1
|
Abstract
Research in the last few years has revealed a sophisticated interaction network between multiple bone marrow cells that regulate different hematopoietic stem cell (HSC) properties such as proliferation, differentiation, localization, and self-renewal during homeostasis. These mechanisms are essential to keep the physiological HSC numbers in check and interfere with malignant progression. In addition to the identification of multiple mutations and chromosomal aberrations driving the progression of myeloid malignancies, alterations in the niche compartment recently gained attention for contributing to disease progression. Leukemic cells can remodel the niche into a permissive environment favoring leukemic stem cell expansion over normal HSC maintenance, and evidence is accumulating that certain niche alterations can even induce leukemic transformation. Relapse after chemotherapy is still a major challenge during treatment of myeloid malignancies, and cure is only rarely achieved. Recent progress in understanding the niche-imposed chemoresistance mechanisms will likely contribute to the improvement of current therapeutic strategies. This article discusses the role of different niche cells and their stage- and disease-specific roles during progression of myeloid malignancies and in response to chemotherapy.
Collapse
|
2
|
Baba T, Naka K, Morishita S, Komatsu N, Hirao A, Mukaida N. MIP-1α/CCL3-mediated maintenance of leukemia-initiating cells in the initiation process of chronic myeloid leukemia. J Exp Med 2013; 210:2661-2673. [PMID: 24166712 PMCID: PMC3832924 DOI: 10.1084/jem.20130112] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 09/26/2013] [Indexed: 12/13/2022] Open
Abstract
In the initiation process of chronic myeloid leukemia (CML), a small number of transformed leukemia-initiating cells (LICs) coexist with a large number of normal hematopoietic cells, gradually increasing thereafter and eventually predominating in the hematopoietic space. However, the interaction between LICs and normal hematopoietic cells at the early phase has not been clearly delineated because of the lack of a suitable experimental model. In this study, we succeeded in causing a marked leukocytosis resembling CML from restricted foci of LICs in the normal hematopoietic system by direct transplantation of BCR-ABL gene-transduced LICs into the bone marrow (BM) cavity of nonirradiated mice. Herein, we observed that BCR-ABL(+)lineage(-)c-kit(-) immature leukemia cells produced high levels of an inflammatory chemokine, MIP-1α/CCL3, which promoted the development of CML. Conversely, ablation of the CCL3 gene in LICs dramatically inhibited the development of CML and concomitantly reduced recurrence after the cessation of a short-term tyrosine kinase inhibitor treatment. Finally, normal hematopoietic stem/progenitor cells can directly impede the maintenance of LICs in BM in the absence of CCL3 signal.
Collapse
MESH Headings
- Animals
- Bone Marrow Transplantation
- Chemokine CCL3/deficiency
- Chemokine CCL3/genetics
- Chemokine CCL3/metabolism
- Disease Models, Animal
- Genes, abl
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Nude
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/transplantation
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Proto-Oncogene Proteins c-kit/metabolism
- Receptors, CCR1/deficiency
- Receptors, CCR1/genetics
- Receptors, CCR1/metabolism
- Receptors, CCR5/deficiency
- Receptors, CCR5/genetics
- Receptors, CCR5/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Tomohisa Baba
- Division of Molecular Bioregulation, Exploratory Project on Cancer Stem Cells, and Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan
| | - Kazuhito Naka
- Division of Molecular Bioregulation, Exploratory Project on Cancer Stem Cells, and Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan
| | - Soji Morishita
- Department of Transfusion Medicine and Stem Cell Regulation and Department of Hematology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Norio Komatsu
- Department of Transfusion Medicine and Stem Cell Regulation and Department of Hematology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Atsushi Hirao
- Division of Molecular Bioregulation, Exploratory Project on Cancer Stem Cells, and Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Exploratory Project on Cancer Stem Cells, and Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan
| |
Collapse
|
3
|
Jung Y, Wang J, Song J, Shiozawa Y, Wang J, Havens A, Wang Z, Sun YX, Emerson SG, Krebsbach PH, Taichman RS. Annexin II expressed by osteoblasts and endothelial cells regulates stem cell adhesion, homing, and engraftment following transplantation. Blood 2007; 110:82-90. [PMID: 17360942 PMCID: PMC1896130 DOI: 10.1182/blood-2006-05-021352] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Differentiation of hematopoietic stem cells (HSCs) after birth is largely restricted to the bone marrow cavity, where HSCs are associated closely with osteoblasts (OBs). How OBs localize HSCs to the endosteal niche remains unclear. To explore adhesive interactions between HSCs and OBs, a cell blot analysis was used that revealed 2 major bands that corresponded to monomers and multimers of annexin II (Anxa2). Immunohistochemistry revealed that OBs and marrow endothelial cells express Anxa2 at high levels. Function-blocking studies confirmed that Anxa2 mediates HSC adhesion mainly via the N-terminal portion of the Anxa2 peptide. Adhesion of HSCs to OBs derived from Anxa2-deficient animals (Anxa2(-/-)) was significantly impaired compared with OBs obtained from wild-type animals (Anxa2(+/+)). Moreover, fewer HSCs were found in the marrow of Anxa2(-/-) versus Anxa2(+/+) animals. Short-term lodging, engraftment, and survival of irradiated mice with whole marrow cells were substantially inhibited by N-terminal peptide fragments of Anxa2 or anti-Anxa2 antibodies. Similar findings were noted in long-term competitive repopulation studies. Collectively, these findings reveal that Anxa2 regulates HSC homing and binding to the bone marrow microenvironment and suggest that Anxa2 is crucial for determining the bone marrow niche of HSCs.
Collapse
Affiliation(s)
- Younghun Jung
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Taichman RS. Blood and bone: two tissues whose fates are intertwined to create the hematopoietic stem-cell niche. Blood 2004; 105:2631-9. [PMID: 15585658 DOI: 10.1182/blood-2004-06-2480] [Citation(s) in RCA: 383] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The mechanisms of bone and blood formation have traditionally been viewed as distinct, unrelated processes, but compelling evidence suggests that they are intertwined. Based on observations that hematopoietic precursors reside close to endosteal surfaces, it was hypothesized that osteoblasts play a central role in hematopoiesis, and it has been shown that osteoblasts produce many factors essential for the survival, renewal, and maturation of hematopoietic stem cells (HSCs). Preceding these observations are studies demonstrating that the disruption or perturbation of normal osteoblastic function has a profound and central role in defining the operational structure of the HSC niche. These observations provide a glimpse of the dimensions and ramifications of HSC-osteoblast interactions. Although more research is required to secure a broader grasp of the molecular mechanisms that govern blood and bone biology, the central role for osteoblasts in hematopoietic stem cell regulation is reviewed herein from the perspectives of (1) historical context; (2) the role of the osteoblast in supporting stem cell survival, proliferation, and maintenance; (3) the participation, if any, of osteoblasts in the creation of a stem cell niche; (4) the molecules that mediate HSC-osteoblast interactions; (5) the role of osteoblasts in stem cell transplantation; and (6) possible future directions for investigation.
Collapse
Affiliation(s)
- Russell S Taichman
- Department of Periodontics, Prevention and Geriatrics, University of Michigan School of Dentistry, 1011 N University Ave, Ann Arbor, MI 48109-1078, USA.
| |
Collapse
|
5
|
Park CC, Henshall-Powell RL, Erickson AC, Talhouk R, Parvin B, Bissell MJ, Barcellos-Hoff MH. Ionizing radiation induces heritable disruption of epithelial cell interactions. Proc Natl Acad Sci U S A 2003; 100:10728-33. [PMID: 12960393 PMCID: PMC196872 DOI: 10.1073/pnas.1832185100] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Indexed: 02/07/2023] Open
Abstract
Ionizing radiation (IR) is a known human breast carcinogen. Although the mutagenic capacity of IR is widely acknowledged as the basis for its action as a carcinogen, we and others have shown that IR can also induce growth factors and extracellular matrix remodeling. As a consequence, we have proposed that an additional factor contributing to IR carcinogenesis is the potential disruption of critical constraints that are imposed by normal cell interactions. To test this hypothesis, we asked whether IR affected the ability of nonmalignant human mammary epithelial cells (HMEC) to undergo tissue-specific morphogenesis in culture by using confocal microscopy and imaging bioinformatics. We found that irradiated single HMEC gave rise to colonies exhibiting decreased localization of E-cadherin, beta-catenin, and connexin-43, proteins necessary for the establishment of polarity and communication. Severely compromised acinar organization was manifested by the majority of irradiated HMEC progeny as quantified by image analysis. Disrupted cell-cell communication, aberrant cell-extracellular matrix interactions, and loss of tissue-specific architecture observed in the daughters of irradiated HMEC are characteristic of neoplastic progression. These data point to a heritable, nonmutational mechanism whereby IR compromises cell polarity and multicellular organization.
