1
|
Moore-Lotridge SN, Hajdu KS, Hou BQ, Gibson BHY, Schoenecker JG. Maintaining the balance: the critical role of plasmin activity in orthopedic surgery injury response. J Thromb Haemost 2023; 21:2653-2665. [PMID: 37558131 PMCID: PMC10926148 DOI: 10.1016/j.jtha.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
The musculoskeletal system plays vital roles in the body, facilitating movement, protecting vital structures, and regulating hematopoiesis and mineral metabolism. Injuries to this system are common and can cause chronic pain, loss of range of motion, and disability. The acute phase response (APR) is a complex process necessary for surviving and repairing injured musculoskeletal tissue. To conceptualize the APR, it is useful to divide it into 2 distinct phases, survival and repair. During the survival-APR, a "damage matrix" primarily composed of fibrin, via thrombin activity, is produced to contain the zone of injury. Once containment is achieved, the APR transitions to the repair phase, where reparative inflammatory cells use plasmin to systematically remove the damage matrix and replace it with new permanent matrices produced by differentiated mesenchymal stem cells. The timing of thrombin and plasmin activation during their respective APR phases is crucial for appropriate regulation of the damage matrix. This review focuses on evidence indicating that inappropriate exuberant activation of plasmin during the survival-APR can result in an overactive APR, leading to an "immunocoagulopathy" that may cause "immunothrombosis" and death. Conversely, preclinical data suggest that too little plasmin activity during the repair-APR may contribute to failed tissue repair, such as a fracture nonunion, and chronic inflammatory degenerative diseases like osteoporosis. Future clinical studies are required to affirm these findings. Therefore, the temporal-spatial functions of plasmin in response to musculoskeletal injury and its pharmacologic manipulation are intriguing new targets for improving orthopedic care.
Collapse
Affiliation(s)
- Stephanie N Moore-Lotridge
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Katherine S Hajdu
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Brian Q Hou
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Breanne H Y Gibson
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan G Schoenecker
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
2
|
Zhen C, Fang X, Ding M, Wang X, Yuan D, Sui X, Liu X, Zhang L, Xu H, Li Y, Wang X. Smoking is an important factor that affects peripheral blood progenitor cells yield in healthy male donors. J Clin Apher 2019; 35:33-40. [PMID: 31693224 DOI: 10.1002/jca.21756] [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: 05/22/2019] [Revised: 08/19/2019] [Accepted: 09/26/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND Smoking could reduce the CD34+ cells in peripheral blood of healthy individual. This study aimed to investigate the correlation between smoking load and the effect of peripheral blood hematopoietic progenitor cells (PBPCs) mobilization by granulocyte colony-stimulating factor (G-CSF) alone in healthy donors. METHODS Retrospective analysis was performed on 145 healthy adult PBPCs donors who underwent PBPCs mobilization and collection. Smoking factors were evaluated and correlated with mobilization responses, as indicated by the collected CD34+ cells concentration. RESULTS The collected CD34+ cells concentration was closely related to pre-CD34 (P < .001) and CD34+ cells collected per volume blood processed (P < .001) which suggested that collected CD34+ cells concentration was a reliable indicator of PBPCs mobilization efficiency. The heavy smoking donors revealed significantly lower collected CD34+ cells concentration, compared to that of the nonsmoking (P < .001) and light smoking donors (P < .05). The levels of collected CD34+ cells in light smoking were also obviously lower than that in nonsmoking donors (P < .05).There were no obvious differences in the collected CD34+ cells concentration, overall processed blood volume and total collected CD34+ cells between nonsmoking and smoking cessation groups (P = .490; P = .464; P = .819). CONCLUSION Cigarette smoking is an important factor that affects the yield of PBPCs in male donors, especially when the smoking load is more than five pack-years. Mobilization of PBMCs could be restored by smoking cessation in chronic smokers.
Collapse
Affiliation(s)
- Changqing Zhen
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Mei Ding
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xianghua Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Dai Yuan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xiaohui Sui
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xin Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Lingyan Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Hongzhi Xu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Ying Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.,Department of Diagnostics, Shandong University School of Medicine, Jinan, Shandong, China
| |
Collapse
|
3
|
Hardy CL. Specificity of Hematopoietic Stem and Progenitor Cell Homing to Bone Marrow: A Perspective. Hematology 2016; 5:391-401. [DOI: 10.1080/10245332.2000.11746535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Cheryl L. Hardy
- G.V. (Sonny) Montgomery Department of Veterans Affairs Medical Center, Department of Medicine, University of Mississippi School of Medicine, Jackson, MS
| |
Collapse
|
4
|
Cao X, Kwek K, Chan JKY, Chan CKH, Lim M. Electrospun nanofibers as a bioadhesive platform for capturing adherent leukemia cells. J Biomed Mater Res A 2013; 102:523-31. [DOI: 10.1002/jbm.a.34716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/26/2013] [Accepted: 03/04/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Xue Cao
- Division of Bioengineering; School of Chemical and Biomedical Engineering, Nanyang Technological University; Singapore
| | - Kenneth Kwek
- Department of Maternal Fetal Medicine; KK Women's and Children's Hospital; Singapore
| | - Jerry K. Y. Chan
- Department of Reproductive Medicine; KK Women's and Children's Hospital; Singapore
- Cancer and Stem Cell Biology Program; Duke-NUS Graduate Medical School; Singapore
- Experimental Fetal Medicine Group; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
| | - Casey K. H. Chan
- Department of Bioengineering; Faculty of Engineering; National University of Singapore; Singapore
- Department of Orthopaedic Surgery; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
| | - Mayasari Lim
- Division of Bioengineering; School of Chemical and Biomedical Engineering, Nanyang Technological University; Singapore
| |
Collapse
|
5
|
Pizzatti L, Panis C, Lemos G, Rocha M, Cecchini R, Souza GHMF, Abdelhay E. Label-free MSE
proteomic analysis of chronic myeloid leukemia bone marrow plasma: disclosing new insights from therapy resistance. Proteomics 2012; 12:2618-31. [DOI: 10.1002/pmic.201200066] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 05/22/2012] [Accepted: 06/11/2012] [Indexed: 01/11/2023]
Affiliation(s)
- Luciana Pizzatti
- Divisão de Laboratórios do CEMO; Instituto Nacional do Câncer; Rio de Janeiro Brazil
- Rede Proteômica do Rio de Janeiro; RJ Brazil
| | - Carolina Panis
- Divisão de Laboratórios do CEMO; Instituto Nacional do Câncer; Rio de Janeiro Brazil
- Laboratório de Fisiopatologia de Radicais Livres; UEL; Londrina PR Brazil
| | - Gabriela Lemos
- Divisão de Laboratórios do CEMO; Instituto Nacional do Câncer; Rio de Janeiro Brazil
| | - Moisés Rocha
- Divisão de Laboratórios do CEMO; Instituto Nacional do Câncer; Rio de Janeiro Brazil
| | - Rubens Cecchini
- Laboratório de Fisiopatologia de Radicais Livres; UEL; Londrina PR Brazil
| | | | - Eliana Abdelhay
- Divisão de Laboratórios do CEMO; Instituto Nacional do Câncer; Rio de Janeiro Brazil
- Rede Proteômica do Rio de Janeiro; RJ Brazil
| |
Collapse
|
6
|
Effect of Subcultivation of Human Bone Marrow Mesenchymal Stem on their Capacities for Chondrogenesis, Supporting Hematopoiesis, and Telomea Length. Cytotechnology 2011; 47:19-27. [PMID: 19003041 DOI: 10.1007/s10616-005-3752-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2004] [Accepted: 03/16/2005] [Indexed: 01/14/2023] Open
Abstract
Effects of subcultivation of human bone marrow mesenchymal stem cells on their capacities for chondrogenesis and supporting hematopoiesis, and telomea length were investigated. Mesenchymal stem cells were isolated from human bone marrow aspirates and subcultivated several times at 37 degrees C under a 5% CO(2) atmosphere employing DMEM medium containing 10% FCS up to the 20th population doubling level (PDL). The ratio of CD45(-) CD105(+) cells among these cells slightly increased as PDL increased. However, there was no marked change in the chondrogenic capacity of these cells, which was confirmed by expression assay of aggrecan mRNA and Safranin O staining after pellet cell cultivation. The change in capacity to support hematopoiesis of cord blood cells was not observed among cells with various PDLs. On the other hand, telomere length markedly decreased as PDL increased at a higher rate than that at which telomere length of primary mesenchymal stem cells decreased as the age of donor increased.
Collapse
|
7
|
Haas R, Bruns I, Kobbe G, Fenk R. High-dose therapy and autologous peripheral blood stem cell transplantation in patients with multiple myeloma. Recent Results Cancer Res 2011; 183:207-238. [PMID: 21509687 DOI: 10.1007/978-3-540-85772-3_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Since its introduction in 1983, high-dose therapy followed by autologous peripheral blood stem cell transplantation is a pillar of the treatment of patients with multiple myeloma. In the last decades, a multitude of clinical trials helped to improve strategies based on high-dose therapy and autologous stem cell transplantation resulting in a continuously prolongation of overall survival of patients. In this chapter we will review the progress, which has been made in order to enhance the mobilisation of autologous stem cells and increase the effectiveness of this treatment.
