1
|
Khong D, Li M, Singleton A, Chin LY, Parekkadan B. Stromalized microreactor supports murine hematopoietic progenitor enrichment. Biomed Microdevices 2018; 20:13. [PMID: 29353324 DOI: 10.1007/s10544-017-0255-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
There is an emerging need to process, expand, and even genetically engineer hematopoietic stem and progenitor cells (HSPCs) prior to administration for blood reconstitution therapy. A closed-system and automated solution for ex vivo HSC processing can improve adoption and standardize processing techniques. Here, we report a recirculating flow bioreactor where HSCs are stabilized and enriched for short-term processing by indirect fibroblast feeder coculture. Mouse 3 T3 fibroblasts were seeded on the extraluminal membrane surface of a hollow fiber micro-bioreactor and were found to support HSPC cell number compared to unsupported BMCs. CFSE analysis indicates that 3 T3-support was essential for the enhanced intrinsic cell cycling of HSPCs. This enhanced support was specific to the HSPC population with little to no effect seen with the Lineagepositive and Lineagenegative cells. Together, these data suggest that stromal-seeded hollow fiber micro-reactors represent a platform to screening various conditions that support the expansion and bioprocessing of HSPCs ex vivo.
Collapse
Affiliation(s)
- Danika Khong
- Department of Surgery, Center for Surgery, Innovation, & Bioengineering, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Matthew Li
- Department of Surgery, Center for Surgery, Innovation, & Bioengineering, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Amy Singleton
- Department of Surgery, Center for Surgery, Innovation, & Bioengineering, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Ling-Yee Chin
- Department of Surgery, Center for Surgery, Innovation, & Bioengineering, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Biju Parekkadan
- Department of Surgery, Center for Surgery, Innovation, & Bioengineering, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospitals for Children, Boston, MA, 02114, USA. .,Department of Biomedical Engineering, Rutgers University and the Department of Medicine, Rutgers Biomedical and Health Sciences, Piscataway, NJ, 08854, USA. .,Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
2
|
Xu Z, Xiong D, Zhang J, Zhang J, Chen X, Chen Z, Zhan R. Bone marrow stromal cells enhance the survival of chronic lymphocytic leukemia cells by regulating HES-1 gene expression and H3K27me3 demethylation. Oncol Lett 2017; 15:1937-1942. [PMID: 29434893 DOI: 10.3892/ol.2017.7450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/09/2017] [Indexed: 11/05/2022] Open
Abstract
The majority of patients with chronic lymphocytic leukemia (CLL) are not cured by traditional chemotherapy. One possible explanation for this is that the microenvironment protects CLL cells from both spontaneous- and cytotoxic-mediated apoptosis. The present study was designed to investigate the mechanisms accounting for these effects, since this information is crucial to understanding CLL physiopathology and identifying potential treatment targets. The CLL cell line L1210 and primary CLL cells were cultured under different conditions: With serum, cyclophosphamide (CTX), or with monolayers and conditioned medium (CM) from the stromal cell line HESS-5. Apoptosis, Hes family BHLH transcription factor 1 (HES-1) gene and protein expression, and histone H3K27me3 DNA demethylation were determined. Co-culture of L1210 cells with HESS-5 cells significantly inhibited serum deprivation- and CTX-induced apoptosis of leukemia cells, and resulted in a significant increase in short-term proliferation. Soluble factors in the CM from HESS-5 cells had a negligible effect. The HESS-5 cell-mediated inhibition of apoptosis of CLL cells was associated with increased HES-1 expression and hypomethylation of the H3K27me3 gene in the leukemia cells. These results indicate that stromal cells enhance the survival of CLL cells by regulating the HES-1 gene and protein expression, as well as H3K27me3 DNA demethylation, and suggest that specific interactions between stromal and leukemia cells may enhance the resistance of leukemia cells to chemotherapy.
Collapse
Affiliation(s)
- Zhenshu Xu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Donglian Xiong
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jushun Zhang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jingyan Zhang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xiuli Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Zhizhe Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Rong Zhan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
3
|
Koller MR, Palsson BO. Review: tissue engineering: reconstitution of human hematopoiesis ex vivo. Biotechnol Bioeng 2010; 42:909-30. [PMID: 18613140 DOI: 10.1002/bit.260420802] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The reconstruction of functioning human tissues ex vivo is becoming an important part of biotechnology. There are compelling scientific, clinical, and biotechnological reasons for fully or partially reconstituting human tissues such as skin, bone marrow, and liver ex vivo. In particular, bone marrow is a tissue of much importance, and there are significant societal and health benefits derived from a successfully constructed ex vivo hematopoietic system. In this article, we review the current status of this effort. The topics covered include the current understanding of the biology of human hematopoiesis, the motivation for reconstructing it ex vivo, the current state of ex vivo human hematopoietic cultures, the development of important metrics to judge culture performance, and an approach based on in vivo mimetics to accomplish this goal. We discuss some applications of functional ex vivo hematopoietic cultures and the biological and engineering challenges that face research in this area.
Collapse
Affiliation(s)
- M R Koller
- Aastrom Biosciences, Inc., Ann Arbor, Michigan 48106, USA
| | | |
Collapse
|
4
|
Tung S, Parmar S, Robinson S, De Lima M, Shpall E. Ex vivo expansion of umbilical cord blood for transplantation. Best Pract Res Clin Haematol 2010; 23:245-57. [DOI: 10.1016/j.beha.2010.06.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
5
|
Abstract
A marked increase in the utilization of umbilical cord blood (UCB) transplantation has been observed in recent years; however, the use of UCB as a hematopoietic stem cell (HSC) source is limited primarily by the number of progenitor cells contained in the graft. Graft failure, delayed engraftment and profound delay in immune reconstitution lead to significant morbidity and mortality in adults. The lack of cells available for post transplant therapies, such as donor lymphocyte infusions, has also been considered to be a disadvantage of UCB. To improve outcomes and extend applicability of UCB transplantation, one potential solution is ex vivo expansion of UCB. Investigators have used several methods, including liquid suspension culture with various cytokines and expansion factors, co-culture with stromal elements and continuous perfusion systems. Techniques combining ex vivo expanded and unmanipulated UCB are being explored to optimize the initial engraftment kinetics as well as the long-term durability. The optimal expansion conditions are still not known; however, recent studies suggest that expanded UCB is safe. It is hoped that by ex vivo expansion of UCB, a resulting decrease in the morbidity and mortality of UCB transplantation will be observed, and that the availability of additional cells may allow adoptive immunotherapy or gene transfer therapies in the UCB setting.
Collapse
Affiliation(s)
- S S Kelly
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA.
| | | | | | | |
Collapse
|
6
|
Dexter TM, Coutinho LH, Spooncer E, Heyworth CM, Daniel CP, Schiro R, Chang J, Allen TD. Stromal cells in haemopoiesis. CIBA FOUNDATION SYMPOSIUM 2007; 148:76-86; discussion 86-95. [PMID: 2180651 DOI: 10.1002/9780470513880.ch6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Stromal cells of the bone marrow can provide the growth-promoting and differentiation-inducing molecules which are necessary for haemopoiesis. While the nature of these stimuli is largely unknown, the development of haemopoietic cells in association with stromal cells requires intimate cell contact. Molecules of the extracellular matrix, such as heparan sulphate, are able to bind growth factors and in this way the stromal cells may form microenvironmental niches which preferentially promote development of multipotent and committed cells along discrete lineages. Cells from some patients with acute and chronic myeloid and lymphoid leukaemias are defective in their ability to interact with stromal cells and consequently cannot survive in stromal cell-mediated long-term marrow cultures. We have exploited this phenomenon to obtain normal haemopoietic cells from patients with leukaemia, and to use these cells for successful autografting in patients with acute and chronic myeloid leukaemias.
