2201
|
Abstract
PURPOSE OF REVIEW Transplantation of hematopoietic stem cells is dependent upon the successful homing, engraftment and repopulation of stem cells in the bone marrow. Stem cell homing through the circulation to the bone marrow is the critical first step in this process. This review discusses the latest progress in defining the molecular processes underlying stem cell homing and the specialized niches where stem cells reside. RECENT FINDINGS Over the past decade, remarkable advances have been made in characterizing the complex sequence of events involved in stem cell homing to the bone marrow. Specifically, the molecular basis of stem cell adhesion and rolling along bone marrow sinusoidal endothelial cells has been defined, and mechanisms underlying endothelial transmigration and enlodgement in bone marrow niches have now been identified. The processes governing hematopoietic stem cell homing to the bone marrow also regulate hematopoietic stem cell migration to extramedullary tissues and the metastasis of cancer stem cells. Improved understanding of these processes has catalyzed the development of therapies to facilitate stem cell mobilization for clinical purposes. SUMMARY Several components of the essential process of stem cell homing have now been characterized. Cell adhesion molecules and their ligands, extracellular matrix components, chemokines, and specialized bone marrow niches all participate in the precise regulation of this process.
Collapse
Affiliation(s)
- John P Chute
- Division of Cellular Therapy, Duke University Medical Center, Durham, North Carolina 27710, USA.
| |
Collapse
|
2202
|
Kacena MA, Gundberg CM, Horowitz MC. A reciprocal regulatory interaction between megakaryocytes, bone cells, and hematopoietic stem cells. Bone 2006; 39:978-984. [PMID: 16860008 DOI: 10.1016/j.bone.2006.05.019] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2006] [Revised: 05/26/2006] [Accepted: 05/27/2006] [Indexed: 11/30/2022]
Abstract
A growing body of evidence suggests that megakaryocytes (MK) or their growth factors play a role in skeletal homeostasis. MK have been shown to express and/or secrete several bone-related proteins including osteocalcin, osteonectin, bone sialoprotein, osteopontin, bone morphogenetic proteins, and osteoprotegerin. In addition, at least 3 mouse models have been described in which MK number was significantly elevated with an accompanying marked increase in bone mineral density. Mice overexpressing thrombopoietin, the major MK growth factor, have an osteosclerotic bone phenotype. Mice deficient in transcription factors GATA-1 and NF-E2, which are required for the differentiation of MK, exhibited a strikingly increased bone mass. Importantly, recent studies have demonstrated that MK can stimulate osteoblast (OB) proliferation and differentiation in vitro and that they can also inhibit osteoclast (OC) formation in vitro. These findings suggest that MK play a dual role in skeletal homeostasis by stimulating formation while simultaneously inhibiting resorption. Conversely, cells of the osteoblast lineage support hematopoiesis, including megakaryopoiesis. Postnatal hematopoiesis occurs almost solely in the bone marrow (BM), close to or on endosteal surfaces. This finding, in conjunction with the observed contact of OB with hematopoietic cells, has lead investigators to explore the molecular and cellular interactions between hematopoietic cells and cells of the OB lineage. Importantly, it has been shown that many of the cytokines that are critical for normal hematopoiesis and megakaryopoiesis are produced by OB. Indeed, culturing osteoblasts with CD34+ BM cells significantly enhances hematopoietic cell number by both enhancing the proliferation of long-term culture initiating cells and the proliferation and differentiation of MK. These data are consistent with cells in the OB lineage playing a critical role in the hematopoietic niche. Overall, these observations demonstrate the importance of MK-bone cell interactions in both skeletal homeostasis and hematopoiesis.
