1
|
Wu CH, Weng TF, Li JP, Wu KH. Biology and Therapeutic Properties of Mesenchymal Stem Cells in Leukemia. Int J Mol Sci 2024; 25:2527. [PMID: 38473775 DOI: 10.3390/ijms25052527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
This comprehensive review delves into the multifaceted roles of mesenchymal stem cells (MSCs) in leukemia, focusing on their interactions within the bone marrow microenvironment and their impact on leukemia pathogenesis, progression, and treatment resistance. MSCs, characterized by their ability to differentiate into various cell types and modulate the immune system, are integral to the BM niche, influencing hematopoietic stem cell maintenance and functionality. This review extensively explores the intricate relationship between MSCs and leukemic cells in acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, and chronic lymphocytic leukemia. This review also addresses the potential clinical applications of MSCs in leukemia treatment. MSCs' role in hematopoietic stem cell transplantation, their antitumor effects, and strategies to disrupt chemo-resistance are discussed. Despite their therapeutic potential, the dual nature of MSCs in promoting and inhibiting tumor growth poses significant challenges. Further research is needed to understand MSCs' biological mechanisms in hematologic malignancies and develop targeted therapeutic strategies. This in-depth exploration of MSCs in leukemia provides crucial insights for advancing treatment modalities and improving patient outcomes in hematologic malignancies.
Collapse
Affiliation(s)
- Cheng-Hsien Wu
- School of Medicine, National Defense Medical Center, Taipei 114, Taiwan
| | - Te-Fu Weng
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Ju-Pi Li
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| |
Collapse
|
2
|
Agarwal P, Li H, Choi K, Hueneman K, He J, Welner RS, Starczynowski DT, Bhatia R. TNF-α-induced alterations in stromal progenitors enhance leukemic stem cell growth via CXCR2 signaling. Cell Rep 2021; 36:109386. [PMID: 34260914 PMCID: PMC8292106 DOI: 10.1016/j.celrep.2021.109386] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/30/2021] [Accepted: 06/21/2021] [Indexed: 11/30/2022] Open
Abstract
Chronic myeloid leukemia (CML) is propagated by leukemia stem cells (LSCs) that are not eradicated by tyrosine kinase inhibitor (TKI) treatment and persist as a source of disease recurrence. Bone marrow (BM) mesenchymal niches play an essential role in hematopoietic stem cell (HSC) and LSC maintenance. Using a murine CML model, we examine leukemia-induced alterations in mesenchymal cell populations. We show that 6C3+ stromal progenitors expand in CML BM and exhibit increased LSC but reduced HSC supportive capacity. Tumor necrosis factor alpha (TNF-α) signaling mediates expansion and higher expression of CXCL1 in CML BM 6C3+ cells and higher expression of the CXCL1 receptor CXCR2 in LSCs. CXCL1 enhances LSC proliferation and self-renewal, whereas CXCR2 inhibition reduces LSC growth and enhances LSC targeting in combination with tyrosine kinase inhibitors (TKIs). We find that TNF-α-mediated alterations in CML BM stromal niches enhance support of LSC maintenance and growth via CXCL1-CXCR2 signaling and that CXCR2 inhibition effectively depletes CML LSCs.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Bone Marrow/pathology
- Cell Cycle/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Chemokines/metabolism
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Humans
- Inflammation/genetics
- Inflammation/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice, Inbred C57BL
- Middle Aged
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Protein Kinase Inhibitors/pharmacology
- Receptors, Interleukin-8B/metabolism
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/metabolism
- Mice
Collapse
Affiliation(s)
- Puneet Agarwal
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA; Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hui Li
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Kwangmin Choi
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kathleen Hueneman
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jianbo He
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Robert S Welner
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ravi Bhatia
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA.
| |
Collapse
|
3
|
Matsuo T, Tashiro H, Sumiyoshi R, Saito S, Shirasaki R, Shirafuji N. Functional expression cloning of molecules inducing CD34 expression in bone marrow-derived stromal myofibroblasts. Biochem Biophys Res Commun 2020; 533:1283-1289. [PMID: 33066959 DOI: 10.1016/j.bbrc.2020.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 01/01/2023]
Abstract
We have previously shown a fraction of stromal fibroblasts/myofibroblasts (Fibs) from leukemic bone marrow cells expresses leukemia-specific transcripts along with hematopoietic and Fib-related markers. Normal bone marrow-derived Fibs (nFibs) do not express CD34 or CD45; however, nFibs may express hematopoietic markers with some specific stimulations. CD34 expression was detected in nFib cultures following the addition of a culture supernatant of blood mononuclear cells stimulated with phytohemagglutinin (PHA)-P. To identify the molecules responsible for inducing CD34 expression in nFibs, cDNA clones were isolated using functional expression cloning with a library constructed from PHA-P-stimulated human blood mononuclear cells. Positive clones inducing CD34 transcription in nFibs were selected. We confirmed that an isolated positive cDNA clone encoded human interleukin (IL)-1 beta (β). CD34 expression was observed in the nFib cultures with recombinant human (rh) IL-1β protein. And CD34 transcription was suppressed when a rhIL-1β neutralizing antibody was added to the IL-1β-stimulated nFib cultures. nFibs expressed gp130 and IL-6 receptors, and CD45 expression was detected in nFibs cultured with rhIL-1β and rhIL-6. Chronic myelogenous leukemia (CML) cells reportedly respond well to IL-1β. When CML-derived Fibs were cultured with rhIL-1β and rhIL-6, CD45-positive cells increased in number. Cell fate may be influenced by an external specific stimulation without gene introduction.