Collapse
Affiliation(s)
- Catherine C Park
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Clarkson B, Strife A, Wisniewski D, Lambek CL, Liu C. Chronic myelogenous leukemia as a paradigm of early cancer and possible curative strategies. Leukemia 2003; 17:1211-62. [PMID: 12835715 DOI: 10.1038/sj.leu.2402912] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The chronological history of the important discoveries leading to our present understanding of the essential clinical, biological, biochemical, and molecular features of chronic myelogenous leukemia (CML) are first reviewed, focusing in particular on abnormalities that are responsible for the massive myeloid expansion. CML is an excellent target for the development of selective treatment because of its highly consistent genetic abnormality and qualitatively different fusion gene product, p210(bcr-abl). It is likely that the multiple signaling pathways dysregulated by p210(bcr-abl) are sufficient to explain all the initial manifestations of the chronic phase of the disease, although understanding of the circuitry is still very incomplete. Evidence is presented that the signaling pathways that are constitutively activated in CML stem cells and primitive progenitors cooperate with cytokines to increase the proportion of stem cells that are activated and thereby increase recruitment into the committed progenitor cell pool, and that this increased activation is probably the primary cause of the massive myeloid expansion in CML. The cooperative interactions between Bcr-Abl and cytokine-activated pathways interfere with the synergistic interactions between multiple cytokines that are normally required for the activation of stem cells, while at the same time causing numerous subtle biochemical and functional abnormalities in the later progenitors and precursor cells. The committed CML progenitors have discordant maturation and reduced proliferative capacity compared to normal committed progenitors, and like them, are destined to die after a limited number of divisions. Thus, the primary goal of any curative strategy must be to eliminate all Philadelphia positive (Ph+) primitive cells that are capable of symmetric division and thereby able to expand the Ph+ stem cell pool and recreate the disease. Several highly potent and moderately selective inhibitors of Bcr-Abl kinase have recently been discovered that are capable of killing the majority of actively proliferating early CML progenitors with minimal effects on normal progenitors. However, like their normal counterparts, most of the CML primitive stem cells are quiescent at any given time and are relatively invulnerable to the Bcr-Abl kinase inhibitors as well as other drugs. We propose that survival of dormant Ph+ stem cells may be the most important reason for the inability to cure the disease during initial treatment, while resistance to the inhibitors and other drugs becomes increasingly important later. An outline of a possible curative strategy is presented that attempts to take advantage of the subtle differences in the proliferative behavior of normal and Ph+ stem cells and the newly discovered selective inhibitors of Bcr-Abl. Leukemia (2003) 17, 1211-1262. doi:10.1038/sj.leu.2402912
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Signal Transduction
- Treatment Outcome
Collapse
Affiliation(s)
- B Clarkson
- Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
7
|
Vuckovic S, Kim M, Khalil D, Turtle CJ, Crosbie GV, Williams N, Brown L, Williams K, Kelly C, Stravos P, Rodwell R, Hill GR, Wright S, Taylor K, Gill D, Marlton P, Bradstock K, Hart DNJ. Granulocyte-colony stimulating factor increases CD123hi blood dendritic cells with altered CD62L and CCR7 expression. Blood 2003; 101:2314-7. [PMID: 12406901 DOI: 10.1182/blood-2002-03-0973] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Changes in blood dendritic cell (BDC) counts (CD123(hi)BDC and CD11c(+)BDC) and expression of CD62L, CCR7, and CD49d were analyzed in healthy donors, multiple myeloma (MM), and non-Hodgkin lymphoma (NHL) patients, who received granulocyte-colony stimulating factor (G-CSF) containing peripheral blood stem cell (PBSC) mobilization protocols. Low-dose G-CSF in healthy donors (8-10 microg/kg/d subcutaneously) and high-dose G-CSF in patients (30 microg/kg/d) increased CD123(hi)BDC (2- to 22-fold, mean 3.7 x 10(6)/L-17.7 x 10(6)/L and 1.9 x 10(6)/L-12.0 x 10(6)/L) in healthy donors and MM but decreased CD11c(+)BDC (2- to 10-fold, mean 5.7 x 10(6)/L-1.6 x 10(6)/L) in NHL patients, on the day of apheresis, compared with steady state. After apheresis, CD123(hi)BDC counts remained high, whereas low CD11c(+)BDC counts tended to recover in the following 2-5 days. Down-regulation of CD62L and up-regulation of CCR7 on CD123(hi)BDC were found in most healthy donors and MM patients. CD49d expression was unchanged. Thus, PBSC mobilization may change BDC counts by altering molecules necessary for BDC homing from blood into tissues.
Collapse
Affiliation(s)
- Slavica Vuckovic
- Mater Medical Research Institute, South Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Salesse S, Verfaillie CM. Mechanisms underlying abnormal trafficking and expansion of malignant progenitors in CML: BCR/ABL-induced defects in integrin function in CML. Oncogene 2002; 21:8605-11. [PMID: 12476307 DOI: 10.1038/sj.onc.1206088] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Stephanie Salesse
- Stem Cell Institute, Cancer Center, Minneapolis, Minnesota, MN 55455, USA
| | | |
Collapse
|
9
|
Peled A, Hardan I, Trakhtenbrot L, Gur E, Magid M, Darash-Yahana M, Cohen N, Grabovsky V, Franitza S, Kollet O, Lider O, Alon R, Rechavi G, Lapidot T. Immature leukemic CD34+CXCR4+ cells from CML patients have lower integrin-dependent migration and adhesion in response to the chemokine SDF-1. Stem Cells 2002; 20:259-66. [PMID: 12004084 DOI: 10.1634/stemcells.20-3-259] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Chronic myelogenous leukemia (CML), a malignant myeloproliferative disorder originating from a pluripotent stem cell expressing the bcr-abl oncogene, is characterized by abnormal release of the expanded, malignant stem cell clone from the bone marrow (BM) into the circulation. Moreover, immature CD34+ CML cells have lower adhesion to stromal cells and fibronectin as well as lower engraftment potential in severe combined immunedeficient (SCID) and nonobese diabetic (NOD)/SCID mice than normal CD34+ cells. We report in this study that leukemic Philadelphia chromosome-positive (Ph+)CD34+ cells from newly diagnosed CML patients that express the chemokine receptor CXCR4 migrate in response to stromal-derived factor-1 (SDF-1). However, normal Ph-CD34+CXCR4+ cells derived from the same patient have significantly higher migration levels toward SDF-1. In contrast to their transwell migration potential, the SDF-1-mediated integrin-dependent polarization and migration of the Ph+CD34+CXCR4+ cells through extracellular matrix-like gels were significantly lower than for normal cells. Concomitantly, binding of these cells to vascular cell adhesion molecule-1 or fibronectin, in the presence of SDF-1, was also substantially lower. These findings suggest a major role for SDF-1-mediated, integrin-dependent BM retention of Ph+CD34+ cells.
Collapse
MESH Headings
- Antigens, CD34
- Cell Adhesion/drug effects
- Cell Movement/drug effects
- Chemokine CXCL12
- Chemokines, CXC/pharmacology
- Chemokines, CXC/physiology
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/physiology
- Humans
- Integrin alpha4beta1
- Integrins/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood
- Philadelphia Chromosome
- Receptors, CXCR4/metabolism
- Receptors, Fibronectin/metabolism
- Receptors, Lymphocyte Homing/metabolism
- Receptors, Very Late Antigen/metabolism
Collapse
Affiliation(s)
- Amnon Peled
- Hadassah University Hospital, Gene Therapy Institutem, Jerusalem, Israel.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kataoka I, Shinagawa K, Shiro Y, Okamoto S, Watanabe R, Mori T, Ito D, Harada M. Multiple sclerosis associated with interferon-alpha therapy for chronic myelogenous leukemia. Am J Hematol 2002; 70:149-53. [PMID: 12111789 DOI: 10.1002/ajh.10090] [Citation(s) in RCA: 24] [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
We report a chronic myelogenous leukemia (CML) patient in chronic phase (CP) who developed multiple sclerosis (MS) in association with interferon-alpha (IFN-alpha) administration. In our patient, recombinant IFN-alpha2b therapy induced hematologically complete and cytogenetically major partial response for CML first, and sequential central nervous system dysfunction evolved, which subsided shortly after the cessation of its administration. Restarting IFN-alpha therapy by changing to a natural type of IFN-alpha resulted in rapid exacerbation of MS. The patient's neurological symptoms progressed gradually, but partial hematologic response persisted without any IFN-alpha derivatives or anti-cancer agents until a matched unrelated donor transplant procedure was performed. Myeloablative therapy led to lasting stable state of MS and finally to complete cytogenetic remission of CML. This patient's presenting clinical course and laboratory data suggest that both exertion of anti-leukemic activity and autoimmune process of MS might be mediated by mutual mechanisms, such as enhancement of specific cellular immunity induced by IFN-alpha.
Collapse
Affiliation(s)
- Itaru Kataoka
- Department of Internal Medicine II, Okayama University Medical School, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Verma A, Platanias LC. Signaling via the interferon-alpha receptor in chronic myelogenous leukemia cells. Leuk Lymphoma 2002; 43:703-9. [PMID: 12153154 DOI: 10.1080/10428190290016782] [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: 01/03/2023]
Abstract
It is well established that IFNalpha has significant clinical activity in the treatment of chronic myelogenous leukemia (CML). This cytokine has been used for many years in the management of patients in the chronic phase of the disease, but the mechanisms by which it induces growth inhibitory effects in CML-cells have not been elucidated. Understanding the signaling mechanisms by which the Type I IFN receptor transduces growth inhibitory signals in BCR-ABL expressing cells should prove very valuable, as it may result in the design of new, more specific pharmacological compounds that target the same cellular cascades. Recent evidence indicates that, in addition to the classic IFN-activated Jak-Stat pathway, the Type I IFN receptor engages in its signaling cascade the CrkL-adapter protein, which is also a substrate for the kinase activity of the BCR-ABL oncogene. In addition, it appears that activation of a member of the Map kinase (MAPK) family of proteins, the p38 MAPK, is essential for the generation of the antileukemic effects of IFNalpha. This review summarizes the recent advances in the-field of interferon signaling in CML cells and discusses the implications of identifying signaling proteins that mediate IFNalpha-induced growth inhibition.