Collapse
Affiliation(s)
- Rainer Haas
- Department of Hematology, Heinrich-Heine-University, Düsseldorf, Germany.
| | | | | | | |
Collapse
|
8
|
Zhang C, Zhang X, Chen XH. Granulocyte-colony stimulating factor-mobilized mesenchymal stem cells: A new resource for rapid engraftment in hematopoietic stem cell transplantation. Med Hypotheses 2010; 76:241-3. [PMID: 21050671 DOI: 10.1016/j.mehy.2010.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 10/07/2010] [Accepted: 10/09/2010] [Indexed: 02/02/2023]
Abstract
The bone marrow (BM) microenvironment plays an important role in regulating hematopoietic stem cell self-renewal and differentiation. Mesenchymal stem cells (MSCs), which constitute approximately 0.01-0.0001% of the nucleated cells in the adult human BM, are an important component of the BM stroma that supports hematopoiesis. The BM stroma system is often damaged in patients who have undergone high-dose chemotherapy and/or radiation treatment. Thus, the BM stroma should be reconstructed during hematopoietic stem cell transplantation (HSCT). Granulocyte-colony stimulating factor (G-CSF) is a potent hematopoietic cytokine that regulates neutrophil generation within the BM by modulating the mobility, proliferation and maturation of neutrophil progenitor cells. The results from our study here show that G-CSF markedly increased the number of donor-derived MSCs in the BM and the peripheral blood. Engraftment was faster in HSCTs with bone marrow that was treated with G-CSF (G-BM) or with G-BM- and G-CSF-treated peripheral blood stem cells compared to stead-state bone marrow (SS-BM). Based on these findings, we hypothesize that G-CSF-mobilized treatment of MSCs may accelerate engraftment in HSCT.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, People's Republic of China
| | | | | |
Collapse
|
9
|
Nair RR, Tolentino J, Hazlehurst LA. The bone marrow microenvironment as a sanctuary for minimal residual disease in CML. Biochem Pharmacol 2010; 80:602-12. [PMID: 20382130 DOI: 10.1016/j.bcp.2010.04.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 03/29/2010] [Accepted: 04/01/2010] [Indexed: 12/15/2022]
Abstract
Bcr-abl kinase inhibitors have provided proof of principal that targeted therapy holds great promise for the treatment of cancer. However, despite the success of these agents in treating chronic myelogenous leukemia (CML), the majority of patients continue to present with minimal residual disease contained within the bone marrow microenvironment. These clinical observations suggest that the bone marrow microenvironment may provide survival signals that contribute to the failure to eliminate minimal residual disease. The bone marrow microenvironment is comprised of multiple sub-domains which vary in cellular composition and gradients of soluble factors and matrix composition. Experimental evidence indicate that exposure of tumor cells to either bone marrow derived soluble factors or the extracellular matrix can confer a multi-drug resistance phenotype. Together, these data indicate that targeting such pathways may be a viable approach for increasing the efficacy of chemotherapy. Moreover, we propose that personalized medicine must go beyond understanding predictive models inherent to tumors but rather build predictive models that consider diversity in response due to interactions with the tumor microenvironment. Although review will focus on CML, understanding the contribution of the bone marrow microenvironment could contribute to rationale combination therapy in other types of leukemia, multiple myeloma and solid tumors which metastasize to the bone.
Collapse
Affiliation(s)
- Rajesh R Nair
- Molecular Oncology Program, H Lee Moffitt Cancer Center, Tampa, FL 33612, United States
| | | | | |
Collapse
|
10
|
Neumann F, Zohren F, Haas R. The role of natalizumab in hematopoietic stem cell mobilization. Expert Opin Biol Ther 2009; 9:1099-106. [PMID: 19566481 DOI: 10.1517/14712590903055011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The humanized monoclonal very late antigen 4 (VLA-4) antibody natalizumab is FDA approved for the treatment of patients with multiple sclerosis and Crohn's disease. In this review we focus on its role in the context of hematopoietic stem cell transplantation and stem cell diseases. The use of natalizumab alone or in combination with either cytotoxic drugs or other antibodies might be a new modality for stem cell mobilization and a therapeutic option for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Frank Neumann
- Heinrich-Heine-University, Department of Hematology, Oncology and Clinical Immunology, Duesseldorf, Germany.
| | | | | |
Collapse
|
11
|
Higuchi A, Yang ST, Li PT, Chang Y, Tsai EM, Chen YH, Chen YJ, Wang HC, Hsu ST. Polymeric Materials for Ex vivo Expansion of Hematopoietic Progenitor and Stem Cells. POLYM REV 2009. [DOI: 10.1080/15583720903048185] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
12
|
Semina OV, Semenets TN, Zamulaeva IA, Selivanova EI, Il'ina TP, Malyutina YV, Semin DY, Deigin VI, Saenko AS. Thymodepressin inhibits migration of CD34+ cells from bone marrow in normal and granulocyte CSF-stimulated hemopoiesis. Bull Exp Biol Med 2008; 144:831-4. [PMID: 18856212 DOI: 10.1007/s10517-007-0442-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
We studied the effect of thymodepressin on migration and adhesion of mouse hemopoietic CD34+ cells under normal conditions and under the effect of granulocytic CSF. It was found that the peptide reduced the absolute number of CD34+ hemopoietic cells in the peripheral blood, increased the percent of cells bound to fibronectin and expressing receptor for integrin beta1 (CD29+) in the bone marrow of mice under normal conditions and after stimulation with granulocytic CSF, and reduced the relative number of cells carrying CXCR4 receptor for stromal factor-1 (CD184+) in the bone marrow (CD34+CD184+) and blood (CD184+) of mice stimulated with granulocytic CSF. The results suggest that thymodepressin can inhibit migration of CD34+ cells from bone marrow into peripheral blood under conditions of normal and granulocytic CSF-stimulated hemopoiesis.
Collapse
Affiliation(s)
- O V Semina
- Medical Radiological Research Center, Russian Academy of Medical Sciences, Obninsk.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Peripheral blood-derived multipotent mesenchymal stromal cells circulate in low number. They share, most although not all, of the surface markers with bone marrow-derived multipotent mesenchymal stromal cells, possess diverse and complicated gene expression characteristics, and are capable of differentiating along and even beyond mesenchymal lineages. Although their origin and physio-pathological function are still unclear, their presence in the adult peripheral blood might relate to some interesting but controversial subjects in the field of adult stem cell biology, such as systemic migration of bone marrow-derived multipotent mesenchymal stromal cells and the existence of common hematopoietic-mesenchymal precursors. In this review, current studies/knowledge about peripheral blood-derived multipotent mesenchymal stromal cells is summarized, and the above-mentioned topics are discussed.
Collapse
Affiliation(s)
- Qiling He
- Centre for Cancer Research and Cell Biology, Musculoskeletal Education and Research Unit, School of Biomedical Sciences, Queen's University of Belfast, Musgrave Park Hospital, Belfast, United Kingdom
| | | | | |
Collapse
|
14
|
Jiang XS, Chai C, Zhang Y, Zhuo RX, Mao HQ, Leong KW. Surface-immobilization of adhesion peptides on substrate for ex vivo expansion of cryopreserved umbilical cord blood CD34+ cells. Biomaterials 2006; 27:2723-32. [PMID: 16376984 PMCID: PMC2376800 DOI: 10.1016/j.biomaterials.2005.12.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Accepted: 12/05/2005] [Indexed: 01/16/2023]
Abstract
The interaction between integrins and extracellular matrix proteins play an important role in the regulation of hematopoiesis. Human hematopoietic progenitor cells express very late antigen-4 (VLA-4) and VLA-5, which mediate their interaction with fibronectin by recognizing the connecting segment-1 (CS-1 and RGD motifs, respectively. In this study, we investigated the ex vivo expansion of human umbilical cord blood (UCB) CD34+ cells on synthetic substrates surface-immobilized with peptides containing the CS-1 binding motif (EILDVPST) and the RGD motif (GRGDSPC). These peptides were covalently conjugated to poly(ethylene terephthalate) (PET) film at a surface density of 2.0-2.3 nmol/cm2. UCB CD34+ cells were cultured for 10 days in serum-free medium supplemented with recombinant human thrombopoietin, stem cell factor, flt3-ligand and interleukin 3. The highest cell expansion fold was observed on the CS-1 peptide-modified surface, where total nucleated cells, total colony forming unit, and long-term culture initiating cells were expanded by 589.6+/-58.6 (mean+/-s.d.), 76.5+/-8.8, and 3.2+/-0.9-fold, respectively, compared to unexpanded cells. All substrates surface-immobilized with peptides, including the control peptides, were more efficient in supporting the expansion of CD34+, CFU-GEMM and LTC-ICs than tissue culture polystyrene surface. Nevertheless, after 10-days of ex vivo expansion from 600 CD34+ cells, only cells cultured on CS-1-immobilized surface yielded positive engraftment, even though the frequency was low. PET surface immobilized with RGD peptide was less efficient than that with CS-1 peptide. Our results suggest that covalently immobilized adhesion peptides can significantly influence the proliferation characteristics of cultured UCB CD34+ cells.
Collapse
Affiliation(s)
- Xue-Song Jiang
- Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Tissue & Therapeutic Engineering Lab, Division of Johns Hopkins in Singapore Ltd, Singapore, 138669
| | - Chou Chai
- Tissue & Therapeutic Engineering Lab, Division of Johns Hopkins in Singapore Ltd, Singapore, 138669
| | - Yue Zhang
- Division of Bioengineering, National University of Singapore, Singapore
| | - Ren-Xi Zhuo
- Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Hai-Quan Mao
- Tissue & Therapeutic Engineering Lab, Division of Johns Hopkins in Singapore Ltd, Singapore, 138669
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kam W. Leong
- Tissue & Therapeutic Engineering Lab, Division of Johns Hopkins in Singapore Ltd, Singapore, 138669
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Corresponding author. Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. E-mail address: (K.W. Leong)
| |
Collapse
|
15
|
Tomte LT, Annatshah Y, Schlüter NK, Miosge N, Herken R, Quondamatteo F. Hematopoietic Cells Are a Source of Nidogen-1 and Nidogen-2 during Mouse Liver Development. J Histochem Cytochem 2006; 54:593-604. [PMID: 16618944 DOI: 10.1369/jhc.5a6810.2006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nidogen-1 and −2 are key components of basement membranes (BMs). Despite the presence of nidogen molecules in the parenchyma of the developing liver, no BMs are formed therein. This suggests that, in the liver, nidogens may also have functions other than BM formation. As a first step toward the elucidation of the possible cell biological functions of nidogens in the developing liver, we aimed to study their cellular origin. We localized expression of nidogen-1 and nidogen-2 on prenatal days 12, 14, and 16 in the developing mouse liver using in situ hybridization at the light and electron microscopic level and light microscopic immunohistochemistry. Our results show that nidogens are produced both in portal anlagen and in the parenchyma during liver development. In the parenchyma, transcripts can be found in hepatocytes, precursors of stellate cells, endothelial cells and, most interestingly, hematopoietic cells. Using real-time PCR, we found that the gene expression for both proteins shows a decrease from day 14 to day 16 concomitant with a decrease in the hepatic hematopoiesis. We suggest that nidogens may, to some extent, take part in the regulation of hepatic hematopoiesis. (J Histochem Cytochem 54:593-604, 2006)
Collapse
Affiliation(s)
- Laurice T Tomte
- Department of Histology, University of Goettingen, Goettingen, Germany.