Collapse
Affiliation(s)
- Takuji Matsuo
- Department of Hematology/Oncology, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Haruko Tashiro
- Department of Hematology/Oncology, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Ritsu Sumiyoshi
- Department of Hematology/Oncology, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Sumiko Saito
- Department of Hematology/Oncology, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Ryosuke Shirasaki
- Department of Hematology/Oncology, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Naoki Shirafuji
- Department of Hematology/Oncology, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8606, Japan.
| |
Collapse
|
4
|
Saki N, Farshchi N, Azandeh S, Jalali M. Biologic profile evaluation of mesenchymal stem cells in co-culture with K562 cells. CLINICAL CANCER INVESTIGATION JOURNAL 2020. [DOI: 10.4103/ccij.ccij_24_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
5
|
Li H, Li J, Cheng J, Chen X, Zhou L, Li Z. AML‑derived mesenchymal stem cells upregulate CTGF expression through the BMP pathway and induce K562‑ADM fusiform transformation and chemoresistance. Oncol Rep 2019; 42:1035-1046. [PMID: 31322275 PMCID: PMC6667869 DOI: 10.3892/or.2019.7237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 07/12/2019] [Indexed: 12/27/2022] Open
Abstract
Bone marrow‑derived mesenchymal stem cells (MSCs), are the basic cellular components that make up the bone marrow microenvironment (BMM). In acute myeloid leukemia (AML), the morphology and function of MSCs changes in accordance with the transformation of the BMM. Moreover, the transformation of MSCs into osteoblasts is determined through the bone morphogenetic protein (BMP) pathway, ultimately leading to an altered expression of the downstream adhesion molecule, connective tissue growth factor (CTGF). In this study, we aimed to explore the interaction of possible pathways in AML‑derived mesenchymal stem cells (AML‑MSCs) co‑cultured with the K562 and K562‑ADM cell lines. AML‑MSCs were co‑cultured with K562/K562‑ADM cells, and the interactions between the cells were verified by morphological detection, peroxidase staining (POX), reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and fluorescence in situ hybridization (FISH). The proliferation of K562/K562‑ADM cells under co‑culture conditions was detected by flow cytometry. The expression levels of BMP4 and CTGF were examined by RT‑qPCR and western blot (WB) analysis. The detection of interleukin (IL)‑6 and IL‑32 was also determined by enzyme linked immunosorbent assay (ELISA). In the co‑culture system, the K562‑ADM cells underwent fusiform transformation. The occurrence of this transformation was associated with an increased expression of CTGF due to the dysregulation of the BMP pathway. The AML‑MSCs promoted the proliferation of the K562‑ADM cell, but inhibited that of the K562 cells. These findings were confirmed by changes in the expression of the soluble cytokines, IL‑6 and IL‑32. On the whole, the findings of this study demonstrate that AML‑MSCs regulate the expression of CTGF through the BMP pathway. In addition, they affect cytokine production, induce spindle‑shaped transformation, and increase drug resistance in the K562‑ADM cells. Thus, the morphological transformation through the BMP pathway provides us with a novel target with which to circumvent tumor occurrence, development, drug resistance, invasion and metastasis.
Collapse
Affiliation(s)
- Haiying Li
- Department of Central Laboratory, The First Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Juan Li
- Department of Central Laboratory, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Juan Cheng
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xuan Chen
- Department of Central Laboratory, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Lanxia Zhou
- Department of Central Laboratory, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhao Li
- Department of Central Laboratory, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
6
|
Xie J, Chen J, Wang B, He X, Huang H. Bone mesenchymal stromal cells exhibit functional inhibition but no chromosomal aberrations in chronic myelogenous leukemia. Oncol Lett 2019; 17:999-1007. [PMID: 30655859 PMCID: PMC6312938 DOI: 10.3892/ol.2018.9681] [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: 02/20/2018] [Accepted: 09/06/2018] [Indexed: 11/21/2022] Open
Abstract
Chronic myelogenous leukemia (CML) is a myeloproliferative neoplasia characterized by the presence of the Philadelphia (Ph) chromosome in hematopoietic cells (HCs). As one of the most important components of the bone marrow microenvironment (BMM), bone mesenchymal stromal cells (BMSCs) are critical in the development of leukemia and essential in the regulation of hematopoiesis. However, little is known regarding the alterations of BMSCs in CML. The current study performed Cell Counting Kit-8 and colony-forming unit fibroblast assays to evaluate the proliferative ability of BMSCs. The percentage of senescent BMSCs was evaluated by a senescence-associated β-galactosidase staining assay. Subsequently, a long-term culture-initiating cell assay was designed to explore the HC-supporting capacity of the BMSCs. Furthermore, cytogenetics were detected by conventional cytogenetic analysis and fluorescence in situ hybridization analysis. The current results revealed that CML-BMSCs exhibited decreased cell proliferation and impaired HC-support capacity, as well as increased susceptibility to senescence. No chromosomal aberrations, including the absence of the Ph chromosome, were noted in all CML-BMSCs. In conclusion, the current study demonstrated functional inhibition of CML-BMSCs; however, no signs of chromosomal aberrations were observed, thereby providing insight into the changes occurring in the CML-BMM.
Collapse
Affiliation(s)
- Jieqiong Xie
- Central Laboratory, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jiadi Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Bin Wang
- Central Laboratory, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xuchun He
- Department of Medical Technology, Fujian Health Career Technical College, Fuzhou, Fujian 350101, P.R. China
| | - Huifang Huang
- Central Laboratory, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
7
|
Galán-Díez M, Cuesta-Domínguez Á, Kousteni S. The Bone Marrow Microenvironment in Health and Myeloid Malignancy. Cold Spring Harb Perspect Med 2018; 8:a031328. [PMID: 28963115 PMCID: PMC6027930 DOI: 10.1101/cshperspect.a031328] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hematopoietic stem cells (HSCs) interact dynamically with an intricate network of cells in the bone marrow (BM) microenvironment or niche. These interactions provide instructive cues that influence the production and lineage determination of different types of blood cells and maintenance of HSC quiescence. They also contribute to hematopoietic deregulation and hematological myeloid malignancies. Alterations in the BM niche are commonly observed in myeloid malignancies and contribute to the aberrant function of myelodysplastic and leukemia-initiating stem cells. In this work, we review how different components of the BM niche affect normal hematopoiesis, the molecular signals that govern this interaction, and how genetic changes in stromal cells or alterations in remodeled malignant BM niches contribute to myeloid malignancies. Understanding the intricacies between normal and malignant niches and their modulation may provide insights into developing novel therapeutics for blood disorders.
