1
|
Qin R, Wang T, He W, Wei W, Liu S, Gao M, Huang Z. Jak2/STAT6/c-Myc pathway is vital to the pathogenicity of Philadelphia-positive acute lymphoblastic leukemia caused by P190 BCR-ABL. Cell Commun Signal 2023; 21:27. [PMID: 36721266 PMCID: PMC9887777 DOI: 10.1186/s12964-023-01039-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/02/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The Philadelphia chromosome encodes the BCR-ABL fusion protein, which has two primary subtypes, P210 and P190. P210 and P190 cause Philadelphia-positive chronic myeloid leukemia (Ph+ CML) and Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL), respectively. The Ph+ ALL is more malignant than Ph+ CML in disease phenotype and progression. This implies the key pathogenic molecules and regulatory mechanisms caused by BCR-ABL in two types of leukemia are different. It is reported that STAT6 was significantly activated only in P190 transformed cells. However, the potential role and the mechanism of STAT6 activation in Ph+ ALL and its activation mechanism by P190 are still unknown. METHODS The protein and mRNA levels of STAT6, c-Myc, and other molecules were measured by western blot and quantitative real-time PCR. The STAT6 inhibitor AS1517499 was used to specifically inhibit p-STAT6. The effect of p-STAT6 inhibition on Ph+ CML and Ph+ ALL cells was identified by CCK-8 and FCM assay. Dual luciferase reporter and ChIP assay were performed to confirm the direct binding between STAT6 and c-Myc. The impact of STAT6 inhibition on tumor progression was detected in Ph+ CML and Ph+ ALL mouse models. RESULTS Our results demonstrated that P210 induced CML-like disease, and P190 caused the more malignant ALL-like disease in mouse models. STAT6 was activated in P190 cell lines but not in P210 cell lines. Inhibition of STAT6 suppressed the malignancy of Ph+ ALL in vitro and in vivo, whereas it had little effect on Ph+ CML. We confirmed that p-STAT6 regulated the transcription of c-Myc, and STAT6 was phosphorylated by p-Jak2 in P190 cell lines, which accounted for the discrepant expression of p-STAT6 in P190 and P210 cell lines. STAT6 inhibition synergized with imatinib in Ph+ ALL cells. CONCLUSIONS Our study suggests that STAT6 activation plays an essential role in the development of Ph+ ALL and may be a potential therapeutic target in Ph+ ALL. Video abstract.
Collapse
Affiliation(s)
- Run Qin
- grid.203458.80000 0000 8653 0555Key Laboratory of Laboratory Medical Diagnostics Designated By the Ministry of Education, Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Teng Wang
- grid.412461.40000 0004 9334 6536Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei He
- grid.203458.80000 0000 8653 0555Key Laboratory of Laboratory Medical Diagnostics Designated By the Ministry of Education, Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Wei Wei
- grid.203458.80000 0000 8653 0555Key Laboratory of Laboratory Medical Diagnostics Designated By the Ministry of Education, Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Suotian Liu
- grid.203458.80000 0000 8653 0555Key Laboratory of Laboratory Medical Diagnostics Designated By the Ministry of Education, Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Miao Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuzhong District, Chongqing, 400016, China.
| | - Zhenglan Huang
- Key Laboratory of Laboratory Medical Diagnostics Designated By the Ministry of Education, Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Ueda M, Stefan T, Stetson L, Ignatz-Hoover JJ, Tomlinson B, Creger RJ, Cooper B, Lazarus HM, de Lima M, Wald DN, Caimi PF. Phase I Trial of Lithium and Tretinoin for Treatment of Relapsed and Refractory Non-promyelocytic Acute Myeloid Leukemia. Front Oncol 2020; 10:327. [PMID: 32211336 PMCID: PMC7076174 DOI: 10.3389/fonc.2020.00327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/25/2020] [Indexed: 11/13/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK3) inhibitors induce differentiation and growth inhibition of acute myeloid leukemia (AML) cells. Our pre-clinical studies showed GSK3 inhibition leads to sensitization of AML cells to tretinoin-mediated differentiation. We conducted a phase I trial of lithium, a GSK3 inhibitor, plus tretinoin for relapsed, refractory non-promyelocytic AML. Nine patients with median (range) age 65 (42–82) years were enrolled. All subjects had relapsed leukemia after prior therapy, with a median (range) of 3 (1–3) prior therapies. Oral lithium carbonate 300 mg was given 2–3 times daily and adjusted to meet target serum concentration (0.6 to 1.0 mmol/L); tretinoin 22.5 or 45 mg/m2/day (two equally divided doses) was administered orally on days 1–7 and 15–21 of a 28-day cycle. Four patients attained disease stability with no increase in circulating blasts for ≥4 weeks. Median (range) survival was 106 days (60–502). Target serum lithium concentration was achieved in all patients and correlated with GSK3 inhibition in leukemic cells. Immunophenotypic changes associated with myeloid differentiation were observed in five patients. The combination treatment led to a reduction in the CD34+ CD38– AML stem cell population both in vivo and in vitro. The combination of lithium and tretinoin is well-tolerated, induces differentiation of leukemic cells, and may target AML stem cells, but has limited clinical activity in the absence of other antileukemic agents. The results of this clinical trial suggest GSK3 inhibition can result in AML cell differentiation and may be a novel therapeutic strategy in this disease, particularly in combination with other antileukemic agents. Lithium is a weak GSK3 inhibitor and future strategies in AML treatment will probably require more potent agents targeting this pathway or combinations with other antileukemic agents. This trial is registered at ClinicalTrials.gov NCT01820624.