Collapse
Affiliation(s)
- T M Dexter
- Department of Experimental Haematology, Paterson Institute for Cancer Research, Withington, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Robinson S, Niu T, de Lima M, Ng J, Yang H, McMannis J, Karandish S, Sadeghi T, Fu P, del Angel M, O'Connor S, Champlin R, Shpall E. Ex vivo expansion of umbilical cord blood. Cytotherapy 2006; 7:243-50. [PMID: 16081350 DOI: 10.1080/14653240510027172] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The efficacy of cord blood (CB) transplantation is limited by the low cell dose available. Low cell doses at transplant are correlated with delayed engraftment, prolonged neutropenia and thrombocytopenia and elevated risk of graft failure. To potentially improve the efficacy of CB transplantation, approaches have been taken to increase the cell dose available. One approach is the transplantation of multiple cord units, another the use of ex vivo expansion. Evidence for a functional and phenotypic heterogeneity exists within the HSC population and one concern associated with ex vivo expansion is that the expansion of lower 'quality' hematopoietic progenitor cells (HPC) occurs at the expense of higher 'quality' HPC, thereby impacting the reserve of the graft. There is evidence that this is a valid concern while other evidence suggests that higher quality HPC are preserved and not exhausted. Currently, ex vivo expansion processes include: (1) liquid expansion: CD34+ or CD133+ cells are selected and cultured in medium containing factors targeting the proliferation and self-renewal of primitive hematopoietic progenitors; (2) co-culture expansion: unmanipulated CB cells are cultured with stromal components of the hematopoietic microenvironment, specifically mesenchymal stem cells (MSC), in medium containing growth factors; and (3) continuous perfusion: CB HPC are cultured with growth factors in 'bioreactors' rather than in static cultures. These approaches are discussed. Ultimately, the goal of ex vivo expansion is to increase the available dose of the CB cells responsible for successful engraftment, thereby reducing the time to engraftment and reducing the risk of graft failure.
Collapse
Affiliation(s)
- S Robinson
- University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Wang L, Fortney JE, Gibson LF. Stromal cell protection of B-lineage acute lymphoblastic leukemic cells during chemotherapy requires active Akt. Leuk Res 2004; 28:733-42. [PMID: 15158095 DOI: 10.1016/j.leukres.2003.10.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2003] [Accepted: 10/29/2003] [Indexed: 10/26/2022]
Abstract
Several studies document ALL cell response to survival signals from bone marrow stromal cells. The current study suggests a requirement for active Akt in ALL cells for optimal stromal cell protection during chemotherapy. ALL cells expressing dominant negative Akt were not efficiently rescued from Ara-C or etoposide-induced apoptosis by stromal cell co-culture. In addition, inhibition of ALL cell PI-3 kinase activity diminished stromal cells support of tumor cells during treatment. ALL cell lines co-cultured with bone marrow stromal cells during chemotherapy maintained higher levels of phosphorylated Akt protein and reduced PP2A activity when compared to ALL cells treated in medium alone. Chemotherapy-induced PARP and Bcl-2 cleavage was reduced in ALL cells cultured with a stromal cell layer compared to tumor cells exposed to drug in medium alone. However, interaction with stromal cells was not able to efficiently block treatment-induced PARP or Bcl-2 cleavage in leukemic cells with blunted Akt activity. These data suggest a pivotal role for Akt in mediating stromal cell regulation of ALL cell apoptosis.
Collapse
Affiliation(s)
- Lin Wang
- Department of Pediatrics, West Virginia University School of Medicine, Morgantown, WV, USA
| | | | | |
Collapse
|
9
|
Dormady SP, Zhang XM, Basch RS. Hematopoietic progenitor cells grow on 3T3 fibroblast monolayers that overexpress growth arrest-specific gene-6 (GAS6). Proc Natl Acad Sci U S A 2000; 97:12260-5. [PMID: 11050245 PMCID: PMC17329 DOI: 10.1073/pnas.97.22.12260] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pluripotential hematopoietic stem cells grow in close association with bone marrow stromal cells, which play a critical role in sustaining hematopoiesis in long-term bone marrow cultures. The mechanisms through which stromal cells act to support pluripotential hematopoietic stem cells are largely unknown. This study demonstrates that growth arrest-specific gene-6 (GAS6) plays an important role in this process. GAS6 is a ligand for the Axl (Ufo/Ark), Sky (Dtk/Tyro3/Rse/Brt/Tif), and Mer (Eyk) family of tyrosine kinase receptors and binds to these receptors via tandem G domains at its C terminus. After translation, GAS6 moves to the lumen of the endoplasmic reticulum, where it is extensively gamma-carboxylated. The carboxylation process is vitamin K dependent, and current evidence suggests that GAS6 must be gamma-carboxylated to bind and activate any of the cognate tyrosine kinase receptors. Here, we show that expression of GAS6 is highly correlated with the capacity of bone marrow stromal cells to support hematopoiesis in culture. Nonsupportive stromal cell lines express little to no GAS6, whereas supportive cell lines express high levels of GAS6. Transfection of the cDNA encoding GAS6 into 3T3 fibroblasts is sufficient to render this previously nonsupportive cell line capable of supporting long-term hematopoietic cultures. 3T3 cells, genetically engineered to stably express GAS6 (GAS6-3T3), produce a stromal layer that supports the generation of colony-forming units in culture (CFU-c) for up to 6 wk. Hematopoietic support by genetically engineered 3T3 is not vitamin K dependent, and soluble recombinant GAS6 does not substitute for coculturing the hematopoietic progenitors with genetically modified 3T3 cells.
Collapse
Affiliation(s)
- S P Dormady
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | | | | |
Collapse
|
10
|
Qiang YW, Kitagawa M, Higashi M, Ishii G, Morimoto C, Harigaya K. Activation of mitogen-activated protein kinase through alpha5/beta1 integrin is required for cell cycle progression of B progenitor cell line, Reh, on human marrow stromal cells. Exp Hematol 2000; 28:1147-57. [PMID: 11027833 DOI: 10.1016/s0301-472x(00)00521-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Attachment to bone marrow (BM) stromal cells is crucial for the normal growth and development of B-cell progenitors (pro-B). However, the molecular mechanisms by which contact facilitates the proliferation of pro-B cells are not completely understood. This study was performed to investigate this interaction. MATERIALS AND METHODS A model pro-B cell line (Reh) and a human BM stromal cell line (KM102) were used. Flow cytomery was used for cell cycle analysis. Western Blotting and immunoprecipitation were utilized to examine the levels of cyclin-dependent kinase (cdk) and p27(Kip1). RESULTS Attachment to both KM102 and normal BM stromal cells significantly promoted the growth of Reh cells. Pretreatment of Reh cells with anti-integrin beta1 or alpha5 monoclonal antibody (mAb), but not alpha4 or ICAM-1 mAb, abrogated this enhancement of proliferation. Furthermore, stroma attachment resulted in shortening of the G(1) phase of cell cycle, significant increases cdk2 activity, degradation of cdk inhibitor p27-GST protein, and decrease in levels of p27(Kip1) protein. In addition, solid-phase cross-linking of alpha5 via immobilized antibody also resulted in extracellular signal-regulated (ERK)-2 kinase phosphorylation, increase in cdk2 activity, decrease in levels of p27(Kip1) protein, and enhanced proliferation that was inhibited by treatment with PD98059, a specific ERK inhibitor. CONCLUSION Integrin alpha5beta1-mediated stroma contact promotes the proliferation of B-cell progenitors through the activation of ERK-2, which in turn modulates cell cycle regulation machinery including induction of cdk2 activity and degradation of p27(Kip1) and contributing to acceleration of the G(1) phase of cell cycle progression.
Collapse
Affiliation(s)
- Y W Qiang
- Division of Molecular and Tumor Pathology, Department of Pathology, Chiba University School of Medicine, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
Abstract
Approximately 20% of B-lineage acute lymphoblastic leukemias are not cured by traditional chemotherapy. The possibility was examined that residual leukemic cells that potentially contribute to relapse are harbored in association with fibroblastic stromal cells in the bone marrow. Modulation of cytarabine (Ara-C) and etoposide (VP-16) efficacy by bone marrow stromal cells in vitro was investigated. Stromal cell coculture was shown to sustain the proliferation of B-lineage leukemic cells and to reduce leukemic cell apoptosis when exposed to Ara-C or VP-16. Direct contact with stromal cells was essential for the protection of leukemic cells during chemotherapy, whereas soluble factors had negligible effect. Specifically, signaling mediated through interaction with the stromal cell adhesion molecule VCAM-1 was required to maintain the maximum viability of leukemic cells during Ara-C and VP-16 exposure. In contrast, the interaction of leukemic cells with fibronectin did not confer significant resistance to either chemotherapeutic agent. These observations suggest a role for the bone marrow microenvironment in modulating the response of B-lineage leukemic cells to Ara-C or VP-16, and they indicate specific molecular interactions that may be important in determining the sensitivity of leukemic cells to treatment.
Collapse
|
12
|
Abstract
Approximately 20% of B-lineage acute lymphoblastic leukemias are not cured by traditional chemotherapy. The possibility was examined that residual leukemic cells that potentially contribute to relapse are harbored in association with fibroblastic stromal cells in the bone marrow. Modulation of cytarabine (Ara-C) and etoposide (VP-16) efficacy by bone marrow stromal cells in vitro was investigated. Stromal cell coculture was shown to sustain the proliferation of B-lineage leukemic cells and to reduce leukemic cell apoptosis when exposed to Ara-C or VP-16. Direct contact with stromal cells was essential for the protection of leukemic cells during chemotherapy, whereas soluble factors had negligible effect. Specifically, signaling mediated through interaction with the stromal cell adhesion molecule VCAM-1 was required to maintain the maximum viability of leukemic cells during Ara-C and VP-16 exposure. In contrast, the interaction of leukemic cells with fibronectin did not confer significant resistance to either chemotherapeutic agent. These observations suggest a role for the bone marrow microenvironment in modulating the response of B-lineage leukemic cells to Ara-C or VP-16, and they indicate specific molecular interactions that may be important in determining the sensitivity of leukemic cells to treatment.