Collapse
Affiliation(s)
- Marta Galán-Díez
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, New York 10032
| | - Álvaro Cuesta-Domínguez
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, New York 10032
| | - Stavroula Kousteni
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, New York 10032
| |
Collapse
|
8
|
BCR-ABL1-positive microvesicles malignantly transform human bone marrow mesenchymal stem cells in vitro. Acta Pharmacol Sin 2017; 38:1475-1485. [PMID: 28836580 DOI: 10.1038/aps.2017.116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 05/19/2017] [Indexed: 12/21/2022] Open
Abstract
The intercellular communication between leukemia cells and bone marrow mesenchymal stem cells (BM-MSCs) plays more important role in chronic myeloid leukemia (CML) than we previously understood. Recently, we found that microvesicles released from human leukemia cell line K562 (K562-MVs) containing BCR-ABL1 mRNA malignantly transformed normal hematopoietic transplants. Here, we investigated whether K562-MVs contribute to the transformation of human bone marrow mesenchymal stem cells (BM-MSCs). We showed that K562-MVs could be integrated into co-cultured normal BM-MSCs and dose-dependently enhanced the proliferation of BM-MSCs. Meanwhile, K562-MVs (400 ng/mL) significantly increased the expression of BCR-ABL1 in these BM-MSCs, accompanied by the enhanced secretion of TGF-β1. These BM-MSCs in turn could trigger the TGF-β1-dependent proliferation of K562 cells. Moreover, we confirmed the presence of BCR-ABL1 in circulating MVs from 11 CML patients. Compared to the normal BM-MSCs, the BM-MSCs from CML patients more effectively increased the BCR-ABL1 expression and TGF-β1 secretion in K562 cells as well as the proliferation of K562 cells. Our findings enrich the mechanisms involved in the interaction between leukemia cells and BM-MSCs and provide novel ways to monitor minimal residual disease and worthwhile approaches to treat CML.
Collapse
|
9
|
Desbourdes L, Javary J, Charbonnier T, Ishac N, Bourgeais J, Iltis A, Chomel JC, Turhan A, Guilloton F, Tarte K, Demattei MV, Ducrocq E, Rouleux-Bonnin F, Gyan E, Hérault O, Domenech J. Alteration Analysis of Bone Marrow Mesenchymal Stromal Cells from De Novo Acute Myeloid Leukemia Patients at Diagnosis. Stem Cells Dev 2017; 26:709-722. [PMID: 28394200 DOI: 10.1089/scd.2016.0295] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Bone marrow (BM)-derived mesenchymal stromal cells (MSCs) frequently display alterations in several hematologic disorders, such as acute lymphoid leukemia, acute myeloid leukemia (AML), and myelodysplastic syndromes. In acute leukemias, it is not clear whether MSC alterations contribute to the development of the malignant clone or whether they are simply the effect of tumor expansion on the microenvironment. We extensively investigated the characteristics of MSCs isolated from the BM of patients with de novo AML at diagnosis (L-MSCs) in terms of phenotype (gene and protein expression, apoptosis and senescence levels, DNA double-strand break formation) and functions (proliferation and clonogenic potentials, normal and leukemic hematopoiesis-supporting activity). We found that L-MSCs show reduced proliferation capacity and increased apoptosis levels compared with MSCs from healthy controls. Longer population doubling time in L-MSCs was not related to the AML characteristics at diagnosis (French-American-British type, cytogenetics, or tumor burden), but was related to patient age and independently associated with poorer patient outcome, as was cytogenetic prognostic feature. Analyzing, among others, the expression of 93 genes, we found that proliferative deficiency of L-MSCs was associated with a perivascular feature at the expense of the osteo-chondroblastic lineage with lower expression of several niche factors, such as KITLG, THPO, and ANGPT1 genes, the cell adhesion molecule VCAM1, and the developmental/embryonic genes, BMI1 and DICER1. L-MSC proliferative capacity was correlated positively with CXCL12, THPO, and ANGPT1 expression and negatively with JAG1 expression. Anyway, these changes did not affect their in vitro capacity to support normal hematopoiesis and to modify leukemic cell behavior (protection from apoptosis and quiescence induction). Our findings indicate that BM-derived MSCs from patients with newly diagnosed AML display phenotypic and functional alterations such as proliferative deficiency that could be attributed to tumor progression, but does not seem to play a special role in the leukemic process.
Collapse
Affiliation(s)
- Laura Desbourdes
- 1 CNRS UMR 7292, LNOx Team, François Rabelais University , Tours, France
| | - Joaquim Javary
- 1 CNRS UMR 7292, LNOx Team, François Rabelais University , Tours, France
| | - Thomas Charbonnier
- 2 Department of Biological Hematology, University Hospital of Tours , Tours, France
| | - Nicole Ishac
- 1 CNRS UMR 7292, LNOx Team, François Rabelais University , Tours, France
| | - Jerome Bourgeais
- 1 CNRS UMR 7292, LNOx Team, François Rabelais University , Tours, France
| | - Aurore Iltis
- 2 Department of Biological Hematology, University Hospital of Tours , Tours, France .,3 Department of Hematology and Cell Therapy, University Hospital of Tours , Tours, France
| | - Jean-Claude Chomel
- 4 INSERM U935, University of Poitiers , Poitiers, France .,5 Department of Biological Oncology, University Hospital of Poitiers , Poitiers, France
| | - Ali Turhan
- 6 INSERM U935, University of Paris-Sud 11 , Paris, France .,7 Department of Hematology, University Hospitals of Paris-Sud , Le Kremlin Bicêtre, France
| | | | - Karin Tarte
- 8 INSERM U917, University of Rennes 1 , Rennes, France .,9 Department of Immunology, Cellular Therapy and Hematopoiesis, University Hospital of Rennes , Rennes, France .,10 CNRS GDR 3697, MicroNiT National Network, Tours , France
| | - Marie-Veronique Demattei
- 11 CNRS UMR 7292, Telomeres and Genome Stability Team, François Rabelais University , Tours, France
| | - Elfi Ducrocq
- 1 CNRS UMR 7292, LNOx Team, François Rabelais University , Tours, France
| | | | - Emmanuel Gyan
- 1 CNRS UMR 7292, LNOx Team, François Rabelais University , Tours, France .,3 Department of Hematology and Cell Therapy, University Hospital of Tours , Tours, France
| | - Olivier Hérault
- 1 CNRS UMR 7292, LNOx Team, François Rabelais University , Tours, France .,2 Department of Biological Hematology, University Hospital of Tours , Tours, France .,10 CNRS GDR 3697, MicroNiT National Network, Tours , France
| | - Jorge Domenech
- 1 CNRS UMR 7292, LNOx Team, François Rabelais University , Tours, France .,2 Department of Biological Hematology, University Hospital of Tours , Tours, France .,10 CNRS GDR 3697, MicroNiT National Network, Tours , France
| |
Collapse
|
10
|
Abstract
Research in the last few years has revealed a sophisticated interaction network between multiple bone marrow cells that regulate different hematopoietic stem cell (HSC) properties such as proliferation, differentiation, localization, and self-renewal during homeostasis. These mechanisms are essential to keep the physiological HSC numbers in check and interfere with malignant progression. In addition to the identification of multiple mutations and chromosomal aberrations driving the progression of myeloid malignancies, alterations in the niche compartment recently gained attention for contributing to disease progression. Leukemic cells can remodel the niche into a permissive environment favoring leukemic stem cell expansion over normal HSC maintenance, and evidence is accumulating that certain niche alterations can even induce leukemic transformation. Relapse after chemotherapy is still a major challenge during treatment of myeloid malignancies, and cure is only rarely achieved. Recent progress in understanding the niche-imposed chemoresistance mechanisms will likely contribute to the improvement of current therapeutic strategies. This article discusses the role of different niche cells and their stage- and disease-specific roles during progression of myeloid malignancies and in response to chemotherapy.