Collapse
Affiliation(s)
- Masumi Ueda
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, United States
| | - Tammy Stefan
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Lindsay Stetson
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - James J Ignatz-Hoover
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Benjamin Tomlinson
- Stem Cell Transplant and Hematologic Malignancies Program, University Hospitals Seidman Cancer Center, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Richard J Creger
- Stem Cell Transplant and Hematologic Malignancies Program, University Hospitals Seidman Cancer Center, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Brenda Cooper
- Stem Cell Transplant and Hematologic Malignancies Program, University Hospitals Seidman Cancer Center, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Hillard M Lazarus
- Stem Cell Transplant and Hematologic Malignancies Program, University Hospitals Seidman Cancer Center, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Marcos de Lima
- Stem Cell Transplant and Hematologic Malignancies Program, University Hospitals Seidman Cancer Center, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - David N Wald
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Paolo F Caimi
- Stem Cell Transplant and Hematologic Malignancies Program, University Hospitals Seidman Cancer Center, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
3
|
Kapoor S, Shenoy SP, Bose B. CD34 cells in somatic, regenerative and cancer stem cells: Developmental biology, cell therapy, and omics big data perspective. J Cell Biochem 2019; 121:3058-3069. [PMID: 31886574 DOI: 10.1002/jcb.29571] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
The transmembrane phosphoglycoprotein protein CD34 has conventionally been regarded as a marker for hematopoietic progenitors. Its expression on these cells has been leveraged for cell therapy applications in various hematological disorders. More recently, the expression of CD34 has also been reported on cells of nonhematopoietic origin. The list includes somatic cells such as endothelial cells, fibrocytes and interstitial cells and regenerative stem cells such as corneal keratocytes, muscle satellite cells, and muscle-derived stem cells. Furthermore, its expression on some cancer stem cells (CSCs) has also been reported. Till date, the functional roles of this molecule have been implicated in a multitude of cellular processes including cell adhesion, signal transduction, and maintenance of progenitor phenotype. However, the complete understanding about this molecule including its developmental origins, its embryonic connection, and associated functions is far from complete. Here, we review our present understanding of the structure and putative functions of the CD34 molecule based upon our literature survey. We also probed various biological databases to retrieve data related to the expression and associated molecular functions of CD34. Such information, upon synthesis, is hence likely to provide the suitability of such cells for cell therapy. Moreover, we have also covered the existing cell therapy and speculated cell therapy applications of CD34+ cells isolated from various lineages. We have also attempted here to speculate the role(s) of CD34 on CSCs. Finally, we discuss number of large-scale proteomics and transcriptomics studies that have been performed using CD34+ cells.
Collapse
Affiliation(s)
- Saketh Kapoor
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Sudheer P Shenoy
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| |
Collapse
|
4
|
Li Z, Shu Q, Li L, Ge M, Zhang Y. Sequential expression of cyclooxygenase-2, glutamate receptor-2, and platelet activating factor receptor in rat hippocampal neurons after fluid percussion injury. Neural Regen Res 2014; 9:978-85. [PMID: 25206921 PMCID: PMC4146214 DOI: 10.4103/1673-5374.133151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2014] [Indexed: 01/09/2023] Open
Abstract
Traumatic brain injury causes gene expression changes in different brain regions. Occurrence and development of traumatic brain injury are closely related, involving expression of three factors, namely cyclooxygenase-2, glutamate receptor-2, and platelet activating factor receptor. However, little is known about the correlation of these three factors and brain neuronal injury. In this study, primary cultured rat hippocampal neurons were subjected to fluid percussion injury according to Scott's method, with some modifications. RT-PCR and semi-quantitative immunocytochemical staining was used to measure the expression levels of cyclooxygenase-2, glutamate receptor-2, and platelet activating factor receptor. Our results found that cyclooxygenase-2 expression were firstly increased post-injury, and then decreased. Both mRNA and protein expression levels reached peaks at 8 and 12 hours post-injury, respectively. Similar sequential changes in glutamate receptor 2 were observed, with highest levels mRNA and protein expression at 8 and 12 hours post-injury respectively. On the contrary, the expressions of platelet activating factor receptor were firstly decreased post-injury, and then increased. Both mRNA and protein expression levels reached the lowest levels at 8 and 12 hours post-injury, respectively. Totally, our findings suggest that these three factors are involved in occurrence and development of hippocampal neuronal injury.