Collapse
|
13
|
The Use of Granulocyte Colony-Stimulating Factor During Retroviral Transduction on Fibronectin Fragment CH-296 Enhances Gene Transfer Into Hematopoietic Repopulating Cells in Dogs. Blood 1999. [DOI: 10.1182/blood.v94.7.2287.419k29_2287_2292] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A competitive repopulation assay in the dog was used to develop improved gene transfer protocols for hematopoietic stem cell gene therapy. Using this assay, we previously showed improved gene transfer into canine hematopoietic repopulating cells when CD34-enriched marrow cells were cocultivated on gibbon ape leukemia virus (GALV)–based retrovirus vector-producing cells. In the present study, we have investigated the use of fibronectin fragment CH-296 and 2 growth factor combinations to further improve gene transfer efficiency. CD34-enriched marrow cells from each dog were prestimulated for 24 hours and then divided into 3 equal fractions. Two fractions were placed into flasks coated with either CH-296 or bovine serum albumin (BSA) and virus-containing medium supplemented with growth factors, and protamine sulfate was replaced 4 times over a 48-hour period. One fraction was cocultivated on irradiated PG13 (GALV-pseudotype) packaging cells for 48 hours. In 2 animals, cells of the different fractions were transduced in the presence of human FLT-3 ligand (FLT3L), canine stem cell factor (cSCF), and human megakaryocyte growth and development factor (MGDF), and in 2 other dogs, transduction was performed in the presence of FLT3L, cSCF, and canine granulocyte-colony stimulating factor (cG-CSF). The vectors used contained small sequence differences, allowing differentiation of cells genetically marked by the different vectors. After transduction, nonadherent and adherent cells from all 3 fractions were pooled and infused into lethally irradiated dogs. Polymerase chain reaction and Southern blot analysis were used to determine the persistence of the transferred vectors in the peripheral blood and marrow cells after transplantation. The highest levels of gene transfer were obtained when cells were transduced in the presence of FLT3L, cSCF, and cG-CSF (gene transfer levels of more than 10% for more than 8 months so far). Compared with the 2 animals that received cells transduced with FLT3L, cSCF, and MGDF, gene transfer levels were significantly higher when dogs received cells that were transduced in the presence of cG-CSF. Transduction on CH-296 resulted in gene transfer levels that were at least as high as transduction by cocultivation. In summary, the overall levels of gene transfer obtained with these conditions should be sufficiently high to allow stem cell gene therapy studies aimed at correcting genetic diseases in dogs as a model for human gene therapy.
Collapse
|
14
|
Abstract
The precursors of bone, cartilage, fat and muscle cells are likely to be derived from more primitive mesenchymal cells which exhibit some of the characteristics of stem cells. Despite extensive study of stromal cell differentiation, neither mesenchymal stem cells or the more committed, tissue-specific progenitors have been well characterized. Here we describe the use of flow cytometry to isolate from fetal rat periosteum a population of small, relatively agranular cells (S cells) that display stem cell characteristics. After plating, S cells demonstrated extensive self-renewal with osteogenic potential. Electron microscopy showed that S cells have high nuclear:cytoplasmic ratios with large condensed nuclei and a paucity of cytoplasmic organelles. Freshly sorted suspensions of immunocytochemically stained S cells did not express differentiation-associated markers such as type I, II, and III collagens, alkaline phosphatase or osteopontin. However following attachment, S cells became immunopositive for collagens I, II, III, osteopontin and also for the cell surface receptor CD44, which mediates cell attachment to hyaluronan and osteopontin. S-cells showed two discrete populations of surface-stained protein by sulforhodamine, wheat germ agglutinin and Thy-1. In contrast, large (L) cells that did not exhibit stem cell characteristics exhibited low staining levels for Thy-1 and for wheat germ agglutinin. These studies demonstrate that viable osteogenic precursor cells with the stem cell characteristics of self-renewal, high proliferative capacity and multipotentiality can be enriched from heterogeneous stromal cell populations with simple flow cytometric methods.
Collapse
Affiliation(s)
- R Ghilzon
- MRC Group in Periodontal Physiology, Faculty of Dentistry University of Toronto, Ontario, Canada
| | | | | |
Collapse
|
15
|
Mutual Education Between Hematopoietic Cells and Bone Marrow Stromal Cells Through Direct Cell-to-Cell Contact: Factors That Determine the Growth of Bone Marrow Stroma-Dependent Leukemic (HB-1) Cells. Blood 1998. [DOI: 10.1182/blood.v92.3.834] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
A stroma-dependent cell line (HB-1) was established from myelogenous leukemic cells of CBA/N mouse. Characterization of the cells showed that HB-1 proliferated on hematopoietic supportive stromal cells (MS-10), but did not survive or proliferate on hematopoietic nonsupportive cells (MS-K). Direct contact between HB-1 and MS-10 appears to be necessary for HB-1 to proliferate on MS-10. We found that interleukin-1α (IL-1α) produced by MS-10 plays a major role in the survival and proliferation of HB-1. IL-11 did not support the proliferation of HB-1 cells by itself, but enhanced the proliferation of HB-1 cells in the presence of IL-1α. The expression of IL-1α and IL-11 was induced in MS-10 by the direct contact with HB-1 cells, and the expression of IL-1 receptor type I (IL-1RI) and interleukin-11 receptor (IL-11R) was induced in HB-1 cells by the attachment of the cells to MS-10. These findings show the existence of two-way interactions between HB-1 and MS-10.
© 1998 by The American Society of Hematology.
Collapse
|
16
|
Mutual Education Between Hematopoietic Cells and Bone Marrow Stromal Cells Through Direct Cell-to-Cell Contact: Factors That Determine the Growth of Bone Marrow Stroma-Dependent Leukemic (HB-1) Cells. Blood 1998. [DOI: 10.1182/blood.v92.3.834.415k04_834_841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A stroma-dependent cell line (HB-1) was established from myelogenous leukemic cells of CBA/N mouse. Characterization of the cells showed that HB-1 proliferated on hematopoietic supportive stromal cells (MS-10), but did not survive or proliferate on hematopoietic nonsupportive cells (MS-K). Direct contact between HB-1 and MS-10 appears to be necessary for HB-1 to proliferate on MS-10. We found that interleukin-1α (IL-1α) produced by MS-10 plays a major role in the survival and proliferation of HB-1. IL-11 did not support the proliferation of HB-1 cells by itself, but enhanced the proliferation of HB-1 cells in the presence of IL-1α. The expression of IL-1α and IL-11 was induced in MS-10 by the direct contact with HB-1 cells, and the expression of IL-1 receptor type I (IL-1RI) and interleukin-11 receptor (IL-11R) was induced in HB-1 cells by the attachment of the cells to MS-10. These findings show the existence of two-way interactions between HB-1 and MS-10.© 1998 by The American Society of Hematology.
Collapse
|
17
|
Abstract
Abstract
The cellular and molecular mechanisms that regulate the most primitive hematopoietic stem cell are not well understood. We have undertaken a systematic dissection of the complex hematopoietic microenvironment to define some of these mechanisms. An extensive panel of immortalized stromal cell lines from murine fetal liver were established and characterized. Collectively, these cell lines display extensive heterogeneity in their in vitro hematopoietic supportive capacity. In the current studies, we describe a long-term in vitro culture system using a single stromal cell clone (AFT024) that qualitatively and quantitatively supports transplantable stem cell activity present in highly purified populations. We show multilineage reconstitution in mice that received the equivalent of as few as 100 purified bone marrow and fetal liver stem cells cultured for 4 to 7 weeks on AFT024. The cultured stem cells meet all functional criteria currently ascribed to the most primitive stem cell population. The levels of stem cell activity present after 5 weeks of coculture with AFT024 far exceed those present in short-term cytokine-supported cultures. In addition, maintenance of input levels of transplantable stem cell activity is accompanied by expansion of other classes of stem/progenitor cells. This suggests that the stem/progenitor cell population is actively proliferating in culture and that the AFT024 cell line provides a milieu that stimulates progenitor cell proliferation while maintaining in vivo repopulating activity.