Collapse
Affiliation(s)
- Amit Verma
- Department of Medicine, University of Illinois at Chicago and West Side Veterans Adminstration Medical Center, IL 60607, USA
| | | |
Collapse
|
12
|
Cashman J, Clark-Lewis I, Eaves A, Eaves C. Stromal-derived factor 1 inhibits the cycling of very primitive human hematopoietic cells in vitro and in NOD/SCID mice. Blood 2002; 99:792-9. [PMID: 11806978 DOI: 10.1182/blood.v99.3.792] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Stromal-derived factor 1 (SDF-1) is a -CXC- chemokine that plays a critical role in embryonic and adult hematopoiesis, and its specific receptor, CXCR4, has been implicated in stem cell homing. In this study, it is shown that the addition of SDF-1 to long-term cultures (LTCs) of normal human marrow can selectively, reversibly, and specifically block the S-phase entry of primitive quiescent erythroid and granulopoietic colony-forming cells (CFCs) present in the adherent layer. Conversely, addition of anti-SDF-1 antibody or SDF-1(G2), a specific CXCR4 antagonist, to preactivated human LTCs prevented both types of primitive CFCs from re-entering a quiescent state, demonstrating that endogenous SDF-1 contributes to the control of primitive CFC proliferation in the LTC system. Interestingly, SDF-1 failed to arrest the proliferation of primitive chronic myeloid leukemia CFCs in the adherent layer of LTCs containing normal marrow stromal cells. In vivo, injection of SDF-1 arrested the cycling of normal human LTC-initiating cells as well as primitive CFCs in the marrow of nonobese diabetic/severe combined immunodeficient mice engrafted with human cord blood cells. Conversely, injection of the antagonist, SDF-1(G2), reactivated the cycling of quiescent primitive human CFCs present in the marrow of mice engrafted with human marrow cells. These studies are the first to demonstrate a potential physiological role of SDF-1 in regulating the cell-cycle status of primitive hematopoietic cells and suggest that the deregulated cycling activity of primitive chronic myeloid leukemia (CML) cells is due to the BCR-ABL-mediated disruption of a pathway shared by multiple chemokine receptors.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/cytology
- Bone Marrow Cells/drug effects
- Bone Marrow Transplantation
- Cell Cycle/drug effects
- Cell Division/drug effects
- Cells, Cultured
- Chemokine CXCL12
- Chemokines, CXC/administration & dosage
- Chemokines, CXC/antagonists & inhibitors
- Chemokines, CXC/pharmacology
- Fetal Blood/cytology
- Fetal Blood/drug effects
- Graft Survival/drug effects
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mice, Inbred NOD/blood
- Mice, SCID/blood
- Stem Cells/cytology
- Stem Cells/drug effects
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Johanne Cashman
- Terry Fox Laboratory, British Columbia Cancer Agency, 601 West 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | | | | | | |
Collapse
|
13
|
Thiele J, Kvasnicka HM, Schmitt-Graeff A, Goekce D, Diehl V, Zankovich R, Schaefer HE. Therapy-related changes of CD34+ progenitor cells in chronic myeloid leukemia: a morphometric study on sequential trephine biopsies. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2001; 10:827-36. [PMID: 11798509 DOI: 10.1089/152581601317210926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In chronic myeloid leukemia (CML), it has been assumed that the number of CD34(+) progenitor cells (PGCs) provides useful diagnostic and prognostic information regarding the evolution of accelerated phase and blastic crisis. However, until now no information is available about changes of this peculiar precursor cell population during therapy or possible associations with the other bone marrow constituents. For this reason, a retrospective clinicopathological study was performed on 83 patients with CML including 209 sequential bone marrow biopsies (intervals ranging between 6 and 143 months) and immunohistological staining of CD34(+) cells (QBEND10), megakaryocyte precursors (CD61), and erythropoiesis (Ret 40f). According to treatment modalities, three different groups of patients could be distinguished that received either monotherapy by interferon-alpha2b (IFN-alpha2b) or hydroxyurea (HU) and a combination of both. In comparison with a control group, morphometry revealed a significant increase in the quantity of CD34(+) PGCs per hematopoiesis (cellularity) in the CML bone marrow before treatment. Independently of treatment modalities and presentation of clinical findings nonresponding patients were generally characterized by a higher amount of progenitors in the initial biopsy specimens. Furthermore, calculation of the CD34(+) cell growth index showed a significant and rapid progression in nonresponding patients and in those developing an accelerated or blastic phase during therapy. This feature was prominently expressed following IFN treatment and related to a failing regeneration of nucleated erythroid precursors. In patients with a myelofibrotic bone marrow at onset no differences in the number of CD34(+) PGCs were recognizable in the pretreatment biopsies. This finding contrasted a significant and gradual change in progenitor cell frequency under treatment and evolving myelofibrosis. Opposed to HU therapy, the latter feature was explicitly detectable in the IFN group. In conclusion, the incidence of CD34(+) PGCs in the CML bone marrow reflects therapeutic efficacy. By demonstrating a significant relationship between fiber content and quantity of CD34(+) cells during treatment, experimental findings concerning the complex functional interactions between the fibrous stroma compartment and progenitor cell differentiation and proliferation are elucidated.
Collapse
Affiliation(s)
- J Thiele
- Institute of Pathology, University of Cologne, Germany.
| | | | | | | | | | | | | |
Collapse
|
14
|
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
|
15
|
Verfaillie CATHERINEM, Gupta PANKAJ, Prosper FELIPE, Hurley RANDY, Lundell BEVERLY, Bhatia RAVI. The Hematopoietic Microenvironment: Stromal Extracellular Matrix Components As Growth Regulators For Human Hematopoietic Progenitors. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2001; 4:321-333. [PMID: 11399573 DOI: 10.1080/10245332.1999.11746456] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
16
|
Lindauer M, Fischer T. Interferon-alpha combined with cytarabine in chronic myelogenous leukemia - clinical benefits. Leuk Lymphoma 2001; 41:523-33. [PMID: 11378570 DOI: 10.3109/10428190109060343] [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: 11/13/2022]
Abstract
During the last decade, several studies have evaluated the treatment of chronic phase chronic myeloid leukemia (CML) with a combination of interferon (IFN)-alpha and low- dose cytarabine (Ara-C). This combination therapy has been shown to be superior compared to monotherapy with IFN-alpha in randomized studies with regard to hematologic and cytogenetic remissions. However, the survival benefit is small, and the toxicity of the combination therapy is high. This paper reviews the published studies on IFN-alpha/low-dose Ara-C for the treatment of chronic phase CML and discusses the value of the combination therapy.
Collapse
Affiliation(s)
- M Lindauer
- 3rd Medical Department, Johannes Gutenberg-University, Mainz, Germany.
| | | |
Collapse
|
17
|
Park CC, Bissell MJ, Barcellos-Hoff MH. The influence of the microenvironment on the malignant phenotype. MOLECULAR MEDICINE TODAY 2000; 6:324-9. [PMID: 10904250 DOI: 10.1016/s1357-4310(00)01756-1] [Citation(s) in RCA: 296] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Normal tissue homeostasis is maintained by dynamic interactions between epithelial cells and their microenvironment. As tissue becomes cancerous, there are reciprocal interactions between neoplastic cells, adjacent normal cells such as stroma and endothelium, and their microenvironments. The current dominant paradigm wherein multiple genetic lesions provide both the impetus for, and the Achilles heel of, cancer might be inadequate to understand cancer as a disease process. In the following brief review, we will use selected examples to illustrate the influence of the microenvironment in the evolution of the malignant phenotype. We will also discuss recent studies that suggest novel therapeutic interventions might be derived from focusing on microenvironment and tumor cells interactions.
Collapse
Affiliation(s)
- C C Park
- Joint Center for Radiation Therapy, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
18
|
Senyukov VV, Seledtsov VI, Poveshchenko OV, Taraban VY, Kozlov VA. Soluble factor and cell-cell interaction in cytostasis induced by bone marrow cells. Bull Exp Biol Med 2000; 129:559-61. [PMID: 11022249 DOI: 10.1007/bf02434876] [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] [Received: 03/22/2000] [Indexed: 11/30/2022]
Abstract
Cell-cell interaction and soluble low-molecular-weight products are probably involved in in vitro inhibition of leukemic cell growth by bone marrow cells.