Collapse
Affiliation(s)
- Melissa A Kacena
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, P.O. Box 208071, New Haven, CT 06520-0871, USA.
| | - Caren M Gundberg
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, P.O. Box 208071, New Haven, CT 06520-0871, USA
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, P.O. Box 208071, New Haven, CT 06520-0871, USA
| |
Collapse
|
2203
|
Abe A, Kiyoi H, Ninomiya M, Yamazaki T, Murase T, Ozeki K, Suzuki M, Hayakawa F, Katsumi A, Emi N, Naoe T. Establishment of a Stroma-Dependent Human Acute Myelomonocytic Leukemia Cell Line, NAMO-2, with FLT3 Tandem Duplication. Int J Hematol 2006; 84:328-36. [PMID: 17118759 DOI: 10.1532/ijh97.06056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have established a stroma-dependent myelomonocytic cell line, NAMO-2, with FLT3 internal tandem duplication (FLT3/ITD). Leukemia cells from a patient with acute myelomonocytic leukemia were administered to form subcutaneous tumors in nude mice, which were maintained successively, although we failed to establish continuously growing cells from the original leukemia cell culture. In the cultures of cells from subcutaneous tumors, there were stroma cells that had originated from the nude mice and showed continuous growth. The leukemia cells showed continuous growth dependent on this stroma, and this cell line was named NAMO-2. Detection of FLT3/ITD by the reverse transcriptase polymerase chain reaction (PCR) and genomic PCR showed that NAMO-2 was homozygous for FLT3/ITD. Constitutive activation of FLT3 was detected by Western blotting, and the phosphorylation of Akt, MEK, and STAT5 was also observed. FLT3 kinase inhibitor AG1296 specifically inhibited cell growth. NAMO-2 provides a useful tool to analyze adherence-dependent survival signaling of leukemia with FLT3/ITD and a model for the screening of FLT3 kinase inhibitors.
Collapse
Affiliation(s)
- Akihiro Abe
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2204
|
Beeton CA, Bord S, Ireland D, Compston JE. Osteoclast formation and bone resorption are inhibited by megakaryocytes. Bone 2006; 39:985-990. [PMID: 16870519 DOI: 10.1016/j.bone.2006.06.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 04/26/2006] [Accepted: 06/01/2006] [Indexed: 10/24/2022]
Abstract
It has been previously reported that addition of megakaryocytes (MKs) to osteoblasts in vitro results in increased osteoblastic collagen and osteoprotegerin (OPG) production, suggesting a role for MKs in bone formation. To further investigate this role, we have studied the effects of MKs on osteoclast formation and activity. Human osteoclasts were generated from CD14 monocytes isolated from peripheral blood and cultured in the presence of M-CSF and sRANKL on dentine and calcium phosphate substrates. MKs were generated from CD34+ cells isolated from either human peripheral blood or cord blood and cultured in liquid medium for 6 days, after which time maturing MKs (CD61-positive cells) were isolated and added to monocyte cultures. After 6 and 9 days of culture, the number of osteoclasts identified morphologically and by TRAP staining was counted. Cells were removed and the area of resorption was identified by von Kossa staining and quantitatively assessed by image analysis. The addition of MKs to osteoclast cultures at day 0 inhibited the number of osteoclasts formed 1.9-fold (p>0.003), whereas addition at 3 days had no effect on osteoclast number. The presence of MKs inhibited resorption 8.7-fold when co-cultured with osteoclasts from day 0 (p>0.004), but only by 3.1-fold when co-cultured from day 3 (p>0.01). In dose-response experiments, it was found that 1-10% of MKs added to monocyte cultures elicited the greatest inhibition of resorption. Similar osteoclast cultures were treated with CD61-negative cells (non-MKs) to confirm that the inhibition of osteoclast formation and activity was specifically due to MKs. Experiments with a cell-impermeable membrane indicated that both cell to cell contact and release of soluble factor(s) were involved in mediating these effects. These results show that MKs inhibit osteoclast formation and activity. The most pronounced effects were seen when MKs and osteoclasts were co-cultured from day 0, suggesting that MKs act primarily on osteoclast precursors.
Collapse
Affiliation(s)
- C A Beeton
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Addenbrooke's Hospital, Box 157, Hills Road, Cambridge, CB2 2QQ, UK.
