1
|
Larsen MC, Almeldin A, Tong T, Rondelli CM, Maguire M, Jaskula-Sztul R, Jefcoate CR. Cytochrome P4501B1 in bone marrow is co-expressed with key markers of mesenchymal stem cells. BMS2 cell line models PAH disruption of bone marrow niche development functions. Toxicol Appl Pharmacol 2020; 401:115111. [PMID: 32553695 PMCID: PMC7293885 DOI: 10.1016/j.taap.2020.115111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/27/2020] [Accepted: 06/07/2020] [Indexed: 12/13/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous pollutants that are metabolized to carcinogenic dihydrodiol epoxides (PAHDE) by cytochrome P450 1B1 (CYP1B1). This metabolism occurs in bone marrow (BM) mesenchymal stem cells (MSC), which sustain hematopoietic stem and progenitor cells (HSPC). In BM, CYP1B1-mediated metabolism of 7, 12-dimethylbenz[a]anthracene (DMBA) suppresses HSPC colony formation within 6 h, whereas benzo(a)pyrene (BP) generates protective cytokines. MSC, enriched from adherent BM cells, yielded the bone marrow stromal, BMS2, cell line. These cells express elevated basal CYP1B1 that scarcely responds to Ah receptor (AhR) inducers. BMS2 cells exhibit extensive transcriptome overlap with leptin receptor positive mesenchymal stem cells (Lepr+ MSC) that control the hematopoietic niche. The overlap includes CYP1B1 and the expression of HSPC regulatory factors (Ebf3, Cxcl12, Kitl, Csf1 and Gas6). MSC are large, adherent fibroblasts that sequester small HSPC and macrophage in the BM niche (Graphic abstract). High basal CYP1B1 expression in BMS2 cells derives from interactions between the Ah-receptor enhancer and proximal promoter SP1 complexes, boosted by autocrine signaling. PAH effects on BMS2 cells model Lepr+MSC niche activity. CYP1B1 metabolizes DMBA to PAHDE, producing p53-mediated mRNA increases, long after the in vivo HSPC suppression. Faster, direct p53 effects, favored by stem cells, remain possible PAHDE targets. However, HSPC regulatory factors remained unresponsive. BP is less toxic in BMS2 cells, but, in BM, CYP1A1 metabolism stimulates macrophage cytokines (Il1b > Tnfa> Ifng) within 6 h. Although absent from BMS2 and Lepr+MSC, their receptors are highly expressed. The impact of this cytokine signaling in MSC remains to be determined. BMS2 and Lepr+MSC cells co-express CYP1B1 and 12 functional niche activity markers. CYP1B1 mRNA in BMS2 cells depends on activation of SP1 coupled to an AhR enhancer unit. DMBA metabolism by CYP1B1 activates p53 gene targets in BMS2 cells far more than BP. HSPC suppression by CYP1B1 generation of PAHDE requires rapid, non-genomic targets. BMS2 and Lepr+MSC share receptors activated by BP stimulation of macrophage cytokines.
Collapse
Affiliation(s)
- Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705, United States of America
| | - Ahmed Almeldin
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705, United States of America; Physiology Department, Faculty of Medicine, Tanta University, Egypt
| | - Tiegang Tong
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705, United States of America
| | - Catherine M Rondelli
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, United States of America
| | - Meghan Maguire
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53705, United States of America
| | - Renata Jaskula-Sztul
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, United States of America
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705, United States of America; Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, United States of America; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53705, United States of America.
| |
Collapse
|
2
|
Murine Bone Marrow Niches from Hematopoietic Stem Cells to B Cells. Int J Mol Sci 2018; 19:ijms19082353. [PMID: 30103411 PMCID: PMC6121419 DOI: 10.3390/ijms19082353] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 01/03/2023] Open
Abstract
After birth, the development of hematopoietic cells occurs in the bone marrow. Hematopoietic differentiation is finely tuned by cell-intrinsic mechanisms and lineage-specific transcription factors. However, it is now clear that the bone marrow microenvironment plays an essential role in the maintenance of hematopoietic stem cells (HSC) and their differentiation into more mature lineages. Mesenchymal and endothelial cells contribute to a protective microenvironment called hematopoietic niches that secrete specific factors and establish a direct contact with developing hematopoietic cells. A number of recent studies have addressed in mouse models the specific molecular events that are involved in the cellular crosstalk between hematopoietic subsets and their niches. This has led to the concept that hematopoietic differentiation and commitment towards a given hematopoietic pathway is a dynamic process controlled at least partially by the bone marrow microenvironment. In this review, we discuss the evolving view of murine hematopoietic–stromal cell crosstalk that is involved in HSC maintenance and commitment towards B cell differentiation.
Collapse
|
3
|
Bone Marrow-Derived Stem Cell Populations Are Differentially Regulated by Thyroid or/and Ovarian Hormone Loss. Int J Mol Sci 2017; 18:ijms18102139. [PMID: 29048335 PMCID: PMC5666821 DOI: 10.3390/ijms18102139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/19/2017] [Accepted: 09/28/2017] [Indexed: 02/08/2023] Open
Abstract
Bone marrow-derived stem cells (BMDSCs) play an essential role in organ repair and regeneration. The molecular mechanisms by which hormones control BMDSCs proliferation and differentiation are unclear. Our aim in this study was to investigate how a lack of ovarian or/and thyroid hormones affects stem cell number in bone marrow lineage. To examine the effect of thyroid or/and ovarian hormones on the proliferative activity of BMDSCs, we removed the thyroid or/and the ovaries of adult female rats. An absence of ovarian and thyroid hormones was confirmed by Pap staining and Thyroid Stimulating Hormone (TSH) measurement, respectively. To obtain the stem cells from the bone marrow, we punctured the iliac crest, and aspirated and isolated cells by using a density gradient. Specific markers were used by cytometry to identify the different BMDSCs types: endothelial progenitor cells (EPCs), precursor B cells/pro-B cells, and mesenchymal stem cells (MSCs). Interestingly, our results showed that hypothyroidism caused a significant increase in the percentage of EPCs, whereas a lack of ovarian hormones significantly increased the precursor B cells/pro-B cells. Moreover, the removal of both glands led to increased MSCs. In conclusion, both ovarian and thyroid hormones appear to have key and diverse roles in regulating the proliferation of cells populations of the bone marrow.
Collapse
|
4
|
Tsunokuma N, Yamane T, Matsumoto C, Tsuneto M, Isono K, Imanaka-Yoshida K, Yamazaki H. Depletion of Neural Crest–Derived Cells Leads to Reduction in Plasma Noradrenaline and Alters B Lymphopoiesis. THE JOURNAL OF IMMUNOLOGY 2016; 198:156-169. [DOI: 10.4049/jimmunol.1502592] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 10/24/2016] [Indexed: 11/19/2022]
|
5
|
Hill M, Wu X, Sullivan M, King B, Webb C, Gimble J. Expression of acute phase proteins by bone marrow stromal cells. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199600300506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The current work examines the expression of acute phase genes in a murine-derived bone marrow stromal cell model (BMS2) which exhibits adipocyte and osteoblast characteristics and supports lymphopoiesis in vitro. Each of these physiologic processes is responsive to inflammatory events such as endotoxemia. Exposure of BMS2 cells to pro-inflammatory cytokines induced the expression of the serum amyloid A and complement factor B. During adipocyte differentiation, expression of complement C3, complement factor D (adipsin), and angiotensinogen increased in a time dependent manner. The bone metabolic steroid, 1,25 dihydroxy vitamin D3, specifically induced complement C3 expression in a time- and dose-dependent manner. Based on gel retention analysis, BMS2 nuclear extract contained proteins recognizing specific response elements from the complement C3, angiotensinogen, and complement factor B promoters. These results suggest that the bone marrow's repertoire of acute phase proteins is dependent on the stromal cell's phenotype or activation state.
Collapse
Affiliation(s)
- M.R. Hill
- Department of Radiologic Technology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA, Department of Natural Sciences, Oklahoma Christian University, Oklahoma City, Oklahoma, USA
| | - X. Wu
- Immunobiology & Cancer Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - M. Sullivan
- Immunobiology & Cancer Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - B.O. King
- Department of Radiologic Technology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - C.F. Webb
- Immunobiology & Cancer Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA, Department of Microbiology & Immunology, University of Oklahma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - J.M. Gimble
- Immunobiology & Cancer Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA, Department of Microbiology & Immunology, University of Oklahma Health Sciences Center, Oklahoma City, Oklahoma, USA, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA, Department of Zoology, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
6
|
Seke Etet PF, Vecchio L, Nwabo Kamdje AH. Signaling pathways in chronic myeloid leukemia and leukemic stem cell maintenance: key role of stromal microenvironment. Cell Signal 2012; 24:1883-1888. [PMID: 22659137 DOI: 10.1016/j.cellsig.2012.05.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 05/15/2012] [Accepted: 05/24/2012] [Indexed: 12/11/2022]
Abstract
Chronic myeloid leukemia (CML) is caused by the malignant transformation of hematopoietic stem cells in leukemic stem cells. From the introduction of the anti-cancer drug imatinib, the therapy of CML has been positively transformed. However, following treatment most patients display a residual CML disease attributed to the presence of quiescent leukemic stem cells intrinsically resistant to imatinib. Considering that the later cancer cells lose their chemoresistance in vitro, it appears that the stromal microenvironment plays a crucial role in CML-affected cell chemoresistance. In the present review, we summarize and discuss the recent findings on signaling pathways through which stromal cells sustain CML leukemogenesis, as well as leukemic stem cell maintenance and chemoresistance.