Collapse
Affiliation(s)
- V V Senyukov
- Institute of Clinical Immunology, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk.
| | | | | | | | | |
Collapse
|
19
|
Abstract
Clinical observation and laboratory evidence suggest that immune mechanisms play an important role in the natural control of evolution of the Ph+ clone in chronic phase as well as during progression of chronic myelogenous leukemia (CML). The understanding of these mechanisms could facilitate development of innovative therapeutic approaches. Due to bcr-abl translocation, CML cells carry an intrinsic resistance to apoptotic signals. However, resistance to apoptosis is not absolute and can be overcome through enhancement of immune-mediated pathways, e.g., during graft vs. leukemia reaction after allogeneic bone marrow transplantation, or during interferon-alpha (IFN-alpha) therapy. Among the effector mechanisms, T-lymphocyte-mediated killing of target cells via Fas-receptor (Fas-R) triggering plays an important role in the elimination of malignant cells, including CML cells. Although CML Ph+ progenitor cells express Fas-R, the expression levels are variable and do not correlate with clinical parameters. In addition, CML progenitor cells also express functional Fas-ligand (Fas-L), which may be an important immune surveillance escape factor. IFN-alpha can greatly upmodulate Fas-R expression, an effect that seems to be more pronounced in CML compared to normal cells, while Fas-L expression levels are not affected by IFN-alpha, thereby improving their susceptibility to elimination by the immune system. Responsiveness to Fas-induced apoptosis following stimulation with IFN-alpha correlates with the clinical effects of IFN-alpha therapy. This effect seems to be associated with decreased bcr-abl protein levels, which are influenced by Fas via posttranscriptional modulation. In comparison to the chronic phase, CML cells derived from patients in blast crisis are refractory to Fas-mediated apoptosis, regardless of the expression levels of Fas, suggesting that an immune-mediated selection pressure could result in acquisition of Fas-resistance. In the future, enhancement of immunological recognition and elimination of CML cells may prove to be an effective therapeutic approach directed towards the cure of CML.
Collapse
Affiliation(s)
- C Selleri
- Division of Hematology, Federico II University of Naples, Italy
| | | |
Collapse
|
20
|
Budak-Alpdoğan T, Alpdoğan O, Akoğlu T. Morphological and functional characteristics of short-term and long-term bone marrow cultures in chronic myelogenous leukemia. Am J Hematol 1999; 62:212-20. [PMID: 10589076 DOI: 10.1002/(sici)1096-8652(199912)62:4<212::aid-ajh3>3.0.co;2-s] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Clonogenic capacity of bone marrow progenitors and stromal layers established from bone marrow of 12 patients with CML and 13 healthy controls were evaluated. The initial BFU-E and CFU-GM contents were slightly higher in the CML patients (p > 0.05) in contrast to CFU-GEMM. CFU-GEMM was lower in the patients compared to healthy controls (p < 0.001). In long-term cultures, the number of non-adherent cell population and total clonogenic progenitor cell content decreased gradually in both groups. Weekly evaluation of stromal confluency of adherent cells revealed that establishment of adherent stromal layer was slower in CML patients than in control samples (p < 0.05). At the end of fourth week, the number of samples presenting confluency was 41.7% in the CML group compared with 92.3% in the controls. The initial CD34 positive cell content of the bone marrow samples was similar in both groups. Although CD34 positive cell number in the adherent stromal layer was well preserved in the control group at the end of 4 weeks, this figure decreased significantly in the CML group. The numbers of total adherent cells as well as the total clonogenic progenitor content of adherent layer were also lower in the CML group (3.03% vs 98.2%). When normal CD34+ cells were cultured on IFN-alpha-treated stromal layer followed by the assessment of the long-term culture initiating cells, a reduced capacity to support hemopoietic growth was observed with IFN-alpha-treated normal stroma. This reduction was even higher when CML stroma was treated with IFN-alpha followed by the seeding of the normal CD34+ cells on this stromal layer (26.9% vs 42.8%). These findings show that stromal cells are abnormal in CML patients as well as the progenitor cells, and IFN-alpha treatment causes further defects of the stromal cells.
Collapse
Affiliation(s)
- T Budak-Alpdoğan
- Department of Hematology and Immunology, Marmara University Hospital, Istanbul, Turkey
| | | | | |
Collapse
|
21
|
Affiliation(s)
- R Chopra
- Christie Hospital and Paterson Institute for Cancer Research, Manchester, UK
| | | | | |
Collapse
|
22
|
Bellucci R, Sala R, De Propris MS, Cordone I, de Fabritiis P. Interferon-alpha and bcr-abl antisense oligodeoxynucleotides in combination enhance the antileukemic effect and the adherence of CML progenitors to preformed stroma. Leuk Lymphoma 1999; 35:471-81. [PMID: 10609784 DOI: 10.1080/10428199909169611] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
We have studied the in vitro effect of IFN-alpha and bcr-abl antisense oligodeoxynucleotides (As ODN) alone and in combination with the aim of enhancing the antileukemic activity of the two single agents and evaluating whether the two agents in combination might restore the adherence capacity of chronic myeloid leukemia (CML) progenitors to preformed stroma. We have also correlated the increased adhesion found after in vitro treatment with the expression of adhesion molecules on leukemic progenitors. Incubation of the BV173 cell line with escalating doses of IFN-alpha (100-10000 U/ml) showed a colony growth inhibition between 10 and 30%. IFN-alpha and junction-specific As ODN in combination showed a greater antiproliferative effect compared to that observed with the two agents used alone. In particular, As ODN at a concentration of 40 microg/ml in combination with IFN-alpha at 100 and 1000 U/ml showed a greater inhibitory effect compared to that obtained with IFN-alpha only. Addition of As ODN to IFN-alpha at 10000 U/ml did not result in a greater BV173 inhibition. In a further set of experiments, primary cells from 16 CML patients at diagnosis were incubated with 40 microg/ml of J-spec As ODN, several control ODNs and IFN-alpha at 1000 U/ml alone and in combination. A significantly greater elimination of CML progenitors was found after treatment with the combination of IFN-alpha and J-spec As ODN, compared to any other treatment group, confirmed also by a more marked effect on p210 expression. The deficient adhesion of CML progenitors on human preformed stroma was restored at levels similar to that of normal bone marrow cells after treatment with IFN-alpha and/or J-spec As ODN, while the phenotypic analysis showed that the combined treatment increased significantly the expression of CD49b and CD62L on CML CD34+ cells. However, when the expression of adhesion molecules was blocked with specific monoclonal antibodies, only CD49d (expressed on more than 90% of CML CD34+ cells) appeared to influence the functional activity of adhesion molecules. In conclusion, IFN-alpha and bcr-abl As ODN in combination exert a marked in vitro antileukemic activity and could be a useful approach for in vitro purging of CML cells prior to autologous transplantation.
Collapse
MESH Headings
- Antigens, CD34/analysis
- Blast Crisis/pathology
- Bone Marrow Purging/methods
- Cell Adhesion/drug effects
- Cell Culture Techniques/methods
- Cell Division/drug effects
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- Drug Synergism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/biosynthesis
- Fusion Proteins, bcr-abl/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/drug effects
- Humans
- Immunologic Factors/administration & dosage
- Immunologic Factors/pharmacology
- Immunophenotyping
- Interferon-alpha/administration & dosage
- Interferon-alpha/pharmacology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Oligodeoxyribonucleotides, Antisense/administration & dosage
- Oligodeoxyribonucleotides, Antisense/pharmacology
- Stromal Cells/cytology
- Tumor Cells, Cultured/drug effects
- Tumor Stem Cell Assay
Collapse
Affiliation(s)
- R Bellucci
- Dipartimento di Biotecnologie Cellulari ed Ematologia, University La Sapienza, Rome, Italy
| | | | | | | | | |
Collapse
|
23
|
Hadida F, De Maeyer E, Cremer I, Autran B, Baggiolini M, Debré P, Vieillard V. Acquired constitutive expression of interferon beta after gene transduction enhances human immunodeficiency virus type 1-specific cytotoxic T lymphocyte activity by a RANTES-dependent mechanism. Hum Gene Ther 1999; 10:1803-10. [PMID: 10446920 DOI: 10.1089/10430349950017482] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CTL lines directed against HIV-1 antigens were generated from infected individuals and were transduced by the HMB-K(b)HuIFNbeta vector, resulting in low, constitutive expression of interferon beta (IFN-beta). The IFN-beta-transduced cells showed markedly increased HIV-1-specific, MHC class I-restricted CTL activity against HIV-1-LAI Gag, Pol, or Env antigens. This effect of IFN-beta was correlated with an overexpression of RANTES and completely abrogated by RANTES-blocking antibody. The present results provide the first evidence that IFN-beta transduction of CTL lines enhances HIV-specific cytotoxic activities through an upregulation of RANTES production. The efficient elimination of HIV-infected cells by IFN-beta-transduced CTL lines makes this gene therapy approach an attractive treatment for AIDS.