Collapse
Affiliation(s)
- Zhiqiang Li
- Second Department of Medicine, Inner Mongolia Corps Hospital, Chinese People's Armed Police Forces, Huhhot, Inner Mongolia Autonomous Region, China
| | - Qingming Shu
- Department of Pathology, General Hospital of Chinese People's Armed Police Forces, Beijing, China
| | - Lingzhi Li
- Section of Pharmaceutical Chemistry, Department of Rescue Medicine, Logistics University of Chinese People's Armed Police Force, Tianjin, China
| | - Maolin Ge
- Second Department of Surgery, Inner Mongolia Corps Hospital, Chinese People's Armed Police Forces, Huhhot, Inner Mongolia Autonomous Region, China
| | - Yongliang Zhang
- Training Department, Logistics University of Chinese People's Armed Police Force, Tianjin Key Laboratory for Biomarkers of Occupational and Environmental Hazard, Tianjin, China
| |
Collapse
|
5
|
Abstract
Human embryonic stem cells potentially represent an unlimited source of cells and tissues for regenerative medicine. Understanding signaling events that drive proliferation and specialization of these cells into various differentiated derivatives is of utmost importance for controlling their behavior in vitro. Major progress has been made in unraveling these signaling events with large-scale studies at the transcriptional level, but analysis of protein expression, interaction and modification has been more limited, since it requires different strategies. Recent advances in mass spectrometry-based proteomics indicate that proteome characterization can contribute significantly to our understanding of embryonic stem cell biology. In this article, we review mass spectrometry-based studies of human and mouse embryonic stem cells and their differentiated progeny, as well as studies of conditioned media that have been reported to support self-renewal of the undifferentiated cells in the absence of the more commonly used feeder cells. In addition, we make concise comparisons with related transcriptome profiling reports.
Collapse
Affiliation(s)
- Dennis Van Hoof
- Netherlands Institute of Developmental Biology, Hubrecht Laboratory, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
6
|
Proteomic Profiling of Ex Vivo Expanded CD34-Positive Haematopoetic Cells Derived from Umbilical Cord Blood. Stem Cells Int 2013; 2013:245695. [PMID: 23606859 PMCID: PMC3622389 DOI: 10.1155/2013/245695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 01/25/2013] [Accepted: 02/07/2013] [Indexed: 11/17/2022] Open
Abstract
Ex vivo expansion of haematopoetic cells by application of specific cytokines is one approach to overcome boundaries in cord blood transplantation due to limited numbers of haematopoetic stem cells. While many protocols describe an effective increase of total cell numbers and the amount of CD34-positive cells, it still remains unclear if and how the procedure actually affects the cells' properties. In the presented publications, CD34-positive cells were isolated from cord blood and expanded for up to 7 days in media supplemented with stem cell factor (SCF), thrombopoietin (THPO), interleukin 6 (IL-6), and fms-related tyrosine kinase 3 ligand (FLT3lg). At days 3 and 7, expanded cells were harvested and analyzed by flow cytometry and quantitative proteomics. 2970 proteins were identified, whereof proteomic analysis showed 440 proteins significantly changed in abundance during ex vivo expansion. Despite the fact that haematopoetic cells still expressed CD34 on the surface after 3 days, major changes in regard to the protein profile were observed, while further expansion showed less effect on the proteome level. Enrichment analysis of biological processes clearly showed a proteomic change toward a protein biosynthesis phenotype already within the first three days of expression.
Collapse
|
7
|
Guo H, Isserlin R, Chen X, Wang W, Phanse S, Zandstra PW, Paddison PJ, Emili A. Integrative network analysis of signaling in human CD34(+) hematopoietic progenitor cells by global phosphoproteomic profiling using TiO2 enrichment combined with 2D LC-MS/MS and pathway mapping. Proteomics 2013; 13:1325-33. [PMID: 23401153 DOI: 10.1002/pmic.201200369] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 11/30/2012] [Accepted: 12/10/2012] [Indexed: 01/01/2023]
Abstract
Protein kinase signaling regulates human hematopoietic stem/progenitor cell (HSPC) fate, yet little is known about critical pathway substrates. To address this, we have developed and applied a large-scale, empirically optimized phosphopeptide affinity enrichment strategy with high-throughput 2D LC-MS/MS screening to evaluate the phosphoproteome of an isolated human CD34(+) HSPC population. We first used hydrophilic interaction chromatography as a first dimension separation to separate and simplify protein digest mixtures into discrete fractions. Phosphopeptides were then enriched off-line using TiO2 -coated magnetic beads and subsequently detected online by C18 RP nanoflow HPLC using data-dependent MS/MS high-energy collision-activated dissociation fragmentation on a high-performance Orbitrap hybrid tandem mass spectrometer. We identified 15 533 unique phosphopeptides in 3574 putative phosphoproteins. Systematic computational analysis revealed biological pathways and phosphopeptide motifs enriched in CD34(+) HSPC that are markedly different from those observed in an analogous parallel analysis of isolated human T cells, pointing to the possible involvement of specific kinase-substrate relationships within activated cascades driving hematopoietic renewal, commitment, and differentiation.