Collapse
|
18
|
Moore KA, Pytowski B, Witte L, Hicklin D, Lemischka IR. Hematopoietic activity of a stromal cell transmembrane protein containing epidermal growth factor-like repeat motifs. Proc Natl Acad Sci U S A 1997; 94:4011-6. [PMID: 9108096 PMCID: PMC20559 DOI: 10.1073/pnas.94.8.4011] [Citation(s) in RCA: 166] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Primitive hematopoietic stem cells are closely associated with discrete in vivo microenvironments. These "niches" are thought to provide the molecular signals that mediate stem cell differentiation and self-renewal. We have dissected the fetal liver microenvironment into distinct cellular components by establishing an extensive panel of stromal cell lines. One particular cell line maintains repopulating stem cells for prolonged in vitro culture periods. A subtraction cloning strategy has yielded a cDNA that encodes a cell surface glycoprotein with a restricted pattern of expression among stromal cell lines. This molecule, previously identified as delta-like/preadipocyte factor-1, contains epidermal growth factor-like repeats that are related to those in the notch/delta/serrate family of proteins. We have investigated the potential role of this molecule in hematopoietic stem/progenitor cell regulation. We show that the delta-like protein displays activity on purified stem cells by promoting the formation of "cobblestone areas" of proliferation. These cobblestone areas contain both primitive high-proliferative potential progenitors and in vivo repopulating stem cells.
Collapse
Affiliation(s)
- K A Moore
- Department of Molecular Biology, Princeton University, NJ 08544, USA.
| | | | | | | | | |
Collapse
|
19
|
Affiliation(s)
- R S Selvan
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
20
|
Ogasawara H, Tsuji T, Hirano D, Aoki Y, Nakamura M, Kodama H. Induction of IL-6 production by bone marrow stromal cells on the adhesion of IL-6-dependent hematopoietic cells. J Cell Physiol 1996; 169:209-16. [PMID: 8841437 DOI: 10.1002/(sici)1097-4652(199610)169:1<209::aid-jcp21>3.0.co;2-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cellular interactions between hematopoietic cells and stromal cells play crucial roles in the proliferation and differentiation of the hematopoietic cells. Interleukin-6 (IL-6)-dependent 7TD1 cells markedly proliferated without IL-6 when they were co-cultured with hematopoietic-supportive bone marrow stromal cells, HESS-5 cells and HESS-1 CL.3 cells, which can support long-term hematopoiesis in vitro with but not without direct cell contact, cell contact being prevented with a microporous membrane. The production of IL-6 and the amount of IL-6 mRNA in hematopoietic-supportive stromal cells but not 7TD1 cells significantly increased only when the stromal cells were co-cultured in direct contact with 7TD1 cells. Furthermore, the amount of IL-6 mRNA increased according to the number of 7TD1 cells co-cultured. These inductions were not observed on co-culture with a murine myeloid cell line, M1 cells, or on the addition of the co-culture supernatant. These results suggest that 7TD1 cells transmit the signal to stromal cells that enhances IL-6 production by stromal cells via direct cell contact. A certain specific molecule for transduction of the signals may exist on the surface membrane of stromal cells and hematopoietic cells.
Collapse
Affiliation(s)
- H Ogasawara
- Life Science Research Laboratory, JT Inc, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
Intimate contact between haemopoietic progenitor cells and elements of the bone marrow stroma is required for progenitor cell proliferation and differentiation. It is believed that the stroma provides particular niches for the development of haemopoietic cells of different lineages. Cytokines, stromal cell surface molecules and molecules of the stromal extracellular matrix all contribute to defining these microenvironmental niches. Data obtained using an in vitro model of haemopoiesis support the view that progenitor cell adhesion to stroma is mediated by multiple receptor-ligand interactions. The possibility of a tethering step, mediated by the engagement of stromal cell heparan sulphate with its ligands on the progenitor cells, preceding stable cell adhesion is discussed. The role of stromal cell heparan sulphate is likely to include cytokine presentation to progenitors as well as the tethering of progenitors to stroma. It is proposed that intracellular signals induced by progenitor cell adhesion to stroma act in association with cytokine induced signals to regulate progenitor cell proliferation and differentiation.
Collapse
Affiliation(s)
- D R Coombe
- TVW Telethon Institute for Child Health Research, West Perth, Western Australia
| |
Collapse
|
22
|
Abstract
Interactions between stromal cells and thymocytes play a crucial role in T cell development. The thymic stroma is complex and consists of epithelial cells derived from the pharyngeal region during development, together with macrophages and dendritic cells of bone marrow origin. In addition, fibroblasts and matrix molecules permeate the whole framework. It is now apparent that these individual stromal components play specialized roles at different stages of T cell differentiation. Thus, at the early CD4-8- stage of development, T cell precursors require fibroblast as well as epithelial cell interactions. Later, at the CD4+8+ stage, as well as providing low avidity TCR/MHC-peptide interactions, thymic epithelial cells have been shown to possess unique properties essential for positive selection. Dendritic cells, on the other hand, are probably efficient mediators of negative selection, but they may not be solely responsible for this activity. Alongside the functional roles of stromal cells, considerable progress is being made in unraveling the nature of the signaling pathways involved in T cell development. Identification of the pre-T cell receptor (pre-TCR) and associated signaling molecules marks an important advance in understanding the mechanisms that control gene rearrangement and allelic exclusion. In addition, a better understanding of the signaling pathways that lead to positive selection on the one hand and negative selection on the other is beginning to emerge. Many issues remain unresolved, and some are discussed in this review. What, for example, is the nature of the chemotactic factor(s) that attract stem cells to the thymus? What is the molecular basis of the essential interactions between early thymocytes and fibroblasts, and early thymocytes and epithelial cells? What is special about cortical epithelial cells in supporting positive selection? These and other issues are ripe for analysis and can now be approached using a combination of modern molecular and cellular techniques.
Collapse
Affiliation(s)
- G Anderson
- Department of Anatomy, Medical School, University of Birmingham, United Kingdom
| | | | | | | |
Collapse
|
23
|
Hurley RW, McCarthy JB, Verfaillie CM. Direct adhesion to bone marrow stroma via fibronectin receptors inhibits hematopoietic progenitor proliferation. J Clin Invest 1995; 96:511-9. [PMID: 7542285 PMCID: PMC185225 DOI: 10.1172/jci118063] [Citation(s) in RCA: 174] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
In long-term bone marrow cultures, stroma-adherent progenitors proliferate significantly less than nonadherent progenitors. Thus, close progenitor-stroma interactions may serve to regulate or restrict rather than promote hematopoietic progenitor proliferation. We hypothesized that signaling through adhesion receptors on hematopoietic cells may contribute to the inhibition of proliferation observed when progenitors are in contact with stroma. We demonstrate that progenitors cultured physically separated from stroma in a transwell proliferate significantly more than progenitors adherent to stroma. Furthermore, proliferation of colony forming cells (CFC) is reduced after specific adhesion to stroma, metabolically inactivated glutaraldehyde-fixed stroma, stromal-extracellular matrix, or the COOH-terminal heparin-binding domain of fibronectin. Nonspecific adhesion to poly-L-lysine fails to inhibit CFC proliferation. That the VLA-4 integrin is one of the receptors that transfers proliferation inhibitory signals was shown using blocking anti-alpha 4 monomeric F(ab) fragments. Furthermore, when synthetic peptides representing specific cell attachment sites within the heparin-binding domain of fibronectin were added to Dexter-type marrow cultures, significantly increased recovery and proliferation of CFC was observed, suggesting that these peptides disrupt adhesion-mediated proliferation inhibitory events. Thus, negative regulation of hematopoiesis may not only depend on the action of growth inhibitory cytokines but also on growth inhibitory signals resulting from direct adhesive interactions between progenitors and marrow stroma.
Collapse
Affiliation(s)
- R W Hurley
- Department of Medicine, University of Minnesota, Minneapolis 55455, USA
| | | | | |
Collapse
|
24
|
Just U, Kan O, Fennelly J, Dexter TM, Spooncer E. The Hox-2.4 gene is not involved in the generation of IL-3 dependent multipotent FDCP-mix cell lines. Growth Factors 1995; 12:173-7. [PMID: 8619923 DOI: 10.3109/08977199509036877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The establishment of IL-3-dependent multipotent progenitor cell lines from Hox-2.4-expressing bone marrow cells suggests that homeobox genes may contribute to immortalization of early myeloid cells. A survey of 20 independently derived multipotent IL-3-dependent cell lines established from either src-virus-infected long-term bone marrow cultures (FDCP-mix) or Multi-CSF-virus (M3MuV)-infected bone marrow revealed that Hox-2.4 was not expressed in any of these cell lines. In addition DNA rearrangements were not observed. We conclude that activation of Hox-2.4 is not an obligatory event in the immortalization of early myeloid cells.