Collapse
Affiliation(s)
- P F Seke Etet
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, 51452 Al-Qaseem, Saudi Arabia
| | | | | |
Collapse
|
7
|
Gimble JM, Nuttall ME. The relationship between adipose tissue and bone metabolism. Clin Biochem 2012; 45:874-9. [PMID: 22429519 DOI: 10.1016/j.clinbiochem.2012.03.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 02/28/2012] [Accepted: 03/01/2012] [Indexed: 01/06/2023]
Abstract
OBJECTIVES The authors have set out to evaluate the literature relevant to the dynamic regulation of adipogenesis and osteogenesis. DESIGN AND METHODS A detailed search of the past and recent literature was conducted on Pubmed using a combination of keywords including: adipogenesis, bone marrow, hematopoiesis, mesenchymal stromal/stem cell, and osteogenesis. RESULTS Throughout one's lifespan, the bone marrow microenvironment provides a unique niche for mesenchymal stromal/stem cells (BMSCs) and hematopoietic stem cells (HSCs). The marrow changes as a function of biological age and pathophysiology. Historically, clinical biochemistry has observed these changes from an HSC and hematological perspective. Nevertheless, these changes also reflect the balance between BMSC adipogenic and osteogenic processes which can display an inverse or reciprocal relationship. Multiple hormonal factors and nuclear hormone receptor ligands and drugs are responsible for BMSC lineage selection. Data from a number of laboratories now implicates endocrine feedback loops between extramedullary adipose depots and the central nervous system. CONCLUSIONS This concise review provides a perspective on the mechanisms regulating BMSC differentiation in the context of biological aging, obesity, and osteoporosis.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA.
| | | |
Collapse
|
8
|
|
9
|
Application of mesenchymal stem cells derived from bone marrow and umbilical cord in human hair multiplication. J Dermatol Sci 2010; 60:74-83. [DOI: 10.1016/j.jdermsci.2010.08.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 08/22/2010] [Accepted: 08/26/2010] [Indexed: 11/20/2022]
|
10
|
Smith TJ. Insulin-like growth factor-I regulation of immune function: a potential therapeutic target in autoimmune diseases? Pharmacol Rev 2010; 62:199-236. [PMID: 20392809 PMCID: PMC2879913 DOI: 10.1124/pr.109.002469] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This topically limited review explores the relationship between the immune system and insulin-like growth factors (IGF-I and IGF-II) and the proteins through which they act, including IGF-I receptor (IGF-IR) and the IGF-I binding proteins. The IGF/IGF-IR pathway plays important and diverse roles in tissue development and function. It regulates cell cycle progression, apoptosis, and the translation of proteins. Many of the consequences ascribed to IGF-IR activation result from its association with several accessory proteins that are either identical or closely related to those involved in insulin receptor signaling. Relatively recent awareness that IGF-I and IGF-IR regulate immune function has cast this pathway in an unexpected light; it may represent an important switch governing the quality and amplitude of immune responses. IGF-I/IGF-IR signaling may also participate in the pathogenesis of autoimmune diseases, although its relationship with these processes seems complex and relatively unexplored. On the one hand, IGF-I seems to protect experimental animals from developing insulin-deficient diabetes mellitus. In contrast, activating antibodies directed at IGF-IR have been detected in patients with Graves' disease, where the receptor is overexpressed by multiple cell types. The frequency of IGF-IR+ B and T cells is substantially increased in patients with that disease. Potential involvement of IGF-I and IGF-IR in the pathogenesis of autoimmune diseases suggests that this pathway might constitute an attractive therapeutic target. IGF-IR has been targeted in efforts directed toward drug development for cancer, employing both small-molecule and monoclonal antibody approaches. These have been generally well-tolerated. Recognizing the broader role of IGF-IR in regulating both normal and pathological immune responses may offer important opportunities for therapeutic intervention in several allied diseases that have proven particularly difficult to treat.
Collapse
Affiliation(s)
- Terry J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA.
| |
Collapse
|
11
|
Gimble JM, Floyd ZE, Bunnell BA. The 4th dimension and adult stem cells: Can timing be everything? J Cell Biochem 2009; 107:569-78. [PMID: 19384905 DOI: 10.1002/jcb.22153] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The rotation of the earth on its axis influences the physiology of all organisms. A highly conserved set of genes encoding the core circadian regulatory proteins (CCRP) has evolved across species. The CCRP acts through transcriptional and post-transcriptional mechanisms to direct the oscillatory expression of genes essential for key metabolic events. In addition to the light:dark cycle, the CCRP expression can be entrained by changes in feeding and physical activity patterns. While mammalian CCRP were originally associated with the central clock located within the suprachiasmatic nucleus of the brain, there is a growing body of evidence documenting the presence of the CCRP in peripheral tissues. It is now evident that the CCRP play a role in regulating the proliferation, differentiation, and function of adult stem cells in multiple organs. This concise review highlights findings concerning the role of the CCRP in modulating the adult stem cell activities. Although the manuscript focuses on hematopoietic stem cells (HSCs), bone marrow-derived mesenchymal stem cells (BMSCs), adipose-derived stem cells (ASCs) and cancer stem cells, it is likely that the contribution of the CCRP merits consideration and evaluation in all stem cell pathways.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA.
| | | | | |
Collapse
|
12
|
Treml LS, Quinn WJ, Treml JF, Scholz JL, Cancro MP. Manipulating B cell homeostasis: a key component in the advancement of targeted strategies. Arch Immunol Ther Exp (Warsz) 2008; 56:153-64. [PMID: 18512030 DOI: 10.1007/s00005-008-0017-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 01/04/2008] [Indexed: 11/28/2022]
Abstract
Understanding the homeostatic mechanisms governing lymphocyte pools achieves critical importance as lymphocyte-targeted therapies expand in use and scope. The primacy of B lymphocyte stimulator (BLyS) family ligands and receptors in governing B lymphocyte homeostasis has become increasingly clear in recent years, affording insight into novel opportunities and potential pitfalls for targeted B cell therapeutics. Interclonal competition for BLyS-BR3 interactions determines the size of naïve B cell pools and can regulate the stringency of selection applied as cells complete maturation. Thus one of the predicted consequences of ablative therapies targeting primary pools is relaxed negative selection. This suggests that BLyS levels and B cell reconstitution rates may serve useful prognostic roles and that BLyS itself might be targeted to circumvent relapse. Alternatively, manipulations that allow rare, minimally autoreactive specificities to survive and mature may lead to opportunities in cases where antibody-based vaccine development has heretofore been unsuccessful. BLyS family ligands and receptors also play a role in activated and memory B cell pools, suggesting they might likewise be targeted to promote or delete particular antigen-experienced subpopulations in a similar way.
Collapse
Affiliation(s)
- Laura S Treml
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6082, USA
| | | | | | | | | |
Collapse
|
13
|
Gimble JM, Guilak F, Nuttall ME, Sathishkumar S, Vidal M, Bunnell BA. In vitro Differentiation Potential of Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2008; 35:228-238. [PMID: 21547120 DOI: 10.1159/000124281] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2007] [Accepted: 03/07/2008] [Indexed: 12/19/2022]
Abstract
SUMMARY: Mesenchymal stem cells (MSCs) represent a class of multipotent progenitor cells that have been isolated from multiple tissue sites. Of these, adipose tissue and bone marrow offer advantages in terms of access, abundance, and the extent of their documentation in the literature. This review focuses on the in vitro differentiation capability of cells derived from adult human tissue. Multiple, independent studies have demonstrated that MSCs can commit to mesodermal (adipocyte, chondrocyte, hematopoietic support, myocyte, osteoblast, tenocyte), ectodermal (epithelial, glial, neural), and endodermal (hepatocyte, islet cell) lineages. The limitations and promises of these studies in the context of tissue engineering are discussed.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | | | | | | | | |
Collapse
|
14
|
Dorshkind K, Witte ON. Linking the hematopoietic microenvironment to imatinib-resistant Ph+ B-ALL. Genes Dev 2007; 21:2249-52. [PMID: 17875661 DOI: 10.1101/gad.1600307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Kenneth Dorshkind
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA.
| | | |
Collapse
|
15
|
Kilroy GE, Foster SJ, Wu X, Ruiz J, Sherwood S, Heifetz A, Ludlow JW, Stricker DM, Potiny S, Green P, Halvorsen YDC, Cheatham B, Storms RW, Gimble JM. Cytokine profile of human adipose-derived stem cells: expression of angiogenic, hematopoietic, and pro-inflammatory factors. J Cell Physiol 2007; 212:702-9. [PMID: 17477371 DOI: 10.1002/jcp.21068] [Citation(s) in RCA: 448] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adipose tissue serves as a source of adipokines and cytokines with both local and systemic actions in health and disease. In this study, we examine the hypothesis that multipotent human adipose-derived stem cells (ASCs), capable of differentiating along the adipocyte, chondrocyte, and osteoblast pathways, contribute to adipose tissue-derived cytokine secretion. Following exposure to basic fibroblast growth factor (bFGF) or epidermal growth factor (EGF), the ASCs significantly increase their secretion of hepatocyte growth factor (HGF), a cytokine implicated in hematopoiesis, vasculogenesis, and mammary epithelial duct formation. Ascorbic acid synergizes with these inductive factors, further increasing HGF levels. Following exposure to lipopolysaccharide, ASCs increase their secretion of both hematopoietic (granulocyte/monocyte, granulocyte, and macrophage colony stimulating factors, interleukin 7) and proinflammatory (interleukins 6, 8, and 11, tumor necrosis factor alpha) cytokines based on ELISA and RT-PCR. In co-cultures established with umbilical cord blood-derived CD34(+) cells, the ASCs support long-term hematopoiesis in vitro. Furthermore, in short-term 12-day co-cultures, the ASC maintain and expand the numbers of both myeloid and lymphoid progenitors. These observations are consistent with the functionality of the secreted cytokines and confirm recent reports by other laboratories concerning the hematopoietic supportive capability of ASCs. We conclude that the ASCs display cytokine secretory properties similar to those reported for bone marrow-derived mesenchymal stem cells (MSCs).