Collapse
Affiliation(s)
- Hongbo Guo
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Kim J, Jeon YJ, Kim HE, Shin JM, Chung HM, Chae JI. Comparative proteomic analysis of endothelial cells progenitor cells derived from cord blood- and peripheral blood for cell therapy. Biomaterials 2012; 34:1669-85. [PMID: 23218840 DOI: 10.1016/j.biomaterials.2012.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/10/2012] [Indexed: 11/29/2022]
Abstract
Vasculopathy due to ischemia in damaged tissues is a major cause of morbidity and mortality. To treat these conditions, endothelial progenitor cells (EPCs) from various sources, such as umbilical cord or peripheral blood, have been the focus of the regenerative medicine field due to their proliferative and vasculogenic activities. However, the fundamental, molecular-level differences between EPCs obtained from different cellular sources have rarely been studied. In this study, we established endothelial progenitor cells derived from cord blood- and peripheral blood (CB- and PB-EPCs) and investigated their fundamental differences at the cellular and molecular levels through a combination of stem cell biology techniques and proteomic analysis. Our results suggest that specifically up-regulated factors such as STMIN 1, CFL 1, PARK 7, NME 1, GLO 1, HSP 27 and PRDX 2 in CB-EPCs as key elements that could be functionally active in ischemic regions. We also discussed functional behaviors important for inducing and maintaining long-lasting blood vessels under ischemic conditions. As a result, CB-EPCs retained a higher anti-oxidant and migration ability than PB-EPCs in vitro. Furthermore, CB-EPCs retained a higher therapeutic efficacy than PB-EPCs in a hindlimb ischemic disease model. The up-regulated expression pattern of STMIN 1, CFL 1, PARK 7, NME 1, GLO 1, HSP 27 and PRDX 2 was confirmed under several conditions in vitro and in vivo, indicating that the up-regulation of these molecules in CB-EPCs may be critical to the mechanism of healing in ischemic conditions and that CB-EPCs may be more appropriate for inducing neo-vessels. Thus, these results may aid in predetermining which cell sources will be of value for cell-based therapies of pathological conditions and identify several candidate molecules that may be involved in the therapeutic mechanism for ischemia.
Collapse
Affiliation(s)
- Jumi Kim
- CHA Bio & Diostech Co., Ltd., 606-16 Yeoksam 1 dong, Gangnam gu, Seoul 135-907, Republic of Korea
| | | | | | | | | | | |
Collapse
|
9
|
Genomic and proteomic analysis of the impact of mitotic quiescence on the engraftment of human CD34+ cells. PLoS One 2011; 6:e17498. [PMID: 21408179 PMCID: PMC3049784 DOI: 10.1371/journal.pone.0017498] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 02/07/2011] [Indexed: 12/25/2022] Open
Abstract
It is well established that in adults, long-term repopulating hematopoietic stem cells (HSC) are mitotically quiescent cells that reside in specialized bone marrow (BM) niches that maintain the dormancy of HSC. Our laboratory demonstrated that the engraftment potential of human HSC (CD34+ cells) from BM and mobilized peripheral blood (MPB) is restricted to cells in the G0 phase of cell cycle but that in the case of umbilical cord blood (UCB) -derived CD34+ cells, cell cycle status is not a determining factor in the ability of these cells to engraft and sustain hematopoiesis. We used this distinct in vivo behavior of CD34+ cells from these tissues to identify genes associated with the engraftment potential of human HSC. CD34+ cells from BM, MPB, and UCB were fractionated into G0 and G1 phases of cell cycle and subjected in parallel to microarray and proteomic analyses. A total of 484 target genes were identified to be associated with engraftment potential of HSC. System biology modeling indicated that the top four signaling pathways associated with these genes are Integrin signaling, p53 signaling, cytotoxic T lymphocyte-mediated apoptosis, and Myc mediated apoptosis signaling. Our data suggest that a continuum of functions of hematopoietic cells directly associated with cell cycle progression may play a major role in governing the engraftment potential of stem cells. While proteomic analysis identified a total of 646 proteins in analyzed samples, a very limited overlap between genomic and proteomic data was observed. These data provide a new insight into the genetic control of engraftment of human HSC from distinct tissues and suggest that mitotic quiescence may not be the requisite characteristic of engrafting stem cells, but instead may be the physiologic status conducive to the expression of genetic elements favoring engraftment.
Collapse
|
10
|
Wang W, Gou L, Yang J. What can proteomics teach us about bone marrow aging? Expert Rev Proteomics 2010; 7:799-802. [PMID: 21142879 DOI: 10.1586/epr.10.96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
11
|
Liumbruno G, D’Alessandro A, Grazzini G, Zolla L. How has proteomics informed transfusion biology so far? Crit Rev Oncol Hematol 2010; 76:153-72. [DOI: 10.1016/j.critrevonc.2010.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2009] [Revised: 12/22/2009] [Accepted: 01/07/2010] [Indexed: 01/06/2023] Open
|
12
|
D’Alessandro A, Grazzini G, Giardina B, Zolla L. In Silico Analyses of Proteomic Data Suggest a Role for Heat Shock Proteins in Umbilical Cord Blood Hematopoietic Stem Cells. Stem Cell Rev Rep 2010; 6:532-47. [DOI: 10.1007/s12015-010-9180-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
13
|
Lee MW, Jang IK, Yoo KH, Sung KW, Koo HH. Stem and progenitor cells in human umbilical cord blood. Int J Hematol 2010; 92:45-51. [PMID: 20577840 DOI: 10.1007/s12185-010-0619-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 05/17/2010] [Accepted: 05/25/2010] [Indexed: 12/28/2022]
Abstract
Both stem cells and progenitor cells are present in umbilical cord blood (UCB) at a high frequency, making these cells a major target population for experimental and clinical studies. As the use of autologous or allogeneic hematopoietic stem cell transplantation in the treatment of various diseases has grown rapidly in recent years, the concept of UCB banking for future use has drawn increasing interest. Stem and progenitor cells derived from UCB offer multiple advantages over adult stem cells, such as their immaturity (which may play a significant role in reducing rejection after transplantation into a mismatched host) and ability to produce large quantities of homogeneous tissue or cells. These cells can also differentiate across tissue lineage boundaries into neural, cardiac, epithelial, hepatic, and dermal tissues. Human UCB provides an alternative cell source that is ethically acceptable and widely supported by the public. This paper summarizes the characteristics of human UCB-derived stem and progenitor cells and their potential therapeutic use for tissue and cell regeneration.