Collapse
|
11
|
Gaafar TM, Raafat II, Aly AA, Mohamed NAEG, Farid RJ, Saad NE, El-Hawary R, Mostafaa N, Ahmed MM. Detection of BCR/ABL Translocation in Bone Marrow Derived Mesenchymal Stem Cells in Egyptian CML Patients. Open Access Maced J Med Sci 2015; 3:231-6. [PMID: 27275226 PMCID: PMC4877858 DOI: 10.3889/oamjms.2015.040] [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: 10/12/2014] [Revised: 01/09/2015] [Accepted: 03/31/2015] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND: Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder of hematopoietic stem cells. It is characterized at the cytogenetic level by Philadelphia (ph) chromosome and at the molecular level by the BCR/ABL gene rearrangement. Bone marrow derived mesenchymal stem cells (MSCs) are pluripotent stem cells that can differentiate into several mesenchymal tissues. AIM: To observe the biological characteristics of MSCS from CML patients and to determine whether MSCs harbor the abnormal BCR/ABL translocation similar to CML bone marrow cells. SUBJECTS AND METHODS: Characterized MSCs were isolated from 12 newly diagnosed Philadelphia positive untreated CML patients. RESULTS: MSCs can be readily isolated from CML marrow and exhibit major expansion. Flow cytometry analysis revealed the typical MSC phenotype. Moreover; MSCs do not harbor the BCR/ABL translocation confirmed by karyotype and real time PCR. CONCLUSION: MSCs from CML patients express the typical MSC phenotype; and do not express the BCR/ABL gene. Since; MSCs are able to support engraftment of hematopoietic stem cells in stem cell transplantation(SCT) as well as suppress alloreactive T cells causing graft versus –host disease, this current study provides evidence that in a SCT setting of CML patients, autologous MSCs could be a source of stem cell support in future cell therapy applications.
Collapse
Affiliation(s)
- Taghrid Mohamed Gaafar
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Inas Ismail Raafat
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Azza Ahmed Aly
- Department of Clinical and Chemical Pathology, National Research Center, Egypt
| | | | - Reem Jan Farid
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Neveen Ezzat Saad
- Department of Clinical and Chemical Pathology, National Research Center, Egypt
| | - Rabab El-Hawary
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Naglaa Mostafaa
- Department of Clinical and Chemical Pathology, National Cancer Institute, Cairo, Egypt
| | | |
Collapse
|
12
|
Hamzic E, Whiting K, Gordon Smith E, Pettengell R. Characterization of bone marrow mesenchymal stromal cells in aplastic anaemia. Br J Haematol 2015; 169:804-13. [PMID: 25819548 DOI: 10.1111/bjh.13364] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 01/05/2015] [Indexed: 12/26/2022]
Abstract
In aplastic anaemia (AA), haemopoietic activity is significantly reduced and generally attributed to failure of haemopoietic stem cells (HSC) within the bone marrow (BM). The regulation of haemopoiesis depends on the interaction between HSC and various cells of the BM microenvironment, including mesenchymal stromal cells (MSC). MSC involvement in the functional restriction of HSC in AA is largely unknown and therefore, the physical and functional properties of AA MSC were studied in vitro. MSC were characterized by their phenotype and ability to form adherent stromal layers. The functional properties of AA MSC were assessed through proliferative, clonogenic and cross-over culture assays. Results indicate that although AA MSC presented typical morphology and distinctive mesenchymal markers, stromal formation was reduced, with 50% of BM samples failing to produce adherent layers. Furthermore, their proliferative and clonogenic capacity was markedly decreased (P = 0·03 and P = 0·04 respectively) and the ability to sustain haemopoiesis was significantly reduced, as assessed by total cell proliferation (P = 0·032 and P = 0·019 at Week 5 and 6, respectively) and clonogenic potential of HSC (P = 0·02 at Week 6). It was concluded that the biological characteristics of AA MSC are different from those of control MSC and their in vitro haemopoiesis-supporting ability is significantly reduced.
Collapse
Affiliation(s)
- Edita Hamzic
- Department of Infection and Immunity, St George's University of London, London, UK.,Department of Life Sciences, Kingston University, Kingston upon Thames, UK
| | - Karen Whiting
- Department of Life Sciences, Kingston University, Kingston upon Thames, UK
| | - Edward Gordon Smith
- Department of Infection and Immunity, St George's University of London, London, UK
| | - Ruth Pettengell
- Department of Infection and Immunity, St George's University of London, London, UK
| |
Collapse
|
13
|
Isidori A, Salvestrini V, Ciciarello M, Loscocco F, Visani G, Parisi S, Lecciso M, Ocadlikova D, Rossi L, Gabucci E, Clissa C, Curti A. The role of the immunosuppressive microenvironment in acute myeloid leukemia development and treatment. Expert Rev Hematol 2014; 7:807-18. [PMID: 25227702 DOI: 10.1586/17474086.2014.958464] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Functional interplay between acute myeloid leukemia (AML) cells and the bone marrow microenvironment is a distinctive characteristic of this hematological cancer. Indeed, a large body of evidence suggests that proliferation, survival and drug resistance of AML are sustained and modulated by the bone marrow immunosuppressive microenvironment, where both innate and adaptive immune responses are profoundly deregulated. Furthermore, the presence of a number of different immunosuppressive mechanisms results in massive immune deregulation, which causes the eventual escape from natural immune control. Modulating the immune system, as documented by 40 years of stem cell transplantation, may improve survival of AML patients, as the immune system is clearly able to recognize and attack leukemic cells. The understanding of the factors responsible for the escape from immune destruction in AML, which becomes more prominent with disease progression, is necessary for the development of innovative immunotherapeutic treatment modalities in AML.