Collapse
Affiliation(s)
- Gail E Kilroy
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
In adult mammals, bone marrow pluripotent hematopoietic stem cells generate B lymphoid-specified progeny that progress through a series of well-characterized stages before generating B-cell receptor expressing B lymphocytes. These functionally immature B lymphocytes then migrate to the spleen wherein they differentiate through transitional stages into follicular or marginal zone B lymphocytes capable of responding to T-dependent and -independent antigens, respectively. During the terminal stages of B lymphocyte development in the bone marrow, as well as immediately following egress into the peripheral compartments, B lymphocytes are counterselected to eliminate B lymphocytes with potentially dangerous self-reactivity. These developmental and selection events in the bone marrow and periphery are dependent on the integration of intrinsic genetic programs with extrinsic microenvironmental signals that drive progenitors toward increasing B lineage commitment and maturation. This chapter provides a comprehensive overview of the various stages of primary and secondary B lymphocyte development with an emphasis on the selection processes that affect decisions at critical checkpoints. Our intent is to stress the concept that at many steps in the developmental process leading to a mature immunocompetent B lymphocyte, B lineage cells are integrating multiple and different signaling inputs that are translated into specific and appropriate cell fate decisions.
Collapse
MESH Headings
- Aging
- Animals
- Antigens, Differentiation, B-Lymphocyte/analysis
- B-Lymphocyte Subsets/cytology
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/physiology
- B-Lymphocytes/cytology
- B-Lymphocytes/immunology
- B-Lymphocytes/physiology
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Bone Marrow Cells/physiology
- Cell Lineage
- Humans
- Lymphopoiesis/genetics
- Models, Immunological
- Precursor Cells, B-Lymphoid/cytology
- Precursor Cells, B-Lymphoid/immunology
- Precursor Cells, B-Lymphoid/physiology
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Recombination, Genetic
- Signal Transduction
Collapse
Affiliation(s)
- John G Monroe
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
17
|
Abstract
Once hematopoiesis is established in the bone marrow, a continuous egress of hematopoietic stem cells (HSCs) to the periphery occurs at a low frequency. It has been proposed that this phenomenon is part of a regenerative homeostatic mechanism that ensures the maintenance of hematopoiesis through the life of the individual. The administration of certain cytotoxic drugs or cytokines can enhance the mobilization of hematopoietic progenitors to the periphery. During the past 15 years, granulocyte-colony stimulating factor (G-CSF) has been used as a standard cytokine for mobilization protocols in experimental models and in humans. Despite extensive efforts by multiple groups, a definitive mechanism explaining its role in mobilization has not been provided. In a recent paper, Katayama et al., through a series of clever associations supported by well-defined experimental systems, proposed that signals through the sympathetic nervous system modify the activity of the hematopoietic niche, acting as regulators of the mobilization of hematopoietic progenitors. This surprising finding adds a new level of complexity to the cellular milieu responsible for generation and maintenance of the hematopoietic niche.
Collapse
Affiliation(s)
- Hector Leonardo Aguila
- Department of Immunology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA.
| |
Collapse
|
18
|
Mauney JR, Volloch V, Kaplan DL. Role of adult mesenchymal stem cells in bone tissue engineering applications: current status and future prospects. ACTA ACUST UNITED AC 2006; 11:787-802. [PMID: 15998219 DOI: 10.1089/ten.2005.11.787] [Citation(s) in RCA: 208] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) have been demonstrated as an attractive cell source for tissue-engineering applications because of their ability to be easily isolated and expanded from adult bone marrow aspirates and their versatility for pluripotent differentiation into mesenchymal tissues. This review highlights advances and progress in bone reconstruction techniques for both the repair of site-specific bone defects and the attenuation of musculoskeletal disease symptoms associated with osteoporosis and osteogenesis imperfecta. Despite the enormous potential benefits of MSCs within these approaches, conventional tissue culture methods limit the clinical utility of these cells because of the gradual loss of both their proliferative and differentiation potential during ex vivo expansion. Novel strategies to overcome these limitations are discussed including cultivation in the presence of basic fibroblastic growth factor 2, induction of ectopotic telomerase expression, and ex vivo expansion on various collagenous biomaterials. In addition, this review also outlines mechanistic theories on the potential role of MSC-extracellular matrix interactions in mediating the retention of MSC proliferative and differentiation capacity after ex vivo expansion on collagenous biomaterials.
Collapse
Affiliation(s)
- Joshua R Mauney
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | | | | |
Collapse
|
19
|
Abstract
The relationship between bone and fat formation within the bone marrow microenvironment is complex and remains an area of active investigation. Classical in vitro and in vivo studies strongly support an inverse relationship between the commitment of bone marrow-derived mesenchymal stem cells or stromal cells to the adipocyte and osteoblast lineage pathways. In this review, we focus on the recent literature exploring the mechanisms underlying these differentiation events and discuss their implications relevant to osteoporosis and regenerative medicine.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Stem Cell Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, 70808, USA.
| | | | | | | | | |
Collapse
|
20
|
Lai L, Zeff RA, Goldschneider I. A recombinant single-chain IL-7/HGFbeta hybrid cytokine induces juxtacrine interactions of the IL-7 and HGF (c-Met) receptors and stimulates the proliferation of CFU-S12, CLPs, and pre-pro-B cells. Blood 2005; 107:1776-84. [PMID: 16304052 PMCID: PMC1895697 DOI: 10.1182/blood-2005-08-3470] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A novel recombinant interleukin-7/hepatocyte growth factor beta-chain (IL-7/HGFbeta) hybrid cytokine was constructed as a single chain (sc) composed of IL-7 and HGFbeta connected by a flexible linker. Unlike recombinant (r) IL-7, which stimulated pro-B cells and pre-B cells only, scIL-7/HGFbeta stimulated the proliferation of pre-pro-B cells, common lymphoid progenitors (CLPs), and colony-forming unit (CFU)-S12 in cultures of IL-7-/- mouse BM cells. When injected in vivo, 3- to 4-fold more splenic B-lineage cells appeared in recipients of bone marrow (BM) cells from the scIL-7/HGFbeta-stimulated cultures than from rIL-7-stimulated cultures. Moreover, on a per-cell basis, scIL-7/HGFbeta culture-generated cells produced 16- to 20-fold more BM and splenic B-lineage cells than did normal BM cells. Antibody blocking, receptor phosphorylation, and confocal microscopy demonstrated that scIL-7/HGFbeta signals though both the IL-7 and HGF (c-Met) receptors, which form IL-7R/c-Met complexes on the surface of CLPs and pre-pro-B cells. In addition, the IL-7Ralpha chain, gammac chain, and c-Met were coisolated from purified CLPs and pre-pro-B cells on scIL-7/HGFbeta affinity gels, indicating that they are major components of the IL-7/HGFbeta receptor. Hence, the present results demonstrate that the IL-7/HGFbeta hybrid cytokine efficiently and selectively stimulates the most primitive B-lineage precursors in BM by inducing juxtacrine interactions between the IL-7 and c-Met receptors.
Collapse
Affiliation(s)
- Laijun Lai
- Department of Immunology, School of Medicine, University of Connecticut, 263 Farmington Ave, Farmington, CT 06030-3710, USA
| | | | | |
Collapse
|
21
|
Hubin F, Humblet C, Belaid Z, Lambert C, Boniver J, Thiry A, Defresne MP. Murine Bone Marrow Stromal Cells Sustain In Vivo the Survival of Hematopoietic Stem Cells and the Granulopoietic Differentiation of More Mature Progenitors. Stem Cells 2005; 23:1626-33. [PMID: 16293584 DOI: 10.1634/stemcells.2005-0041] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The study of the human hematopoietic system would be facilitated by availability of a relevant animal model. Because the medullar microenvironment is made of different types of cells, interactions between hematopoietic cells and stromal cells are difficult to analyze in detail. As an approach for establishing an in vivo model to dissect these interactions, we grafted murine bone marrow fibroblastic cells (MS-5 cell line) with hematopoietic cells into the kidney capsule of syngenic mice. To identify the origin of cells present in the graft, we used green fluorescent protein-stable transfected MS-5 cells for the transplantation. To analyze the evolution of stromal cells and identify hematopoietic cells able to develop in these conditions, we performed morphology, histochemistry, and immunohistology on tissue sections at different times after transplantation. When injected alone, MS-5 cells differentiate into adipocytes. When injected with a bone marrow suspension or with isolated CD45+ cells (leukocytes), the stromal cells keep their fibroblastic morphology and their alkaline phosphatase expression and sustain granulopoiesis. When injected with hematopoietic stem cells called c-kit+ Sca-1+ Lin- suspension, clusters of hematopoietic cells are also observed: They do not present any granulopoietic activity and do not belong to B or T population nor to erythroid lineage. They are quiescent, induce bone marrow recovery and survival of lethally irradiated recipients, are able to form macroscopic colonies in the spleen, and are able to form very few colonies in vitro, suggesting that they are hematopoietic stem cells. In conclusion, our results show that reticular fibroblastic stromal cells MS-5 sustain the survival of stem cells and are not able to induce their differentiation. However, they can control differentiation, proliferation, and/or survival of hematopoietic cells engaged in myeloid lineage.