Collapse
Affiliation(s)
- Myoung Woo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul, Korea
| | | | | | | | | |
Collapse
|
14
|
Ly L, Wasinger VC. Mass and charge selective protein fractionation for the differential analysis of T-cell and CD34+ stem cell proteins from cord blood. J Proteomics 2010; 73:571-8. [DOI: 10.1016/j.jprot.2009.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/11/2009] [Accepted: 09/02/2009] [Indexed: 12/11/2022]
|
15
|
Monti E, Bonten; E, D'Azzo A, Bresciani R, Venerando B, Borsani G, Schauer R, Tettamanti G. Sialidases in Vertebrates. Adv Carbohydr Chem Biochem 2010; 64:403-79. [DOI: 10.1016/s0065-2318(10)64007-3] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
16
|
D'Alessandro A, Liumbruno G, Grazzini G, Pupella S, Lombardini L, Zolla L. Umbilical cord blood stem cells: Towards a proteomic approach. J Proteomics 2010; 73:468-82. [DOI: 10.1016/j.jprot.2009.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 06/04/2009] [Accepted: 06/16/2009] [Indexed: 02/07/2023]
|
17
|
Abstract
The field of cord blood transplantation has come a long way since the first transplant more than 20 years ago. Advancements in the field will require continuing efforts to better understand hematopoietic stem and progenitor cell function and engraftment. Cautious optimism is inherent in the potential relevance and applicability of nonhematopoietic stem and progenitor cell types found in cord blood, and induced pluripotent stem cells generated from cord blood cells. Rigorous investigations and close interactions between scientific and clinical investigators are required to translate human in vitro and animal in vivo findings into clinical utility.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA.
| |
Collapse
|
18
|
Gadhoum SZ, Sackstein R. CD15 expression in human myeloid cell differentiation is regulated by sialidase activity. Nat Chem Biol 2008; 4:751-7. [PMID: 18953356 PMCID: PMC2597094 DOI: 10.1038/nchembio.116] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 09/08/2008] [Indexed: 01/29/2023]
Abstract
The glycan determinant CD15 (also known as Lewis x, or Le(x)) is a distinguishing marker for human myeloid cells and mediates neutrophil adhesion to dendritic cells. Despite broad interest in this structure, the mechanisms underlying CD15 expression remain relatively uncharacterized. Accordingly, we investigated the molecular basis of increasing CD15 expression associated with human myeloid cell differentiation. Flow cytometric analysis of differentiating cells together with biochemical studies using inhibitors of glycan synthesis and of sialidases showed that increased CD15 expression is not due to de novo biosynthesis of CD15, but results predominantly from induction of alpha(2-3)-sialidase activity, which yields CD15 from cell-surface sialyl-CD15 (also known as sialyl-Lewis x, sLe(x) or CD15s). This differentiation-associated conversion of surface CD15s to CD15 occurs mainly on glycoproteins. Until now, modulation of post-translational glycan modifications has been attributed solely to dynamic variations in glycosyltransferase expression. Our results unveil a new paradigm by demonstrating a critical role for post-Golgi membrane glycosidase activity in the 'biosynthesis' of a key glycan determinant.
