signaling are highly conserved intercellular communication pathways involved in developmental processes, such as hematopoiesis. Even though data from literature support a role for these two pathways in both physiological hematopoiesis and leukemia, there are still many controversies concerning the nature of their contribution. Early studies, strengthened by findings from T-cell acute lymphoblastic leukemia (T-ALL), have focused their investigation on the mutations in genes encoding for components of the pathways, with limited results except for B-cell chronic lymphocytic leukemia (CLL); in because in other leukemia the two pathways could be hyper-expressed without genetic abnormalities. As normal and malignant hematopoiesis require close and complex interactions between hematopoietic cells and specialized bone marrow (BM) niche cells, recent studies have focused on the role of Notch and Wnt signaling in the context of normal crosstalk between hematopoietic/leukemia cells and stromal components. Amongst the latter, mesenchymal stromal/stem cells (MSCs) play a pivotal role as multipotent non-hematopoietic cells capable of giving rise to most of the BM niche stromal cells, including fibroblasts, adipocytes, and osteocytes. Indeed, MSCs express and secrete a broad pattern of bioactive molecules, including Notch and Wnt molecules, that support all the phases of the hematopoiesis, including self-renewal, proliferation and differentiation. Herein, we provide an overview on recent advances on the contribution of MSC-derived Notch and Wnt signaling to hematopoiesis and leukemia development.
Collapse
Affiliation(s)
- Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
- EA4340-BCOH, Biomarker in Cancerology and Onco-Haematology, UVSQ, Université Paris Saclay, Boulogne-Billancourt, France
| | - Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Giada Dal Collo
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Adriana Cassaro
- Hematology Unit, Department of Oncology, Niguarda Hospital, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Ilaria Tanasi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Anna Russignan
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Cristina Tecchio
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
6
|
Pelullo M, Nardozza F, Zema S, Quaranta R, Nicoletti C, Besharat ZM, Felli MP, Cerbelli B, d'Amati G, Palermo R, Capalbo C, Talora C, Di Marcotullio L, Giannini G, Checquolo S, Screpanti I, Bellavia D. Kras/ADAM17-Dependent Jag1-ICD Reverse Signaling Sustains Colorectal Cancer Progression and Chemoresistance. Cancer Res 2019; 79:5575-5586. [PMID: 31506332 DOI: 10.1158/0008-5472.can-19-0145] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/17/2019] [Accepted: 09/06/2019] [Indexed: 11/16/2022]
Abstract
Colorectal cancer is characterized by well-known genetic defects and approximately 50% of cases harbor oncogenic Ras mutations. Increased expression of Notch ligand Jagged1 occurs in several human malignancies, including colorectal cancer, and correlates with cancer progression, poor prognosis, and recurrence. Herein, we demonstrated that Jagged1 was constitutively processed in colorectal cancer tumors with mutant Kras, which ultimately triggered intrinsic reverse signaling via its nuclear-targeted intracellular domain Jag1-ICD. This process occurred when Kras/Erk/ADAM17 signaling was switched on, demonstrating that Jagged1 is a novel target of the Kras signaling pathway. Notably, Jag1-ICD promoted tumor growth and epithelial-mesenchymal transition, enhancing colorectal cancer progression and chemoresistance both in vitro and in vivo. These data highlight a novel role for Jagged1 in colorectal cancer tumor biology that may go beyond its effect on canonical Notch activation and suggest that Jag1-ICD may behave as an oncogenic driver that is able to sustain tumor pathogenesis and to confer chemoresistance through a noncanonical mechanism. SIGNIFICANCE: These findings present a novel role of the transcriptionally active Jag1-ICD fragment to confer and mediate some of the activity of oncogenic KRAS.
Collapse
Affiliation(s)
- Maria Pelullo
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | | | - Sabrina Zema
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Roberta Quaranta
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Carmine Nicoletti
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| | | | - Maria Pia Felli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Bruna Cerbelli
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University, Rome, Italy
| | - Giulia d'Amati
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University, Rome, Italy
| | - Rocco Palermo
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Carlo Capalbo
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Claudio Talora
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia, Italy
| | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University, Latina, Italy.
| | | | - Diana Bellavia
- Department of Molecular Medicine, Sapienza University, Rome, Italy.