Collapse
Affiliation(s)
- Frédérique Hubin
- Department of Cytology and Histology, University of Liège, Liège, 4000, Belgium.
| | | | | | | | | | | | | |
Collapse
|
22
|
Kim SJ, Cho HH, Kim YJ, Seo SY, Kim HN, Lee JB, Kim JH, Chung JS, Jung JS. Human adipose stromal cells expanded in human serum promote engraftment of human peripheral blood hematopoietic stem cells in NOD/SCID mice. Biochem Biophys Res Commun 2005; 329:25-31. [PMID: 15721268 DOI: 10.1016/j.bbrc.2005.01.092] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2005] [Indexed: 01/18/2023]
Abstract
Human mesenchymal stem cells (hMSC), that have been reported to be present in bone marrow, adipose tissues, dermis, muscles, and peripheral blood, have the potential to differentiate along different lineages including those forming bone, cartilage, fat, muscle, and neuron. Therefore, hMSC are attractive candidates for cell and gene therapy. The optimal conditions for hMSC expansion require medium supplemented with fetal bovine serum (FBS). Some forms of cell therapy will involve multiple doses, raising a concern over immunological reactions caused by medium-derived FBS proteins. In this study, we cultured human adipose stromal cells (hADSC) and bone marrow stroma cells (HBMSC) in human serum (HS) during their isolation and expansion, and demonstrated that they maintain their proliferative capacity and ability for multilineage differentiation and promote engraftment of peripheral blood-derived CD34(+) cells mobilized from bone marrow in NOD/SCID mice. Our results indicate that hADSC and hBMSC cultured in HS can be used for clinical trials of cell and gene therapies, including promotion of engraftment after allogeneic HSC transplantation.
Collapse
Affiliation(s)
- Su Jin Kim
- Department of Physiology, College of Medicine, Pusan National University, Pusan 602-739, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Seo MJ, Suh SY, Bae YC, Jung JS. Differentiation of human adipose stromal cells into hepatic lineage in vitro and in vivo. Biochem Biophys Res Commun 2005; 328:258-64. [PMID: 15670778 DOI: 10.1016/j.bbrc.2004.12.158] [Citation(s) in RCA: 393] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2004] [Indexed: 12/11/2022]
Abstract
Embryonic stem cells (ES cells), bone marrow-derived mesenchymal stem cells, umbilical cord blood-derived mesenchymal stem cells, and hepatic stem cells in liver have been known as a useful source that can induce to differentiate into hepatocytes. In this study, we examined whether human adipose tissue-derived stromal cells (hADSC) can differentiate into hepatic lineage in vitro. hADSC, that were induced to differentiate into hepatocyte-like cells by the treatment of HGF and OSM, had morphology similar to hepatocytes. Addition of DMSO enhanced differentiation into hepatocytes. RT-PCR and immunocytochemical analysis showed that hADSC express albumin and alpha-fetoprotein during differentiation. Differentiated hADSC showed LDL uptake and production of urea. Additionally, transplanted hADSC to CCl4-injured SCID mouse model were able to be differentiated into hepatocytes and they expressed albumin in vivo. Mesenchymal stem cells isolated from human adipose tissue are immunocompatible and are easily isolated. Therefore, hADSC may become an alternative source to hepatocyte regeneration or liver cell transplantation.
Collapse
Affiliation(s)
- Min Jeong Seo
- Department of Physiology, College of Medicine, Pusan National University, Pusan 602-739, Republic of Korea
| | | | | | | |
Collapse
|
24
|
Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila HL. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood 2004; 103:3258-64. [PMID: 14726388 DOI: 10.1182/blood-2003-11-4011] [Citation(s) in RCA: 547] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously reported a transgenic mouse model expressing herpesvirus thymidine kinase (TK) gene under the control of a 2.3-kilobase fragment of the rat collagen alpha1 type I promoter (Col2.3 Delta TK). This construct confers lineage-specific expression in developing osteoblasts, allowing the conditional ablation of osteoblast lineage after treatment with ganciclovir (GCV). After GCV treatment these mice have profound alterations on bone formation leading to a progressive bone loss. In addition, treated animals also lose bone marrow cellularity. In this report we characterized hematopoietic parameters in GCV-treated Col2.3 Delta TK mice, and we show that after treatment transgenic animals lose lymphoid, erythroid, and myeloid progenitors in the bone marrow, followed by decreases in the number of hematopoietic stem cells (HSCs). Together with the decrease in bone marrow hematopoiesis, active extramedullary hematopoiesis was observed in the spleen and liver, as measured by an increase in peripheral HSCs and active primary in vitro hematopoiesis. After withdrawal of GCV, osteoblasts reappeared in the bone compartment together with a recovery of medullary and decrease in extramedullary hematopoiesis. These observations directly demonstrate the role of osteoblasts in hematopoiesis and provide a model to study the interactions between the mesenchymal and hematopoietic compartments in the marrow.
Collapse
Affiliation(s)
- Dora Visnjic
- Department of Genetics and Developmental Biolgoy, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | |
Collapse
|
25
|
Ryu HY, Mann KK, Schlezinger JJ, Jensen B, Sherr DH. Environmental chemical-induced pro/pre-B cell apoptosis: analysis of c-Myc, p27Kip1, and p21WAF1 reveals a death pathway distinct from clonal deletion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4897-904. [PMID: 12734332 DOI: 10.4049/jimmunol.170.10.4897] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAH) are common environmental pollutants that suppress the immune system in part by inducing pro/pre-B cell apoptosis. The PAH-induced death signaling pathway resembles the signaling cascade activated during clonal deletion and modeled by B cell receptor cross-linking or by dexamethasone exposure of immature surface Ig(+) B cells in that apoptosis is mediated by NF-kappa B down-regulation. Because a PAH-induced, clonally nonrestricted deletion of B cells would have important implications for B cell repertoire development, the nature of the PAH-induced intracellular death signal was studied further. Particular emphasis was placed on the roles of growth arrest and c-Myc, p27(Kip1), and p21(WAF1) expression, because all of these elements contribute to clonal deletion. As in clonal deletion models, and as predicted by the down-regulation of NF-kappa B, PAH-induced death of pro/pre-B cells was at least partially dependent on c-Myc down-regulation. Furthermore, whereas dexamethasone induced a G(0)/G(1) cell cycle arrest, PAH had no effect on pro/pre-B cell growth, indicating that growth arrest and apoptosis occur by separable signaling pathways in this early phase of B cell development. Finally, in contrast to clonal deletion, PAH-induced pro/pre-B cell death was not dependent on p27(Kip1) or p21(WAF1) up-regulation but did coincide with p53 induction. These results distinguish the PAH-induced apoptosis pathway from that activated during clonal deletion and indicate that signaling cascades leading to growth arrest and/or apoptosis in pro/pre-B cells differ from those active at later B cell developmental stages.
Collapse
Affiliation(s)
- Heui-Young Ryu
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
26
|
Abstract
Circulating lymphocytes normally migrate through extravascular spaces in relatively low numbers as important members of the immunosurveillance process. That is until signals are received by endothelial cells that there is an underlying infection or inflammatory condition. These vascular surface cells in turn overexpress and present ligands to circulating lymphocyte adhesion molecules. Upon encountering this higher density of ligands, lymphocytes, which had been leisurely rolling along the vascular surface, now become more firmly attached, change shape, and migrate through tight junctions to the sites of infection or inflammation. If the initiating events are not resolved and the condition becomes chronic, there can be a sustained extravasation of lymphocytes that can exacerbate the inflammatory condition, which in turn will continue to recruit more inflammatory cells resulting in unwanted tissue destruction. It is for the attenuation of this cycle of sustained inflammatory cell recruitment that very late activating antigen-4 (VLA-4) antagonists are being developed. Most lymphocytes, except neutrophils, express VLA-4 on their surface and they interact with endothelial vascular cell adhesion molecule-1 (VCAM-1). It is this interaction that VLA-4 antagonists are intended to disrupt, thus, putting an end to the cycle of chronic inflammation, which is the hallmark of many diseases. This review will provide an update of VLA-4 antagonists that have appeared since early 2001 and will discuss some of the issues, both positive and negative, that may be encountered in their development.