Collapse
Affiliation(s)
- Samah Zeineb Gadhoum
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts 02115
- Harvard Skin Disease Research Center, Harvard Medical School, Boston, Massachusetts, 02115
| | - Robert Sackstein
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts 02115
- Harvard Skin Disease Research Center, Harvard Medical School, Boston, Massachusetts, 02115
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts 02115
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| |
Collapse
|
19
|
Liumbruno G, D'Amici GM, Grazzini G, Zolla L. Transfusion medicine in the era of proteomics. J Proteomics 2008; 71:34-45. [DOI: 10.1016/j.jprot.2008.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 02/18/2008] [Accepted: 02/18/2008] [Indexed: 12/21/2022]
|
20
|
Liumbruno GM. Proteomics: applications in transfusion medicine. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2008; 6:70-85. [PMID: 18946951 PMCID: PMC2626841 DOI: 10.2450/2008.0038-07] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Accepted: 12/20/2007] [Indexed: 12/30/2022]
|
21
|
Beyer S, Mix E, Hoffrogge R, Lünser K, Völker U, Rolfs A. Neuroproteomics in stem cell differentiation. Proteomics Clin Appl 2007; 1:1513-23. [PMID: 21136647 DOI: 10.1002/prca.200700324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Indexed: 12/31/2022]
Abstract
The term "proteome" is used to describe the entire complement of proteins in a given organism or in a system at a given time. Proteome analysis in neuroscience, also called "neuroproteomics" or "neuromics" is in its initial stage, and shows a deficit of studies in the context of brain development. It is the main objective of this review to illustrate the potential of neuroproteomics as a tool to unravel the differentiation of neural stem or progenitor cells to terminally differentiated neurons. Experimental results regarding the rat striatal progenitor model cell line ST14A are presented to illustrate the large rearrangements of the proteome during the differentiation process of neural progenitor cells and their modification by neurotrophic factors like the glial cell line-derived neurotrophic factor (GDNF). Thereby native stem cells and cells transfected with GDNF gene were investigated at the proliferative state and at seven time points up to 72 h after induction of differentiation. In addition, the immortalized human fetal midbrain stem cell line ReNcell VM was analyzed in order to detect stem cell differentiation associated changes of the protein profile. This review gives also an outlook on technical improvements and perspectives of application of neural stem cell proteomics.
Collapse
Affiliation(s)
- Susanne Beyer
- Neurobiological Laboratory, Department of Neurology, Medical Faculty, University of Rostock, Rostock, Germany
| | | | | | | | | | | |
Collapse
|
22
|
Abstract
Gene expression analyses of stem cells (SCs) will help to uncover or further define signaling pathways and molecular mechanisms involved in the maintenance of self-renewal, pluripotency, and/or multipotency. In recent years, proteomic approaches have produced a wealth of data identifying proteins and mechanisms involved in SC proliferation and differentiation. Although many proteomics techniques have been developed and improved in peptide and protein separation, as well as mass spectrometry, several important issues, including sample heterogeneity, post-translational modifications, protein-protein interaction, and high-throughput quantification of hydrophobic and low-abundance proteins, still remain to be addressed and require further technical optimization. This review summarizes the methodologies used and the information gathered with proteome analyses of SCs, and it discusses biological and technical challenges for proteomic study of SCs. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
|
23
|
Hoffrogge R, Beyer S, Hübner R, Mikkat S, Mix E, Scharf C, Schmitz U, Pauleweit S, Berth M, Zubrzycki IZ, Christoph H, Pahnke J, Wolkenhauer O, Uhrmacher A, Völker U, Rolfs A. 2-DE profiling of GDNF overexpression-related proteome changes in differentiating ST14A rat progenitor cells. Proteomics 2007; 7:33-46. [PMID: 17146836 DOI: 10.1002/pmic.200600614] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Targeted differentiation of neural progenitor cells (NPCs) is a challenge for treatment of neurodegenerative diseases by cell replacement therapy and cell signalling manipulation. Here, we applied a proteome profiling approach to the rat striatal progenitor model cell line ST14A in order to elucidate cellular differentiation processes. Native cells and cells transfected with the glial cell line-derived neurotrophic factor (GDNF) gene were investigated at the proliferative state and at seven time points up to 72 h after induction of differentiation. 2-DE combined with MALDI-MS was used to create a reference 2-DE-map of 652 spots of which 164 were identified and assigned to 155 unique proteins. For identification of protein expression changes during cell differentiation, spot patterns of triplicate gels were matched to the 2-DE-map. Besides proteins that display expression changes in native cells, we also noted 43 protein-spots that were differentially regulated by GDNF overexpression in more than four time points of the experiment. The expression patterns of putative differentiation markers such as annexin 5 (ANXA5), glucosidase II beta subunit (GLU2B), phosphatidylethanolamine-binding protein 1 (PEBP1), myosin regulatory light chain 2-A (MLRA), NASCENT polypeptide-associated complex alpha (NACA), elongation factor 2 (EF2), peroxiredoxin-1 (PRDX1) and proliferating cell nuclear antigen (PCNA) were verified by Western blotting. The results reflect the large rearrangements of the proteome during the differentiation process of NPCs and their strong modification by neurotrophic factors like GDNF.