Collapse
Affiliation(s)
- Alessandro Isidori
- Haematology and Haematopoietic Stem Cell Transplant Center, AORMN Hospital, Via Lombroso, 1, 61122, Pesaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Han I, Yun M, Kim EO, Kim B, Jung MH, Kim SH. Umbilical cord tissue-derived mesenchymal stem cells induce apoptosis in PC-3 prostate cancer cells through activation of JNK and downregulation of PI3K/AKT signaling. Stem Cell Res Ther 2014; 5:54. [PMID: 24739733 PMCID: PMC4055109 DOI: 10.1186/scrt443] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 03/21/2014] [Indexed: 12/15/2022] Open
Abstract
Introduction Although mesenchymal stem cells (MSCs) have antitumor potential in hepatocellular carcinoma and breast cancer cells, the antitumor mechanism of human umbilical cord mesenchymal stem cells (hUCMSCs) in prostate cancer cells still remains unclear. Thus, in the present study, we elucidated the antitumor activity of hUCMSCs in PC-3 prostate cancer cells in vitro and in vivo. Methods hUCMSCs were isolated from Wharton jelly of umbilical cord and characterized via induction of differentiations, osteogenesis, and adipogenesis. Antitumor effects of UCMSCs on tumor growth were evaluated in a co-culture condition with PC-3 prostate cancer cells. PC-3 cells were subcutaneously (sc) injected into the left flank of nude mice, and UCMSCs were sc injected into the right flank of the same mouse. Results We found that hUCMSCs inhibited the proliferation of PC-3 cells in the co-culture condition. Furthermore, co-culture of hUCMSCs induced the cleavage of caspase 9/3 and PARP, activated c-jun NH2-terminal kinase (JNK), and Bax, and attenuated the phosphorylation of phosphatidylinositol 3-kinase (PI3K)/ AKT, extracellular signal-regulated kinase (ERK), and the expression of survival genes such as Bcl-2, Bcl-xL, Survivin, Mcl-1, and cIAP-1 in PC-3 cells in Western blotting assay. Conversely, we found that treatment of specific JNK inhibitor SP600125 suppressed the cleavages of caspase 9/3 and PARP induced by hUCMSCs in PC-3 cells by Western blotting and immunofluorescence assay. The homing of hUCMSCs to, and TUNEL-positive cells on, the K562 xenograft tumor region were detected in Nu/nu-BALB/c mouse. Conclusions These results suggest that UCMSCs inhibit tumor growth and have the antitumor potential for PC-3 prostate cancer treatment.
Collapse
|
15
|
Estrada-González PK, Gómez-Ceja L, Montesinos JJ, Mayani H, Chávez-González A, Meillón L, Delgado N, Sánchez-Nava E, Flores-Figueroa E. Decreased frequency, but normal functional integrity of mesenchymal stromal cells derived from untreated and Imatinib-treated chronic myeloid leukemia patients. Leuk Res 2014; 38:594-600. [PMID: 24661629 DOI: 10.1016/j.leukres.2014.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 02/07/2023]
Abstract
In vitro, Imatinib inhibits the proliferation and stimulates the osteogenic and adipogenic differentiation of mesenchymal stromal cells (MSC). However, it is unknown whether Imatinib affects the biology of MSC in vivo. We asked whether MSC from long-term Imatinib-treated CML patients were affected by the in vivo treatment. MSC from untreated and Imatinib-treated patients displayed normal functional properties (i.e. proliferation, immunophenotype, differentiation and hematopoietic supportive capacity) - but a decreased frequency. In vitro, Imatinib lost its effect when discontinued; which suggest that it has a reversible effect on MSC. Therefore it might lose its effect on MSC after discontinuation in vivo.
Collapse
Affiliation(s)
- P K Estrada-González
- Niche and Microenvironment Laboratory, Oncology Research Unit, Oncology Hospital, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Coyoacán, México City, Mexico
| | - L Gómez-Ceja
- Niche and Microenvironment Laboratory, Oncology Research Unit, Oncology Hospital, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Coyoacán, México City, Mexico
| | - J J Montesinos
- Mesenchymal Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, Mexico
| | - H Mayani
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, Mexico
| | - A Chávez-González
- Leukemic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, Mexico
| | - L Meillón
- Hematology Service, Bernardo Sepulveda Hospital, National Medical Center, IMSS, México City, Mexico
| | - N Delgado
- Hematology Service, Bernardo Sepulveda Hospital, National Medical Center, IMSS, México City, Mexico
| | - E Sánchez-Nava
- Hematology Service, Bernardo Sepulveda Hospital, National Medical Center, IMSS, México City, Mexico
| | - E Flores-Figueroa
- Niche and Microenvironment Laboratory, Oncology Research Unit, Oncology Hospital, Mexico.