Collapse
Affiliation(s)
- U Just
- Department of Experimental Haematology, Paterson Institute for Cancer Research, Manchester, UK
| | | | | | | | | |
Collapse
|
25
|
Takashita E, Sugimoto K, Adachi Y, Mori KJ. Induction of bcl-2 gene expression by intercellular information from hemopoietic supportive stromal cells to DA-1 cells. J Cell Physiol 1994; 161:367-73. [PMID: 7962121 DOI: 10.1002/jcp.1041610222] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
When interleukin-3 (IL-3) dependent DA-1 cells were cultured on hemopoietic supportive stromal cells (MS-5), DA-1 cells survived and proliferated in the absence of detectable IL-3. Although IL-3 was not produced by the MS-5 cells, their production of granulocyte-macrophage colony-stimulating factor (GM-CSF) was increased when they were co-cultured with DA-1 cells. This suggests that DA-1 cells transmit signals to stromal cells that enhance growth factor(s) production. Expression of bcl-2 by DA-1 cells was induced when they were co-cultured with MS-5 cells, suggesting that DA-1 cells express bcl-2 strongly in response to a signal produced by MS-5 cells. These data indicate the existence of a two-way interaction between DA-1 cells and hemopoietic supportive stromal cells.
Collapse
Affiliation(s)
- E Takashita
- Department of Biology, Faculty of Science, Niigata University, Japan
| | | | | | | |
Collapse
|
26
|
Abstract
The factors that induce proliferation of the human hematopoietic stem cell are ill defined. Further characterization of such growth factors will be needed to develop ex vivo culture systems that induce prolonged proliferation and expansion of human hematopoietic stem cells. Human or murine hematopoietic progenitors that can initiate and sustain long-term culture systems (LTC-IC) represent a population of very primitive hematopoietic progenitors. When cultured in direct contact with stromal layers, we and others have demonstrated that a fraction of such LTC-IC can be maintained. In addition, stroma-free long-term cultures supplemented with two to nine cytokines can induce proliferation and differentiation of immature human hematopoietic progenitors. However, 70-90% of primitive LTC-IC are lost after five weeks in such cultures. We describe a "stroma-non-contact" culture system, in which progenitors are cultured separated from stroma by a 0.4 micron microporous membrane which prevents cell stroma contact but allows free passage of diffusible factors. Primitive progenitors in such cultures can not only differentiate into committed progenitors but also are maintained to a greater extent than in Dexter cultures. We will discuss the relative contribution of 1) direct contact between hematopoietic progenitors and bone marrow stroma, 2) soluble stroma-derived factors and 3) previously characterized growth promoting and presumed growth inhibitory cytokines in the in vitro maintenance and potential expansion of LTC-IC.
Collapse
Affiliation(s)
- C M Verfaillie
- Division of Hematology, University of Minnesota, Minneapolis 55455
| |
Collapse
|
27
|
Benya RV, Duncan MD, Mishra L, Bass BL, Voyles NR, Korman LY. Extracellular matrix composition influences insulinlike growth factor I receptor expression in rat IEC-18 cells. Gastroenterology 1993; 104:1705-11. [PMID: 8500730 DOI: 10.1016/0016-5085(93)90649-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The composition of the extracellular matrix (ECM) as well as insulinlike growth factor I (IGF-I) receptor density vary along the crypt-villus axis. We determined whether components of the ECM influence IGF-I receptor expression in IEC-18 rat small intestine crypt cells. METHODS IEC-18 cells were cultured on plastic, collagen type IV, Matrigel, and laminin at the plateau and proliferative growth phases. Receptor affinity (Kd) and number (Bmax) were determined by competitive binding of 125I-IGF-I in the presence of increasing concentrations of unlabeled IGF-I. Receptor isolation was performed by affinity cross linking. Messenger RNA (mRNA) for IGF-I receptor was quantified by Northern analysis. RESULTS Specific binding of IGF-I > IGF-II > insulin was observed. A 130,000-molecular weight protein was identified by cross-linking, consistent with the alpha subunit of the IGF-I receptor. Scatchard analysis revealed no effect of ECM on IGF-I binding affinity. In contrast, the Bmax was 18% lower for plateau-phase cells cultured on Matrigel vs. plastic and was 42% lower for cells cultured on laminin vs. collagen type IV. The Bmax for proliferative growth phase cells was decreased when cultured on Matrigel vs. plastic and was 10-fold less than for cells cultured at the plateau growth phase. Northern analysis revealed that IEC-18 cells cultured on Matrigel had less mRNA for IGF-I receptor than cells cultured on plastic. CONCLUSIONS The rate of cell proliferation and the composition of the ECM influence IGF-I receptor expression in IEC-18 cells.
Collapse
Affiliation(s)
- R V Benya
- Research Service, Veterans Administration Medical Center, Washington, D.C
| | | | | | | | | | | |
Collapse
|
28
|
Alvarez-Silva M, da Silva LC, Borojevic R. Cell membrane-associated proteoglycans mediate extramedullar myeloid proliferation in granulomatous inflammatory reactions to schistosome eggs. J Cell Sci 1993; 104 ( Pt 2):477-84. [PMID: 8505374 DOI: 10.1242/jcs.104.2.477] [Citation(s) in RCA: 28] [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
In chronic murine schistosomiasis, extramedullar myelopoiesis was observed, with proliferation of myeloid cells in liver parenchyma and in periovular granulomas. We have studied the question of whether cells obtained from granulomatous connective tissue may act as myelopoietic stroma, supporting long-term myeloid proliferation. Primary cell lines (GR) were obtained in vitro from periovular granulomas, induced in mouse livers by Schistosoma mansoni infection. These cells were characterized as myofibroblasts, and represent liver connective tissue cells involved in fibro-granulomatous reactions. They were able to sustain survival and proliferation of the multipotent myeloid cell lines FDC-P1 and DA-1 (dependent on interleukin-3 and/or granulocyte-macrophage colony stimulating factor, GM-CSF) without the addition of exogenous growth factors. This stimulation was dependent upon myeloid cell attachment to the GR cell layer; GR cell-conditioned medium had no activity. Primary murine skin fibroblasts could not sustain myelopoiesis. The endogenous growth-factor was identified as GM-CSF by neutralization assays with monoclonal antibodies. The stimulation of myelopoiesis occurred also when GR cells had been fixed with glutardialdehyde. The observed stimulatory activity was dependent upon heparan sulphate proteoglycans (HSPGs) associated with GR cell membranes. It could be dislodged from the cell layer with heparin or a high salt buffer. Our results indicate a molecular interaction between endogenous growth-factor and HSPGs; this interaction may be responsible for the stabilization and presentation of growth factors in myelopoietic stromas, mediating extramedullar proliferation of myeloid cells in periovular granulomas.
Collapse
Affiliation(s)
- M Alvarez-Silva
- Departamento de Bioquímica, Instituto de Química, Universidade Federal de Rio de Janeiro, Brazil
| | | | | |
Collapse
|
29
|
Just U, Katsuno M, Stocking C, Spooncer E, Dexter M. Targeted in vivo infection with a retroviral vector carrying the interleukin-3 (multi-CSF) gene leads to immortalization and leukemic transformation of primitive hematopoietic progenitor cells. Growth Factors 1993; 9:41-55. [PMID: 8102237 DOI: 10.3109/08977199308991581] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
To measure the effect of endogenous IL-3 (Multi-CSF) expression on hematopoietic cells in vivo, we have infected several kinds of hematopoietic cell populations with retroviral vectors carrying the IL-3 gene (M3MuV) in vitro and injected the virus-producing cells into mice to "target" the virus to sites of hematopoiesis. Mast cell lines (Elut cells) or multipotent cell lines (FDC-Pmix) were infected with MPSV-based replication defective retroviral vectors carrying either the neomycin resistance gene alone (M3neoV) or the neomycin gene plus the IL-3 gene (M3MuV). These cell lines produced infective retroviral particles consisting of the replication defective vectors and helper virus constitutively produced by the target cell populations. Irradiated and non-irradiated virus-producing Elut cells and the virus-producing FDC-Pmix cells were transplanted into syngeneic mice to "target" virus infection to the sites of hemopoiesis. Control mice injected with M3neoV-producing cells did not develop a disease up to 6 months following transplantation, whereas mice injected with M3MuV-producing cells developed a myeloproliferative disease within 3 months. Hematopoietic cell lines were rescued from diseased and control mice. In all cases these cell lines were of host origin. Cell lines derived from control mice were of basophil/mast cell morphology only, and required IL-3 for their continued proliferation (similar to cell lines derived from uninfected animals), whereas the cell lines generated from spleen and bone marrow cells of host mice with myeloproliferative disease carried the M3MuV vector, were G418 resistant and IL-3 independent. The biologic properties of M3MuV infected host derived cell lines varied considerably. Some were multipotential and could be induced to differentiate in response to stromal cells and serum factors, others were more restricted to the granulocyte/macrophage lineage but were also differentiation inducible, and some were blocked in differentiation at the myeloblast/promyelocyte stage. We conclude that the injected donor cells acted as "infectious centers" to facilitate the infection of host hematopoietic cells with the M3MuV vector. Our results indicate that the "targeted" in vivo infection of primitive hematopoietic cells with M3MuV can initiate the immortalization and leukaemogenesis of multipotential and lineage restricted progenitor cells.