| | | | | | | | | | | |
Collapse
|
16
|
Nepomnyashchikh LM, Lushnikova EL, Goldshtein DV. Whether modern cell technologies can break down biological limitations of tissue-specific regeneration of the myocardium. Bull Exp Biol Med 2005; 139:481-90. [PMID: 16027887 DOI: 10.1007/s10517-005-0328-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The paper reviews modern concepts of physiological and reparative regeneration of the myocardium as a highly specific and highly differentiated tissue system. Special attention was given to evaluation of the proliferative potential of cardiomyocytes, in particular, to the existence of a population of resident cardiac stem cells in the myocardium. Modern approaches to replenishment of massive cardiomyocyte loss via transplantation and transdifferentiation of adult and embryonic stem cells are discussed and the possibilities of using cell technologies for induction of tissue-specific regeneration of the myocardium are analyzed.
Collapse
Affiliation(s)
- L M Nepomnyashchikh
- Research Institute of Regional Pathology and Pathomorphology, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk, Russia.
| | | | | |
Collapse
|
17
|
Mändle T, Einsele H, Schaller M, Neumann D, Vogel W, Autenrieth IB, Kempf VAJ. Infection of human CD34+ progenitor cells with Bartonella henselae results in intraerythrocytic presence of B. henselae. Blood 2005; 106:1215-22. [PMID: 15860668 DOI: 10.1182/blood-2004-12-4670] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although there is evidence that endothelial cells are important targets for human pathogenic Bartonella species, the primary niche of infection is unknown. Here we elucidated whether human CD34+ hematopoietic progenitor cells (HPCs) internalize B. henselae and may serve as a potential niche of the pathogen. We showed that B. henselae does not adhere to or invade human erythrocytes. In contrast, B. henselae invades and persists in HPCs as shown by gentamicin protection assays, confocal laser scanning microscopy (CLSM), and electron microscopy (EM). Fluorescence-activated cell sorting (FACS) analysis of glycophorin A expression revealed that erythroid differentiation of HPCs was unaffected following infection with B. henselae. The number of intracellular B. henselae continuously increased over a 13-day period. When HPCs were infected with B. henselae immediately after isolation, intracellular bacteria were subsequently detectable in differentiated erythroid cells on day 9 and day 13 after infection, as shown by CLSM, EM, and FACS analysis. Our data provide, for the first time, evidence that a bacterial pathogen is able to infect and persist in differentiating HPCs, and suggest that HPCs might serve as a potential primary niche in Bartonella infections.
Collapse
Affiliation(s)
- Tanja Mändle
- Institut für Medizinische Mikrobiologie und Hygiene, Elfriede-Aulhorn-Str 6, D-72076, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
18
|
Han P, Guo X, Story C. Role of beta(1)-integrins and their associated tetraspanin molecules in fibronectin-enhanced megakaryopoiesis. Cytotherapy 2005; 6:465-75. [PMID: 15512913 DOI: 10.1080/14653240410004998] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND We have shown previously that fibronectin (FN) together with megakaryocyte (Mk) growth and development factor (MGDF) enhanced generation of Mk progenitors determined by colony-forming unit (CFU)-Mk assay. MGDF can activate beta(1)-integrins on progenitor cells and increase binding to FN. We studied the role of beta(1)-integrin-tetraspanin complexes by which FN may enhance megakaryopoiesis. METHODS Cord blood CD34(+) cells were cultured for up to 8 days in serum-free medium containing IL-3, IL-6 and SCF with or without MGDF in the presence or absence of FN. Immunofluorescence flow cytometry was used to monitor changes in beta(1)-integrin-tetraspanin complexes. CFU-Mk assay was used to assess Mk commitment. RESULTS The cocktail of cytokines irrespective of the presence of MGDF altered the percentage expression of beta(1)-integrins CD49d and CD49e on CD34(+) cells. CD49d expression fell initially (98% to 15%) and then rose to 75%, whereas CD49e progressively increased over the 8 days of culture, from 5.4% to 22%. However, with the addition of FN, similar changes in the expression of beta(1)-integrins were observed but the expression was maintained at a higher level. MGDF and FN increased expression of tetraspanin molecules, CD63 and CD151, as well as their co-expression with the beta(1)-integrins on both the CD34(+) and CD34(-) cells (e.g. and increase from 0% to 80% co-expression of CD49d and CD151 on CD34(+)). Blocking of beta(1)-integrins or the tetraspanin CD151 with the respective MAb reduced Mk progenitor generation in a stromal cell model. DISCUSSION FN enhanced Mk progenitor generation through modulation of beta(1)-integrin-tetraspanin complexes, such as CD151/CD49d.
Collapse
Affiliation(s)
- P Han
- Department of Haematology, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | | | | |
Collapse
|
19
|
Takagi M. Cell processing engineering for ex-vivo expansion of hematopoietic cells. J Biosci Bioeng 2005; 99:189-96. [PMID: 16233777 DOI: 10.1263/jbb.99.189] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2004] [Accepted: 12/14/2004] [Indexed: 12/14/2022]
Abstract
The cell processing engineering for ex vivo expansion of hematopoietic cells is reviewed. All hematopoietic cells of different lineages and/or at various stages of differentiation are derived from the same precursor, pluripotent hematopoietic stem cells. Bone marrow stromal cells promote and regulate the self-renewal, commitment, differentiation, and proliferation of stem cells and progenitors through their secreted extracellular matrices and cytokine environment in the hematopoietic microenvironment. Although stroma-mediated hematopoiesis has been studied in vitro using the Dexter culture system in tissue culture flasks, hematopoiesis in the Dexter culture system is almost limited to a granulocyte lineage and the system could not expand primitive cells. The addition of large amounts of cytokines to the culture of hematopoietic cells enabled their expansion, but is too expensive. Some clonal stromal cell lines have been established from the Dexter culture of murine bone marrow cells in order to simplify and stimulate the ex vivo expansion of hematopoietic cells. In order to solve the problem regarding the usage of exogeneic stromal cell lines, a novel membrane-separated coculture system, in which stromal cells adhere onto the lower surface of a porous membrane and hematopoietic cells are incubated on the upper surface of the membrane, was proposed. In order to mimic the contact between stromal and hematopoietic cells in vivo in the bone marrow, several types of three-dimensional (3-D) culture of hematopoietic cells were developed. The 3-D coculture of hematopoietic cells with spatial development of stromal cells in nonwoven fabrics enabled the expansion of progenitors without cytokine addition. Progenitors in cord blood mononucleated cells were also successfully expanded without the addition in the 3-D coculture with primary human bone marrow stromal cells in 3-D. Heparin addition to the 3-D coculture and coating the nonwoven fabrics with N-(O-beta-(6-O-sulfogalactopyranosyl)-6-oxyhexyl)-3,5-bis(dodecyloxy)-benzamide further increased the number of progenitors.
Collapse
Affiliation(s)
- Mutsumi Takagi
- Division of Molecular Chemistry, Graduate School of Engineering, Hokkaido University, N13W8, Kita-ku, Sapporo 060-8628, Japan.
| |
Collapse
|
20
|
Dörmer P, Spitzer E, Möller W. EDR is a stress-related survival factor from stroma and other tissues acting on early haematopoietic progenitors (E-Mix). Cytokine 2005; 27:47-57. [PMID: 15242693 DOI: 10.1016/j.cyto.2004.03.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2003] [Revised: 03/12/2004] [Accepted: 03/30/2004] [Indexed: 01/16/2023]
Abstract
The erythroid differentiation regulator (EDR) is a highly conserved autocrine factor produced in many tissues. Its haemoglobin synthesis-inducing activity for human and murine erythroleukaemia cell lines had been detected in WEHI-3 conditioned medium. EDR functions were analysed in detail. It is released from cells immediately in response to various stressful conditions and enhances cell survival particularly at a lower concentration range and low cell density. At high cell density and high EDR concentration the opposite effect of an increase in cell death was observed. Its essential function within a tissue is considered to be the maintenance of growth homeostasis. Cells kept in culture for weeks show a decreasing responsiveness to EDR supply. This was also noted in freshly cloned EDR-responsive mouse erythroleukaemia cells pointing to a molecular adaptation process. Human haematopoietic progenitors were amplified 7-fold by EDR when kept at low cytokine levels. At saturating levels progenitors giving rise to at least two lineages in semisolid medium (E-mix) respond to EDR with an average 1.87-fold increase in colony numbers and a bell-shaped dose-response curve. Of the more mature BFU-E compartment a response was observed particularly in cases with low colony numbers. Given the release from irradiated stromal cells and the ability to partly substitute for stromal cells in the Burkitt's lymphoma cell line BL-70, EDR functions as a stromal survival factor for stroma-responsive cells.