Collapse
Affiliation(s)
- F Hadida
- Laboratoire d'Immunologie Cellulaire, UMR 7627 CNRS, Hôpital Pitié-Salpétrière, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
24
|
Verfaillie CM. Chronic myelogenous leukemia: from pathogenesis to therapy. JOURNAL OF HEMATOTHERAPY 1999; 8:3-13. [PMID: 10192297 DOI: 10.1089/106161299320523] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
MESH Headings
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Oncogenes
- Stem Cells/metabolism
- Stem Cells/pathology
Collapse
Affiliation(s)
- C M Verfaillie
- Department of Medicine, University of Minnesota Cancer Center, Minneapolis 55455, USA
| |
Collapse
|
25
|
Lane CM, Guo XY, Macaluso LH, Yung KC, Deisseroth AB. Presence of P210bcrabl is associated with decreased expression of a beta chemokine C10 gene in a P210bcrabl-positive myeloid leukemia cell line. Mol Med 1999; 5:55-61. [PMID: 10072448 PMCID: PMC2230372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Chronic myelogenous leukemia (CML) is thought to start with the acquisition of the t(9;22) chromosomal translocation that codes for the P210bcrabl tyrosine-specific protein kinase. The CML cells exhibit anchorage-independent cell growth and genetic instability. After the initial phase, the cells acquire the phenotype of growth factor-independent growth. After the chronic phase, the disease evolves into the accelerated and blastic phases through the process of sequential random mutation. MATERIALS AND METHODS To identify some of the genetic changes that contribute to the phenotype of blastic and accelerated phase cells, we used differential display PCR to compare levels of cDNA reverse transcripts of mRNA in 32Dc13 cells and 32Dc13 cells that were stably transfected with a bcrabl cDNA plasmid in a constitutively expressed transcription unit. These cells were designated 32Dc13P210bcrabl. For these studies, we used the 32D myeloid leukemia cell line, which depends on IL-3 for growth. RESULTS Following introduction of the bcr-abl cDNA through transfection, the cell line became growth factor independent, mimicking the change in phenotype that occurs during the later phases of CML. These differential display screening assays detected altered levels of transcripts for 28 genes. Of interest to the biology of growth factor-independent growth in the bcrabl-positive 32D cells was the fact that the C10 beta chemokine gene was expressed at higher levels in the 32Dc13 cells than in the 32Dc13P210bcrabl cells. CONCLUSIONS These studies show that a C10 beta chemokine gene was expressed at different levels with or without P210bcrabl.
Collapse
MESH Headings
- Base Sequence
- Chemokines, CC/genetics
- DNA Primers/genetics
- DNA, Complementary/genetics
- DNA, Neoplasm/genetics
- Fusion Proteins, bcr-abl/genetics
- Gene Expression
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Phenotype
- Polymerase Chain Reaction
- RNA, Messenger/genetics
- RNA, Neoplasm/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- C M Lane
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
26
|
Presence of P210bcrabl Is Associated with Decreased Expression of a Beta Chemokine C10 Gene in a P210bcrabl-Positive Myeloid Leukemia Cell Line. Mol Med 1999. [DOI: 10.1007/bf03402139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
27
|
Expression of Macrophage Inflammatory Protein-1 Receptors in Human CD34+ Hematopoietic Cells and Their Modulation by Tumor Necrosis Factor- and Interferon-γ. Blood 1998. [DOI: 10.1182/blood.v92.9.3073] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Macrophage inflammatory protein-1 (MIP-1) can stimulate growth inhibitory and potent chemotactic functions in hematopoietic cells. To investigate whether the action of MIP-1 may be regulated at the cellular receptor level, we studied the expression and modulation of MIP-1 receptors on CD34+ cells isolated from normal bone marrow (NBM), umbilical cord blood (CB), and leukapheresis products (LP). Expression of MIP-1 receptors on CD34+cells was analyzed by two-color flow cytometry using a biotinylated MIP-1 molecule. The mean percentage of LP CD34+ cells expressing the MIP-1 receptors was 67.7 ± 7.2% (mean ± SEM; n = 22) as compared with 89.9 ± 2.6% (n = 10) and 74.69 ± 7.04% (n = 10) in CB and NBM, respectively (P = .4). The expression of the MIP-1 receptor subtypes on LP CD34+ cells was studied by indirect immunofluorescence using specific antibodies for the detection of CCR-1, CCR-4, and CCR-5. Microscopical examination revealed a characteristic staining of the cytoplasmic cell membrane for all three receptor subtypes. Detailed analysis of two LP samples showed that 65.8%, 4.4%, and 30.5% of CD34+ cells express CCR-1, CCR-4, and CCR-5, respectively. Culture of LP CD34+ cells for 24 to 36 hours in the presence of tumor necrosis factor- (TNF-) and interferon-γ (IFN-γ) resulted in a significant increase in MIP-1 receptor expression. TNF- induced MIP-1 receptor upregulation in a time- and concentration-dependent manner. Our results suggest that inhibitory cytokines produced by the bone marrow microenvironment are likely to be involved in the regulation of MIP-1 receptor expression on hematopoietic cells.
© 1998 by The American Society of Hematology.
Collapse
|
28
|
Expression of Macrophage Inflammatory Protein-1 Receptors in Human CD34+ Hematopoietic Cells and Their Modulation by Tumor Necrosis Factor- and Interferon-γ. Blood 1998. [DOI: 10.1182/blood.v92.9.3073.421k42_3073_3081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Macrophage inflammatory protein-1 (MIP-1) can stimulate growth inhibitory and potent chemotactic functions in hematopoietic cells. To investigate whether the action of MIP-1 may be regulated at the cellular receptor level, we studied the expression and modulation of MIP-1 receptors on CD34+ cells isolated from normal bone marrow (NBM), umbilical cord blood (CB), and leukapheresis products (LP). Expression of MIP-1 receptors on CD34+cells was analyzed by two-color flow cytometry using a biotinylated MIP-1 molecule. The mean percentage of LP CD34+ cells expressing the MIP-1 receptors was 67.7 ± 7.2% (mean ± SEM; n = 22) as compared with 89.9 ± 2.6% (n = 10) and 74.69 ± 7.04% (n = 10) in CB and NBM, respectively (P = .4). The expression of the MIP-1 receptor subtypes on LP CD34+ cells was studied by indirect immunofluorescence using specific antibodies for the detection of CCR-1, CCR-4, and CCR-5. Microscopical examination revealed a characteristic staining of the cytoplasmic cell membrane for all three receptor subtypes. Detailed analysis of two LP samples showed that 65.8%, 4.4%, and 30.5% of CD34+ cells express CCR-1, CCR-4, and CCR-5, respectively. Culture of LP CD34+ cells for 24 to 36 hours in the presence of tumor necrosis factor- (TNF-) and interferon-γ (IFN-γ) resulted in a significant increase in MIP-1 receptor expression. TNF- induced MIP-1 receptor upregulation in a time- and concentration-dependent manner. Our results suggest that inhibitory cytokines produced by the bone marrow microenvironment are likely to be involved in the regulation of MIP-1 receptor expression on hematopoietic cells.
© 1998 by The American Society of Hematology.
Collapse
|
29
|
Abstract
Interferons (IFN) have had increasing clinical usage in the treatment of a variety of disorders, at times being used in combination with chemo- or radiotherapy. However, interferons may have inhibitory effects on hematopoietic stem cell proliferation and the effects of this cytokine's use on long-term hematologic function have not been studied. We performed the competitive repopulation assay in the murine system, using cells exposed to irradiation or single-dose chemotherapy with or without concomitant IFN-gamma use. IFN-gamma alone had no deleterious effects on hematopoietic stem cell productivity. We measured the repopulating ability of exhaustible multilineage precursors that were present at early stages of marrow repopulation after competitive repopulation (30 days). These progenitors were minimally impacted by cyclophosphamide (CTX) with or without IFN-gamma. Irradiation (XRT) and CTX alone produced significant repopulating defects in the most primitive hematopoietic stem cell, PHSC. Addition of IFN to either treatment regimen resulted in protection of PHSC, with improved repopulating ability, although the levels of donor marrow reached control levels only when CTX and IFN were used together. The results of multiple use of IFN with chemotherapy must be studied further, but IFN may offer hematologic radio- and chemoprotection, in addition to its antitumor properties in clinical protocols for treatment of cancers.
Collapse
Affiliation(s)
- R V Gardner
- LSU Medical Center, New Orleans, Louisiana 70112, USA
| |
Collapse
|
30
|
Prosper F, Stroncek D, McCarthy JB, Verfaillie CM. Mobilization and homing of peripheral blood progenitors is related to reversible downregulation of alpha4 beta1 integrin expression and function. J Clin Invest 1998; 101:2456-67. [PMID: 9616217 PMCID: PMC508835 DOI: 10.1172/jci188] [Citation(s) in RCA: 124] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Despite the wide use of mobilized peripheral blood (PB) progenitor cells (PBPC) for clinical transplantation the mechanism(s) underlying their mobilization and subsequent engraftment are still unknown. We compared the adhesive phenotype of CD34(+) colony-forming cells (CFC) in bone marrow (BM) and PB of normal donors before and after administration of granulocyte colony-stimulating factor (G-CSF) for 5 d. G-CSF-mobilized PB CFC cells adhered significantly less to BM stroma, fibronectin, and to the alpha4 beta1 binding fibronectin peptide, CS1, because of decreased expression of the alpha4 integrin. Since incubation of BM CD34(+) cells for 4 d with G-CSF at concentrations found in serum of G-CSF- treated individuals did not affect alpha4-dependent adhesion, G-CSF may not be directly responsible for the decreased alpha4-mediated adhesion of PB CFC. Culture of G-CSF-mobilized PB CD34(+) cells with cytokines at concentrations found in BM stromal cultures upregulated alpha4 expression and restored adhesion of mobilized PB CFC to stroma, fibronectin, and CS1. Adhesion of cultured, mobilized PB CFC to stroma and CS1 could not be further upregulated by the beta1 activating antibody, 8A2. This indicates acquisition of a maximally activated alpha4 beta1 integrin once PB CFC have been removed from the in vivo mobilizing milieu. Thus, decreased alpha4 expression on CD34(+) CFC in PB may be responsible for the aberrant circulation of mobilized PB CD34(+) cells. Reexpression of a maximally activated alpha4 beta1 integrin on mobilized PB CFC removed from the mobilizing in vivo milieu may contribute to the early engraftment of mobilized PBPC.