| | - S Bord
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Addenbrooke's Hospital, Box 157, Hills Road, Cambridge, CB2 2QQ, UK
| | - D Ireland
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Addenbrooke's Hospital, Box 157, Hills Road, Cambridge, CB2 2QQ, UK
| | - J E Compston
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Addenbrooke's Hospital, Box 157, Hills Road, Cambridge, CB2 2QQ, UK
| |
Collapse
|
2205
|
Whitfield JF. Parathyroid hormone: A novel tool for treating bone marrow depletion in cancer patients caused by chemotherapeutic drugs and ionizing radiation. Cancer Lett 2006; 244:8-15. [PMID: 16540235 DOI: 10.1016/j.canlet.2006.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Revised: 02/07/2006] [Accepted: 02/08/2006] [Indexed: 12/28/2022]
Abstract
Between 1958 and the late 1970s it was learned that PTH (the parathyroid hormone) could directly stimulate the initiation of DNA replication by murine CFU-S (colony-forming unit-spleen) cells via cyclic AMP, stimulate the proliferation of normal and X-irradiated murine and rat bone marrow cells, control hematopoiesis, and increase the survival of X-irradiated mice and rats when injected any time between 18h before and 3h after X-irradiation. Since then, it has been shown that the hematopoietic stem cell niche consists of PTH receptor-bearing, osteoblastic trabecular bone-lining cells that maintain the stem cells' (HSCs') proliferatively quiescent 'stemness' by various gene up-regulating and down-regulating signals caused by the tight adhesion of the HSCs to the osteoblastic niche-lining cells. Stimulating the osteoblastic lining cells with recombinant human PTH-(1-34) (Forteo) causes a cyclic AMP-mediated enlargement of the HSC pool and promotes bone marrow transplant engraftment and growth and the survival of lethally irradiated mice. But this is only the beginning of the exploitation of the PTHs for marrow engraftment. It must now be determined whether the marrow engraftment-enhancing action of this potent bone growth-stimulating PTH can be extended from mice to rats and monkeys. It must be determined whether two other PTH peptides, rhPTH-(1-84) [Preos]and [Leu(27)]cyclo(Glu(22)-Lys(26))hPTH-(1-31)NH(2) [Ostabolin-C]) are as effective as or better than rhPTH-(1-34)(Forteo). Since, all three peptides are on the market, or nearing the market, for safely and strongly stimulating bone growth and treating osteoporosis one or all of them may become valuable tools for safely promoting the engraftment of peripherally harvested HSCs in cancer patients whose bone marrows have been 'emptied' by chemotherapeutic drugs or ionizing radiation.
Collapse
Affiliation(s)
- James F Whitfield
- Institute for Biological Sciences, National Research Council of Canada, Building M-54, Montreal Road Campus, Ottawa, Ont., Canada K1A 0R6.
| |
Collapse
|
2206
|
Zeng L, Rahrmann E, Hu Q, Lund T, Sandquist L, Felten M, O'Brien TD, Zhang J, Verfaillie C. Multipotent Adult Progenitor Cells from Swine Bone Marrow. Stem Cells 2006; 24:2355-66. [PMID: 16931778 DOI: 10.1634/stemcells.2005-0551] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We show that multipotent adult progenitor cells (MAPCs) can be derived from both postnatal and fetal swine bone marrow (BM). Although swine MAPC (swMAPC) cultures are initially mixed, cultures are phenotypically homogenous by 50 population doublings (PDs) and can be maintained as such for more than 100 PDs. swMAPCs are negative for CD44, CD45, and major histocompatibility complex (MHC) classes I and II; express octamer binding transcription factor 3a (Oct3a) mRNA and protein at levels close to those seen in human ESCs (hESCs); and have telomerase activity preventing telomere shortening even after 100 PDs. Using quantitative-reverse transcription-polymerase chain reaction (Q-RT-PCR), immunofluorescence, and functional assays, we demonstrate that swMAPCs differentiate into chondrocytes, adipocytes, osteoblasts, smooth muscle cells, endothelium, hepatocyte-like cells, and neuron-like cells. Consistent with what we have shown for human and rodent MAPCs, Q-RT-PCR demonstrated a significant upregulation of transcription factors and other lineage-specific transcripts in a time-dependent fashion similar to development. When swMAPCs were passaged for 3-6 passages at high density (2,000-8,000 cells per cm(2)), Oct3a mRNA levels were no longer detectable, cells acquired the phenotype of mesenchymal stem cells (CD44(+), MHC class I(+)), and could differentiate into typical mesenchymal lineages (adipocytes, osteoblasts, and chondroblasts), but not endothelium, hepatocyte-like cells, or neuron-like cells. Even if cultures were subsequently replated at low density (approximately 100-500 cells per cm(2)) for >20 PDs, Oct3a was not re-expressed, nor were cells capable of differentiating to cells other than mesenchymal-type cells. This suggests that the phenotype and functional characteristics of swMAPCs may not be an in vitro culture phenomenon.