Collapse
Affiliation(s)
- Peter Dörmer
- Department of Experimental Hematology, GSF-National Research Center for Environment and Health, Munich, Germany.
| | | | | |
Collapse
|
21
|
Delforge M, Raets V, Van Duppen V, Vandenberghe P, Boogaerts M. CD34+ marrow progenitors from MDS patients with high levels of intramedullary apoptosis have reduced expression of α4β1 and α5β1 integrins. Leukemia 2004; 19:57-63. [PMID: 15510209 DOI: 10.1038/sj.leu.2403551] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Excessive intramedullary apoptosis is central in the pathogenesis of myelodysplastic syndromes (MDS). Growth-inhibiting cytokines, the Fas/FasLigand pathway, and autoreactive cytotoxic T-lymphocytes have been identified to be important proapoptotic factors in MDS. In normal hematopoiesis, alpha4beta1 and alpha5beta1 integrin-mediated interactions between progenitors and fibronectin are critical for progenitor cell survival. In this study, we have used flow cytometry to quantify the expression levels of members of the beta1 integrin family on CD34(+) marrow progenitors in 27 untreated patients with MDS, three with s-AML, and 25 control subjects. In MDS, we observed that nonapoptotic progenitors significantly downregulate cell surface expression levels of alpha4 and beta1 integrin chains compared with healthy controls. Downregulation of alpha4, beta1, and also alpha5 was present in MDS patients with > or =25% apoptotic progenitors, irrespective of their French, American, British subcategory. Reduced cell surface expression levels of alpha4, alpha5, and beta1 did also correlate with decreased in vitro adhesiveness to fibronectin fragments. Therefore, our observations suggest that downregulation of alpha4beta1 and alpha5beta1 integrins on CD34(+) progenitors could be a newly identified proapoptotic mechanism in MDS.
Collapse
Affiliation(s)
- M Delforge
- Department of Hematology, University Hospital Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium.
| | | | | | | | | |
Collapse
|
22
|
Yoshida T, Takagi M. Cell processing engineering for ex vivo expansion of hematopoietic cells: a review. Biochem Eng J 2004. [DOI: 10.1016/j.bej.2003.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
23
|
Jensen MK, Holten-Andersen MN, Riisbro R, de Nully Brown P, Larsen MB, Kjeldsen L, Heickendorff L, Brünner N, Hasselbalch HC. Elevated plasma levels of TIMP-1 correlate with plasma suPAR/uPA in patients with chronic myeloproliferative disorders. Eur J Haematol 2003; 71:377-84. [PMID: 14667201 DOI: 10.1034/j.1600-0609.2003.00096.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Chronic myeloproliferative disorders (MPD) are characterized by progressive remodelling of bone marrow stroma as evidenced by increased deposition of extracellular matrix proteins, neoangiogenesis and displacement of normal haematopoietic cells by fibrotic tissue. The family of metalloproteinases (MMPs) and tissue inhibitors of metalloproteinase (TIMPs) serve to facilitate and inhibit matrix degradation processes, respectively. In an attempt to investigate potential markers for bone marrow remodelling processes, we investigated plasma levels of total-, free- and complexed TIMP-1, TIMP-2, MMP-2 and MMP-9 in a patient cohort comprising 17 with myelofibrosis (MF), 17 with polycythaemia vera (PV), 15 with essential thrombocythaemia (ET), 1 with a transitional MPD and 30 controls. Compared with controls, total- (P < 0.0001) (median: 132.6 microg/L vs. 80.8 microg/L), free- (P < 0.0001) (median: 126.4 microg/L vs. 65.8 microg/L) and complexed TIMP-1 (P = 0.0009) (median: 17.7 microg/L vs. 10.7 microg/L) concentration was significantly higher in the patients. TIMP-1 was significantly correlated with plasma soluble urokinase plasminogen activator receptor (P = 0.003) and urokinase plasminogen activator (P < 0.0001), respectively, suggesting a common cellular origin. No statistical significant difference between TIMP-2 and MMP-2 levels was observed between patients and controls. Furthermore, a significant correlation between free TIMP-1 and TIMP-2 levels was detected (r = 0.56; P < 0.0001). Median MMP-9 concentration was significantly higher among PV patients compared with controls (P = 0.0015), and 41% of patients with PV (7/17) had MMP-9 values that were above the mean + 2SD of plasma MMP-9 levels found in controls. The ratio of total TIMP-1/MMP-9 was significantly higher in patients with MF compared with controls (P = 0.0004). These findings suggest that a disturbed TIMP-1/MMP ratio may reflect an imbalance of the extracellular homeostasis towards an increased matrix deposition promoting fibrosis.
Collapse
Affiliation(s)
- Morten Krogh Jensen
- Department of Haematology L, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Scortegagna M, Morris MA, Oktay Y, Bennett M, Garcia JA. The HIF family member EPAS1/HIF-2alpha is required for normal hematopoiesis in mice. Blood 2003; 102:1634-40. [PMID: 12750163 DOI: 10.1182/blood-2003-02-0448] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Hypoxic stress plays a role in pathophysiologic states such as myocardial infarction and cerebral vascular events as well as in normal physiologic conditions including development and hematopoiesis. Members of the hypoxia inducible factor (HIF) family function as transcriptional regulators of genes involved in the hypoxic response. After generating adult mice that globally lack endothelial PAS domain protein 1 (EPAS1, also known as HIF-2alpha/HRF/HLF/MOP3), the second member of the HIF family, characterization of the hematopoietic cell population indicated that the loss of EPAS1/HIF-2alpha resulted in pancytopenia. Using bone marrow reconstitution experiments of lethally irradiated hosts, we have defined the extent and site of hematopoietic impairment in the EPAS1/HIF-2alpha null mice. These data suggest a critical role for EPAS1/HIF-2alpha in maintaining a functional microenvironment in the bone marrow for effective hematopoiesis.
Collapse
Affiliation(s)
- Marzia Scortegagna
- University of Texas Southwestern Medical Center, Department of Internal Medicine, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | | | | | | | | |
Collapse
|
25
|
Giles FJ, Keating A, Goldstone AH, Avivi I, Willman CL, Kantarjian HM. Acute myeloid leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2003:73-110. [PMID: 12446420 DOI: 10.1182/asheducation-2002.1.73] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In this chapter, Drs. Keating and Willman review recent advances in our understanding of the pathophysiology of acute myeloid leukemia (AML) and allied conditions, including the advanced myelodysplastic syndromes (MDS), while Drs. Goldstone, Avivi, Giles, and Kantarjian focus on therapeutic data with an emphasis on current patient care and future research studies. In Section I, Dr. Armand Keating reviews the role of the hematopoietic microenvironment in the initiation and progression of leukemia. He also discusses recent data on the stromal, or nonhematopoietic, marrow mesenchymal cell population and its possible role in AML. In Section II, Drs. Anthony Goldstone and Irit Avivi review the current role of stem cell transplantation as therapy for AML and MDS. They focus on data generated on recent Medical Research Council studies and promising investigation approaches. In Section III, Dr. Cheryl Willman reviews the current role of molecular genetics and gene expression analysis as tools to assist in AML disease classification systems, modeling of gene expression profiles associated with response or resistance to various interventions, and identifying novel therapeutic targets. In Section IV, Drs. Hagop Kantarjian and Francis Giles review some promising agents and strategies under investigation in the therapy of AML and MDS with an emphasis on novel delivery systems for cytotoxic therapy and on targeted biologic agents.
Collapse
Affiliation(s)
- Francis J Giles
- M.D. Anderson Cancer Center, Department of Leukemia, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
26
|
Han P, Guo XH, Story CJ. Enhanced expansion and maturation of megakaryocytic progenitors by fibronectin. Cytotherapy 2003; 4:277-83. [PMID: 12194724 DOI: 10.1080/146532402320219790] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Megakaryopoiesis occurs as a result of a complex interaction between hemopoietic cells, stromal cells, hemopoietic growth factors and extracellular matrix (ECM). Megakaryocyte growth and development factor (MGDF) or thrombopoietin is the main growth factor that induces megakaryocyte commitment. The role of adhesion proteins in the generation of megakaryocytic progenitors has not been well studied. METHODS We isolated CD34(+) cells from umbilical cord (UC) blood and cultured them in serum-free medium containing IL-3, IL-6, SCF and MGDF, with or without fibronectin. Cultures were sampled at Days 4 and 8 for cell count, immunophenotyping by flow cytometry, and megakaryocyte colony-forming units (CFU-Mk) assay. RESULTS Fibronectin in synergy with MGDF increased both total and CD34(+) cell count. Immunophenotyping of the CD34(+) and CD34(-) cells showed that the percentage expression of CD61 and CD41a on CD34(-) cells was increased by fibronectin compared with CD34(+) cells by Day 8 of culture. The CFU-Mk assay confirmed that fibronectin increased generation of megakaryocytic progenitors by 2.4-fold by Day 8 of culture, but the absolute number was less than that of Day 4 culture. The cells in the CFU-Mk colonies derived from culture containing fibronectin were larger than those from cytokine-only culture. CONCLUSION It was concluded that the addition of fibronectin to the culture system enhanced MGDF, induced ex vivo expansion of megakaryocytic progenitors from CD34(+) cells, as well as increasing their maturation towards later stages of megakaryocyte differentiation.
Collapse
Affiliation(s)
- P Han
- Department of Haematology, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | | | | |
Collapse
|
27
|
Sasaki T, Takagi M, Soma T, Yoshida T. 3D culture of murine hematopoietic cells with spatial development of stromal cells in nonwoven fabrics. Cytotherapy 2003; 4:285-91. [PMID: 12194725 DOI: 10.1080/146532402320219808] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND The in vivo hematopoietic microenvironment is composed of stromal cells and extracellular matrix in a 3D configuration. We have created a 3D microenvironment in vitro, employing spatial development of stromal cells in a nonwoven fabric porous carrier, Fibra-cel (FC). We compared its performance with that of a 2D microenvironment. METHODS Primary murine BM cells were inoculated on the layers of stromal cells prepared in FC (3D) or on a dish (2D) and cultured for 7-21 days. The hematopoietic cells harvested from the cultures were evaluated by colony-forming unit (CFU) assay and transplantation to sub lethally irradiated mice. RESULTS The maximum stromal cell concentration in the 2D culture was higher than that in the 3D culture. However, the hematopoietic cell concentration in the 3D culture was kept at a higher level than that in the 2D culture. The number of CFU-mix increased five times during 3D cultivation, but decreased in the 2D culture. The engraftment percentage of 3D cultured cells was comparable with that of fresh cells, and markedly higher than that of 2D cultured cells. DISCUSSION The 3D culture constructed with FC and stromal cells was clearly superior to 2D culture because hematopoietic progenitor cells were expanded without the addition of cytokines and the content of hematopoietic stem cells was maintained.