| |
Collapse
|
16
|
Chandia M, Sayagués JM, Gutiérrez ML, Chillón MC, Aristizábal JA, Corrales A, Castellanos M, Melón A, Sánchez ML, Bárcena P, Matarraz S, González-González M, Barrena S, López A, del Cañizo MC, Sánchez-Guijo F, Orfao A. Involvement of primary mesenchymal precursors and hematopoietic bone marrow cells from chronic myeloid leukemia patients by BCR-ABL1 fusion gene. Am J Hematol 2014; 89:288-94. [PMID: 24779036 DOI: 10.1002/ajh.23626] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
For decades now, it is well established that chronic myeloid leukemia (CML) is a hematopoietic stem cell(HPC) disorder. However, it remains to be determined whether BCR-ABL1 gene rearrangement occurs in a HPC or at an earlier stem cell and whether the degree of involvement of hematopoiesis by the BCR-ABL1 fusion gene relates to the response to therapy. Here, we have investigated by interphase fluorescence in situ hybridization (iFISH) the distribution of BCR-ABL1 fusion gene in FACS-sorted bone marrow (BM) populations of mesenchymal precursor cells (MPC) and other hematopoietic cell populations from 18 newly diagnosed CML patients. Overall, our results showed systematic involvement at relatively high percentages of BM maturing neutrophils (97%615%), basophils (95%612%), eosinophils (90%68%), CD341 precursors cells (90%67%),monocytes (84%630%), nucleated red blood cells (87%624%), and mast cells (77%633%). By contrast, MPC(30%634%), B-cells (15%627%), T-lymphocytes (50%626%), and NK-cells (35%634%) were involved at lower percentages. In 8/18 CML patients, 2 tumor BCR-ABL11 subclones were detected by iFISH. Of note, all tumor cell subclones were systematically detected in CD341 cells, whereas MPC were only involved by the ancestral tumor cell subclone. In summary, here we confirm the presence at diagnosis of the BCR-ABL1 fusion gene inMPC, CD341 precursors, and other different BM hematopoietic myeloid cell lineages from CML patients,including also in a significant fraction of cases, a smaller percentage of T, B, and NK lymphocytes.Interestingly, involvement of MPC was restricted to the ancestral BCR-ABL11 subclone.
Collapse
Affiliation(s)
- Mauricio Chandia
- Servicio de Hematología; IBSAL-Hospital Universitario de Salamanca; Paseo de San Vicente, 58-182 37007 Salamanca Spain
- Becario Fundación Carolina-Fundación BBVA; Salamanca Spain
| | | | | | - María-Carmen Chillón
- Servicio de Hematología; IBSAL-Hospital Universitario de Salamanca; Paseo de San Vicente, 58-182 37007 Salamanca Spain
| | - José-Alejandro Aristizábal
- Servicio de Hematología; IBSAL-Hospital Universitario de Salamanca; Paseo de San Vicente, 58-182 37007 Salamanca Spain
| | - Alejandro Corrales
- Servicio de Hematología; Hospital Virgen de la Concha; Zamora Spain
- Grupo de estudio de patología mieloide; Sociedad Castellano-Leonesa de Hematología y Hemoterapia (SCLHH); Salamanca Spain
| | - Marta Castellanos
- Grupo de estudio de patología mieloide; Sociedad Castellano-Leonesa de Hematología y Hemoterapia (SCLHH); Salamanca Spain
- Servicio de Hematología; Hospital Clínico Universitario de Valladolid; Valladolid Spain
| | - Alberto Melón
- Servicio de Hematología; IBSAL-Hospital Universitario de Salamanca; Paseo de San Vicente, 58-182 37007 Salamanca Spain
| | - María-Luz Sánchez
- Servicio General de Citometría; Universidad de Salamanca; Salamanca Spain
| | - Paloma Bárcena
- Servicio General de Citometría; Universidad de Salamanca; Salamanca Spain
| | - Sergio Matarraz
- Servicio General de Citometría; Universidad de Salamanca; Salamanca Spain
| | | | - Susana Barrena
- Servicio General de Citometría; Universidad de Salamanca; Salamanca Spain
| | - Antonio López
- Servicio General de Citometría; Universidad de Salamanca; Salamanca Spain
| | - María-Consuelo del Cañizo
- Servicio de Hematología; IBSAL-Hospital Universitario de Salamanca; Paseo de San Vicente, 58-182 37007 Salamanca Spain
- Grupo de estudio de patología mieloide; Sociedad Castellano-Leonesa de Hematología y Hemoterapia (SCLHH); Salamanca Spain
| | - Fermín Sánchez-Guijo
- Servicio de Hematología; IBSAL-Hospital Universitario de Salamanca; Paseo de San Vicente, 58-182 37007 Salamanca Spain
- Grupo de estudio de patología mieloide; Sociedad Castellano-Leonesa de Hematología y Hemoterapia (SCLHH); Salamanca Spain
| | - Alberto Orfao
- Servicio General de Citometría; Universidad de Salamanca; Salamanca Spain
| |
Collapse
|
17
|
Chronic Myelogenous Leukemia Cells Contribute to the Stromal Myofibroblasts in Leukemic NOD/SCID Mouse In Vivo. JOURNAL OF ONCOLOGY 2012; 2012:901783. [PMID: 22848221 PMCID: PMC3403094 DOI: 10.1155/2012/901783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/13/2012] [Accepted: 06/14/2012] [Indexed: 01/19/2023]
Abstract
We recently reported that chronic myelogenous leukemia (CML) cells converted into myofibroblasts to create a microenvironment for proliferation of CML cells in vitro. To analyze a biological contribution of CML-derived myofibroblasts in vivo, we observed the characters of leukemic nonobese diabetes/severe combined immunodeficiency (NOD/SCID) mouse. Bone marrow nonadherent mononuclear cells as well as human CD45-positive cells obtained from CML patients were injected to the irradiated NOD/SCID mice. When the chimeric BCR-ABL transcript was demonstrated in blood, human CML cells were detected in NOD/SCID murine bone marrow. And CML-derived myofibroblasts composed with the bone marrow-stroma, which produced significant amounts of human vascular endothelial growth factor A. When the parental CML cells were cultured with myofibroblasts separated from CML cell-engrafted NOD/SCID murine bone marrow, CML cells proliferated significantly. These observations indicate that CML cells make an adequate microenvironment for their own proliferation in vivo.