Collapse
Affiliation(s)
- Ginger X Yang
- Merck Research Laboratories, Rahway, New Jersey 07090, USA.
| | | |
Collapse
|
27
|
Abstract
Hematopoiesis is regulated by a variety of signals that either originate within a developing cell or are supplied by the surrounding environment in secreted- or contact-dependent forms. This review discusses the effects of one secreted factor, interleukin-7, on the development of B lymphocytes. We describe a molecular mechanism for a crucial checkpoint during B lineage maturation, based on the integration of signals mediated by the pre-B cell receptor, the interleukin-7 receptor, and the environment in which these signals are received.
Collapse
Affiliation(s)
- Heather E Fleming
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Department of Immunology, University of Toronto, 610 University Avenue, 7-504 Toronto, Ont., Canada M5G 2M9.
| | | |
Collapse
|
28
|
Lee Y, Gotoh A, Kwon HJ, You M, Kohli L, Mantel C, Cooper S, Hangoc G, Miyazawa K, Ohyashiki K, Broxmeyer HE. Enhancement of intracellular signaling associated with hematopoietic progenitor cell survival in response to SDF-1/CXCL12 in synergy with other cytokines. Blood 2002; 99:4307-17. [PMID: 12036856 DOI: 10.1182/blood.v99.12.4307] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Stromal cell-derived factor 1 (SDF-1/CXCL12) is a multifunctional cytokine. We previously reported that myelopoiesis was enhanced in SDF-1 alpha transgenic mice, probably due in part to SDF-1 alpha enhancement of myeloid progenitor cell (MPC) survival. To understand signaling pathways involved in this activity, we studied the effects on factor-dependent cell line MO7e cells incubated with SDF-1 alpha alone or in combination with other cytokines. SDF-1 alpha induced transient activation of extracellular stress-regulated kinase (ERK1/2), ribosomal S6 kinase (p90RSK) and Akt, molecules implicated in cell survival. Moreover, ERK1/2, p90RSK, and Akt were synergistically activated by SDF-1 alpha in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), Steel factor (SLF), or thrombopoietin (TPO). Similar effects were seen after pretreatment of MO7e cells with SDF-1 alpha followed by stimulation with the other cytokines, suggesting a priming effect of SDF-1 alpha. Nuclear factor-kappa B (NF-kappa B) did not appear to be involved in SDF-1 alpha actions, alone or in combination with other cytokines. These intracellular effects were consistent with enhanced myeloid progenitor cell survival by SDF-1 alpha after delayed addition of growth factors. SDF-1 alpha alone supported survival of highly purified human cord blood CD34(+++) cells, less purified human cord blood, and MO7e cells; this effect was synergistically enhanced when SDF-1 alpha was combined with low amounts of other survival-promoting cytokines (GM-CSF, SLF, TPO, and FL). SDF-1 may contribute to maintenance of MPCs in bone marrow by enhancing cell survival alone and in combination with other cytokines.
Collapse
Affiliation(s)
- Younghee Lee
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, 46202, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wyman A, Lavin AL, Wilding GE, Gasiewicz TA. 2,3,7,8-tetrachlorodibenzo-p-dioxin does not directly alter the phenotype of maturing B cells in a murine coculture system. Toxicol Appl Pharmacol 2002; 180:164-77. [PMID: 12009856 DOI: 10.1006/taap.2002.9396] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), acting through the aromatic hydrocarbon receptor (AhR), elicits numerous toxicological effects, including immunosuppression. Previous work from our laboratory has suggested that TCDD exposure in mice is associated with altered lymphopoietic development, in particular altered B-cell phenotype in the bone marrow. It remains to be determined which specific hematopoietic populations or subpopulations within the marrow cavity are directly targeted by TCDD. To examine the effects of TCDD on developing B cells in vitro, a stromal coculture model was used. Primary bone marrow cells from male, 6- to 7-week-old C57Bl/6 mice were cocultured separately on two AhR-containing stromal cell lines (M2-10B4 and S17) that support B-cell development in the presence of IL-7. The cocultures were treated with 0 to 10 nM TCDD. Shifts in phenotype were quantified by cell surface marker staining and flow cytometry. Four populations in the maturing B cell (very early pre-pro-B, pre-pro-B, pro-B, and pre-B) were defined for quantification. The results show that the only statistically significant effect of TCDD was within the pre-pro-B population in cultures with the S17 stromal cell line. The increase in number of cells with this phenotype was seen in cultures with both wild-type and AhR-/- primary bone marrow cells. These results suggest that the maturing B220+ B cell is not the direct target for TCDD-induced bone marrow B-cell alterations.
Collapse
Affiliation(s)
- Amber Wyman
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|
30
|
Tulin EE, Onoda N, Hasegawa M, Nosaka T, Nomura H, Kitamura T. Genetic approach and phenotype-based complementation screening for identification of stroma cell-derived proteins involved in cell proliferation. Exp Cell Res 2002; 272:23-31. [PMID: 11740862 DOI: 10.1006/excr.2001.5394] [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: 11/22/2022]
Abstract
The functional capacities of stromal cell lines to support stem cell activity are heterogeneous and the mechanism of how they support bone marrow cultures remains unclear. Recently, we reported a strategy of functional analysis in which a genetic approach is combined with phenotype-based complementation screening to search for a novel secreted growth factor from mouse bone marrow stroma called ShIF that supported proliferation of bone marrow cells. To investigate the role of stromal cells in hemopoiesis, we extended this strategy to search for stroma-derived proteins that induce cell proliferation by establishing stroma-dependent Ba/F3 mutants of three stroma cell lines from two mouse tissues. Seven stroma-dependent Ba/F3 mutants were used as responder cells to identify cDNAs from stroma cell lines whose products supported proliferation not only to the mutant cells but also to hemopoietic progenitor cells in vitro.
Collapse
Affiliation(s)
- Edgardo E Tulin
- Chugai Research Institute for Molecular Medicine, Inc., 153-2 Nagai, Niihari, Ibaraki, 300-4101, Japan.
| | | | | | | | | | | |
Collapse
|
31
|
Fleming HE, Paige CJ. Pre-B cell receptor signaling mediates selective response to IL-7 at the pro-B to pre-B cell transition via an ERK/MAP kinase-dependent pathway. Immunity 2001; 15:521-31. [PMID: 11672535 DOI: 10.1016/s1074-7613(01)00216-3] [Citation(s) in RCA: 137] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
B lymphocyte development is regulated at multiple checkpoints, mediated by signals originating both inside and outside the cell. Two signaling pathways known to be essential in this process are interleukin-7 (IL-7) and the pre-B cell receptor (pBCR). We have shown previously that these signaling pathways intersect functionally. Specifically, response to low concentrations of IL-7 requires pBCR expression. In this report, we identify the ERK/MAP kinase pathway as a key regulatory component of this response. We propose a molecular mechanism for the selective expansion of pBCR(+) precursors and for the culling of inappropriately rearranged pro-B cells.
Collapse
Affiliation(s)
- H E Fleming
- Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
32
|
Gronthos S, Franklin DM, Leddy HA, Robey PG, Storms RW, Gimble JM. Surface protein characterization of human adipose tissue-derived stromal cells. J Cell Physiol 2001; 189:54-63. [PMID: 11573204 DOI: 10.1002/jcp.1138] [Citation(s) in RCA: 995] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human bone marrow stromal cells are a multipotent population of cells capable of differentiating into a number of mesodermal lineages as well as supporting hematopoeisis. Their distinct protein and gene expression phenotype is well characterized in the literature. Human adipose tissue presents an alternative source of multipotent stromal cells. In this study, we have defined the phenotype of the human adipose tissue-derived stromal cells in both the differentiated and undifferentiated states. Flow cytometry and immunohistochemistry show that human adipose tissue-derived stromal cells have a protein expression phenotype that is similar to that of human bone marrow stromal cells. Expressed proteins include CD9, CD10, CD13, CD29, CD34, CD44, CD 49(d), CD 49(e), CD54, CD55, CD59, CD105, CD106, CD146, and CD166. Expression of some of these proteins was further confirmed by PCR and immunoblot detection. Unlike human bone marrow-derived stromal cells, we did not detect the STRO-1 antigen on human adipose tissue-derived stromal cells. Cells cultured under adipogenic conditions uniquely expressed C/EBPalpha and PPARdelta, two transcriptional regulators of adipogenesis. Cells cultured under osteogenic conditions were more likely to be in the proliferative phases of the cell cycle based on flow cytometric analysis of PCNA and Ki67. The similarities between the phenotypes of human adipose tissue-derived and human bone marrow-derived stromal cells could have broad implications for human tissue engineering.