Collapse
Affiliation(s)
- Lepeng Zeng
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
2207
|
Ninomiya M, Abe A, Katsumi A, Xu J, Ito M, Arai F, Suda T, Ito M, Kiyoi H, Kinoshita T, Naoe T. Homing, proliferation and survival sites of human leukemia cells in vivo in immunodeficient mice. Leukemia 2006; 21:136-42. [PMID: 17039228 DOI: 10.1038/sj.leu.2404432] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cellular components of the hematopoietic stem cell niche have been gradually identified. However, the niche for malignant hematopoiesis remains to be elucidated. Here, using human leukemia cells, which could be transplanted to immunodeficient mice, we studied the in vivo homing, proliferation and survival sites by immunohistopathology, compared with the corresponding sites for cord blood CD34(+) (CBCD34(+)) cells. The human leukemia cells initially localized on the surface of osteoblasts in the epiphysial region, and expanded to the inner vascular and diaphysial regions within 4 weeks. The percentage of CD34(+) leukemia cells in the bone marrow was transiently increased up to 50%. In vivo 5-bromo-2'-deoxyuridine labeling revealed that the epiphysis was the most active site for leukemia cell proliferation. CBCD34(+) cells showed the similar pattern of homing and proliferation to leukemia cells. After high-dose administration of cytosine-1-beta-D-arabinofuranoside, residual leukemia cells were localized in the perivascular endothelium as well as in contact with the trabecular endosteum. These findings suggest that xenotransplantation into immunodeficient mice provides a useful model to study the leukemia niche.
Collapse
Affiliation(s)
- M Ninomiya
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2208
|
Abstract
Niche has become the most important issue in stem cell biology, but it is still a hypothetical notion that cannot be defined in a better way than the microenvironment surrounding stem cells. Using a melanocyte stem cell system as a model, we have analyzed the cellular and molecular requirements for differentiation of quiescent stem cells. Our results demonstrate the multiple subsets within the stem cell compartment and thus suggests the complexity of niche.
Collapse
|
2209
|
|
2210
|
Abstract
Hematopoietic stem cells (HSCs) develop during embryogenesis in a complex process that involves multiple anatomical sites. Once HSC precursors have been specified from mesoderm, they have to mature into functional HSCs and undergo self-renewing divisions to generate a pool of HSCs. During this process,developing HSCs migrate through various embryonic niches, which provide signals for their establishment and the conservation of their self-renewal ability. These processes have to be recapitulated to generate HSCs from embryonic stem cells. Elucidating the interactions between developing HSCs and their niches should facilitate the generation and expansion of HSCs in vitro to exploit their clinical potential.
Collapse
Affiliation(s)
- Hanna K A Mikkola
- Department of Molecular, Cell and Developmental Biology, Jonsson Comprehensive Cancer Center, Institute for Stem Cell Biology and Medicine, University of California, Los Angeles, CA 90095, USA.
| | | |
Collapse
|
2211
|
Li Z, Li L. Understanding hematopoietic stem-cell microenvironments. Trends Biochem Sci 2006; 31:589-95. [PMID: 16911868 DOI: 10.1016/j.tibs.2006.08.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Revised: 07/04/2006] [Accepted: 08/02/2006] [Indexed: 01/15/2023]
Abstract
The hematopoietic system is the paradigm for adult mammalian stem-cell research. Recent advances have improved our understanding of the cellular and molecular components of the microenvironment - or niche - that regulates hematopoietic stem cells (HSCs). Here, we summarize the molecular and cellular properties of two types of niche, namely the osteoblastic and the vascular niche, in homeostatic regulation of HSC behavior, including its maintenance, proliferation, differentiation, mobilization and homing. We highlight the most recent findings and point to an important trend to the study of niche activity in cancers. Knowledge of the basic features of the HSC niches, including physical location, cell type and various signaling pathways, should provide insights into other stem-cell systems and benefit clinical applications.