Collapse
|
18
|
Campioni D, Bardi MA, Cavazzini F, Tammiso E, Pezzolo E, Pregnolato E, Volta E, Cuneo A, Lanza F. Cytogenetic and molecular cytogenetic profile of bone marrow-derived mesenchymal stromal cells in chronic and acute lymphoproliferative disorders. Ann Hematol 2012; 91:1563-77. [DOI: 10.1007/s00277-012-1500-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 05/22/2012] [Indexed: 12/31/2022]
|
19
|
McLean K, Gong Y, Choi Y, Deng N, Yang K, Bai S, Cabrera L, Keller E, McCauley L, Cho KR, Buckanovich RJ. Human ovarian carcinoma–associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production. J Clin Invest 2011; 121:3206-19. [PMID: 21737876 DOI: 10.1172/jci45273] [Citation(s) in RCA: 287] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 05/11/2011] [Indexed: 01/01/2023] Open
Abstract
Accumulating evidence suggests that mesenchymal stem cells (MSCs) are recruited to the tumor microenvironment; however, controversy exists regarding their role in solid tumors. In this study, we identified and confirmed the presence of carcinoma-associated MSCs (CA-MSCs) in the majority of human ovarian tumor samples that we analyzed. These CA-MSCs had a normal morphologic appearance, a normal karyotype, and were nontumorigenic. CA-MSCs were multipotent with capacity for differentiating into adipose, cartilage, and bone. When combined with tumor cells in vivo, CA-MSCs promoted tumor growth more effectively than did control MSCs. In vitro and in vivo studies suggested that CA-MSCs promoted tumor growth by increasing the number of cancer stem cells. Although CA-MSCs expressed traditional MSCs markers, they had an expression profile distinct from that of MSCs from healthy individuals, including increased expression of BMP2, BMP4, and BMP6. Importantly, BMP2 treatment in vitro mimicked the effects of CA-MSCs on cancer stem cells, while inhibiting BMP signaling in vitro and in vivo partly abrogated MSC-promoted tumor growth. Taken together, our data suggest that MSCs in the ovarian tumor microenvironment have an expression profile that promotes tumorigenesis and that BMP inhibition may be an effective therapeutic approach for ovarian cancer.
Collapse
Affiliation(s)
- Karen McLean
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Fadini GP, Albiero M, Menegazzo L, Boscaro E, Vigili de Kreutzenberg S, Agostini C, Cabrelle A, Binotto G, Rattazzi M, Bertacco E, Bertorelle R, Biasini L, Mion M, Plebani M, Ceolotto G, Angelini A, Castellani C, Menegolo M, Grego F, Dimmeler S, Seeger F, Zeiher A, Tiengo A, Avogaro A. Widespread increase in myeloid calcifying cells contributes to ectopic vascular calcification in type 2 diabetes. Circ Res 2011; 108:1112-21. [PMID: 21393578 DOI: 10.1161/circresaha.110.234088] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
RATIONALE Acquisition of a procalcific phenotype by resident or circulating cells is important for calcification of atherosclerotic plaques, which is common in diabetes. OBJECTIVE We aim to identify and characterize circulating calcifying cells, and to delineate a pathophysiological role for these cells in type 2 diabetes. METHODS AND RESULTS We demonstrate for the first time that a distinct subpopulation of circulating cells expressing osteocalcin and bone alkaline phosphatase (OC(+)BAP(+)) has procalcific activity in vitro and in vivo. The study of naïve patients with chronic myeloid leukemia indicated that OC(+)BAP(+) cells have a myeloid origin. Myeloid calcifying OC(+)BAP(+) cells (MCCs) could be differentiated from peripheral blood mononuclear cells, and generation of MCCs was closely associated with expression of the osteogenic transcription factor Runx2. In gender-mismatched bone marrow-transplanted humans, circulating MCCs had a much longer half-life compared with OC(-)BAP(-) cells, suggesting they belong to a stable cell repertoire. The percentage of MCCs was higher in peripheral blood and bone marrow of type 2 diabetic patients compared with controls but was lowered toward normal levels by optimization of glycemic control. Furthermore, diabetic carotid endoarterectomy specimens showed higher degree of calcification and amounts of cells expressing OC and BAP in the α-smooth muscle actin-negative areas surrounding calcified nodules, where CD68(+) macrophages colocalize. High glucose increased calcification by MCCs in vitro, and hypoxia may regulate MCC generation in vitro and in vivo. CONCLUSIONS These data identify a novel type of blood-derived procalcific cells potentially involved in atherosclerotic calcification of diabetic patients.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Clinical and Experimental Medicine, Metabolic Division, University of Padova, Medical School, Padova, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ardianto B, Sugimoto T, Kawano S, Kasagi S, Jauharoh SNA, Kurimoto C, Tatsumi E, Morikawa K, Kumagai S, Hayashi Y. The HPB-AML-I cell line possesses the properties of mesenchymal stem cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:163. [PMID: 21144016 PMCID: PMC3016278 DOI: 10.1186/1756-9966-29-163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Accepted: 12/13/2010] [Indexed: 12/19/2022]
Abstract
Background In spite of its establishment from the peripheral blood of a case with acute myeloid leukemia (AML)-M1, HPB-AML-I shows plastic adherence with spindle-like morphology. In addition, lipid droplets can be induced in HPB-AML-I cells by methylisobutylxanthine, hydrocortisone, and indomethacin. These findings suggest that HPB-AML-I is similar to mesenchymal stem cells (MSCs) or mesenchymal stromal cells rather than to hematopoietic cells. Methods To examine this possibility, we characterized HPB-AML-I by performing cytochemical, cytogenetic, and phenotypic analyses, induction of differentiation toward mesenchymal lineage cells, and mixed lymphocyte culture analysis. Results HPB-AML-I proved to be negative for myeloperoxidase, while surface antigen analysis disclosed that it was positive for MSC-related antigens, such as CD29, CD44, CD55, CD59, and CD73, but not for CD14, CD19, CD34, CD45, CD90, CD105, CD117, and HLA-DR. Karyotypic analysis showed the presence of complicated abnormalities, but no reciprocal translocations typically detected in AML cases. Following the induction of differentiation toward adipocytes, chondrocytes, and osteocytes, HPB-AML-I cells showed, in conjunction with extracellular matrix formation, lipid accumulation, proteoglycan synthesis, and alkaline phosphatase expression. Mixed lymphocyte culture demonstrated that CD3+ T-cell proliferation was suppressed in the presence of HPB-AML-I cells. Conclusions We conclude that HPB-AML-I cells appear to be unique neoplastic cells, which may be derived from MSCs, but are not hematopoietic progenitor cells.