| |
Collapse
|
7
|
Chellini F, Tani A, Vallone L, Nosi D, Pavan P, Bambi F, Zecchi-Orlandini S, Sassoli C. Platelet-Rich Plasma and Bone Marrow-Derived Mesenchymal Stromal Cells Prevent TGF-β1-Induced Myofibroblast Generation but Are Not Synergistic when Combined: Morphological in vitro Analysis. Cells Tissues Organs 2019; 206:283-295. [PMID: 31382258 DOI: 10.1159/000501499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/14/2019] [Indexed: 11/19/2022] Open
Abstract
The persistence of activated myofibroblasts is a hallmark of fibrosis of many organs. Thus, the modulation of the generation/functionality of these cells may represent a strategical anti-fibrotic therapeutic option. Bone marrow-derived mesenchymal stromal cell (MSC)-based therapy has shown promising clues, but some criticisms still limit the clinical use of these cells, including the need to avoid xenogeneic compound contamination for ex vivo cell amplification and the identification of appropriate growth factors acting as a pre-conditioning agent and/or cell delivery vehicle during transplantation, thus enabling the improvement of cell survival in the host tissue microenvironment. Many studies have demonstrated the ability of platelet-rich plasma (PRP), a source of many biologically active molecules, to positively influence MSC proliferation, survival, and functionality, as well as its anti-fibrotic potential. Here we investigated the effects of PRP, murine and human bone marrow-derived MSCs, and of the combined treatment PRP/MSCs on in vitro differentiation of murine NIH/3T3 and human HDFα fibroblasts to myofibroblasts induced by transforming growth factor (TGF)-β1, a well-known pro-fibrotic agent. The myofibroblastic phenotype was evaluated morphologically (cell shape and actin cytoskeleton assembly) and immunocytochemically (vinculin-rich focal adhesion clustering, α-smooth muscle actin and type-1 collagen expression). We found that PRP and MSCs, both as single treatments and in combination, were able to prevent the TGF-β1-induced fibroblast-myofibroblast transition. Unexpectedly, the combination PRP/MSCs had no synergistic effects. In conclusion, within the limitations related to an in vitro experimentation, our study may contribute to providing an experimental background for supporting the anti-fibrotic potential of the combination PRP/MSCs which, once translated "from bench to bedside," could potentially offer advantages over the single treatments.
Collapse
Affiliation(s)
- Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Larissa Vallone
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Paola Pavan
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Franco Bambi
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy,
| |
Collapse
|
8
|
Forghany Z, Robertson F, Lundby A, Olsen JV, Baker DA. Control of endothelial cell tube formation by Notch ligand intracellular domain interactions with activator protein 1 (AP-1). J Biol Chem 2017; 293:1229-1242. [PMID: 29196606 DOI: 10.1074/jbc.m117.819045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/30/2017] [Indexed: 01/08/2023] Open
Abstract
Notch signaling is a ubiquitous signal transduction pathway found in most if not all metazoan cell types characterized to date. It is indispensable for cell differentiation as well as tissue growth, tissue remodeling, and apoptosis. Although the canonical Notch signaling pathway is well characterized, accumulating evidence points to the existence of multiple, less well-defined layers of regulation. In this study, we investigated the function of the intracellular domain (ICD) of the Notch ligand Delta-like 4 (DLL4). We provide evidence that the DLL4 ICD is required for normal DLL4 subcellular localization. We further show that it is cleaved and interacts with the JUN proto-oncogene, which forms part of the activator protein 1 (AP-1) transcription factor complex. Mechanistically, the DLL4 ICD inhibited JUN binding to DNA and thereby controlled the expression of JUN target genes, including DLL4 Our work further demonstrated that JUN strongly stimulates endothelial cell tube formation and that DLL4 constrains this process. These results raise the possibility that Notch/DLL4 signaling is bidirectional and suggest that the DLL4 ICD could represent a point of cross-talk between Notch and receptor tyrosine kinase (RTK) signaling.