Collapse
Affiliation(s)
- T Sasaki
- International Center for Biotechnology, Osaka Univeristy, Suita, Osaka, Japan
| | | | | | | |
Collapse
|
28
|
Hermand P, Gane P, Huet M, Jallu V, Kaplan C, Sonneborn HH, Cartron JP, Bailly P. Red cell ICAM-4 is a novel ligand for platelet-activated alpha IIbbeta 3 integrin. J Biol Chem 2003; 278:4892-8. [PMID: 12477717 DOI: 10.1074/jbc.m211282200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ICAM-4 (LW blood group glycoprotein) is an erythroid-specific membrane component that belongs to the family of intercellular adhesion molecules and interacts in vitro with different members of the integrin family, suggesting a potential role in adhesion or cell interaction events, including hemostasis and thrombosis. To evaluate the capacity of ICAM-4 to interact with platelets, we have immobilized red blood cells (RBCs), platelets, and ICAM-Fc fusion proteins to a plastic surface and analyzed their interaction in cell adhesion assays with RBCs and platelets from normal individuals and patients, as well as with cell transfectants expressing the alpha(IIb)beta(3) integrin. The platelet fibrinogen receptor alpha(IIb)beta(3) (platelet GPIIb-IIIa) in a high affinity state following GRGDSP peptide activation was identified for the first time as the receptor for RBC ICAM-4. The specificity of the interaction was demonstrated by showing that: (i) activated platelets adhered less efficiently to immobilized ICAM-4-negative than to ICAM-4-positive RBCs, (ii) monoclonal antibodies specific for the beta(3)-chain alone and for a complex-specific epitope of the alpha(IIb)beta(3) integrin, and specific for ICAM-4 to a lesser extent, inhibited platelet adhesion, whereas monoclonal antibodies to GPIb, CD36, and CD47 did not, (iii) activated platelets from two unrelated type-I glanzmann's thrombasthenia patients did not bind to coated ICAM-4. Further support to RBC-platelet interaction was provided by showing that dithiothreitol-activated alpha(IIb)beta(3)-Chinese hamster ovary transfectants strongly adhere to coated ICAM-4-Fc protein but not to ICAM-1-Fc and was inhibitable by specific antibodies. Deletion of individual Ig domains of ICAM-4 and inhibition by synthetic peptides showed that the alpha(IIb)beta(3) integrin binding site encompassed the first and second Ig domains and that the G65-V74 sequence of domain D1 might play a role in this interaction. Although normal RBCs are considered passively entrapped in fibrin polymers during thrombus, these studies identify ICAM-4 as the first RBC protein ligand of platelets that may have relevant physiological significance.
Collapse
Affiliation(s)
- Patricia Hermand
- INSERM U76, Institut National de la Transfusion Sanguine, 6 rue Alexandre Cabanel, 75015 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Wertheim JA, Miller JP, Xu L, He Y, Pear WS. The biology of chronic myelogenous leukemia:mouse models and cell adhesion. Oncogene 2002; 21:8612-28. [PMID: 12476308 DOI: 10.1038/sj.onc.1206089] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Chronic myelogenous leukemia (CML) is a biphasic neoplasm of the bone marrow that is precipitated by the Philadelphia chromosome, a t(9;22) balanced translocation that encodes a constitutively activated nonreceptor tyrosine kinase termed P210(BCR-ABL). This oncoprotein has several intracellular functions; however, the most important effect of P210(BCR-ABL) leading to cell transformation is phosphorylation of signaling molecules through a constitutively active tyrosine kinase domain. Despite extensive knowledge of the structure and functional domains of BCR-ABL, its precise function in transformation is not known. Progress has been hampered, in part, by the lack of relevant CML models, as cell culture and in vitro assays do not mimic the pathogenesis of CML. Recently, there has been significant progress toward improving murine models that closely resemble human CML. This has allowed researchers to evaluate critical functions of BCR-ABL and has provided a model to test the efficacy of therapeutic medications that block these pathways. Our laboratory has developed two intersecting research programs to better understand the functioning of P210(BCR-ABL) in leukemogenesis. In one approach, we have developed a murine CML model by transferring HSCs that express BCR-ABL from a retroviral vector. All recipients develop a rapidly fatal MPD that shares several important features with CML. This model has been extremely useful for studying the function of BCR-ABL in the pathogenesis of CML. A second approach utilizes a quantitative cell detachment apparatus capable of measuring small changes in cell adhesion to investigate the mechanism by which P210(BCR-ABL) causes abnormal cell binding. Altered cell adhesion may contribute to the imbalance between proliferation and self-renewal in the hematopoietic progenitor compartment. To better understand the role abnormal adhesion may play in the development of leukemia, we have attempted to correlate the effects of functional P210(BCR-ABL) mutants in regulating adhesion and oncogenicity.
Collapse
Affiliation(s)
- Jason A Wertheim
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6160, USA
| | | | | | | | | |
Collapse
|
30
|
Siler U, Rousselle P, Müller CA, Klein G. Laminin gamma2 chain as a stromal cell marker of the human bone marrow microenvironment. Br J Haematol 2002; 119:212-20. [PMID: 12358928 DOI: 10.1046/j.1365-2141.2002.03800.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Laminins are large heterotrimeric molecules consisting of alpha, beta and gamma chains. At present, five alpha chains, three beta chains and three gamma chains have been characterized. Laminin-5 (alpha3beta3gamma2) is the only isoform known to date which contains a gamma2 chain. In human bone marrow, non-haematopoietic stromal cells expressed the laminin gamma2 chain, whereas bone marrow mononuclear cells did not. Co-localization of the gamma2 chain was detected with the laminin alpha4 and alpha5 chains, and co-immunoprecipitation studies revealed a new isoform consisting of alpha5, beta2 and gamma2 chains. The laminin gamma2 chain was also co-localized with alpha-sm-actin in bone marrow, but it was not expressed in endothelial cells or megakaryocytes, indicating that the gamma2 chain is exclusively expressed in vivo in bone marrow stromal cells. The laminin gamma2 chain containing isoform LN-5 was shown to be an adhesive substrate for a small subpopulation of bone marrow mononuclear cells and also for peripheral blood platelets. Taken together, these results indicate that (I) laminin isoforms containing the gamma2 chain can act as adhesive substrates for human haematopoietic cells, and (II) the laminin gamma2 chain can be used as a specific marker molecule for human bone-marrow-derived stromal cells.
Collapse
Affiliation(s)
- Ulrich Siler
- University Medical Clinic, Section for Transplantation Immunology and Immunohematology, Tübingen, Germany
| | | | | | | |
Collapse
|
31
|
Luikart S, Masri M, Wahl D, Hinkel T, Beck JM, Gyetko MR, Gupta P, Oegema T. Urokinase is required for the formation of mactinin, an alpha-actinin fragment that promotes monocyte/macrophage maturation. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1591:99-107. [PMID: 12183060 DOI: 10.1016/s0167-4889(02)00255-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have previously shown that lysates from HL-60 myeloid leukemia cells or from peripheral blood monocytes are able to degrade alpha-actinin to form a 31-kDa amino-terminal fragment with monocyte/macrophage maturation promoting activity. In contrast, intact alpha-actinin, which is a 100-kDa actin-binding protein, has no differentiating activity. The aim of this study was to investigate the enzyme responsible for the degradation of alpha-actinin to form this fragment, named mactinin. The ability of cell lysates to degrade [125I]alpha-actinin in the presence of various enzyme inhibitors, including inhibitors of metalloproteinases, cysteine proteinases, and serine proteases, was measured. Phenylmethylsulfonyl fluoride (PMSF) was the only inhibitor able to prevent formation of mactinin by cell lysate degradation of alpha-actinin, suggesting that a serine protease is responsible for the digestion. Of the various serine proteases tested (thrombin, plasmin, and urokinase), only urokinase was able to produce a 31-kDa band. The urokinase-generated 31-kDa band promoted maturation in HL-60 cells. Amiloride, a specific inhibitor of urokinase, inhibited production of the 31-kDa alpha-actinin fragment by HL-60 cell lysates. For in vivo tests, inflammatory fluid (from bronchoalvelolar lavage) was collected from uPA (urokinase) knockout mice and their wild-type counterparts after intratracheal challenge with Pneumocystis carinii. Although most (6 of 8) wild-type mice had mactinin in their inflammatory fluid samples, none (0 of 8) of the uPA knockout mice had mactinin present (P<0.01). These results demonstrate that urokinase is necessary and sufficient for the formation of the monocyte/macrophage maturation promoting fragment, mactinin, in vitro and in vivo. These findings support the role of urokinase in the regulation of monocyte/macrophage functions, such as that occurring in inflammatory reactions.
Collapse
|
32
|
Netelenbos T, Zuijderduijn S, van den Born J, Kessler FL, Zweegman S, Huijgens PC, Dräger AM. Proteoglycans guide SDF‐1‐induced migration of hematopoietic progenitor cells. J Leukoc Biol 2002. [DOI: 10.1189/jlb.72.2.353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Tanja Netelenbos
- Department of Hematology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands and
| | - Suzanne Zuijderduijn
- Department of Hematology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands and
| | - Jacob van den Born
- Department of Molecular Cell Biology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Floortje L. Kessler
- Department of Hematology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands and
| | - Sonja Zweegman
- Department of Hematology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands and
| | - Peter C. Huijgens
- Department of Hematology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands and
| | - Angelika M. Dräger
- Department of Hematology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands and
| |
Collapse
|
33
|
Noll T, Jelinek N, Schmid S, Biselli M, Wandrey C. Cultivation of hematopoietic stem and progenitor cells: biochemical engineering aspects. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2002; 74:111-28. [PMID: 11991176 DOI: 10.1007/3-540-45736-4_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The ex vivo expansion of hematopoietic cells is one of the most challenging fields in cell culture. This is a rapidly growing area of tissue engineering with many potential applications in bone marrow transplantation, transfusion medicine or gene therapy. Over the last few years much progress has been made in understanding hematopoietic differentiation, discovery of cytokines, isolation and identification of cellular subtypes and in the development of a variety of bioreactor concepts. All this has led to a number of (preliminary) clinical trials that gave a hint of the benefits that can be obtained from the use of expanded hematopoietic cells in therapy. Moreover, as we understand the complexity and the regulation of hematopoiesis, it becomes obvious that highly sophisticated cultivation techniques and bioreactor concepts are needed: a new challenge for bioprocess engineering in cell culture.