Collapse
Affiliation(s)
- F Prosper
- Stem Cell Biology Program and Division of Hematology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
31
|
Oostendorp RA, Spitzer E, Brandl M, Eaves CJ, Dörmer P. Evidence for differences in the mechanisms by which antibodies against CD44 promote adhesion of erythroid and granulopoietic progenitors to marrow stromal cells. Br J Haematol 1998; 101:436-45. [PMID: 9633883 DOI: 10.1046/j.1365-2141.1998.00746.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Adhesive interactions between haemopoietic progenitor cells and stromal elements involve a number of different molecules, some of which may be progenitor- lineage- and stage-specific. CD44 is one such molecule, although little is known about the mechanism(s) by which it is involved. In this study, several anti-CD44 monoclonal antibodies (mAb) increased the adherence of clonogenic cells, without affecting the total number of types of progenitors recoverable from the adhesion cultures. All of these mAb recognized epitopes on the globular head of CD44. In contrast, two mAb that recognized other regions of CD44 reduced progenitor adhesion to stroma. The mechanism by which one of the anti-CD44 mAb (L178) enhanced progenitor adhesion did not involve CD44-crosslinking, and was independent of VLA-4-, VLA-5- or LFA-1-mediated interactions, Ca or Mg cations, or accessory cells. In addition, CD44 expression on both progenitors and stromal cells contributed to L178-enhanced progenitor adhesion. Baseline adherence of erythroid progenitors to stroma required tyrosine kinase activity, whereas that of granulopoietic progenitors did not. However, the increase in adhesion did require tyrosine kinase activation. Additional experiments suggested that enhanced adhesion of CFU-GM to stroma may also be adenylate cyclase-dependent. Taken together, the present studies indicate both similarities and differences in the mechanisms of CD44-mediated adhesion of erythroid and granulopoietic progenitors to stromal cells.
Collapse
Affiliation(s)
- R A Oostendorp
- Institute for Experimental Haematology, GSF-National Research Centre for Environment and Health, Munich, Germany
| | | | | | | | | |
Collapse
|
32
|
Abstract
Hematopoietic stem cell differentiation occurs in direct proximity to osteoblasts within the bone marrow cavity. Despite this striking affiliation, surprisingly little is known about the precise cellular and molecular impact of osteoblasts on the bone marrow microenvironment. Recently, it has been proposed that human osteoblasts support the growth of primitive human hematopoietic cells in vitro and possibly in vivo. Evidence to support this hypothesis is reviewed as follows: the influence of osteoblasts on osteoclast development; the participation of osteoblasts in long-term bone marrow cultures; the production of positive hematopoietic regulatory molecules by osteoblasts; the production of cell-cycle inhibitory factors by osteoblasts, and cell-cell interactions between early hematopoietic cells and osteoblasts.
Collapse
Affiliation(s)
- R S Taichman
- Department of Periodontics/Prevention/Geriatrics, University of Michigan Dental School, Ann Arbor 48109-1078, USA
| | | |
Collapse
|
33
|
Abstract
This article reviews the biology of chronic myelogenous leukemia (CML) and its effect on the process of hematopoiesis. The relevance of the BCR-ABL fusion protein as well as murine models are also discussed. CML has been studied more extensively than any other malignancy, yet the correlation between the clinical symptoms of chronic phase CML and the BCR-ABL oncoprotein is poorly understood. Insights from recent efforts both to develop a good in vivo animal model and to characterize the effect of the BCR-ABL oncoprotein on relevant signal molecules may lead to a better understanding of the pathophysiology of chronic phase CML and, thereby, to the development of targeted therapeutic approaches.
Collapse
Affiliation(s)
- C M Verfaillie
- Department of Medicine, University of Minnesota, Minneapolis, USA
| |
Collapse
|
34
|
Ahmed T, Lutton JD, Feldman E, Tani K, Asano S, Abraham NG. Gene transfer of alpha interferon into hematopoietic stem cells. Leuk Res 1998; 22:119-24. [PMID: 9593468 DOI: 10.1016/s0145-2126(97)00133-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gene transfer or gene therapy has advantages in the treatment of a variety of disorders due to its selective expression within specific mammalian cells. IFN-alpha has been used in the management of leukemia, and gene transfer of the IFN-alpha gene into hematopoietic progenitor cells may have great potential for the treatment of chronic myelogenous leukemia (CML). Therefore, we examined the ability of adenovirus (Ad)-IFN-alpha gene construct to transfect normal bone marrow hematopoietic CD34+ stem cells and the production of IFN-alpha protein by these cells. Ad-cytomegalovirus (CMV) promoter-driven IFN-alpha at multiple doses was assessed to transfect highly purified CD34+ cells in liquid culture. Optimal transduction of CD34+ cells with the AdCMV-IFN-alpha construct was achieved using 120 plaque forming units (pfu). Flow cytometric determinations revealed that there was no significant difference in CD34+ cell viability for the 8 or 12-h transfection periods. Immunoassay of IFN-alpha produced by CD34+ cells shows that IFN-alpha levels increased several fold in transfected cells and this was not seen in CD34+ cells transfected with the heme oxygenase gene (HO-1). These in vitro data suggest that adenovirus-mediated gene transfer of IFN-alpha into hematopoietic stem cells can be achieved and that the IFN-alpha protein is produced by viable CD34 progenitor cells.
Collapse
Affiliation(s)
- T Ahmed
- New York Medical College, Department of Pharmacology, Valhalla 10595, USA
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Innovative therapies for chronic myelogenous leukemia (CML) have focused mainly on combining autologous transplantation with another modality of therapy for purging of the graft or treatment of the patient after transplant. Of the three categories of innovative therapies, two are based on studies that demonstrate the bcr/abl gene rearrangement in the pathogenesis of CML, whereas the third is based on the observation that allogeneic disparity is important to maintain remissions in CML. The rationale and data supporting these innovative approaches are reviewed in this article and future strategies are discussed.
Collapse
Affiliation(s)
- J S Miller
- Department of Medicine, University of Minnesota, Minneapolis, USA
| |
Collapse
|
36
|
Jonuleit T, Peschel C, Schwab R, van der Kuip H, Buchdunger E, Fischer T, Huber C, Aulitzky WE. Bcr-Abl kinase promotes cell cycle entry of primary myeloid CML cells in the absence of growth factors. Br J Haematol 1998; 100:295-303. [PMID: 9488616 DOI: 10.1046/j.1365-2141.1998.00564.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cell cycle control of both immature and differentiated primary myeloid normal and chronic-phase chronic myeloid leukaemia (CML) cells to growth factor deprivation was studied. CD34+ cells were cultured in liquid culture. After removal of growth factors for 48 h normal cells were very efficiently arrested with the fraction of cells in S phase reduced by 70.8 +/- 6.5% in CD34+ and 50.5 +/- 4.2% in CD34- cells. In contrast, a significantly higher proportion of leukaemic cells remained in S phase. The fraction of S-phase cells was reduced by only 29.3 +/- 5.7% in CD34+ CML cells and 21.2 +/- 3.8% in CD34- cells. This abnormal negative cell cycle control in leukaemic cells was specific for growth factor deprivation. Reaction to IFN-alpha and TNF-alpha treatment was identical both in normal and CML cells. Equal quantities of the cytokines TNF-alpha, IL-1alpha, IL-1RA and IL-6 were produced by CML and normal cells. However, production of GM-CSF, with a median of 11 +/- 5 pg/ml, was found only in the supernatants of CML cells. But antibodies to GM-CSF did not restore growth factor dependence of the leukaemic cells. The defect was completely corrected by the abl-specific tyrosine kinase inhibitor CGP 57148 without effecting cell cycle control of normal cells. Our results demonstrate a directly Bcr-Abl-dependent defective response of both immature and differentiated primary myeloid CML cells to growth factor deprivation.