Collapse
Affiliation(s)
- U Just
- Cancer Research Campaign Department of Experimental Haematology, Paterson Institute for Cancer Research, Christie Hospital, Manchester, UK
| | | | | | | | | |
Collapse
|
30
|
Graham GJ, Pragnell IB. The haemopoietic stem cell: properties and control mechanisms. SEMINARS IN CELL BIOLOGY 1992; 3:423-34. [PMID: 1489974 DOI: 10.1016/1043-4682(92)90013-l] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
All the cells of the immuno-haemopoietic system derive ultimately from a single pluripotent stem cell through processes of commitment and differentiation. The stem cell is also likely to be capable of extensive self renewal. Many factors which potentially control these processes have been identified and characterised both in vitro and in vivo. We discuss the nature of the haemopoietic stem cell and also the factors which have been identified as potential stem cell regulators. We also draw parallels from embryonal stem cell differentiation to derive a model of intrinsic determination of the haemopoietic stem cell. The possible role of developmental regulators, such as homeobox containing genes, are discussed in the context of differentiation commitment of the haemopoietic stem cell.
Collapse
Affiliation(s)
- G J Graham
- Cancer Research Campaign Beatson Laboratories, Bearsden, Glasgow, UK
| | | |
Collapse
|
31
|
Abstract
In adult mammals, hemopoiesis takes place primarily in the bone marrow. The steady-state production of blood cells depends to a large extent on the interaction between hemopoietic stem/progenitor cells (HPC) and the different components of the microenvironment present in the medullary cavity. During the last three decades, in vivo and in vitro studies have allowed significant advances in understanding of the biology of such a hemopoietic microenvironment. Although not evident in histological sections, it is well known that the hemopoietic microenvironment is a highly organized structure that regulates the location and physiology of HPC. The hemopoietic microenvironment is composed of stromal cells (fibroblasts, macrophages, endothelial cells, adipocytes), accessory cells (T lymphocytes, monocytes), and their products (extracellular matrix and cytokines). Microenvironmental cells can regulate hemopoiesis by interacting directly (cell-to-cell contact) with HPC and/or by secreting regulatory molecules that influence, in a positive or negative manner, HPC growth. Recent in vitro studies suggest that functional abnormalities of the hemopoietic microenvironment may be implicated in the manifestation of certain hematological disorders such as aplastic anemia, and acute and chronic myelogenous leukemia. Thus, the characterization of the structure and function of the human hemopoietic microenvironment may have relevance in understanding and treating different hematological disorders.
Collapse
Affiliation(s)
- H Mayani
- Department of Medicine, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
32
|
van der Sluijs JP, Baert MRM, van Beurden CAJ, Ploemacher RE. Evaluation of in vivo and in vitro repopulation assays for murine haemopoietic stem cells. ACTA ACUST UNITED AC 1992. [DOI: 10.1007/bf00426164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
33
|
van Beusechem VW, Kukler A, Heidt PJ, Valerio D. Long-term expression of human adenosine deaminase in rhesus monkeys transplanted with retrovirus-infected bone-marrow cells. Proc Natl Acad Sci U S A 1992; 89:7640-4. [PMID: 1502175 PMCID: PMC49766 DOI: 10.1073/pnas.89.16.7640] [Citation(s) in RCA: 134] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Gene transfer into hemopoietic stem cells could offer a lasting cure for a variety of congenital disorders. As a preclinical test for such a gene therapy, rhesus monkeys were transplanted with autologous bone-marrow cells infected with helper-free recombinant retroviruses carrying the human adenosine deaminase gene. The in vivo regenerative capacity of the infected bone marrow could be conserved, suggesting survival of repopulating hemopoietic stem cells. In the hemopoietic system of transplanted animals the foreign gene could be observed for as long as the animals were analyzed (in two monkeys greater than 1 yr after transplantation). Genetically modified cell types and tissues included peripheral blood mononuclear cells, granulocytes, bone-marrow cells of various densities, and spleen and lymph nodes. The presence of the provirus in the short-living granulocytes greater than 1 yr after bone-marrow transplantation provided evidence for the transduction of very primitive hemopoietic progenitors. Moreover, the gene transfer resulted in sustained production of functional human adenosine deaminase enzyme in peripheral blood mononuclear cells. These results demonstrate the feasibility of bone-marrow gene-therapy approaches, in particular for treating adenosine deaminase deficiency.
Collapse
Affiliation(s)
- V W van Beusechem
- Department of Gene Therapy, TNO Institute of Applied Radiobiology and Immunology, Rijswijk, The Netherlands
| | | | | | | |
Collapse
|
34
|
Teofili L, Sargiacomo M, Iovino MS, Zini G, Leone G, Bizzi B, Peschle C. Effects of a preformed extracellular matrix on long-term serum-free bone marrow culture. Ann Hematol 1992; 65:22-5. [PMID: 1643156 DOI: 10.1007/bf01715121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The extracellular matrix (ECM) produced by the stromal layer plays a key role in the regulation of commitment and differentiation of hematopoietic cells. Long-term bone marrow culture (LTBMC) allows analysis of the stromal microenvironment. Recently, serum-free LTBMC has been described, but the formation of a classical adherent layer was never observed under these conditions. We have evaluated the effect(s) of a chemically well defined ECM on serum-free and serum-dependent LTBMC. In serum-dependent cultures ECM did not induce a significant increase of hematopoiesis. In serum-free conditions, a marked improvement of hematopoiesis was observed, both in terms of CFU-GM and BFU-E yield and in duration of cultures. A confluent stromal layer was observed only in the presence of ECM. The present results indicate that the addition of ECM to serum-free cultures provides a standardized culture condition, while improving progenitor cell recovery and allowing formation of a confluent stromal layer. Moreover, ECM+LTBMC may provide a model to study the effect(s) of adhesive proteins and hematopoietic growth factors normally present in serum.
Collapse
Affiliation(s)
- L Teofili
- Istituto di Semeiotica Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Umezawa A, Maruyama T, Segawa K, Shadduck RK, Waheed A, Hata J. Multipotent marrow stromal cell line is able to induce hematopoiesis in vivo. J Cell Physiol 1992; 151:197-205. [PMID: 1373147 DOI: 10.1002/jcp.1041510125] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Several murine marrow stromal cells were established from murine bone marrow cultures. Stromal cell lines transfected with a tumor-inducing polyoma virus middle T antigen (MTAg) were inoculated into nude mice subcutaneously. KUSA-MTAg cells, one of these cell lines, led to the rapid local development of bone marrow consisting of trilineage hematopoietic cells and bone; other cell lines produced spindle cell sarcoma or hemangiosarcoma. These results suggested that a single stromal cell line, KUSA-MTAg cells, may induce hematopoietic stem cells or early progenitors of three lineages of hematopoietic cells in vivo. Interestingly, untransfected KUSA cells expressed three new mesenchymal phenotypes, osteocytes, adipocytes, and myotubes, after treatment with 5-azacytidine.
Collapse
Affiliation(s)
- A Umezawa
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Collins DA, Chambers TJ. Prostaglandin E2 promotes osteoclast formation in murine hematopoietic cultures through an action on hematopoietic cells. J Bone Miner Res 1992; 7:555-61. [PMID: 1615762 DOI: 10.1002/jbmr.5650070512] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Osteoclastic differentiation is induced from hematopoietic cells in the presence of 1,25-(OH)2D3 by stromal cells that are present in bone but not in hematopoietic spleen. Recent evidence suggests that prostaglandins (PGs) are essential for this process. In this communication we describe experiments in which we have examined further the role of PGE2 in osteoclast formation. We found a marked reduction in basal, 1,25-(OH)2D3, and IL-3-induced production of calcitonin receptor (CTR)-positive cells and bone resorption by cyclooxygenase inhibitors, which was restored by PGE2 addition. Although some stromal cell types (ST2 cells) that support osteoclast formation from spleen cells produced PGs in response to 1,25-(OH)2D3, others (ts8 and calvarial cells) did not, either alone or in combination with spleen cells. On the other hand, both bone marrow and spleen cells produced amounts of PGE2 in response to 1,25-(OH)2D3 that were sufficient to account for osteoclast formation. Osteoclast-inductive ts8 cells were able to support osteoclast formation from spleen cells in the presence of 1,25-(OH)2D3 or PGE2 even if devitalized. Incubation of ts8 cells in these agents before devitalization did not avoid the requirement for the presence of PGE2 or 1,25-(OH)2D3 during subsequent incubation with spleen cells. Thus, hematopoietic cells produce sufficient PGE2 for osteoclast formation, and the PGE2 thus produced acts on hematopoietic precursors, which can be induced in the presence of PGE2 to express CTR and resorb bone on contact with osteoclast-inductive stromal cells. The ability of osteoclast-inductive cells to support osteoclast formation appears not to rest on their ability to produce, induce, or respond to PGE2.