Collapse
Affiliation(s)
- S Gronthos
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, Bethesda, Maryland, USA
| | | | | | | | | | | |
Collapse
|
33
|
Popernack PM, Truong LT, Kamphuis M, Henderson AJ. Ectopic expression of CCAAT/enhancer binding protein beta (C/EBPbeta) in long-term bone marrow cultures induces granulopoiesis and alters stromal cell function. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2001; 10:631-42. [PMID: 11672509 DOI: 10.1089/152581601753193841] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CCAAT/enhancer binding proteins (C/EBP) have been demonstrated to impact directly the development of multiple hematopoietic lineages. However, the role of C/EBPbeta in the differentiation of various hematopoietic lineages has not been thoroughly examined. We used primary bone marrow cultures to assess directly the ability of C/EBPbeta to influence myelopoiesis. Retroviral expression vectors were used to express C/EBPbeta ectopically in murine primary long-term bone marrow cultures. The differentiation potential of these cells was determined using hematopoietic colony assays and differential staining of cells within the cultures. Bone marrow cultures that overexpressed C/EBPbeta had fewer myeloid progenitors and a significant increase in the number of granulocytes. The ability of C/EBPbeta to alter hematopoiesis in vitro was dependent on the presence of the transcriptional activation domain because LIP, which lacks this functional domain, did not decrease the ability of bone marrow cultures to support myeloid progenitors. These data also show that C/EBPbeta influences hematopoiesis by altering stromal cell function rather than the intrinsic developmental potential of myeloid progenitor cells.
Collapse
Affiliation(s)
- P M Popernack
- Department of Veterinary Science, Immunology Research Laboratories, The Pennsylvania State University, University Park, PA 16802-3500, USA
| | | | | | | |
Collapse
|
34
|
Montecino‐Rodriguez E, Dorshkind K. Regulation of hematopoiesis by gap junction‐mediated intercellular communication. J Leukoc Biol 2001. [DOI: 10.1189/jlb.70.3.341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Encarnacion Montecino‐Rodriguez
- Department of Pathology and Laboratory Medicine and the Jonsson Comprehensive Cancer Center, UCLA School of Medicine, Los Angeles, California
| | - Kenneth Dorshkind
- Department of Pathology and Laboratory Medicine and the Jonsson Comprehensive Cancer Center, UCLA School of Medicine, Los Angeles, California
| |
Collapse
|
35
|
Yamane T, Kunisada T, Tsukamoto H, Yamazaki H, Niwa H, Takada S, Hayashi SI. Wnt signaling regulates hemopoiesis through stromal cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:765-72. [PMID: 11441081 DOI: 10.4049/jimmunol.167.2.765] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hemopoietic cells develop in a complex milieu that is made up of diverse components, including stromal cells. Wnt genes, which are known to regulate the fate of the cells in a variety of tissues, are expressed in hemopoietic organs. However, their roles in hemopoiesis are not well characterized. In this study, we examined the roles of Wnt proteins in hemopoiesis using conditioned medium containing Wnt-3a. This conditioned medium dramatically reduced the production of B lineage cells and myeloid lineage cells, except for macrophages in the long-term bone marrow cultures grown on stromal cells, although the sensitivity to the conditioned medium differed, depending on the hemopoietic lineage. In contrast, the same conditioned medium did not affect the generation of B lineage or myeloid lineage cells in stromal cell-free conditions. These results suggested that Wnt proteins exert their effects through stromal cells. Indeed, these effects were mimicked by the expression of a stabilized form of beta-catenin in stromal cells. In this study, we demonstrated that Wnt signaling regulates hemopoiesis through stromal cells with selectivity and different degrees of the effect, depending on the hemopoietic lineage in the hemopoietic microenvironment.
Collapse
Affiliation(s)
- T Yamane
- Department of Immunology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-machi, Yonago 683-8503, Japan.
| | | | | | | | | | | | | |
Collapse
|
36
|
Khaldoyanidi S, Sikora L, Orlovskaya I, Matrosova V, Kozlov V, Sriramarao P. Correlation between nicotine-induced inhibition of hematopoiesis and decreased CD44 expression on bone marrow stromal cells. Blood 2001; 98:303-12. [PMID: 11435297 DOI: 10.1182/blood.v98.2.303] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This study demonstrates that in vivo exposure to cigarette smoke (CS) and in vitro treatment of long-term bone marrow cultures (LTBMCs) with nicotine, a major constituent of CS, result in inhibition of hematopoiesis. Nicotine treatment significantly delayed the onset of hematopoietic foci and reduced their size. Furthermore, the number of long-term culture-initiating cells (LTC-ICs) within an adherent layer of LTBMCs was significantly reduced in cultures treated with nicotine. Although the production of nonadherent mature cells and their progenitors in nicotine-treated LTBMCs was inhibited, this treatment failed to influence the proliferation of committed hematopoietic progenitors when added into methylcellulose cultures. Bone marrow stromal cells are an integral component of the hematopoietic microenvironment and play a critical role in the regulation of hematopoietic stem cell proliferation and self-renewal. Exposure to nicotine decreased CD44 surface expression on primary bone marrow-derived fibroblastlike stromal cells and MS-5 stromal cell line, but not on hematopoietic cells. In addition, mainstream CS altered the trafficking of hematopoietic stem/progenitor cells (HSPC) in vivo. Exposure of mice to CS resulted in the inhibition of HSPC homing into bone marrow. Nicotine and cotinine treatment resulted in reduction of CD44 surface expression on lung microvascular endothelial cell line (LEISVO) and bone marrow-derived (STR-12) endothelial cell line. Nicotine treatment increased E-selectin expression on LEISVO cells, but not on STR-12 cells. These findings demonstrate that nicotine can modulate hematopoiesis by affecting the functions of the hematopoiesis-supportive stromal microenvironment, resulting in the inhibition of bone marrow seeding by LTC-ICs and interfering with stem cell homing by targeting microvascular endothelial cells.
Collapse
Affiliation(s)
- S Khaldoyanidi
- Division of Vascular Biology, La Jolla Institute for Molecular Medicine, La Jolla, CA 92121, USA
| | | | | | | | | | | |
Collapse
|
37
|
Hahn BRANDIK, Piktel DEBRA, Gibson LAURAF, Landreth KENNETHS. Hematopoiesis: The Role of Stromal Integrin Interactions in Pro-B Cell Proliferation. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2001; 5:153-160. [PMID: 11399610 DOI: 10.1080/10245332.2000.11746501] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Recent studies using long term bone marrow cultures have concluded that adherence of lymphoid precursors to the underlying stromal cells is required for normal B cell development. However, the function of specific integrin interactions in B cell development remains unresolved. In our laboratory, we observed that maximal proliferation of pro-B cells required the presence of stromal cells and that stromal cytokines alone could not replace the requirement for stromal cell contact. For that reason, we questioned whether integrin interactions play a role in regulating pro-B cell proliferation in the bone marrow. Murine pro-B cell line Cl.92 expressed VLA-4, CD44, and fibronectin-receptor. Abrogation of binding of these molecules to stromal cell ligands using blocking antibodies resulted in failure of pro-B cell adherence and significant decreases in pro-B cell proliferation. Disruption of single integrin interactions did not compromise either adhesion of pro-B cells to stromal cells or IL-7 stimulated proliferation. Taken together, these data suggest that normal pro-B cells interact with stromal cells through multiple integrin interactions and that integrin mediated potentiation of pro-B cell proliferation is functionally redundant and not affected by failure of single ligand interactions.
Collapse
Affiliation(s)
- BRANDI K. Hahn
- Blood and Marrow Transplantation Program of the Mary Babb Randolph Cancer Center
| | | | | | | |
Collapse
|
38
|
Simmons PJ, Levesque JP, Haylock DN. Mucin-like molecules as modulators of the survival and proliferation of primitive hematopoietic cells. Ann N Y Acad Sci 2001; 938:196-206; discussion 206-7. [PMID: 11458509 DOI: 10.1111/j.1749-6632.2001.tb03590.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Current data suggest that interplay between two classes of molecules contributes to the regulation of hematopoiesis: hematopoietic growth factors, which regulate the survival, proliferation, and development of primitive hematopoietic cells and cell adhesion molecules (CAMs), which are responsible for the localization of hematopoiesis to the bone marrow (BM) and for mediating physical association between developing hematopoietic cells and marrow stromal tissue. A range of cell surface molecules representing several CAM superfamilies including integrins, selectins, the immunoglobulin gene superfamily and an emerging family of mucin-like molecules (the sialomucins) are involved in supporting cell-cell and cell-extracellular matrix (ECM) interactions between primitive hematopoietic cells and the stromal cell-mediated hematopoietic microenvironment (HM) of the bone marrow. There is abundant evidence in non-hematopoietic tissues that CAMs are signalling molecules which participate in a range of signal transduction events important not only for regulating cell adhesion and motility, but also for cell growth and survival. Although the signalling functions of CAMs have not been studied extensively in primitive hematopoietic progenitors (HPCs), extrapolation from burgeoning data in other systems is consistent with the hypothesis that hematopoiesis within the BM is regulated by interaction between signals generated locally by CAMs and those elicited by cytokines. Evidence in support of this notion was initially provided by studies on normal HPCs demonstrating cross-talk between members of the integrin superfamily and cytokine receptors. In this article we review recent reports that mucin-like molecules are also signalling molecules on primitive hematopoietic cells and that the signals they deliver potently inhibit hematopoiesis.