Collapse
Affiliation(s)
- Raimund Hoffrogge
- Department of Neurology, Medical Faculty, Neurobiological Laboratory, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Blood-based therapeutics are cellular or plasma components derived from human blood. Their production requires appropriate selection and treatment of the donor and processing of cells or plasma proteins. In contrast to clearly defined, chemically synthesized drugs, blood-derived therapeutics are highly complex mixtures of plasma proteins or even more complex cells. Pathogen transmission by the product as well as changes in the integrity of blood constituents resulting in loss of function or immune modulation are currently important issues in transfusion medicine. Protein modifications can occur during various steps of the production process, such as acquisition, enrichment of separate components (e.g. coagulation factors, cell populations), virus inactivation, conservation, and storage. Contemporary proteomic strategies allow a comprehensive assessment of protein modifications with high coverage, offer capabilities for qualitative and even quantitative analysis, and for high-throughput protein identification. Traditionally, proteomics approaches predominantly relied on two-dimensional gel electrophoresis (2-DE). Even if 2-DE is still state of the art, it has inherent limitations that are mainly based on the physicochemical properties of the proteins analyzed; for example, proteins with extremes in molecular mass and hydrophobicity (most membrane proteins) are difficult to assess by 2-DE. These limitations have fostered the development of mass spectrometry centered on non-gel-based separation approaches, which have proven to be highly successful and are thus complementing and even partially replacing 2-DE-based approaches. Although blood constituents have been extensively analyzed by proteomics, this technology has not been widely applied to assess or even improve blood-derived therapeutics, or to monitor the production processes. As proteomic technologies have the capacity to provide comprehensive information about changes occurring during processing and storage of blood products, proteomics can potentially guide improvement of pathogen inactivation procedures and engineering of stem cells, and may also allow a better understanding of factors influencing the immunogenicity of blood-derived therapeutics. An important development in proteomics is the reduction of inter-assay variability. This now allows the screening of samples taken from the same product over time or before and after processing. Optimized preparation procedures and storage conditions will reduce the risk of protein alterations, which in turn may contribute to better recovery, reduced exposure to allogeneic proteins, and increased transfusion safety.
Collapse
Affiliation(s)
- Thomas Thiele
- Institute of Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany
| | | | | | | |
Collapse
|
25
|
Stem cells and proteomics. Chin J Cancer Res 2006. [DOI: 10.1007/s11670-006-0161-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
26
|
Kirouac DC, Zandstra PW. Understanding cellular networks to improve hematopoietic stem cell expansion cultures. Curr Opin Biotechnol 2006; 17:538-47. [PMID: 16899360 DOI: 10.1016/j.copbio.2006.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 07/02/2006] [Accepted: 07/21/2006] [Indexed: 12/16/2022]
Abstract
Efforts to develop culture technologies capable of eliciting robust human blood stem cell growth have met with limited success. Considering that adult stem cell cultures are complex systems, comprising multiple cell types with dynamically changing intracellular signalling environments and cellular compositions, this is not surprising. Typically treated as single-input single-output systems, adult stem cell cultures are better described as complex, non-linear, multiple-input multiple-output systems wherein the proliferation of subpopulations of cells leads to the formation of intercellular endogenously secreted protein interaction networks. Genomic and proteomic tools need to be applied to generate high-throughput (and ideally high-content) biological measurements of stem cell culture evolution. Datasets describing cellular interaction networks need to be integrated into predictive models of in vitro stem cell development. Ultimately, such models will serve as a starting point for the rational design of blood stem cell expansion bioprocesses utilizing dynamic system perturbations to achieve the preferential expansion of target cell populations.
Collapse
Affiliation(s)
- Daniel C Kirouac
- Institute of Biomaterials and Biomedical Engineering, Terrance Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Room 1116 11th Floor, 160 College Street, M5S 3E1 Toronto, Ontario, Canada
| | | |
Collapse
|
27
|
Liu F, Lu J, Fan HH, Wang ZQ, Cui SJ, Zhang GA, Chi M, Zhang X, Yang PY, Chen Z, Han ZG. Insights into human CD34+ hematopoietic stem/progenitor cells through a systematically proteomic survey coupled with transcriptome. Proteomics 2006; 6:2673-92. [PMID: 16596711 DOI: 10.1002/pmic.200500032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Hematopoietic stem cells are capable of self-renewal and differentiation into different hematopoietic lineages. To gain a comprehensive understanding of hematopoietic stem/progenitor cells, a systematic proteomic survey of human CD34+ cells collected from human umbilical cord blood was performed, in which the proteins were separated by 1- and 2-DE, as well as by nano-LC, and subsequently identified by MS. A total of 370 distinct proteins identified from those cells provided new insights into the potential of the stem/progenitor cells because the nerve, gonad, and eye-associated proteins were reliably identified. Interestingly, the transcripts of 133 (35.9%) identified proteins were not found by the prevalent transcriptome approaches, although several selected transcripts could be detected by RT-PCR. Moreover, the heterogeneity of 33 proteins identified from 2-DE was attributable primarily to post-translational processes rather than to alternative splicing at transcriptional level. Furthermore, the biosyntheses of 15 proteins identified in this study appears not to be completely interrupted in spite of the fact that corresponding antisense RNAs were found in the existing transcriptome data. The integrated proteomic and transcriptomic analyses employed here provided a unique view of the human stem/progenitor cells.
Collapse
Affiliation(s)
- Feng Liu
- Chinese National Human Genome Center at Shanghai, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hoffrogge R, Mikkat S, Scharf C, Beyer S, Christoph H, Pahnke J, Mix E, Berth M, Uhrmacher A, Zubrzycki IZ, Miljan E, Völker U, Rolfs A. 2-DE proteome analysis of a proliferating and differentiating human neuronal stem cell line (ReNcell VM). Proteomics 2006; 6:1833-47. [PMID: 16475233 DOI: 10.1002/pmic.200500556] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The proteome of a proliferating human stem cell line was analyzed and then utilized to detect stem cell differentiation-associated changes in the protein profile. The analysis was conducted with a stable human fetal midbrain stem cell line (ReNcell VM) that displays the properties of a neural stem cell. Therefore, acquisition of proteomic data should be representative of cultured human neural stem cells (hNSCs) in general. Here we present a 2-DE protein-map of this cell line with annotations of 402 spots representing 318 unique proteins identified by MS. The subsequent proteome profiling of differentiating cells of this stem cell line at days 0, 4 and 7 of differentiation revealed changes in the expression of 49 identified spots that could be annotated to 45 distinct proteins. This differentiation-associated expression pattern was validated by Western blot analysis for transgelin-2, proliferating cell nuclear antigen, as well as peroxiredoxin 1 and 4. The group of regulated proteins also included NudC, ubiquilin-1, STRAP, stress-70 protein, creatine kinase B, glial fibrillary acidic protein and vimentin. Our results reflect the large rearrangement of the proteome during the differentiation process of the stem cells to terminally differentiated neurons and offer the possibility for further characterization of specific targets driving the stem cell differentiation.