Collapse
Affiliation(s)
- T Jonuleit
- Department of Haematology, Robert Bosch Hospital, Stuttgart, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Bhatia R, Verfaillie CM. The effect of interferon-alpha on beta-1 integrin mediated adhesion and growth regulation in chronic myelogenous leukemia. Leuk Lymphoma 1998; 28:241-54. [PMID: 9517496 DOI: 10.3109/10428199809092680] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is considerable interest in understanding the mechanisms by which interferon-alpha can induce hematological and cytogenetic remissions and prolong survival in patients with chronic myelogenous leukemia (CML). There is evidence that the selective expansion and growth advantage of malignant hematopoietic progenitors in CML may be related, at least in part, to their deficient responsiveness to normal negative regulation by the marrow stromal microenvironment and that interferon-alpha may restore normal hematopoiesis in CML by restoring normal microenvironmental regulation of malignant CML progenitor proliferation. In normal hematopoiesis, beta1 integrin receptors mediate progenitor adhesion to stroma. Stimulation of beta1 integrin receptors on normal progenitors results in transmission of proliferation inhibitory signals. CML progenitors demonstrate defective beta1 integrin receptor-mediated adhesion and regulation of proliferation. We have shown that interferon-alpha can restore both beta1 integrin mediated adhesion as well as integrin-mediated proliferation inhibition of CML progenitors. Interferon-alpha enhances CML integrin function through direct effects on CML progenitors as well as indirect effects on stroma. Restored beta1 integrin-mediated regulation of malignant CML progenitor proliferation may result in restoration of normal hematopoiesis in CML patients treated with interferon-alpha. There is evidence that abnormal integrin mediated signaling in CML progenitors may result from abnormalities in integrin-cytoskeletal interactions induced by the p210BCR/ABL tyrosine kinase. Although the exact mechanisms by which interferon-alpha restores normal integrin signaling in CML are not known, preliminary studies indicate that this may at least partly be related to the restoration of normal integrin-cytoskeletal interactions. The above studies offer some insights into the mechanisms of abnormal hematopoietic regulation in CML and the mechanisms by which interferon-alpha may restore normal hematopoiesis, in this disease.
Collapse
Affiliation(s)
- R Bhatia
- Department of Hematology and Bone Marrow Transplantation, City of Hope National Medical Center, Duarte, California, USA.
| | | |
Collapse
|
38
|
Abstract
This article discusses briefly the molecular consequences of the BCR-ABL fusion gene. It then reviews the current evidence supporting the notion that chronic myelogenous leukemia in its chronic phase is a clonal, hematopoietic, stem cell disease in which malignant hematopoietic stem and progenitor cells respond to "normal" external proliferation and differentiation stimuli, but in which such responses are altered owing to defects in the stem and progenitor cells as a result of the BCR-ABL oncogene.
Collapse
MESH Headings
- Cell Adhesion
- Cell Division
- Cytokines/pharmacology
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/pathology
- Humans
- Immunophenotyping
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology
Collapse
Affiliation(s)
- C M Verfaillie
- Department of Medicine, University of Minnesota, Minneapolis, USA
| |
Collapse
|
39
|
Feldman E, Ahmed T, Lutton JD, Farley T, Tani K, Freund M, Asano S, Abraham NG. Adenovirus mediated alpha interferon (IFN-alpha) gene transfer into CD34+ cells and CML mononuclear cells. Stem Cells 1997; 15:386-95. [PMID: 9323802 DOI: 10.1002/stem.150386] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Gene transfer or gene therapy has advantages in the treatment of a variety of disorders due to its selective expression within specific mammalian cells. Interferon-alpha (IFN-alpha) has been used in the management of leukemia but its diverse adverse activities with multiple potential side effects, possibly unrelated to therapeutic targets, may negatively influence the ability of IFN-alpha to treat this disorder. Therefore, we examined the ability of adenovirus (Ad)-IFN-alpha gene construct to transfect normal (CD34+ cells) and chronic myelogenous leukemia (CML) bone marrow mononuclear cells (BMMNC) and the transient overexpression of IFN-alpha in these cells. Ad-cytomegalovirus promoter driven IFN-alpha (AdCMV-IFN-alpha) at multiple doses was assessed to transfect highly purified CD34+ cells in liquid culture, and optimal transduction of CD34+ cells was achieved using 120 plaque forming units. Flow cytometric determinations revealed that there was no significant difference in cell viability for the 4 h or 24 h transfection periods. Immunoassay of IFN-alpha produced by CD34+ cells shows that IFN-alpha levels increased several fold in transfected cells. Transient expression of the IFN-alpha gene did not suppress proliferation of CD34+ progenitors as indicated by BFU-E or colony forming units-granulocyte-macrophage (CFU-GM) growth. Reverse transcriptase/polymerase chain reaction analysis of RNA from CD34+ harvested CFU-GM progenitor cells demonstrated transient IFN-alpha mRNA expression. Similarly, CML BMMNC were transfected with AdCMV-IFN-alpha under similar conditions as described for CD34+ cells. BMMNC cells exposed to adenovirus for 24 h and 48 h were found to express IFN-alpha at a substantial level. This in vitro data suggest that Ad-mediated gene transfer of IFN-alpha into hematopoietic stem cells can be achieved and that the IFN-alpha gene can be translated into its specific mRNA in CD34 progenitor cells.
Collapse
MESH Headings
- Adenoviridae/genetics
- Antigens, CD34/genetics
- Bone Marrow Cells/cytology
- Colony-Forming Units Assay
- Cytomegalovirus/genetics
- DNA, Complementary/analysis
- DNA, Complementary/genetics
- DNA, Complementary/metabolism
- Dose-Response Relationship, Drug
- Gene Transfer Techniques
- Genetic Vectors
- Humans
- Interferon-alpha/genetics
- Interferon-alpha/therapeutic use
- Interferon-alpha/toxicity
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/physiology
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Stem Cells/drug effects
- Stem Cells/immunology
- Stem Cells/physiology
- Transfection
Collapse
Affiliation(s)
- E Feldman
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
The stromal cells are integral components of the bone marrow (BM) that provide and respond to cytokines, and offer adhesion elements for hematopoietic cell homing. Steady-state hematopoiesis results from a balance between negative and positive acting cytokines, whose expression is in addition the subject of regulation. TGF-beta1 is present in the BM microenvironment and plays a central role in controlling hematopoiesis, by modulating the synthesis of cytokines and cytokine receptors, as well as cell adhesion molecules. We have recently described the TGF-beta1 receptor system expressed on human BM stromal cells. The consequences of signalling through this system, which can affect stromal cell function, and hence, influence the hematopoiesis, is the subject of this review.
Collapse
Affiliation(s)
- M M Robledo
- Centro de Investigaciones Biológicas, Departamento de Immunología Velásquez, Madrid, Spain
| | | |
Collapse
|
41
|
Pierson BA, Miller JS. The role of autologous natural killer cells in chronic myelogenous leukemia. Leuk Lymphoma 1997; 27:387-99. [PMID: 9477121 DOI: 10.3109/10428199709058306] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic myelogenous leukemia (CML) is a lethal disease of the hematopoietic stem cell. Bone marrow transplantation has highlighted the importance of allogeneic disparity in maintaining remissions in CML. However, it has been unclear whether the immune effect against CML is mediated by T cells, natural killer cells (NK) or a combination of both. We have previously demonstrated that autologous activated NK are capable of selectively lysing malignant CML progenitors while sparing benign progenitors. NK effectors may play an important role in CML since NK lytic function, clonogenic frequency and proliferative capacity decrease as CML progresses from chronic phase to advanced phase and blast crisis. Incubation of CML NK with IL-2 is capable of restoring cytolytic activity to normal levels. We hypothesize that activated NK represent a potential therapy against CML to maintain remissions in a minimal residual disease setting induced by autologous transplantation. Clinical trials are in progress to test whether IL-2 based immunotherapy and activated cell infusions play a therapeutic role in CML.
Collapse
MESH Headings
- Disease Progression
- Humans
- Immunity, Cellular
- Immunotherapy, Adoptive
- Interleukin-2/therapeutic use
- Killer Cells, Lymphokine-Activated/drug effects
- Killer Cells, Lymphokine-Activated/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Lymphocyte Activation
- Lymphocyte Subsets
- Remission Induction
Collapse
Affiliation(s)
- B A Pierson
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, USA
| | | |
Collapse
|
42
|
Primitive Long-Term Culture Initiating Cells (LTC-ICs) in Granulocyte Colony-Stimulating Factor Mobilized Peripheral Blood Progenitor Cells Have Similar Potential for Ex Vivo Expansion as Primitive LTC-ICs in Steady State Bone Marrow. Blood 1997. [DOI: 10.1182/blood.v89.11.3991] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
We have recently shown that more than 90% of long-term culture initiating cells (LTC-IC) mobilized in the peripheral blood (PB) of normal individuals express HLA-DR and CD38 antigens and can sustain hematopoiesis for only 5 weeks. However, 10% of LTC-IC in mobilized PB are CD34+HLA-DR− and CD34+CD38− and can sustain hematopoiesis for at least 8 weeks. We now examine the ex vivo expansion potential of CD34+HLA-DR+ cells (rich in mature LTC-IC) and CD34+HLA-DR− cells (rich in primitive LTC-IC) in granulocyte colony-stimulating factor (G-CSF ) mobilized PB progenitor cells (PBPC). Cells were cultured in contact with M2-10B4 cells (contact) or in transwells above M2-10B4 (noncontact) without and with interleukin-3 (IL-3) and macrophage inflammatory protein (MIP-1α) for 2 and 5 weeks. Progeny were evaluated for the presence of colony-forming cells (CFC) and LTC-IC. When CD34+HLA-DR+ PB cells were cultured in contact cultures without cytokines, a threefold expansion of CFC was seen at 2 weeks, but an 80% decrease in CFC was seen at week 5. Further, the recovery of LTC-IC at week 2 was only 17% and 1% at week 5. This confirms our previous observation that although CD34+HLA-DR+ mobilized PB cells can initiate long-term cultures, they are relatively mature and cannot sustain long-term hematopoiesis. In contrast, when CD34+HLA-DR− mobilized PB cells were cultured in contact cultures without cytokines, CFC expansion persisted until week 5 and 49% and 11% of LTC-IC were recovered at week 2 and 5, respectively. As we have shown for steady state bone marrow (BM) progenitors, recovery of LTC-IC was threefold higher when CD34+HLA-DR− PBPC were cultured in noncontact rather than contact cultures, and improved further when IL-3 and MIP-1α were added to noncontact cultures (96 ± 2% maintained at week 5). We conclude that although G-CSF mobilizes a large population of “mature” CD34+HLA-DR+ LTC-IC with a limited proliferative capacity, primitive CD34+HLA-DR− LTC-IC present in mobilized PB have similar characteristics as LTC-IC from steady state BM: (1) they can be maintained in noncontact cultures containing IL-3 and MIP-1α for at least 5 weeks; (2) they are subject to the same proliferation inhibitory influences of contact with stroma. Since the absolute number of primitive LTC-IC (week 8 LTC-IC) per mL of G-CSF mobilized PB is similar to that per mL of steady state BM, these studies further confirm that G-CSF mobilized PBPC may have similar long-term repopulating abilities as steady state BM.