Collapse
Affiliation(s)
- Zhixing Li
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.
| | | |
Collapse
|
2212
|
Matsumoto T, Kawamoto A, Kuroda R, Ishikawa M, Mifune Y, Iwasaki H, Miwa M, Horii M, Hayashi S, Oyamada A, Nishimura H, Murasawa S, Doita M, Kurosaka M, Asahara T. Therapeutic potential of vasculogenesis and osteogenesis promoted by peripheral blood CD34-positive cells for functional bone healing. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:1440-57. [PMID: 17003498 PMCID: PMC1698844 DOI: 10.2353/ajpath.2006.060064] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/20/2006] [Indexed: 12/20/2022]
Abstract
Failures in fracture healing are mainly caused by a lack of vascularization. Adult human circulating CD34+ cells, an endothelial/hematopoietic progenitor-enriched cell population, have been reported to differentiate into osteoblasts in vitro; however, the therapeutic potential of CD34+ cells for fracture healing is still unclear. Therefore, we performed a series of experiments to test our hypothesis that functional fracture healing is supported by vasculogenesis and osteogenesis via regenerative plasticity of CD34+ cells. Peripheral blood CD34+ cells, isolated from total mononuclear cells of adult human volunteers, showed gene expression of osteocalcin in 4 of 20 freshly isolated cells by single cell reverse transcriptase-polymerase chain reaction analysis. Phosphate-buffered saline, mononuclear cells, or CD34+ cells were intravenously transplanted after producing nonhealing femoral fractures in nude rats. Reverse transcriptase-polymerase chain reaction and immunohistochemical staining at the peri-fracture site demonstrated molecular and histological expression of human-specific markers for endothelial cells and osteoblasts at week 2. Functional bone healing assessed by biomechanical as well as radiological and histological examinations was significantly enhanced by CD34+ cell transplantation compared with the other groups. Our data suggest circulating human CD34+ cells have therapeutic potential to promote an environment conducive to neovascularization and osteogenesis in damaged skeletal tissue, allowing the complete healing of fractures.
Collapse
Affiliation(s)
- Tomoyuki Matsumoto
- Stem Cell Translational Research, Kobe Institute of Biomedical Research and Innovation/Riken Center for Developmental Biology, 2-2 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2213
|
Hayashi R, Yamato M, Sugiyama H, Sumide T, Yang J, Okano T, Tano Y, Nishida K. N-Cadherin is expressed by putative stem/progenitor cells and melanocytes in the human limbal epithelial stem cell niche. Stem Cells 2006; 25:289-96. [PMID: 17008425 DOI: 10.1634/stemcells.2006-0167] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Corneal epithelial stem cells are known to be localized to the basal layer of the limbal epithelium, providing a model system for epithelial stem cell biology; however, the mechanisms regarding the maintenance of these stem cells in their specialized niche remain poorly understood. N-cadherin is a member of the classic cadherin family and has previously been demonstrated to be expressed by hematopoietic stem cells. In the present study, we demonstrate that N-cadherin is expressed by putative stem/progenitor cells, as well as melanocytes, in the human limbal epithelial stem cell niche. In addition, we demonstrate that upon in vitro culture using 3T3 feeder layers, loss of N-cadherin expression occurs with cell proliferation. These results indicate that N-cadherin may be a critical cell-to-cell adhesion molecule between corneal epithelial stem/progenitor cells and their corresponding niche cells in the limbal epithelium.