Collapse
MESH Headings
- Blotting, Western
- Cell Differentiation
- Cell Line
- Cell Line, Transformed
- Cell Proliferation
- Cell Transformation, Viral
- Computational Biology
- Culture Media/chemistry
- Culture Media/pharmacology
- Databases, Protein
- Electrophoresis, Gel, Two-Dimensional
- Epidermal Growth Factor/pharmacology
- Fibroblast Growth Factor 2/pharmacology
- Genetic Markers
- Humans
- Mass Spectrometry
- Mesencephalon/cytology
- Mesencephalon/embryology
- Microfilament Proteins/analysis
- Microfilament Proteins/isolation & purification
- Microfilament Proteins/metabolism
- Muscle Proteins/analysis
- Muscle Proteins/isolation & purification
- Muscle Proteins/metabolism
- Neoplasm Proteins/analysis
- Neoplasm Proteins/isolation & purification
- Neoplasm Proteins/metabolism
- Neurons/cytology
- Peptide Mapping
- Peroxidases/analysis
- Peroxidases/isolation & purification
- Peroxidases/metabolism
- Peroxiredoxins
- Proliferating Cell Nuclear Antigen/analysis
- Proliferating Cell Nuclear Antigen/isolation & purification
- Proliferating Cell Nuclear Antigen/metabolism
- Proteome/analysis
- Retroviridae/genetics
- Selection, Genetic
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Stem Cells/cytology
- Stem Cells/physiology
- Transduction, Genetic
- Transgenes
- Trypsin/pharmacology
Collapse
Affiliation(s)
- Raimund Hoffrogge
- Neurobiological Laboratory, Department of Neurology, Medical Faculty, University of Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Thadikkaran L, Siegenthaler MA, Crettaz D, Queloz PA, Schneider P, Tissot JD. Recent advances in blood-related proteomics. Proteomics 2005; 5:3019-34. [PMID: 16041673 DOI: 10.1002/pmic.200402053] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Blood is divided in two compartments, namely, plasma and cells. The latter contain red blood cells, leukocytes, and platelets. From a descriptive medical discipline, hematology has evolved towards a pioneering discipline where molecular biology has permitted the development of prognostic and diagnostic indicators for disease. The recent advance in MS and protein separation now allows similar progress in the analysis of proteins. Proteomics offers great promise for the study of proteins in plasma/serum, indeed a number of proteomics databases for plasma/serum have been established. This is a very complex body fluid containing lipids, carbohydrates, amino acids, vitamins, nucleic acids, hormones, and proteins. About 1500 different proteins have recently been identified, and a number of potential new markers of diseases have been characterized. Here, examples of the enormous promise of plasma/serum proteomic analysis for diagnostic/prognostic markers and information on disease mechanism are given. Within the blood are also a large number of different blood cell types that potentially hold similar information. Proteomics of red blood cells, until now, has not improved our knowledge of these cells, in contrast to the major progresses achieved while studying platelets and leukocytes. In the future, proteomics will change several aspects of hematology.
Collapse
Affiliation(s)
- Lynne Thadikkaran
- Service régional vaudois de transfusion sanguine, Rue du Bugnon 27, CH-1005 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Cord blood (CB) has served as a clinically beneficial source of hematopoietic stem (HSC) and progenitor (HPC) cells for transplantation and correction of a large number of malignant and non-malignant disorders. The capacity of CB to perform these functions is intimately related to the quality and quantity of HSC and HPC present in CB. This review covers the biology of HSC and HPC, efforts to expand these cells ex vivo for enhanced clinical utility that has thus far not been very successful, and recent studies on attempts to enhance the homing and engrafting capability of HSC as an alternative means for more effective use of the limited numbers of CB cells collected. This review also highlights the presence in CB of mesenchymal stem cells, unrestricted somatic stem cells, endothelial progenitor cells and immune cells. The presence and biology of these non-HSC/HPC may open up future possibilities for additional clinical benefit of CB, a product considered mainly for discard before its clinical transplantation potential was realized in the late 1980s.
Collapse
Affiliation(s)
- H E Broxmeyer
- Department of Microbiology and Immunology, and the Walther Oncology Center, Indiana univrsity School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Current Awareness on Comparative and Functional Genomics. Comp Funct Genomics 2005. [PMCID: PMC2447482 DOI: 10.1002/cfg.421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|