Collapse
Affiliation(s)
- D A Collins
- Department of Histopathology, St. George's Hospital Medical School, London, England
| | | |
Collapse
|
37
|
Affiliation(s)
- C Chabannon
- Department of Transplantation Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98104
| | | |
Collapse
|
38
|
Yaeger PC, Stiles CD, Rollins BJ. Human keratinocyte growth-promoting activity on the surface of fibroblasts. J Cell Physiol 1991; 149:110-6. [PMID: 1939339 DOI: 10.1002/jcp.1041490114] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
To proliferate in serum-containing medium, normal human keratinocytes must be co-cultured with fibroblast feeder cells. Conditioned medium from feeder cell cultures cannot substitute for the cells themselves. We tested the hypothesis that fibroblasts display a keratinocyte growth-promoting activity on their outer cell surface. The results of our investigation showed that (1) glutaraldehyde-fixed fibroblast feeder cells promote keratinocyte growth, (2) the growth-promoting effect requires contact between fixed fibroblasts and keratinocytes, and (3) feeder activity is highly enriched within the plasma membrane fraction of fibroblasts. We conclude that at least part of the fibroblast "feeder" activity involves a keratinocyte growth-promoting factor which is bound to the outer surface of fibroblast plasma membranes.
Collapse
Affiliation(s)
- P C Yaeger
- Department of Cellular and Molecular Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115
| | | | | |
Collapse
|
39
|
Chambers TJ, Hall TJ. Cellular and molecular mechanisms in the regulation and function of osteoclasts. VITAMINS AND HORMONES 1991; 46:41-86. [PMID: 1660641 DOI: 10.1016/s0083-6729(08)60682-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- T J Chambers
- Department of Histopathology, St. George's Hospital Medical School, London, England
| | | |
Collapse
|
40
|
Rios M, Williams DA. Systematic analysis of the ability of stromal cell lines derived from different murine adult tissues to support maintenance of hematopoietic stem cells in vitro. J Cell Physiol 1990; 145:434-43. [PMID: 1703166 DOI: 10.1002/jcp.1041450307] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hematopoietic stem cells interact with a complex microenvironment both in vivo and in vitro. In association with this microenvironment, murine stem cells are maintained in vitro for several months. Fibroblast-like stromal cells appear to be important components of the microenvironment, since several laboratories have demonstrated that cloned stromal cell lines support hematopoiesis in vitro. The importance of the tissue of origin of such cell lines remains unknown, since systematic generation of stromal cell lines from adult tissues has never been accomplished. In addition, the capacity of stromal cell lines to support reconstituting stem cell has not been examined. We have previously described an efficient and rapid method for the immortalization of primary bone marrow stromal cell lines (Williams et al., Mol. Cell. Biol. 8:3864-3871, 1988) which can be used to systematically derive cell lines from multiple tissues of the adult mouse. Here we report the immortalization of primary murine lung, kidney, skin, and bone marrow stromal cells using a recombinant retrovirus vector (U19-5) containing the simian virus large T antigen (SV40 LT) and the neophosphotransferase gene. The interaction of these stromal cells with factor-dependent cells Patterson-Mix (FDCP-Mix), colony forming units-spleen (CFU-S), and reconstituting hematopoietic stem cells was studied in order to analyze the ability of such lines to support multipotent stem cells in vitro. These studies revealed that stromal cell lines from these diverse tissues were morphologically and phenotypically similar and that they quantitatively bound CFU-S and FDCP-Mix cells equally well. However, only those cell lines derived from bone marrow-supported maintenance of day 12 CFU-S in vitro. One lung-derived stromal cell line, ULU-3, supported the survival of day 8 CFU-S, but not the more primitive CFU-S12. A bone marrow-derived stromal cell line, U2, supported the survival of long-term reconstituting stem cells for up to 3 weeks in vitro as assayed by reconstitution 1 year post-transplant. These studies suggest that adherence of HSC to stromal cells is necessary but not sufficient for maintenance of these stem cell populations and that bone marrow provides specific signals relating to hematopoietic stem cell survival and proliferation.
Collapse
Affiliation(s)
- M Rios
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115
| | | |
Collapse
|
41
|
Soligo D, Schiró R, Luksch R, Manara G, Quirici N, Parravicini C, Lambertenghi Deliliers G. Expression of integrins in human bone marrow. Br J Haematol 1990; 76:323-32. [PMID: 2261342 DOI: 10.1111/j.1365-2141.1990.tb06363.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Expression of integrins, a superfamily of glycoprotein alpha/beta heterodimers which integrate the cytoskeleton with the extracellular matrix and/or mediate cell-cell adhesive interactions, was examined on normal and leukaemic bone marrow cells by immunohistochemistry and immunotransmission electron microscopy (immuno-TEM). Among the beta 1/VLA molecules studied, VLA-2 and 6 were expressed on megakaryocytes and platelets, while VLA-4 was present on 40% of haemopoietic cells, including monocytes, erythroblasts and immature cells; this molecule was typically localized at sites of intercellular contact, as seen by immuno-TEM, suggesting it may be involved in interactions among haemopoietic cells during differentiation. In human long-term bone marrow cultures (LTBMC), VLA-1 and 3 were present respectively on 35% and 40% of the adherent cells which included fibroblasts and endothelial cells, as shown by double-labelling experiments; VLA-2 was expressed only on a subpopulation of fibroblasts. beta 2/LeuCAM molecules were absent from platelets, megakaryocytes and HLA-DR+/myeloperoxidase- early myeloid precursors, and appeared progressively during maturation in both lymphoid and myeloid cells. Expression of beta 3/cytoadhesin molecules was restricted to megakaryocytes and platelets and, in the adherent layer of LTBMC, to endothelial cells. The regulated expression and specific localization of integrins in the bone marrow suggest that these molecules may have a role in normal haemopoiesis.
Collapse
Affiliation(s)
- D Soligo
- Clinica Pediatrica Ospedale S. Gerardo, University of Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Effect of myogenic and adipogenic differentiation on expression of colony-stimulating factor genes. Mol Cell Biol 1990. [PMID: 1697034 DOI: 10.1128/mcb.10.9.4948] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The influence of cellular differentiation on colony-stimulating factor gene expression was examined in myogenically and adipogenically determined cell lines derived from 5-azacytidine-treated C3H10T1/2 C18 (10T1/2) mouse embryo fibroblasts. These studies demonstrate that colony-stimulating factor gene expression can be modulated by myogenic and adipogenic determination and terminal differentiation.
Collapse
|
43
|
Harrington MA, Falkenburg JH, Daub R, Broxmeyer HE. Effect of myogenic and adipogenic differentiation on expression of colony-stimulating factor genes. Mol Cell Biol 1990; 10:4948-52. [PMID: 1697034 PMCID: PMC361117 DOI: 10.1128/mcb.10.9.4948-4952.1990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The influence of cellular differentiation on colony-stimulating factor gene expression was examined in myogenically and adipogenically determined cell lines derived from 5-azacytidine-treated C3H10T1/2 C18 (10T1/2) mouse embryo fibroblasts. These studies demonstrate that colony-stimulating factor gene expression can be modulated by myogenic and adipogenic determination and terminal differentiation.