Collapse
Affiliation(s)
- P J Simmons
- Stem Cell Laboratory, Peter MacCallum Cancer Institute, St. Andrew's Place, East Melbourne, VIC 3002, Australia.
| | | | | |
Collapse
|
39
|
Tulin EE, Onoda N, Maeda M, Hasegawa M, Nosaka T, Nomura H, Asano S, Kitamura T. A novel secreted form of immune suppressor factor with high homology to vacuolar ATPases identified by a forward genetic approach of functional screening based on cell proliferation. J Biol Chem 2001; 276:27519-26. [PMID: 11375395 DOI: 10.1074/jbc.m101781200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In the search for stromal-derived growth factors, we have identified a novel secreted short form of immune suppressor factor (ISF) using a combination of a genetic approach and retrovirus-mediated functional screening. This protein, which we termed ShIF, was isolated based on its ability to support proliferation of a mutant clone S21, which was established from Ba/F3 cells that are usually interleukin-3-dependent but became dependent on a stroma cell line ST2 after chemical mutagenesis. ISF, a membrane protein harboring six transmembrane domains, was reported to have immunosuppressive functions. The coding region of ShIF started from the third transmembrane domain of ISF. Biochemical analysis demonstrated that ShIF was expressed in both the secreted and membrane-bound forms of 27-kDa protein, which was supposed to have an internal ATG present in the third transmembrane domain of ISF as a start codon. In addition to the full-length form of ISF, a major protein with a molecular size of 27 kDa was also expressed through the proteolytic process of ISF. ShIF resembles this naturally occurring short form of ISF (sISF). Deletion analysis of the major domains of ISF cDNA revealed that ShIF is an active functional domain of ISF with a capability to support proliferation of S21 cells. Enforced expression of ShIF in MS10 cells, bone marrow stroma cells that do not express endogenous ShIF or ISF, conferred on the cells an ability to support the growth of S21 cells as well as bone marrow cells. Interestingly, ShIF shows a high sequence homology to the C-terminal part of a 95-kDa yeast vacuolar H (+)-ATPase subunit, Vph1p (39%), and a 116-kDa proton pump (VPP1) (54%) of the rat and bovine synaptic vesicle. Therefore, it is possible that ShIF also acts as a proton pump and somehow prevents the cells from undergoing apoptosis.
Collapse
Affiliation(s)
- E E Tulin
- Division of Hematopoietic Factors and the Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Sandel PC, Gendelman M, Kelsoe G, Monroe JG. Definition of a novel cellular constituent of the bone marrow that regulates the response of immature B cells to B cell antigen receptor engagement. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:5935-44. [PMID: 11342608 DOI: 10.4049/jimmunol.166.10.5935] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previously we defined a Thy1(dull) bone marrow-derived cell population that regulated fate decisions by immature B cells after Ag receptor signaling. The microenvironmental signals provided by this cell population were shown to redirect the B cell Ag receptor -induced apoptotic response of immature B cells toward continued recombination-activating gene (RAG) expression and secondary light chain recombination (receptor editing). Neither the identity of the cell responsible for this activity nor its role in immature B cell development in vivo were addressed by these previous studies. Here we show that this protective microenvironmental niche is defined by the presence of a novel Thy1(dull), DX5(pos) cell that can be found in close association with immature B cells in vivo. Depletion of this cell eliminates the anti-apoptotic effect of bone marrow in vitro and leads to a significant decrease in the number and frequency of bone marrow immature B cells in vivo. We propose that, just as the bone marrow environment is essential for the survival and progression of pro-B and pre-B cells through their respective developmental checkpoints, this cellular niche regulates the progression of immature stage B cells through negative selection.
Collapse
Affiliation(s)
- P C Sandel
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
41
|
Hall BM, Fortney JE, Gibson LF. Alteration of nuclear factor-kappaB (NF-kappaB) expression in bone marrow stromal cells treated with etoposide. Biochem Pharmacol 2001; 61:1243-52. [PMID: 11322928 DOI: 10.1016/s0006-2952(01)00602-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bone marrow stromal cells are an essential regulatory component in the hematopoietic microenvironment. Regulation of hematopoietic cell development is mediated, in part, through interaction of progenitor cells with stromal cell vascular cell adhesion molecule-1 (VCAM-1). VCAM-1 expression has been shown to be driven primarily by binding of nuclear factor-kappaB (NF-kappaB) to two consensus binding sites in the promoter region. In this study, we show that down-regulation of VCAM-1 by the chemotherapeutic agent etoposide (VP-16) is associated with altered cellular localization of NF-kappaB. We demonstrated that VCAM-1 was diminished at the transcriptional level following treatment of stromal cells with VP-16, without alteration of VCAM-1 stability. Culture of bone marrow stromal cells in VP-16 resulted in reduced nuclear RelA (p65), a modest increase in nuclear NF-kappaB1 (p50), and reduced NF-kappaB binding to its DNA consensus sequence. Total levels of the NF-kappaB inhibitor Ikappa-Balpha were reduced during exposure to VP-16. Following removal of VP-16 from the culture, p65 and p50 nuclear profiles approximated those of untreated stromal cells, and VCAM-1 protein expression was restored. The current study indicates that NF-kappaB is a target molecule that is responsive to VP-16-induced damage in bone marrow stromal cells. As the primary transcription factor that promotes VCAM-1 expression, the observed changes in p65 and p50 cellular localization during treatment have a direct consequence for stromal cell function. The myriad of genes regulated by NF-kappaB, including both adhesion molecules and cytokines that contribute to stromal cell function, make chemotherapy-induced disruption of NF-kappaB biologically significant. Alterations in NF-kappaB activity may provide one measure by which the effects of aggressive treatment strategies on the bone marrow microenvironment can be evaluated.
Collapse
Affiliation(s)
- B M Hall
- Department of Pediatrics, Robert C. Byrd Health Sciences Center, P.O. Box 9214, West Virginia University, 26506, Morgantown, WV, USA
| | | | | |
Collapse
|
42
|
Hess J, Werner A, Wirth T, Melchers F, Jäck HM, Winkler TH. Induction of pre-B cell proliferation after de novo synthesis of the pre-B cell receptor. Proc Natl Acad Sci U S A 2001; 98:1745-50. [PMID: 11172022 PMCID: PMC29328 DOI: 10.1073/pnas.98.4.1745] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The assembly of a pre-B cell receptor (pre-BCR) composed of an Ig mu heavy chain (mu H-chain), the surrogate light (SL) chain, and the Ig alpha/beta dimer is critical for late pro-B cells to advance to the pre-B cell stage. By using a transgenic mouse model, in which mu H-chain synthesis is solely driven by a tetracycline-controlled transactivator, we show that de novo synthesis of mu H-chain in transgenic pro-B cells not only induces differentiation but also proliferation. This positive effect of mu H-chain synthesis on proliferation requires the presence of SL chain and costimulatory signals provided by stromal cells or IL-7. We conclude that pre-BCR signaling induces clonal expansion of early pre-B cells.
Collapse
Affiliation(s)
- J Hess
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Würzburg, Versbacher Strasse 5, D-97078 Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Induction of pre-B cell proliferation after de novo synthesis of the pre-B cell receptor. Proc Natl Acad Sci U S A 2001. [PMID: 11172022 PMCID: PMC29328 DOI: 10.1073/pnas.041492098] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The assembly of a pre-B cell receptor (pre-BCR) composed of an Ig mu heavy chain (mu H-chain), the surrogate light (SL) chain, and the Ig alpha/beta dimer is critical for late pro-B cells to advance to the pre-B cell stage. By using a transgenic mouse model, in which mu H-chain synthesis is solely driven by a tetracycline-controlled transactivator, we show that de novo synthesis of mu H-chain in transgenic pro-B cells not only induces differentiation but also proliferation. This positive effect of mu H-chain synthesis on proliferation requires the presence of SL chain and costimulatory signals provided by stromal cells or IL-7. We conclude that pre-BCR signaling induces clonal expansion of early pre-B cells.
Collapse
|
44
|
Abstract
Abstract
Adult bone marrow is a major site for hematopoiesis, and reduction of the bone marrow cavity induces hematopoiesis in extramarrow tissues. To investigate the rudimentary intramarrow and the compensatory extramarrow hematopoiesis, particularly B lymphopoiesis, we used 3 osteopetrotic mouse strains [op/op, mi/mi, and Fos(−/−)], which are severely deficient in functional osteoclasts and therefore form inadequate bone marrow cavities. We found that bone marrow in these osteopetrotic mice supports myelopoiesis but not B lymphopoiesis, although cells that have the potential to differentiate into B lineage cells are present in the bone marrow. Although B lymphopoiesis normally occurs both in the spleen and liver of newborn mice, compensatory B lymphopoiesis in adultop/op and mi/mi mice is observed only in the liver, while myelopoiesis is enhanced in both organs. Interestingly, mice lacking the Fos proto-oncogene exhibit B lymphopoiesis in the spleen as well as liver. The amounts of expression of steel factor, Flt3/Flk-2 ligand, and interleukin-7 in the bone marrow, spleen, or liver were not significantly affected in these osteopetrotic mutants. These findings suggest that the volume of the bone marrow cavity regulates B lymphopoiesis without affecting the production of certain hematopoietic growth factors. The splenic microenvironments that support both myelopoiesis and B lymphopoiesis in the neonatal stage are lost in adults and are not reactivated even in the osteopetrotic adults unless the Fos gene is disrupted.