Collapse
|
43
|
Oostendorp RA, Dörmer P. VLA-4-mediated interactions between normal human hematopoietic progenitors and stromal cells. Leuk Lymphoma 1997; 24:423-35. [PMID: 9086434 DOI: 10.3109/10428199709055581] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The alpha 4 beta 1 integrin very late activation antigen-4 (VLA-4) has been implicated to play a role in the adhesive interactions between hematopoietic progenitor cells (HPC) and bone marrow stromal cells which express the vascular cell adhesion molecule-1 (VCAM-1) or produce fibronectin (FN). Here, we summarize some of the recent advances made in the elucidation of the role of these particular adhesive interactions for the regulation of normal hematopoiesis. HPC bind to stroma mainly through VLA-4/VCAM-1 interactions. There is evidence which suggests that more primitive HPC constitutively express VLA-4 in a high-affinity state. In vitro studies in the mouse have shown that monoclonal antibodies (mAb) against VLA-4 partly block the development of lymphocytes, myelopoietic cells, and erythropoiesis, whereas in the human system outgrowth of TdT+ B cells is severely retarded by such mAb. In vivo studies revealed that VLA-4 is involved in erythropoietic development, and is particularly important for homing and lodgement of HPC in the bone marrow. Hematopoiesis in mice with deficient expression of alpha 4 integrin or VLA-4's ligand VCAM-1 appears to develop normally. However, chimeras developed from wild-type blastocysts and beta 1 -/- embryonic stem cells do not contain beta 1 -/- hematopoietic cells, although these are present as blood islands in the yolk sac. These beta 1 -/- hematopoietic cells are capable of forming colonies, indicating that beta 1-integrin is not involved in hematopoietic differentiation, but is primarily important for migration of hematopoietic cells into the fetal hematopoietic organs. In addition to the role of VLA-4 in migration, it may also have other regulatory functions. It has been demonstrated that ligation of VLA-4 induces phosphorylation of the protein tyrosine kinase (PTK) pp125FAK as well as other proteins which may be involved in the regulation of ligand affinity. Indeed, it has been shown that tyrosine kinase-dependent stimulation of CD34+ hematopoietic cell lines with c-kit ligand (KL), IL-3 or GM-CSF transiently activates the ability of VLA-4 to bind to VCAM-1 or FN. These events are most probably involved in the induction of quiescence in HPC which adhere to stromal cells. This claim was recently substantiated: when HPC were treated with Fab fragments of an anti-VLA-4 mAb, entry into S-phase of the cell cycle was prevented. Taken together, the present data point to a role for VLA-4 in HPC migration, cell cycle regulation, erythropoiesis and B-lymphopoiesis. Moreover, these insights may explain how defects in adhesive behavior of leukemic HPC through VLA-4 contribute to their dysregulated growth and provide a rationale for therapeutically correcting those defects.
Collapse
Affiliation(s)
- R A Oostendorp
- GSF-Forschungszentrum für Umwelt und Gesundheit, Institut für Experimentelle Hämatologie, München, Germany.
| | | |
Collapse
|
44
|
Involvement of Fas-Mediated Apoptosis in the Inhibitory Effects of Interferon-α in Chronic Myelogenous Leukemia. Blood 1997. [DOI: 10.1182/blood.v89.3.957] [Citation(s) in RCA: 121] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractInterferon-α (IFN-α) is an established treatment for chronic myelogenous leukemia (CML) in chronic phase, but the mechanism of its antileukemic activity is not clear. One possible mechanism of action might include the induction of apoptosis, and especially Fas-mediated cell killing may play an important role in the elimination of malignant cells. We investigated Fas receptor (Fas-R) expression and the consequences of Fas-R triggering in CML patients. Using two-color flow cytometry, we found a significantly higher number of Fas-R–expressing CD34+ cells in the bone marrow (BM) of CML patients compared with normal subjects. We have previously shown that IFN-γ induces Fas-R expression on CD34+ cells; in this study, we investigated whether IFN-α induces Fas-R expression on CML progenitor cells. Dose-dependent induction of Fas-R expression was observed after IFN-α stimulation of CD34+ cells from CML BM. In methylcellulose culture, IFN-α alone at a therapeutic concentration showed only marginal antiproliferative effects on both normal and CML BM progenitors. In contrast, a Fas-R agonist, the anti-CD95 monoclonal antibody CH11, inhibited colony formation from normal progenitors, and the inhibition was even stronger on CML progenitors. When CML BM cells were cultured in the presence of IFN-α, Fas-R–mediated inhibition of colony growth was potentiated in a dose-dependent fashion, consistent with IFN-α induction of Fas-R expression. This functional effect did not require the presence of accessory cells, since similar results were obtained with purified CD34+ cells. In suspension cultures, we demonstrated that suppression of CML hematopoiesis by IFN-α and Fas-R agonist was exerted through Fas-R–mediated induction of apoptosis. Our findings suggest that the Fas-R/Fas-ligand system might be involved in the immunologic regulation of CML progenitor growth and that its effect can be amplified by IFN-α.
Collapse
|
45
|
Johnson GJ, Leis LA, Dunlop PC. Specificity of G alpha q and G alpha 11 gene expression in platelets and erythrocytes. Expressions of cellular differentiation and species differences. Biochem J 1996; 318 ( Pt 3):1023-31. [PMID: 8836152 PMCID: PMC1217719 DOI: 10.1042/bj3181023] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
G alpha q and G alpha 11, members of the Gq family of G-proteins, transduce signals from receptors to the beta isoenzymes of phosphatidyl-inositol-specific phospholipase C (PI-PLC). The receptor specificity of these alpha subunits is unknown. G alpha q and G alpha 11 are ubiquitously expressed in tissues; however, there have been conflicting reports of the presence or absence of G alpha 11 protein in haematopoietic cells. Platelet thromboxane A2/prostaglandin H2 (TXA2/PGH2) receptors activate PI-PLC via G alpha q, but the role of G alpha 11 is uncertain. To define their roles in platelet activation we studied G alpha q and G alpha 11 gene expression by immunotransfer blotting and by reverse transcription of mRNA followed by PCR (RT-PCR) and direct sequencing. An antiserum specific for mouse G alpha 11 failed to identify G alpha 11 in dog or human platelets or in dog liver, a tissue known to contain G alpha 11. RT-PCR performed with gene-specific primers demonstrated G alpha q mRNA, but not G alpha 11 mRNA, in normal human and mouse platelets and in thromboxane-sensitive and thromboxane-insensitive dog platelets. Studies of mouse and dog liver and human retina confirmed that the cDNA, primers and probes used could amplify and recognize G alpha 11 in other tissues. However, species-specific oligonucleotide primers and probes were essential to demonstrate G alpha 11, but not G alpha q, mRNA. Compared with mouse cDNA, dog and human G alpha 11 cDNA had twice as many nucleotide substitutions (approx. 12% compared with approx. 6%) as G alpha q, G alpha q mRNA was also found in mature erythrocytes but G alpha 11 mRNA was not identified, whereas both G alpha q and G alpha 11 mRNAs were found in bone marrow stem cells. Therefore G alpha 11 gene expression in haematopoietic cells is linked with cellular differentiation. The lack of G alpha 11 indicates that signal transduction from platelet TXA2/PGH2 receptors to PI-PLC occurs via G alpha q, and that G alpha 11 deficiency is not responsible for defective activation of PI-PLC in thromboxane-insensitive dog platelets. Despite the high degree of similarity that exists between G alpha q and G alpha 11, significantly greater species-specific variation in nucleotide sequence is present in G alpha 11 than in G alpha q. Cellular specificity and species specificity are important characteristics of these Gq family G-proteins.
Collapse
Affiliation(s)
- G J Johnson
- Department of Medicine, VA Medical Center, Minneapolis, MN, USA
| | | | | |
Collapse
|
46
|
VerFaillie CM. Chemokines as inhibitors of hematopoietic progenitors. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 1996; 127:148-50. [PMID: 8636643 DOI: 10.1016/s0022-2143(96)90073-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|