Collapse
Affiliation(s)
- Thomas Noll
- Institut für Biotechnologie 2, Forschungszentrum Jülich GmbH, Germany.
| | | | | | | | | |
Collapse
|
34
|
Bhatia R, Williams AD, Munthe HA. Contact with fibronectin enhances preservation of normal but not chronic myelogenous leukemia primitive hematopoietic progenitors. Exp Hematol 2002; 30:324-32. [PMID: 11937267 DOI: 10.1016/s0301-472x(01)00799-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVE Coculture with stromal cells enhances preservation and self-renewal of primitive progenitor potential in hematopoietic cells during ex vivo culture with growth factors (GF). However, the respective roles of growth factors, stromal contact, and extracellular matrix (ECM) ligands in this effect are not clear. Here we investigated the role of direct contact with stroma and the ECM protein fibronectin (FN) in these effects, and investigated whether abnormal integrin receptor function in chronic myelogenous leukemia (CML) progenitors was associated with perturbation in these responses. METHODS Normal bone marrow CD34+ cells were cultured in GF-containing medium with or without contact with stromal layers, glutaraldehyde-fixed stromal layers (stroma-contact), or integrin-binding FN fragments for 7 days. Progeny cells were assayed for primitive progenitors in week-6 long-term culture-initiating cell (LTC-IC) and week-10 extended LTC-IC (ELTC-IC) assays. RESULTS Increased LTC-IC and ELTC-IC preservation was seen following coculture with stroma, and was also observed after culture in contact with fixed stromal layers and FN. Both alpha4beta1 and alpha5beta1-integrin binding FN fragments enhanced LTC-IC preservation. Analysis of single CD34+CD38- cells showed that coculture with FN resulted in significantly reduced cell division, but enhanced retention of LTC-IC capacity in divided cells. FN also increased LTC-IC frequency in undivided cells. CML progenitors demonstrate deficient integrin-mediated adhesion, migration, and signaling. Coculture of CML CD34+ cells with stroma and FN failed to enhance LTC-IC preservation. CONCLUSION We conclude that beta1 integrin-FN interactions enhance normal primitive progenitor preservation with or without cell division, and that these mechanisms are impaired in CML primitive progenitors.
Collapse
Affiliation(s)
- Ravi Bhatia
- Division of Hematology and Bone Marrow Transplantation, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA 91010, USA.
| | | | | |
Collapse
|
35
|
Marquez-Curtis LA, Dobrowsky A, Montaño J, Turner AR, Ratajczak J, Ratajczak MZ, Janowska-Wieczorek A. Matrix metalloproteinase and tissue inhibitors of metalloproteinase secretion by haematopoietic and stromal precursors and their production in normal and leukaemic long-term marrow cultures. Br J Haematol 2001; 115:595-604. [PMID: 11736941 DOI: 10.1046/j.1365-2141.2001.03160.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) regulate the turnover of the extracellular matrix and may modulate the biology of haematopoietic cells. We investigated whether MMPs and TIMPs are produced in long-term marrow cultures (LTMCs) established from normal donors and acute myelogenous leukaemia (AML) patients, and by fibroblast- (F), granulocyte macrophage- (GM) and megakaryocyte- (Meg) colony-forming unit (CFU) and erythroid burst-forming unit (BFU-E)-derived precursor cells. ProMMP-9 levels were highest (> 400 ng/ml) at week 1 of normal LTMC, whereas proMMP-2, TIMP-1, TIMP-2 and TIMP-3 levels peaked (up to 1000 ng/ml) after the establishment of the adherent layer. In LTMC from AML patients, these patterns of secretion were reversed. Moreover, we found that after a 24 h incubation in serum-free media, normal CFU-GM-, BFU-E- and CFU-Meg-derived cells secreted proMMP-9 and CFU-F-derived cells proMMP-2, in contrast to cells from LTMC adherent layer which secreted both active and latent forms of MMP-2 and MMP-9 under serum-free conditions. However, when these adherent cells were incubated in 12.5% fetal calf or horse serum or complete LTMC growth media, active forms of MMP-2 and MMP-9 were no longer detectable, and TIMP levels increased. Hence, we concluded that (i) MMPs/TIMPs are secreted by normal human bone marrow haematopoietic and stromal cells and may play an important role in intercellular cross-talk in haematopoiesis; and (ii) only latent forms of MMPs are present under LTMC conditions, indicating that the specific media used for weekly re-feeding of LTMC can block activation of MMP-2 and MMP-9, maintaining the integrity of the stromal layer and supporting haematopoiesis in vitro.
Collapse
|
36
|
Bhatia R, Munthe HA, Forman SJ. Abnormal growth factor modulation of beta1-integrin-mediated adhesion in chronic myelogenous leukaemia haematopoietic progenitors. Br J Haematol 2001; 115:845-53. [PMID: 11843818 DOI: 10.1046/j.1365-2141.2001.03192.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abnormal progenitor circulation and extramedullary haematopoiesis are characteristic features of chronic myelogenous leukaemia (CML). Growth factor (GF) and beta1-integrin interactions play an important role in regulation of progenitor trafficking to and from the marrow space. CML progenitors demonstrate abnormal beta1-integrin-mediated adhesion to fibronectin (FN). In the present study we investigated whether GF modulation of beta1-integrin-mediated adhesion and migration was altered in CML progenitors. Culture with low concentrations of GF enhanced normal progenitor adhesion to FN compared with no GF, but failed to enhance CML progenitor adhesion to FN. Similarly, high concentrations of selected GF rapidly enhanced beta1-integrin-mediated adhesion of normal progenitors to FN through a phosphotidylinositol-3 (PI-3) kinase-dependent mechanism, but failed to increase CML progenitor adhesion. Exposure to a BCR-ABL tyrosine kinase inhibitor restored GF modulation of CML progenitor adhesion. CML colony-forming cells (CFC) demonstrated increased migration across FN-coated transwells compared with normal CFC in the absence of GF. The addition of stem cell factor resulted in enhanced migration of CML and normal CFC on FN. In conclusion, GF stimulation failed to enhance integrin-mediated adhesion but enhanced migration in CML progenitors on FN. BCR-ABL induced abnormalities in GF-integrin interactions could contribute to abnormal circulation and microenvironmental localization of CML progenitors.
Collapse
Affiliation(s)
- R Bhatia
- Division of Hematology and Bone Marrow Transplantation, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA.
| | | | | |
Collapse
|
37
|
Inoue S, Osmond DG. Basement membrane of mouse bone marrow sinusoids shows distinctive structure and proteoglycan composition: a high resolution ultrastructural study. THE ANATOMICAL RECORD 2001; 264:294-304. [PMID: 11596011 DOI: 10.1002/ar.1166] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Venous sinusoids in bone marrow are the site of a large-scale traffic of cells between the extravascular hemopoietic compartment and the blood stream. The wall of the sinusoids consists solely of a basement membrane interposed between a layer of endothelial cells and an incomplete covering of adventitial cells. To examine its possible structural specialization, the basement membrane of bone marrow sinusoids has now been examined by high resolution electron microscopy of perfusion-fixed mouse bone marrow. The basement membrane layer was discontinuous, consisting of irregular masses of amorphous material within a uniform 60-nm-wide space between apposing endothelial cells and adventitial cell processes. At maximal magnifications, the material was resolved as a random arrangement of components lacking the "cord network" formation seen in basement membranes elsewhere. Individual components exhibited distinctive ultrastructural features whose molecular identity has previously been established. By these morphological criteria, the basement membrane contained unusually abundant chondroitin sulfate proteoglycan (CSPG) revealed by 3-nm-wide "double tracks," and moderate amounts of both laminin as dense irregular coils and type IV collagen as 1-1.5-nm-wide filaments, together with less conspicuous amounts of amyloid P forming pentagonal frames. In contrast, 4.5-5-nm-wide "double tracks" characteristic of heparan sulfate proteoglycan (HSPG) were absent. The findings demonstrate that, in comparison with "typical" basement membranes in other tissues, the bone marrow sinusoidal basement membrane is uniquely specialized in several respects. Its discontinuous nature, lack of network organization, and absence of HSPG, a molecule that normally helps to maintain membrane integrity, may facilitate disassembly and reassembly of basement membrane material in concert with movements of adventitial cell processes as maturing hemopoietic cells pass through the sinusoidal wall: the exceptionally large quantity of CSPG may represent a reservoir of CD44 receptor for use in hemopoiesis.