Collapse
Affiliation(s)
- Zary Forghany
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| | - Francesca Robertson
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| | - Alicia Lundby
- Novo Nordisk Foundation Center for Protein Research and.,the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | | | - David A Baker
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| |
Collapse
|
9
|
Mitroulis I, Chen LS, Singh RP, Kourtzelis I, Economopoulou M, Kajikawa T, Troullinaki M, Ziogas A, Ruppova K, Hosur K, Maekawa T, Wang B, Subramanian P, Tonn T, Verginis P, von Bonin M, Wobus M, Bornhäuser M, Grinenko T, Di Scala M, Hidalgo A, Wielockx B, Hajishengallis G, Chavakis T. Secreted protein Del-1 regulates myelopoiesis in the hematopoietic stem cell niche. J Clin Invest 2017; 127:3624-3639. [PMID: 28846069 DOI: 10.1172/jci92571] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 07/11/2017] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cells (HSCs) remain mostly quiescent under steady-state conditions but switch to a proliferative state following hematopoietic stress, e.g., bone marrow (BM) injury, transplantation, or systemic infection and inflammation. The homeostatic balance between quiescence, self-renewal, and differentiation of HSCs is strongly dependent on their interactions with cells that constitute a specialized microanatomical environment in the BM known as the HSC niche. Here, we identified the secreted extracellular matrix protein Del-1 as a component and regulator of the HSC niche. Specifically, we found that Del-1 was expressed by several cellular components of the HSC niche, including arteriolar endothelial cells, CXCL12-abundant reticular (CAR) cells, and cells of the osteoblastic lineage. Del-1 promoted critical functions of the HSC niche, as it regulated long-term HSC (LT-HSC) proliferation and differentiation toward the myeloid lineage. Del-1 deficiency in mice resulted in reduced LT-HSC proliferation and infringed preferentially upon myelopoiesis under both steady-state and stressful conditions, such as hematopoietic cell transplantation and G-CSF- or inflammation-induced stress myelopoiesis. Del-1-induced HSC proliferation and myeloid lineage commitment were mediated by β3 integrin on hematopoietic progenitors. This hitherto unknown Del-1 function in the HSC niche represents a juxtacrine homeostatic adaptation of the hematopoietic system in stress myelopoiesis.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Lan-Sun Chen
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Rashim Pal Singh
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Ioannis Kourtzelis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Matina Economopoulou
- Department of Ophthalmology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Tetsuhiro Kajikawa
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maria Troullinaki
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Athanasios Ziogas
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Klara Ruppova
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Kavita Hosur
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tomoki Maekawa
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Baomei Wang
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pallavi Subramanian
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Torsten Tonn
- Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Panayotis Verginis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and.,Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Malte von Bonin
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Manja Wobus
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Tatyana Grinenko
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and
| | - Marianna Di Scala
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
| | - Andres Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ben Wielockx
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and.,Center for Regenerative Therapies Dresden, Dresden, Germany
| | - George Hajishengallis
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, and.,Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
10
|
Dhawan A, von Bonin M, Bray LJ, Freudenberg U, Pishali Bejestani E, Werner C, Hofbauer LC, Wobus M, Bornhäuser M. Functional Interference in the Bone Marrow Microenvironment by Disseminated Breast Cancer Cells. Stem Cells 2016; 34:2224-35. [PMID: 27090603 DOI: 10.1002/stem.2384] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 03/01/2016] [Accepted: 03/14/2016] [Indexed: 01/14/2023]
Abstract
Skeletal metastasis of breast cancer is associated with a poor prognosis and significant morbidity. Investigations in other solid tumors have revealed an impairment in hematopoietic function upon bone marrow invasion. However, the interaction between disseminated breast cancer cells and the bone marrow microenvironment which harbors them has not been addressed comprehensively. Employing advanced co-culture assays, proteomic studies, organotypic models as well as in vivo xenotransplant models, we define the consequences of this interaction on the stromal compartment of bone marrow, affected molecular pathways and subsequent effects on the hematopoietic stem and progenitor cells (HSPCs). The results showed a basic fibroblast growth factor (bFGF)-mediated, synergistic increase in proliferation of breast cancer cells and mesenchymal stromal cells (MSCs) in co-culture. The stromal induction was associated with elevated phosphoinositide-3 kinase (PI3K) signaling in the stroma, which coupled with elevated bFGF levels resulted in increased migration of breast cancer cells towards the MSCs. The perturbed cytokine profile in the stroma led to reduction in the osteogenic differentiation of MSCs via downregulation of platelet-derived growth factor-BB (PDGF-BB). Long term co-cultures of breast cancer cells, HSPCs, MSCs and in vivo studies in NOD.Cg-Prkdc(scid) Il2rg(tm1Wjl) /SzJ (NSG) mice showed a reduced support for HSPCs in the altered niche. The resultant non- conducive phenotype of the niche for HSPC support emphasizes the importance of the affected molecular pathways in the stroma as clinical targets. These findings can be a platform for further development of therapeutic strategies aiming at the blockade of bone marrow support to disseminated breast cancer cells. Stem Cells 2016;34:2224-2235.