Collapse
Affiliation(s)
- Ryuhei Hayashi
- Department of Ophthalmology, Tohoku University School of Medicine, 1-1 Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | | | | | | | | | | | | | | |
Collapse
|
2214
|
Scadden DT, Muse VV, Hasserjian RP. Case records of the Massachusetts General Hospital. Case 30-2006. A 41-year-old man with dyspnea, fever, and lymphadenopathy. N Engl J Med 2006; 355:1358-68. [PMID: 17005954 DOI: 10.1056/nejmcpc069021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- David T Scadden
- Department of Hematology-Oncology, Massachusetts General Hospital, USA
| | | | | |
Collapse
|
2215
|
Sneddon JB, Zhen HH, Montgomery K, van de Rijn M, Tward AD, West R, Gladstone H, Chang HY, Morganroth GS, Oro AE, Brown PO. Bone morphogenetic protein antagonist gremlin 1 is widely expressed by cancer-associated stromal cells and can promote tumor cell proliferation. Proc Natl Acad Sci U S A 2006; 103:14842-7. [PMID: 17003113 PMCID: PMC1578503 DOI: 10.1073/pnas.0606857103] [Citation(s) in RCA: 218] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although tissue microenvironments play critical roles in epithelial development and tumorigenesis, the factors mediating these effects are poorly understood. In this work, we used a genomic approach to identify factors produced by cells in the microenvironment of basal cell carcinoma (BCC) of the skin, one of the most common human cancers. The global gene expression programs of stromal cell cultures derived from human BCCs showed consistent, systematic differences from those derived from nontumor skin. The gene most consistently expressed at a higher level in BCC tumor stromal cells compared with those from nontumor skin was GREMLIN 1, which encodes a secreted antagonist of the bone morphogenetic protein (BMP) pathway. BMPs and their antagonists are known to play a crucial role in stem and progenitor cell biology as regulators of the balance between expansion and differentiation. Consistent with the hypothesis that BMP antagonists might have a similar role in cancer, we found GREMLIN 1 expression in the stroma of human BCC tumors but not in normal skin in vivo. Furthermore, BMP 2 and 4 are expressed by BCC cells. Ex vivo, BMP inhibits, and Gremlin 1 promotes, proliferation of cultured BCC cells. We further found that GREMLIN 1 is expressed by stromal cells in many carcinomas but not in the corresponding normal tissue counterparts that we examined. Our data suggest that BMP antagonists may be important constituents of tumor stroma, providing a favorable microenvironment for cancer cell survival and expansion in many cancers.
Collapse
Affiliation(s)
| | | | | | | | - Aaron D. Tward
- G. W. Hooper Foundation, University of California, San Francisco, CA 94143
| | | | | | | | | | | | - Patrick O. Brown
- Departments of *Biochemistry
- Howard Hughes Medical Institute, Stanford University Medical Center, Stanford, CA 94305; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
2216
|
Tada T, Widayati DT, Fukuta K. Morphological study of the transition of haematopoietic sites in the developing mouse during the peri-natal period. Anat Histol Embryol 2006; 35:235-40. [PMID: 16836587 DOI: 10.1111/j.1439-0264.2005.00671.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The transition of fetal haematopoietic sites in mice was examined histologically from 12.5-day embryos (E12.5) until 10 days of age (D10), and the expression of the adhesive molecules VCAM-1 and fibronectin was examined immunohistologically. Erythropoiesis occurred in the liver (E12.5-18.5), spleen (E18.5-D4) and bone marrow (D6-10), in that sequence. Even at D10, some erythropoiesis occurred in the liver, and more so in the spleen, although the active haematopoietic site was the bone marrow. Similarly, granulopoiesis of neutrophils occurred in the liver, spleen and bone marrow in turn. Granulopoiesis still occurred in the spleen at D10, but no neutrophils were found in the liver after D4. VCAM-1 appeared in the liver, spleen and bone marrow in parallel with active erythropoiesis and granulopoiesis. The co-expression of VCAM-1 and fibronectin was recognized in the endothelial cells of the sinus at the onset of haematopoiesis. This study showed that haematopoiesis in the liver, spleen and bone marrow overlapped peri-natally, although it shifted sequentially. VCAM-1 appears to be closely associated with erythropoiesis and granulopoiesis, and the co-expression of VCAM-1 and fibronectin plays a role in inducing haematopoietic stem cells to move to the tissues.