Collapse
Affiliation(s)
- M A Harrington
- Department of Medicine (Hematology/Oncology), Indiana University School of Medicine, Indianapolis 46202-5121
| | | | | | | |
Collapse
|
44
|
Brandt J, Srour EF, van Besien K, Briddell RA, Hoffman R. Cytokine-dependent long-term culture of highly enriched precursors of hematopoietic progenitor cells from human bone marrow. J Clin Invest 1990; 86:932-41. [PMID: 1697607 PMCID: PMC296813 DOI: 10.1172/jci114795] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human marrow cells positive for the CD34 antigen but not expressing HLA-DR, CD15, or CD71 antigens were isolated. In a liquid culture system supplemented with 48-hourly additions of recombinant interleukins IL-1 alpha, IL-3, IL-6, or granulocyte/macrophage colony-stimulating factor (GM-CSF), these cells were capable of sustaining in vitro hematopoiesis for up to eight weeks. The establishment of an adherent cell layer was never observed. Cultures containing no exogenous cytokine produced clonogenic cells for only 1 wk. IL-1 alpha and IL-6 were alone able to support hematopoiesis for 2 or 3 wk. Cells maintained with GM-CSF proliferated and contained assayable colony-forming cells for 3 or 4 wk, while maximal cellular expansion and generation of assayable progenitor cells occurred in the presence of IL-3 for 4-5 wk. When IL-3 was combined with IL-1 alpha or IL-6, hematopoiesis was sustained for 8 wks. Basophil numbers were markedly increased in the presence of IL-3. These studies indicate that marrow subpopulations can sustain hematopoiesis in vitro in the presence of repeated additions of cytokines. We conclude that a major function of marrow adherent cells in long-term cultures is that of providing cytokines which promote the proliferation and differentiation of primitive hematopoietic cells.
Collapse
Affiliation(s)
- J Brandt
- Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis 46202
| | | | | | | | | |
Collapse
|
45
|
Lillien LE, Sendtner M, Raff MC. Extracellular matrix-associated molecules collaborate with ciliary neurotrophic factor to induce type-2 astrocyte development. J Cell Biol 1990; 111:635-44. [PMID: 2199462 PMCID: PMC2116219 DOI: 10.1083/jcb.111.2.635] [Citation(s) in RCA: 126] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
O-2A progenitor cells give rise to both oligodendrocytes and type-2 astrocytes in vitro. Whereas oligodendrocyte differentiation occurs constitutively, type-2 astrocyte differentiation requires extracellular signals, one of which is thought to be ciliary neurotrophic factor (CNTF). CNTF, however, is insufficient by itself to induce the development of stable type-2 astrocytes. In this report we show the following: (a) that molecules associated with the extracellular matrix (ECM) cooperate with CNTF to induce stable type-2 astrocyte differentiation in serum-free cultures. The combination of CNTF and the ECM-associated molecules thus mimics the effect of FCS, which has been shown previously to induce stable type-2 astrocyte differentiation in vitro. (b) Both the ECM-associated molecules and CNTF act directly on O-2A progenitor cells and can induce them to differentiate prematurely into type-2 astrocytes. (c) ECM-associated molecules also inhibit oligodendrocyte differentiation, even in the absence of CNTF, but this inhibition is not sufficient on its own to induce type-2 astrocyte differentiation. (d) Whereas the effect of ECM on oligodendrocyte differentiation is mimicked by basic fibroblast growth factor (bFGF), the effect of ECM on type-2 astrocyte differentiation is not. (e) The ECM-associated molecules that are responsible for inhibiting oligodendrocyte differentiation and for cooperating with CNTF to induce type-2 astrocyte differentiation are made by non-glial cells in vitro. (f) Molecules that have these activities and bind to ECM are present in the optic nerve at the time type-2 astrocytes are thought to be developing.
Collapse
Affiliation(s)
- L E Lillien
- Biology Department, University College London, United Kingdom
| | | | | |
Collapse
|
46
|
Gimble JM, Dorheim MA, Cheng Q, Medina K, Wang CS, Jones R, Koren E, Pietrangeli C, Kincade PW. Adipogenesis in a murine bone marrow stromal cell line capable of supporting B lineage lymphocyte growth and proliferation: biochemical and molecular characterization. Eur J Immunol 1990; 20:379-87. [PMID: 2178944 DOI: 10.1002/eji.1830200222] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent advances in long-term bone marrow (BM) culture techniques have allowed investigators to dissect cellular components responsible for lympho hematopoiesis. Consequently, a number of "stromal" cell clones have been developed which are capable of supporting B lineage lymphocyte growth and proliferation in vitro by direct cell-cell interactions and the release of cytokines. While much work has focused on the support function of these cells, questions remain regarding their own differentiation potential. We have examined adipogenesis in the cloned BM stromal cell, BMS2. The presence of hydrocortisone, methylisobutylxanthine, or 30% fetal calf serum each accelerated adipocyte differentiation. This process was accompanied by the accumulation of triglycerides and cholesterol esters along with the induction of adipocyte-specific enzymes. Likewise, the steady-state level of mRNA transcripts increased for genes related to lipid metabolism. However, the pattern of mRNA expression in BMS2 adipocytes differed from that of a well-established, pre-adipocyte cell line, 3T3-L1, with respect to the following genes: glycerol phosphate dehydrogenase, CAAT/enhancer binding protein and angiotensinogen. Adipocyte BMS2 cells retailed the ability to support stromal cell-dependent B lineage lymphocytes in methylcellulose assays. The adipocytes continued to express macrophage-colony-stimulating factor mRNA constitutively and interleukin 6 mRNA in an inducible manner, similar to the BMS2 pre-adipocytes. Together, these data document a close developmental relationship between a specialized fibroblasts and adipocytes in the BM and suggest that adipocyte stromal cells may play an active role in lympho-hematopoiesis.
Collapse
Affiliation(s)
- J M Gimble
- Immunobiology and Cancer Program, Oklahoma Medical Research Foundation, Oklahoma City 73104
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Affiliation(s)
- P W Kincade
- Immunobiology and Cancer Program, Oklahoma Medical Research Foundation, Oklahoma City 73104
| |
Collapse
|
48
|
Stimulation of myeloid colony development elaborated by colony forming cells--fibroblasts from rat developing ossicles. Leuk Res 1990; 14:63-8. [PMID: 2406512 DOI: 10.1016/0145-2126(90)90147-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Subcutaneous implantation of demineralized bone matrix in rats results in endochondral bone formation and bone-marrow development. The cascade of events leading to this process occurs following the accumulation of mesenchymal cells with colony forming cell fibroblasts (CFC-F) potential in the implanted area, a process which commences already 72 h post implantation. It is demonstrated herein that CFC-F from various stages of ossicle development (days, 3, 7, 10, 14 or 18) stimulate hemopoiesis to the same extent as judged by the number of granuloid-macrophage-progenitors (CFU-GM), developed as hemopoietic colonies, and by the ratio of granuloid to macrophagic colonies. High concentrations of CFC-F, however, tend to diminish the stimulatory capacity. Prostaglandin E, CFU-GM, CFC-F, ossicle, growth factors, microenvironment, hemopoiesis, development.
Collapse
|
49
|
Hattersley G, Chambers TJ. Generation of osteoclasts from hemopoietic cells and a multipotential cell line in vitro. J Cell Physiol 1989; 140:478-82. [PMID: 2777887 DOI: 10.1002/jcp.1041400311] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Osteoclasts are the cells that resorb bone. It is generally presumed, on the basis of indirect experiments, that they are derived from the hemopoietic stem cell. However, this origin has never been established. We have developed an assay for osteoclastic differentiation in which bone marrow cells are incubated in liquid culture on slices of cortical bone. The bone slices are inspected in the scanning electron microscope after incubation for the presence of excavations, which are characteristic of osteoclastic activity. We have now incubated bone marrow cells at low density, or a factor-dependent mouse hemopoietic cell line (FDCP-mix A4) with 1,25 dihydroxyvitamin D3 (a hormone which we have previously found induces osteoclastic differentiation) with and without murine bone marrow stromal cells, or with and without 3T3 cells, on bone slices. Neither the bone marrow cells nor the bone marrow stromal cells alone developed osteoclastic function even in the presence of 1,25 dihydroxyvitamin D3. However, extensive excavation of the bone surface was observed, only in the presence of 1,25 dihydroxyvitamin D3, on bone slices on which bone marrow stromal cells were cocultured with low-density bone marrow cells or the hemopoietic cell line. Similar results were obtained when the bone marrow stromal cells were killed by glutaraldehyde fixation; 3T3 cells were unable to substitute for stromal cells. These results are strong evidence that osteoclasts derive from the hemopoietic stem cell and suggest that although mature osteoclasts possess neither receptors for nor responsiveness to 1,25 dihydroxyvitamin D3, the hormone induces osteoclastic function through a direct effect on hemopoietic cells rather than through some accessory cell in the bone marrow stroma. The failure of 3T3 cells, which enable differentiation of other hemopoietic progeny from this cell line, to induce osteoclastic differentiation suggests that bone marrow stroma possesses additional characteristics distinct from those that induce differentiation of other hemopoietic cells that are specifically required for osteoclastic differentiation.
Collapse
Affiliation(s)
- G Hattersley
- Department of Histopathology, St George's Hospital Medical School, London, England
| | | |
Collapse
|
50
|
Affiliation(s)
- P A Hall
- Keratinocyte Laboratory, Imperial Cancer Research Fund, London, UK
| | | |
Collapse
|