Collapse
Affiliation(s)
- Abhishek Dhawan
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany
| | - Malte von Bonin
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany.,German Consortium for Translational Cancer Research (DKTK), partner site, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura J Bray
- Institute of Biofunctional Polymer Materials, Leibniz Institute for Polymer Research, Max Bergmann Center of Biomaterials, Dresden, Germany.,Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Uwe Freudenberg
- Institute of Biofunctional Polymer Materials, Leibniz Institute for Polymer Research, Max Bergmann Center of Biomaterials, Dresden, Germany
| | - Elham Pishali Bejestani
- German Consortium for Translational Cancer Research (DKTK), partner site, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carsten Werner
- Institute of Biofunctional Polymer Materials, Leibniz Institute for Polymer Research, Max Bergmann Center of Biomaterials, Dresden, Germany
| | - Lorenz C Hofbauer
- German Consortium for Translational Cancer Research (DKTK), partner site, Dresden, Germany.,Department of Internal Medicine III, University Clinic, Dresden, Germany
| | - Manja Wobus
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany
| | - Martin Bornhäuser
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany.,German Consortium for Translational Cancer Research (DKTK), partner site, Dresden, Germany
| |
Collapse
|
11
|
Nowakowski A, Walczak P, Janowski M, Lukomska B. Genetic Engineering of Mesenchymal Stem Cells for Regenerative Medicine. Stem Cells Dev 2015; 24:2219-42. [PMID: 26140302 DOI: 10.1089/scd.2015.0062] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs), which can be obtained from various organs and easily propagated in vitro, are one of the most extensively used types of stem cells and have been shown to be efficacious in a broad set of diseases. The unique and highly desirable properties of MSCs include high migratory capacities toward injured areas, immunomodulatory features, and the natural ability to differentiate into connective tissue phenotypes. These phenotypes include bone and cartilage, and these properties predispose MSCs to be therapeutically useful. In addition, MSCs elicit their therapeutic effects by paracrine actions, in which the metabolism of target tissues is modulated. Genetic engineering methods can greatly amplify these properties and broaden the therapeutic capabilities of MSCs, including transdifferentiation toward diverse cell lineages. However, cell engineering can also affect safety and increase the cost of therapy based on MSCs; thus, the advantages and disadvantages of these procedures should be discussed. In this review, the latest applications of genetic engineering methods for MSCs with regenerative medicine purposes are presented.
Collapse
Affiliation(s)
- Adam Nowakowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland
| | - Piotr Walczak
- 2 Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,4 Department of Radiology, Faculty of Medical Sciences, University of Warmia and Mazury , Olsztyn, Poland
| | - Miroslaw Janowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland .,2 Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Barbara Lukomska
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland
| |
Collapse
|
12
|
Bigildeev AE, Zezina EA, Shipounova IN, Drize NJ. Interleukin-1 beta enhances human multipotent mesenchymal stromal cell proliferative potential and their ability to maintain hematopoietic precursor cells. Cytokine 2014; 71:246-54. [PMID: 25461405 DOI: 10.1016/j.cyto.2014.10.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 10/15/2014] [Accepted: 10/28/2014] [Indexed: 12/28/2022]
Abstract
Multipotent mesenchymal stromal cells (MMSCs) have been demonstrated to produce mature stromal cells and maintain hematopoietic progenitor cells (HPC). It was previously demonstrated that interleukin-1 beta (IL-1 beta) stimulates the growth of the stromal microenvironment in vivo. The aim of this study was to investigate the effect of IL-1 beta treatment of human MMSCs on their proliferative potential, gene expression, immunomodulating properties, and their ability to support HPCs in vitro. Human bone marrow-derived MMSCs were cultivated in standard conditions or with IL-1 beta. The cumulative cell production was assessed for five passages. After withdrawal of IL-1 beta, MMSC clonal efficiency was investigated, and the maintenance of HPCs on top of MMSCs layers was estimated using cobblestone area forming cell (CAFC) and long-term culture initiating cell (LTC-IC) assays. The effect of untreated MMSCs or MMSCs pretreated with IL-1 beta on lymphocyte proliferation was studied by CFSE staining. The relative expression level of various genes by MMSCs was analyzed using RT-qPCR. The administration of IL-1 beta elevated MMSCs clonal efficiency and total cell production but did not affect lymphocyte proliferation. MMSCs pretreatment with IL-1 beta enhanced their ability to maintain HPCs, as detected by CAFC assay, and it altered the expression levels of genes participating in HPC regulation by stromal cells, e.g., adhesion molecules (ICAM1) and growth factors (SDF1). This study revealed the ability of IL-1 beta to stimulate MMSCs proliferation and enhance their potential to maintain HPCs. MMSCs are considered a stromal niche component in vitro. The combined in vitro and previous in vivo data suggest that IL-1 beta is a systemic regulator of the stromal microenvironment.