Collapse
|
45
|
Dorshkind K, Horseman ND. The roles of prolactin, growth hormone, insulin-like growth factor-I, and thyroid hormones in lymphocyte development and function: insights from genetic models of hormone and hormone receptor deficiency. Endocr Rev 2000; 21:292-312. [PMID: 10857555 DOI: 10.1210/edrv.21.3.0397] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
An extensive literature suggesting that PRL, GH, IGF-I, and thyroid hormones play an important role in immunity has evolved. Because the use of one or more of these hormones as immunostimulants in humans is being considered, it is of critical importance to resolve their precise role in immunity. This review addresses new experimental evidence from analysis of lymphocyte development and function in mice with genetic defects in expression of these hormones or their receptors that calls into question the presumed role played by some of these hormones and reveals unexpected effects of others. These recent findings from the mutant mouse models are integrated and placed in context of the wider literature on endocrine-immune system interactions. The hypothesis that will be developed is that, with the exception of a role for thyroid hormones in B cell development, PRL, GH, and IGF-I are not obligate immunoregulators. Instead, they apparently act as anabolic and stress-modulating hormones in most cells, including those of the immune system.
Collapse
Affiliation(s)
- K Dorshkind
- Department of Pathology and Laboratory Medicine and The Jonsson Comprehensive Cancer Center, University of California at Los Angeles School of Medicine, 90095-1732, USA
| | | |
Collapse
|
46
|
Abstract
Adult bone marrow is a major site for hematopoiesis, and reduction of the bone marrow cavity induces hematopoiesis in extramarrow tissues. To investigate the rudimentary intramarrow and the compensatory extramarrow hematopoiesis, particularly B lymphopoiesis, we used 3 osteopetrotic mouse strains [op/op, mi/mi, and Fos(−/−)], which are severely deficient in functional osteoclasts and therefore form inadequate bone marrow cavities. We found that bone marrow in these osteopetrotic mice supports myelopoiesis but not B lymphopoiesis, although cells that have the potential to differentiate into B lineage cells are present in the bone marrow. Although B lymphopoiesis normally occurs both in the spleen and liver of newborn mice, compensatory B lymphopoiesis in adultop/op and mi/mi mice is observed only in the liver, while myelopoiesis is enhanced in both organs. Interestingly, mice lacking the Fos proto-oncogene exhibit B lymphopoiesis in the spleen as well as liver. The amounts of expression of steel factor, Flt3/Flk-2 ligand, and interleukin-7 in the bone marrow, spleen, or liver were not significantly affected in these osteopetrotic mutants. These findings suggest that the volume of the bone marrow cavity regulates B lymphopoiesis without affecting the production of certain hematopoietic growth factors. The splenic microenvironments that support both myelopoiesis and B lymphopoiesis in the neonatal stage are lost in adults and are not reactivated even in the osteopetrotic adults unless the Fos gene is disrupted.
Collapse
|
47
|
Herrera L, Farah RA, Pellegrini VA, Aquino DB, Sandler ES, Buchanan GR, Vitetta ES. Immunotoxins against CD19 and CD22 are effective in killing precursor-B acute lymphoblastic leukemia cells in vitro. Leukemia 2000; 14:853-8. [PMID: 10803517 DOI: 10.1038/sj.leu.2401779] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Monoclonal antibodies (Mabs) conjugated to toxins or their subunits (immunotoxins or ITs) are undergoing clinical testing in adults with a variety of malignancies. The potential impact of this form of therapy in pediatric precursor B-lineage acute lymphoblastic leukemia (pre-B ALL) has yet to be determined. Mabs directed against the cell surface antigens, CD19 and CD22 conjugated to deglycosylated ricin A chain (dgRTA) have been tested in patients with non-Hodgkin's lymphoma (NHL), but not in patients with pre-B ALL. Because of the encouraging performance of these ITs in phase I trials, we evaluated the specific cytotoxicity of anti-CD19 (HD37-dgRTA) and anti-CD22 (RFB4-dgRTA) ITs or their combination (Combotox) on patient-derived pre-B ALL cells maintained in vitro on a stromal feeder layer. After 48 h in culture, cytotoxicity to tumor cells was determined by flow cytometry using propidium iodide (PI) and fluorescein isothiocyanate (FITC)-conjugated anti-CD10, 19, and 22. Both RFB4-dgRTA and HD37-dgRTA induced a statistically significant reduction in the number of viable leukemic cells, and Combotox was even more effective. Our results demonstrate that these ITs are specifically cytotoxic to primary pre-B ALL cells and that they should be further evaluated for the therapy of B-lineage ALL.
Collapse
Affiliation(s)
- L Herrera
- Cancer Immunobiology Center, The University of Texas Southwestern Medical Center at Dallas, 75235-8576, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Foster MP, Montecino-Rodriguez E, Dorshkind K. Proliferation of Bone Marrow Pro-B Cells Is Dependent on Stimulation by the Pituitary/Thyroid Axis. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.11.5883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The frequency and absolute number of pro-B, pre-B, and B cells in the bone marrow of the hypothyroid strain of mice are significantly reduced compared with those of their normal littermates. To investigate why this is the case, various B cell developmental processes were examined in the thyroid hormone-deficient mice. These studies revealed that the frequency of pro-B cells in the S-G2/M phase of the cell cycle was significantly reduced in hypothyroid mice. That thyroid hormone deficiency was responsible for this proliferation defect was established by demonstrating that treatment of hypothyroid mice with thyroxine resulted in a specific increase in the frequency and total number of cycling pro-B cells. The latter effect was paralleled by increases in the frequency and number of bone marrow B lineage cells. Additional in vitro experiments revealed that at least some thyroid hormone effects were directly mediated on the bone marrow. Taken together, these data demonstrate that thyroid hormones are required for normal B cell production in the bone marrow through regulation of pro-B cell proliferation and establish a role for the pituitary/thyroid axis in B cell development.
Collapse
Affiliation(s)
- Melanie P. Foster
- Department of Pathology and Laboratory Medicine and Jonsson Comprehensive Cancer Center, School of Medicine, University of California, Los Angeles, CA 90095
| | - Encarnacion Montecino-Rodriguez
- Department of Pathology and Laboratory Medicine and Jonsson Comprehensive Cancer Center, School of Medicine, University of California, Los Angeles, CA 90095
| | - Kenneth Dorshkind
- Department of Pathology and Laboratory Medicine and Jonsson Comprehensive Cancer Center, School of Medicine, University of California, Los Angeles, CA 90095
| |
Collapse
|
49
|
Lagneaux L, Delforge A, De Bruyn C, Bernier M, Bron D. Adhesion to bone marrow stroma inhibits apoptosis of chronic lymphocytic leukemia cells. Leuk Lymphoma 1999; 35:445-53. [PMID: 10609782 DOI: 10.1080/10428199909169609] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
B-cell chronic lymphocytic leukemia (B-CLL) is characterized by the accumulation of monoclonal long-lived B cells which are apparently resistant to normal apoptotic regulation. Since bone marrow stromal cells play an essential role in B lymphopoiesis, we have investigated whether stromal cells influence B-CLL cell survival. Our results indicate that intimate contact with stromal cells reduces B-CLL cell apoptosis and prevents the loss of bcl-2 protein expression. Binding of B-CLL cells to stromal cells requires simultaneous action of beta1 and beta2 integrins. The interaction between B-CLL cells and other cell types seems important for their survival and may represent an important mechanism underlying accumulation of malignant cells in B-CLL patients.
Collapse
Affiliation(s)
- L Lagneaux
- Service de Médecine Interne et Laboratoire d'investigation Clinique Henri Tagnon, Institut J. Bordet, Brussels, Belgium.
| | | | | | | | | |
Collapse
|
50
|
Tomita T, Takeuchi E, Toyosaki-Maeda T, Oku H, Kaneko M, Takano H, Sugamoto K, Ohzono K, Suzuki R, Ochi T. Establishment of nurse-like stromal cells from bone marrow of patients with rheumatoid arthritis: indication of characteristic bone marrow microenvironment in patients with rheumatoid arthritis. Rheumatology (Oxford) 1999; 38:854-63. [PMID: 10515647 DOI: 10.1093/rheumatology/38.9.854] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To investigate the microenvironment of bone marrow (BM) of patients with rheumatoid arthritis (RA). METHODS Nurse cell-like BM stromal cell lines were established from BM mononuclear cells of patients with RA. We examined the various characteristics of these cell lines, including morphology, pseudoemperipolesis activity, cell surface markers, cytokine production and hyaluronan (HA) production. RESULTS These RA BM nurse cell-like lines (RA-BMNC) were of mesenchymal origin and positive for CD44, CD54 and HLA-DR. They were defined as nurse cells because of pseudoemperipolesis activity that allowed lymphocytes to migrate underneath. RA-BMNC lines produced HA and multiple cytokines including interleukin (IL)-6, IL-7, IL-8 and granulocyte-macrophage colony-stimulating factor (GM-CSF). HA production by BM stromal cells was correlated with pseudoemperipolesis activity. RA-BMNC produced significantly higher levels of IL-6, IL-8 and GM-CSF by co-culture with lymphocytes. The cells also produced IL-1beta, G-CSF and tumour necrosis factor only when co-cultured with lymphocytes. The RA-BMNC maintained the growth of CD14+ myeloid cells unique to severe RA. CONCLUSION The present results both indicate that RA-BMNC are nurse cells and suggest that they may play an important role in the pathogenesis of RA.
Collapse
Affiliation(s)
- T Tomita
- Department of Orthopaedic Surgery, Osaka University Medical School, Suita, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|