Collapse
Affiliation(s)
- Bambang Ardianto
- Division of Molecular Medicine and Medical Genetics, Department of Pathology, Graduate School of Medicine, Kobe University, Kobe, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kurt Yüksel M, Topçuoğlu P, Kurdal M, Ilhan O. The clonogenic potential of hematopoietic stem cells and mesenchymal stromal cells in various hematologic diseases: a pilot study. Cytotherapy 2010; 12:38-44. [PMID: 19878078 DOI: 10.3109/14653240903313958] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSC) are the most popular cells used in regenerative medicine and biotechnology. The clonogenic potential of these cells is defined by colony-forming unit-fibroblasts (CFU-F). It is well known that there is an interaction between hematopoietic cells and stromal cells in disease formation pathogenesis. Therefore we hypothesized that there should be a quantitative and qualitative relationship between MSC colonies (CFU-F) and hematopoietic stem cell colonies (colony-forming unit-granulocyte-macrophages; CFU-GM) among patients with and without hematologic diseases. METHODS Forty-two patients were included in this study. Patients were divided into three groups: group A, patients with hematologic malignancies (n =20); group B, patients with bone marrow (BM) failure (n =11); group C, patients without hematologic diseases (n =11). BM aspirates were plated in different densities for CFU-F culture. The plating density was the same for CFU-GM culture. RESULTS CFU-GM colonies grew in 90% of group A cells and all of group B and C cells (P= 0.0001). CFU-F colonies became visible on the ninth day of plating in group A and on the eight day in groups B and C. There was no statistically significant difference between the groups for the duration of CFU-F colony formation (P= 0.12). There were differences in the morphology of the colonies among the groups. CONCLUSIONS This is the first study that has compared the clonogenic potential of stromal cells and hematopoietic stem cells in the same subjects with and without hematologic diseases. No correlation was shown between the clonogenic potential of stromal cells and hematopoietic cells.
Collapse
Affiliation(s)
- Meltem Kurt Yüksel
- Department of Hematology, Ankara Oncology Research and Education Hospital, Ankara University School of Medicine, Ankara, Turkey.
| | | | | | | |
Collapse
|
23
|
Multilineage potential of adult human mesenchymal stromal cells derived from bone marrow of patients with polycytaemia vera. Biologia (Bratisl) 2010. [DOI: 10.2478/s11756-010-0012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
24
|
Schulenburg A, Brämswig K, Herrmann H, Karlic H, Mirkina I, Hubmann R, Laffer S, Marian B, Shehata M, Krepler C, Pehamberger H, Grunt T, Jäger U, Zielinski CC, Valent P. Neoplastic stem cells: current concepts and clinical perspectives. Crit Rev Oncol Hematol 2010; 76:79-98. [PMID: 20185329 DOI: 10.1016/j.critrevonc.2010.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 12/29/2009] [Accepted: 01/06/2010] [Indexed: 12/20/2022] Open
Abstract
Neoplastic stem cells have initially been characterized in myeloid leukemias where NOD/SCID mouse-repopulating progenitors supposedly reside within a CD34+/Lin- subset of the malignant clone. These progenitors are considered to be self-renewing cells responsible for the in vivo long-term growth of neoplastic cells in leukemic patients. Therefore, these cells represent an attractive target of therapy. In some lymphoid leukemias, NOD/SCID mouse-repopulating cells were also reported to reside within the CD34+/Lin- subfraction of the clone. More recently, several attempts have been made to transfer the cancer stem cell concept to solid tumors and other non-hematopoietic neoplasms. In several of these tumors, the cell surface antigens AC133 (CD133) and CD44 are considered to indicate the potential of a cell to initiate permanent tumor formation in vivo. However, several questions concerning the phenotype, self-renewal capacity, stroma-dependence, and other properties of cancer- or leukemia-initiating cells remain to be solved. The current article provides a summary of our current knowledge on neoplastic (cancer) stem cells, with special emphasis on clinical implications and therapeutic options as well as a discussion about conceptual and technical limitations.
Collapse
Affiliation(s)
- Axel Schulenburg
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bacher U, Asenova S, Badbaran A, Zander AR, Alchalby H, Fehse B, Kröger N, Lange C, Ayuk F. Bone marrow mesenchymal stromal cells remain of recipient origin after allogeneic SCT and do not harbor the JAK2V617F mutation in patients with myelofibrosis. Clin Exp Med 2009; 10:205-8. [PMID: 19629639 DOI: 10.1007/s10238-009-0058-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Accepted: 06/29/2009] [Indexed: 10/20/2022]
Abstract
The close association of the myeloproliferative neoplasms with the activating non-receptor tyrosine kinase JAK2V617F mutation is well established. To further clarify the pathomechanisms of this mutation in patients with myelofibrosis, we performed screening with quantitative real-time PCR for the respective mutation in in vitro expanded bone marrow (BM) mesenchymal stromal cells (MSCs) and compared the results with BM/peripheral blood (PB). Eight patients with primary/secondary myelofibrosis were investigated before (n = 4) or after allogeneic stem cell transplantation (n = 4). All patients had systemic evidence of the JAK2V617F mutation in BM/PB (mutation ratios 0.2-23.5) at the time of investigation in contrast to negative results in the MSCs (n = 7) or a very low (0.004) mutation ratio (n = 1) which was probably due to hematopoietic contamination. The four patients post-transplant had systemic donor chimerism between 96.5 and 100% in BM/PB, while MSCs showed no evidence of donor-specific alleles. In conclusion, in myelofibrosis, the JAK2V617F mutation is restricted to hematopoietic cells, and cannot explain the stromal alterations being observed in this disorder. Further, the MSCs remain of recipient origin after allogeneic SCT, which might contribute to the increased risk of graft dysfunction or failure in myelofibrosis patients after allogeneic transplantation.
Collapse
Affiliation(s)
- Ulrike Bacher
- Interdisciplinary Clinic for Stem Cell Transplantation, University Cancer Center Hamburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Valent P, Deininger M. Clinical perspectives of concepts on neoplastic stem cells and stem cell-resistance in chronic myeloid leukemia. Leuk Lymphoma 2009; 49:604-9. [DOI: 10.1080/10428190801923212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
27
|
Mayani H, Flores-Figueroa E, Chávez-González A. In vitro biology of human myeloid leukemia. Leuk Res 2009; 33:624-37. [DOI: 10.1016/j.leukres.2008.11.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 11/04/2008] [Accepted: 11/08/2008] [Indexed: 11/27/2022]
|
28
|
Flores-Figueroa E, Montesinos JJ, Flores-Guzmán P, Gutiérrez-Espíndola G, Arana-Trejo RM, Castillo-Medina S, Pérez-Cabrera A, Hernández-Estévez E, Arriaga L, Mayani H. Functional analysis of myelodysplastic syndromes-derived mesenchymal stem cells. Leuk Res 2008; 32:1407-16. [DOI: 10.1016/j.leukres.2008.02.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 02/06/2008] [Accepted: 02/07/2008] [Indexed: 12/20/2022]
|