Collapse
Affiliation(s)
- Alexey E Bigildeev
- Laboratory Physiology of Hematopoiesis, Hematological Research Center, Ministry of Health, Noviy Zikovskiy proezd 4, Moscow 125167, Russian Federation.
| | - Ekaterina A Zezina
- Laboratory Physiology of Hematopoiesis, Hematological Research Center, Ministry of Health, Noviy Zikovskiy proezd 4, Moscow 125167, Russian Federation; MSU im. Lomonosov, Biology Department, Subdepartment Molecular Immunology, Leninskie Gory, 1, 12, Moscow 119991, Russian Federation.
| | - Irina N Shipounova
- Laboratory Physiology of Hematopoiesis, Hematological Research Center, Ministry of Health, Noviy Zikovskiy proezd 4, Moscow 125167, Russian Federation.
| | - Nina J Drize
- Laboratory Physiology of Hematopoiesis, Hematological Research Center, Ministry of Health, Noviy Zikovskiy proezd 4, Moscow 125167, Russian Federation.
| |
Collapse
|
13
|
Li D, Masiero M, Banham AH, Harris AL. The notch ligand JAGGED1 as a target for anti-tumor therapy. Front Oncol 2014; 4:254. [PMID: 25309874 PMCID: PMC4174884 DOI: 10.3389/fonc.2014.00254] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/04/2014] [Indexed: 12/26/2022] Open
Abstract
The Notch pathway is increasingly attracting attention as a source of therapeutic targets for cancer. Ligand-induced Notch signaling has been implicated in various aspects of cancer biology; as a consequence, pan-Notch inhibitors and therapeutic antibodies targeting one or more of the Notch receptors have been investigated for cancer therapy. Alternatively, Notch ligands provide attractive options for therapy in cancer treatment due to their more restricted expression and better-defined functions, as well as their low rate of mutations in cancer. One of the Notch ligands, Jagged1 (JAG1), is overexpressed in many cancer types, and plays an important role in several aspects of tumor biology. In fact, JAG1-stimulated Notch activation is directly implicated in tumor growth through maintaining cancer stem cell populations, promoting cell survival, inhibiting apoptosis, and driving cell proliferation and metastasis. In addition, JAG1 can indirectly affect cancer by influencing tumor microenvironment components such as tumor vasculature and immune cell infiltration. This article gives an overview of JAG1 and its role in tumor biology, and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Demin Li
- Radcliffe Department of Medicine, Nuffield Division of Clinical Laboratory Sciences, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford , UK
| | - Massimo Masiero
- Radcliffe Department of Medicine, Nuffield Division of Clinical Laboratory Sciences, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford , UK
| | - Alison H Banham
- Radcliffe Department of Medicine, Nuffield Division of Clinical Laboratory Sciences, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford , UK
| | - Adrian L Harris
- Cancer Research UK Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford , UK
| |
Collapse
|
14
|
Ferrell PI, Hexum MK, Kopher RA, Lepley MA, Gussiaas A, Kaufman DS. Functional assessment of hematopoietic niche cells derived from human embryonic stem cells. Stem Cells Dev 2014; 23:1355-63. [PMID: 24517837 DOI: 10.1089/scd.2013.0497] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To evaluate hematopoietic niche cell populations isolated from human embryonic stem cells (hESCs), we tested the ability of hESC-derived stromal lines to support CD34(+) umbilical cord blood (UCB)- and hESC-derived CD34(+)45(+) cells in long-term culture initiating cell (LTC-IC) assays. Specifically, these hematopoietic populations were cocultured with hESC-derived mesenchymal stromal cells (hESC-MSCs) and hESC-derived endothelial cells (hESC-ECs), and then assessed for their LTC-IC potential in comparison to coculture with bone marrow (BM)-derived MSCs and the mouse stromal line M2-10B4. We found that the hESC-derived stromal lines supported LTC-ICs from UCB similar to M2-10B4 cells and better than BM-MSCs. However, none of the stromal populations supported LTC-IC from hESC-derived CD34(+)45(+) cells. Engraftment data using the output from LTC-IC assays showed long-term repopulation (12 weeks) of NSG mice to correlate with LTC-IC support on a given stromal layer. Therefore, hESC-derived stromal lines can be used to efficiently evaluate putative hematopoietic stem/progenitor cells derived from hESCs or other cell sources.
Collapse
Affiliation(s)
- Patrick I Ferrell
- 1 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | | | | | | | | | | |
Collapse
|