Collapse
Affiliation(s)
- S Inoue
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada H3A 2B2.
| | | |
Collapse
|
38
|
Frimberger AE, Stering AI, Quesenberry PJ. An in vitro model of hematopoietic stem cell homing demonstrates rapid homing and maintenance of engraftable stem cells. Blood 2001; 98:1012-8. [PMID: 11493446 DOI: 10.1182/blood.v98.4.1012] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cell (HSC) homing is believed to rely heavily on adhesion interactions between stem cells and stroma. An in vitro assay was developed for adhesion of engraftable HSCs in bone marrow suspensions to pre-established Dexter-type long-term bone marrow culture stromal layers. The cell numbers in the adherent layer and supernatant were examined, along with the engraftment capability of adherent layer cells to indicate the number of HSCs that homed to in vitro stroma. The cell number in the supernatant declined over the 24-hour period. The number of test cells adhering to the stromal layer increased during the first hour and then fell at 6 and 24 hours. The number of test HSCs adhering to the stromal layer was substantial at 20 minutes, increased during the first hour, and then remained constant at 1, 6, and 24 hours of adhesion. These data indicate that adhesion of engraftable HSCs occurs quickly and increases during the first hour of contact with pre-established stroma, that adhesion plateaus within 1 hour of contact, and that HSCs maintain their engraftment capability for at least 24 hours of stromal adhesion. Long-term engraftment from test cells at more than 1 hour of adhesion represents 70.7% of the predicted engraftment from equivalent numbers of unmanipulated marrow cells, indicating that 2 of 3 test engraftable HSCs adhered. These findings demonstrate the usefulness of this model system for studying stem-stromal adhesion, allowing further dissection of the mechanism of HSC homing and exploration of possible manipulations of the process. (Blood. 2001;98:1012-1018)
Collapse
Affiliation(s)
- A E Frimberger
- The University of Massachusetts Cancer Center, Worcester, USA.
| | | | | |
Collapse
|
39
|
Khaldoyanidi S, Sikora L, Orlovskaya I, Matrosova V, Kozlov V, Sriramarao P. Correlation between nicotine-induced inhibition of hematopoiesis and decreased CD44 expression on bone marrow stromal cells. Blood 2001; 98:303-12. [PMID: 11435297 DOI: 10.1182/blood.v98.2.303] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This study demonstrates that in vivo exposure to cigarette smoke (CS) and in vitro treatment of long-term bone marrow cultures (LTBMCs) with nicotine, a major constituent of CS, result in inhibition of hematopoiesis. Nicotine treatment significantly delayed the onset of hematopoietic foci and reduced their size. Furthermore, the number of long-term culture-initiating cells (LTC-ICs) within an adherent layer of LTBMCs was significantly reduced in cultures treated with nicotine. Although the production of nonadherent mature cells and their progenitors in nicotine-treated LTBMCs was inhibited, this treatment failed to influence the proliferation of committed hematopoietic progenitors when added into methylcellulose cultures. Bone marrow stromal cells are an integral component of the hematopoietic microenvironment and play a critical role in the regulation of hematopoietic stem cell proliferation and self-renewal. Exposure to nicotine decreased CD44 surface expression on primary bone marrow-derived fibroblastlike stromal cells and MS-5 stromal cell line, but not on hematopoietic cells. In addition, mainstream CS altered the trafficking of hematopoietic stem/progenitor cells (HSPC) in vivo. Exposure of mice to CS resulted in the inhibition of HSPC homing into bone marrow. Nicotine and cotinine treatment resulted in reduction of CD44 surface expression on lung microvascular endothelial cell line (LEISVO) and bone marrow-derived (STR-12) endothelial cell line. Nicotine treatment increased E-selectin expression on LEISVO cells, but not on STR-12 cells. These findings demonstrate that nicotine can modulate hematopoiesis by affecting the functions of the hematopoiesis-supportive stromal microenvironment, resulting in the inhibition of bone marrow seeding by LTC-ICs and interfering with stem cell homing by targeting microvascular endothelial cells.
Collapse
Affiliation(s)
- S Khaldoyanidi
- Division of Vascular Biology, La Jolla Institute for Molecular Medicine, La Jolla, CA 92121, USA
| | | | | | | | | | | |
Collapse
|
40
|
Xu YX, Talati BR, Janakiraman N, Chapman RA, Gautam SC. Growth Factors: Production of Monocyte Chemotactic Protein-1 (MCP-1/JE) by Bone Marrow Stromal Cells: Effect on the Migration and Proliferation of Hematopoietic Progenitor Cells. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2001; 4:345-356. [PMID: 11399575 DOI: 10.1080/10245332.1999.11746458] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Recombinant chemotactic cytokines (chemokines) have been shown to modulate in vitro proliferation of hematopoietic progenitor cells. Whether bone marrow stromal cells produce chemokines and the physiological role they may have in the regulation of hematopoiesis has largely remained unexamined. We have examined the expression of monocyte chemoattractant protein-1 (MCP-1/JE) in bone marrow stromal cells and its effect on the migration and proliferation of murine hematopoietic progenitor cells. Freshly derived murine bone marrow stromal cells were found to secrete abundant amounts of MCP-1/JE, which was further increased upon stimulation of stromal cells with pro-inflammatory agents LPS, IL1-alpha, IFN-gamma, or TNF-alpha. Although culture supernatant conditioned by stromal cells exhibited chemotactic activity toward hematopoietic progenitor cells, the chemotactic activity was not due to MCP-1/JE. Furthermore, rMCP-1/JE also failed to induce migration of progenitor cells. MCP-1/JE, however, caused 20 to 30% increase in the clonal expansion of progenitor cells. Thus, although MCP-1/JE does not chemoattract hematopoietic progenitor cells it may have a role in their proliferation and clonal expansion.
Collapse
Affiliation(s)
- Y. X. Xu
- Division of Hematology/Oncology, Henry Ford Hospital, Detroit, Michigan
| | | | | | | | | |
Collapse
|
41
|
Armeanu S, MÜller CA, Klein G. Involvement of E-cadherin in the Development of Erythroid Cells; Subject Heading. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2001; 5:307-316. [PMID: 11399629 DOI: 10.1080/10245332.2000.11746524] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The cadherins represent a large family of structurally and functionally related cell adhesion molecules involved in morphogenesis of multicellular organisms and maintenance of solid tissues. In the hematopoietic system, however, almost nothing was known about the involvement of this family. PCR screening of RNA of human bone marrow mononuclear cells with specific primers for different classical cadherins revealed that members of this family are also expressed by bone marrow cells. Here we report that E-cadherin, which is mainly expressed by cells of epithelial origin, plays a critical role in the development of human erythrocytes. FACS analysis with human E-cadherin-specific antibodies and the use of immunoaffinity columns revealed that expression of E-cadherin is restricted to defined maturation stages of the erythropoietic cell lineage. Erythroblasts and normoblasts express E-cadherin, but mature erythrocytes do not. Lymphoid and all the other myeloid cell lineages do not express E-cadherin at any developmental stage. The differentiation of the erythroid lineage in vitro could be influenced by addition of anti-E-cadherin antibodies in a concentration dependent manner indicating a direct involvement of this cell adhesion molecule in the differentiation process. In line with these in vitro data is the finding that E-cadherin is down regulated during erythroleukemia on the developing erythroid cells. Our results suggest an unanticipated function of E-cadherin in the hematopoietic system.
Collapse
Affiliation(s)
- Sorin Armeanu
- Medical University Clinic, Section for Transplantation Immunology and Immunohematology, 72072 Tübingen, FRG
| | | | | |
Collapse
|
42
|
Abstract
Biochemical and molecular genetic studies have revealed that blood group antigens are present on cell surface molecules of wide structural diversity, including carbohydrate epitopes on glycoproteins and/or glycolipids, and peptide antigens on proteins inserted within the membrane via single or multi-pass transmembrane domains, or via glycosylphosphatidylinositol linkages. These studies have also shown that some blood group antigens are carried by complexes consisting of several membrane components which may be lacking or severely deficient in rare blood group 'null' phenotypes. In addition, although all blood group antigens are serologically detectable on red blood cells (RBCs), most of them are also expressed in non-erythroid tissues, raising further questions on their physiological function under normal and pathological conditions. In addition to their structural diversity, blood group antigens also possess wide functional diversity, and can be schematically subdivided into five classes: i) transporters and channels; ii) receptors for ligands, viruses, bacteria and parasites; iii) adhesion molecules; iv) enzymes; and v) structural proteins. The purpose of this review is to summarize recent findings on these molecules, and in particular to illustrate the existing structure-function relationships.
Collapse
MESH Headings
- Animals
- Anion Exchange Protein 1, Erythrocyte/chemistry
- Anion Exchange Protein 1, Erythrocyte/physiology
- Antigens, Protozoan
- Blood Group Antigens/chemistry
- Blood Group Antigens/classification
- Blood Group Antigens/genetics
- Blood Group Antigens/immunology
- Blood Group Antigens/physiology
- Blood Proteins/chemistry
- Blood Proteins/genetics
- Blood Proteins/immunology
- Blood Proteins/physiology
- Carrier Proteins/chemistry
- Carrier Proteins/genetics
- Carrier Proteins/immunology
- Carrier Proteins/physiology
- Cell Adhesion Molecules/chemistry
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/physiology
- Chromosomes, Human/genetics
- Enzymes/chemistry
- Enzymes/genetics
- Enzymes/immunology
- Enzymes/physiology
- Erythrocyte Membrane/chemistry
- Erythrocyte Membrane/immunology
- Erythrocytes/enzymology
- Erythrocytes/microbiology
- Erythrocytes/parasitology
- Erythrocytes/virology
- Genes
- Humans
- Integrins/chemistry
- Integrins/genetics
- Integrins/immunology
- Integrins/physiology
- Ion Channels/chemistry
- Ion Channels/genetics
- Ion Channels/immunology
- Ion Channels/physiology
- Models, Molecular
- Organ Specificity
- Protein Conformation
- Protozoan Proteins
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/physiology
- Receptors, HIV/physiology
- Rh-Hr Blood-Group System/chemistry
- Rh-Hr Blood-Group System/genetics
- Rh-Hr Blood-Group System/immunology
- Rh-Hr Blood-Group System/physiology
- Species Specificity
- Structure-Activity Relationship
Collapse
|
43
|
Puch S, Armeanu S, Kibler C, Johnson KR, Müller CA, Wheelock MJ, Klein G. N-cadherin is developmentally regulated and functionally involved in early hematopoietic cell differentiation. J Cell Sci 2001; 114:1567-77. [PMID: 11282032 DOI: 10.1242/jcs.114.8.1567] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cadherins, an important family of cell adhesion molecules, are known to play major roles during embryonic development and in the maintenance of solid tissue architecture. In the hematopoietic system, however, little is known of the role of this cell adhesion family. By RT-PCR, western blot analysis and immunofluorescence staining we show that N-cadherin, a classical type I cadherin mainly expressed on neuronal, endothelial and muscle cells, is expressed on the cell surface of resident bone marrow stromal cells. FACS analysis of bone marrow mononuclear cells revealed that N-cadherin is also expressed on a subpopulation of early hematopoietic progenitor cells. Triple-color FACS analysis defined a new CD34(+) CD19(+) N-cadherin(+) progenitor cell population. During further differentiation, however, N-cadherin expression is lost. Treatment of CD34(+) progenitor cells with function-perturbing N-cadherin antibodies drastically diminished colony formation, indicating a direct involvement of N-cadherin in the differentiation program of early hematopoietic progenitors. N-cadherin can also mediate adhesive interactions within the bone marrow as demonstrated by inhibition of homotypic interactions of bone-marrow-derived cells with N-cadherin antibodies. Together, these data strongly suggest that N-cadherin is involved in the development and retention of early hematopoietic progenitors within the bone marrow microenvironment.
Collapse
Affiliation(s)
- S Puch
- University Medical Clinic, Section for Transplantation Immunology and Immunohematology, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
Prosper F, Verfaillie CM. Regulation of hematopoiesis through adhesion receptors. J Leukoc Biol 2001. [DOI: 10.1189/jlb.69.3.307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Felipe Prosper
- Department of Hematology and Medical Oncology, Hospital Clinico Universitario, University of Valencia, Spain
| | - Catherine M. Verfaillie
- Stem Institute, and Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis
| |
Collapse
|
45
|
Abstract
Laminins are a family of disulfide-linked heterotrimeric proteins consisting of 3 different subunits termed α, β, and γ chains. Combinations of 11 characterized laminin subunits (α1-α5, β1-β3, and γ1-γ3) generate at least 12 laminin isoforms, which can serve different functions. Although expression of laminin in the hematopoietic microenvironment has been known for many years, the nature of the laminin isoforms present in the human bone marrow is poorly characterized. The present study attempts to clarify this issue. Reverse transcriptase–polymerase chain reaction analysis of human bone marrow stromal cells suggested the expression of many laminin isoforms in the marrow. Northern blot and immunoblot analysis, however, showed that laminin-8/9 and laminin-10/11 are the most abundant laminin isoforms synthesized by human bone marrow stromal cells. Other isoforms, if present, certainly play a minor role in the hematopoietic microenvironment. Functionally, laminin-10/11 preparations showed strong adhesive interactions with human CD34+ cell lines. Antibodies against the β1 integrin subunit inhibited these interactions. Other laminin isoforms, especially laminin-1 and laminin-2/4, showed only weak or no adhesive interactions with the hematopoietic cell lines tested, explaining former negative results. In addition to its adhesion-mediating properties, laminin-10/11 preparations also showed a mitogenic activity for human hematopoietic progenitor cells. Taken together, these data suggest that laminin in the bone marrow plays a hitherto unexpected important function in the development of hematopoietic progenitor cells.
Collapse
|
46
|
Abstract
AbstractLaminins are a family of disulfide-linked heterotrimeric proteins consisting of 3 different subunits termed α, β, and γ chains. Combinations of 11 characterized laminin subunits (α1-α5, β1-β3, and γ1-γ3) generate at least 12 laminin isoforms, which can serve different functions. Although expression of laminin in the hematopoietic microenvironment has been known for many years, the nature of the laminin isoforms present in the human bone marrow is poorly characterized. The present study attempts to clarify this issue. Reverse transcriptase–polymerase chain reaction analysis of human bone marrow stromal cells suggested the expression of many laminin isoforms in the marrow. Northern blot and immunoblot analysis, however, showed that laminin-8/9 and laminin-10/11 are the most abundant laminin isoforms synthesized by human bone marrow stromal cells. Other isoforms, if present, certainly play a minor role in the hematopoietic microenvironment. Functionally, laminin-10/11 preparations showed strong adhesive interactions with human CD34+ cell lines. Antibodies against the β1 integrin subunit inhibited these interactions. Other laminin isoforms, especially laminin-1 and laminin-2/4, showed only weak or no adhesive interactions with the hematopoietic cell lines tested, explaining former negative results. In addition to its adhesion-mediating properties, laminin-10/11 preparations also showed a mitogenic activity for human hematopoietic progenitor cells. Taken together, these data suggest that laminin in the bone marrow plays a hitherto unexpected important function in the development of hematopoietic progenitor cells.
Collapse
|
47
|
Kronenwett R, Martin S, Haas R. The role of cytokines and adhesion molecules for mobilization of peripheral blood stem cells. Stem Cells 2000; 18:320-30. [PMID: 11007916 DOI: 10.1634/stemcells.18-5-320] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
CD34(+) hematopoietic stem cells from peripheral blood are commonly used for autologous or allogeneic transplantation following high-dose therapy in malignant diseases. The introduction of hematopoietic growth factors such as G-CSF has greatly facilitated the mobilization of CD34(+) cells. The mechanism of stem cell mobilization is not yet clear. It seems to be a multistep process with a crosstalk between cytokines and adhesion molecules. In this review, the role of hematopoietic growth factors, chemokines, and adhesion molecules for mobilization and homing of CD34(+) cells is summarized. In addition, factors influencing the cytokine-induced mobilization in patients and healthy donors are described. The review closes with an overview of new classes of mobilizing drugs such as monoclonal antibodies, specific peptides, or antisense oligonucleotides targeting adhesion molecules.
Collapse
Affiliation(s)
- R Kronenwett
- Klinik für Hämatologie, Onkologie und klinische Immunologie, Heinrich-Heine-Universität Düsseldorf, Germany
| | | | | |
Collapse
|
48
|
Abstract
Stem cell trafficking between extravascular marrow sites and circulating blood is an essential part of the blood stem cell transplantation technology. Recombinant human G-CSF (rHuG-CSF) is widely used for stem cell peripheralization alone or together with chemopriming mobilizing early and pluripotent CD34+ cell subsets. New cytokine/chemokine mobilization regimens are under investigation such as combined rHuG-CSF and rHu thrombopoietin, rHuG-CSF and interleukin 3, rHuG-CSF and rHu stem cell factor, rHuG-CSF and Flt-3 ligand, human macrophage inflammatory protein, interleukin 1, and interleukin 8. Modifying the adherence of CD34+ cells to extracellular matrix molecules is a new mechanism by which hematopoietic progenitor cells are released into the circulating blood. Blocking the alpha4beta1 integrin receptor on CD34+ progenitor cells by using monoclonal antibodies specific for the heterodimeric complex alpha4beta1 has been shown to further increase the circulating stem cell concentration when given following rHuG-CSF priming. The current clinical research is primarily focused on improving stem cell mobilization efficiency in heavily pretreated and poorly mobilizing patients, and to decrease adverse effects of cytokine treatment.
Collapse
Affiliation(s)
- M Körbling
- University of Texas, M.D. Anderson Cancer Center, Houston, USA
| |
Collapse
|
49
|
Oostendorp RA, Ghaffari S, Eaves CJ. Kinetics of in vivo homing and recruitment into cycle of hematopoietic cells are organ-specific but CD44-independent. Bone Marrow Transplant 2000; 26:559-66. [PMID: 11019847 DOI: 10.1038/sj.bmt.1702536] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this study, we investigated the homing and initiation of division of fluorescently labelled adult mouse bone marrow cells after their intravenous injection into lethally irradiated congenic mice. After 2 h, only 3% of the transplanted cells remained in the blood, and approximately 35% could be retrieved from the marrow, liver and spleen in approximately equal numbers. Subsequently, the proportion of injected cells found in blood, liver and spleen decreased further, but increased slightly (to approximately 17%) in the marrow. Homing of progenitors followed a similar pattern. At 22 h post transplant, almost half of the injected cells in the blood, liver and spleen had completed a first mitosis; although these did not include progenitors with in vitro clonogenic ability. At the same time, >90% of the injected cells recovered from the marrow had not yet divided. Parallel studies with CD44-/- mice showed these to contain a numerically and functionally normal stem cell population whose homing and activation in either CD44+/+ or CD44-/- hosts appeared unaltered. These results indicate homing mechanisms that favor more stable retention of transplanted marrow cells in the marrow of the recipient, more rapid activation of some of those cells that home to other sites, and a lack of change in either of these responses when either the transplanted or the recipient cells do not express CD44.
Collapse
Affiliation(s)
- R A Oostendorp
- The Terry Fox Laboratory, British Columbia Cancer Research Center, Vancouver, Canada
| | | | | |
Collapse
|
50
|
Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood 2000. [DOI: 10.1182/blood.v96.5.1723] [Citation(s) in RCA: 889] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AbstractWe investigated the functions of adiponectin, an adipocyte-specific secretory protein and a new member of the family of soluble defense collagens, in hematopoiesis and immune responses. Adiponectin suppressed colony formation from colony-forming units (CFU)—granulocyte-macrophage, CFU-macrophage, and CFU-granulocyte, whereas it had no effect on that of burst-forming units—erythroid or mixed erythroid-myeloid CFU. In addition, adiponectin inhibited proliferation of 4 of 9 myeloid cell lines but did not suppress proliferation of erythroid or lymphoid cell lines except for one cell line. These results suggest that adiponectin predominantly inhibits proliferation of myelomonocytic lineage cells. At least one mechanism of the growth inhibition is induction of apoptosis because treatment of acute myelomonocytic leukemia lines with adiponectin induced the appearance of subdiploid peaks and oligonucleosomal DNA fragmentation. Aside from inhibiting growth of myelomonocytic progenitors, adiponectin suppressed mature macrophage functions. Treatment of cultured macrophages with adiponectin significantly inhibited their phagocytic activity and their lipopolysaccharide-induced production of tumor necrosis factor α. Suppression of phagocytosis by adiponectin is mediated by one of the complement C1q receptors, C1qRp, because this function was completely abrogated by the addition of an anti-C1qRp monoclonal antibody. These observations suggest that adiponectin is an important negative regulator in hematopoiesis and immune systems and raise the possibility that it may be involved in ending inflammatory responses through its inhibitory functions.
Collapse
|