Collapse
Affiliation(s)
- T Tada
- Laboratory of Animal Morphology and Function, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa-ku, Japan
| | | | | |
Collapse
|
2217
|
Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med 2006; 12:1167-74. [PMID: 16998484 DOI: 10.1038/nm1483] [Citation(s) in RCA: 897] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Accepted: 08/15/2006] [Indexed: 12/11/2022]
Abstract
The long-term survival of patients with acute myeloid leukemia (AML) is dismally poor. A permanent cure of AML requires elimination of leukemic stem cells (LSCs), the only cell type capable of initiating and maintaining the leukemic clonal hierarchy. We report a therapeutic approach using an activating monoclonal antibody directed to the adhesion molecule CD44. In vivo administration of this antibody to nonobese diabetic-severe combined immune-deficient mice transplanted with human AML markedly reduced leukemic repopulation. Absence of leukemia in serially transplanted mice demonstrated that AML LSCs are directly targeted. Mechanisms underlying this eradication included interference with transport to stem cell-supportive microenvironmental niches and alteration of AML-LSC fate, identifying CD44 as a key regulator of AML LSCs. The finding that AML LSCs require interaction with a niche to maintain their stem cell properties provides a therapeutic strategy to eliminate quiescent AML LSCs and may be applicable to other types of cancer stem cells.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/biosynthesis
- Animals
- Antibodies, Monoclonal/chemistry
- Antigens, CD34/biosynthesis
- Cell Adhesion
- Cell Line, Tumor
- Cell Movement
- Humans
- Hyaluronan Receptors/biosynthesis
- Immunotherapy/methods
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Stem Cells/cytology
Collapse
Affiliation(s)
- Liqing Jin
- Division of Cell and Molecular Biology, University Health Network Suite 8-355, Toronto Medical Discovery Tower, 101 College Street, Toronto, M5G 1L7, Canada
| | | | | | | | | |
Collapse
|
2218
|
Nagamatsu G, Ohmura M, Mizukami T, Hamaguchi I, Hirabayashi S, Yoshida S, Hata Y, Suda T, Ohbo K. A CTX family cell adhesion molecule, JAM4, is expressed in stem cell and progenitor cell populations of both male germ cell and hematopoietic cell lineages. Mol Cell Biol 2006; 26:8498-506. [PMID: 16982697 PMCID: PMC1636774 DOI: 10.1128/mcb.01502-06] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Stem cells are maintained in an undifferentiated state by interacting with a microenvironment known as the "niche," which is comprised of various secreted and membrane proteins. Our goal was to identify niche molecules participating in stem cell-stem cell and/or stem cell-supporting cell interactions. Here, we isolated genes encoding secreted and membrane proteins from purified male germ stem cells using a signal sequence trap approach. Among the genes identified, we focused on the junctional adhesion molecule 4 (JAM4), an immunoglobulin type cell adhesion molecule. JAM4 protein was actually localized to the plasma membrane in male germ cells. JAM4 expression was downregulated as cells differentiated in both germ cell and hematopoietic cell lineages. To analyze function in vivo, we generated JAM4-deficient mice. Histological analysis of testes from homozygous nulls did not show obvious abnormalities, nor did liver and kidney tissues, both of which strongly express JAM4. The numbers of hematopoietic stem cells in bone marrow were indistinguishable between wild-type and mutant mice, as was male germ cell development. These results suggest that JAM4 is expressed in stem cells and progenitor cells but that other cell adhesion molecules may substitute for JAM4 function in JAM4-deficient mice both in male germ cell and hematopoietic lineages.
Collapse
Affiliation(s)
- Go Nagamatsu
- Sakaguchi Laboratory, Department of Cell Differentiation, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
2219
|
Chadwick N, Nostro MC, Baron M, Mottram R, Brady G, Buckle AM. Notch Signaling Induces Apoptosis in Primary Human CD34+Hematopoietic Progenitor Cells. Stem Cells 2006; 25:203-10. [PMID: 16973835 DOI: 10.1634/stemcells.2005-0303] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Notch signaling regulates diverse cell fate decisions during development and is reported to promote murine hematopoietic stem cell (HSC) self-renewal. The purpose of this study was to define the functional consequences of activating the Notch signaling pathway on self-renewal in human HSCs. Subsets of human umbilical cord blood CD34(+) cells were retrovirally transduced with the constitutively active human Notch 1 intracellular domain (N1ICD). N1ICD-transduced cells proliferated to a lesser extent in vitro than cells transduced with vector alone, and this was accompanied by a reduction in the percentage and absolute number of CD34(+) cell populations, including CD34(+)Thy(+)Lin(-) HSCs. Ectopic N1ICD expression inhibited cell cycle kinetics concurrent with an upregulation of p21 mRNA expression and induced apoptosis. Transduction of cells with HES-1, a known transcriptional target of Notch signaling and a mediator of Notch function, had no effect on HSC proliferation, indicating that the mechanism of the Notch-induced effect is HES-1-independent. The results of this study show that activation of the Notch signaling pathway has an inhibitory effect on the proliferation and survival of human hematopoietic CD34(+) cells populations. These findings have important implications for strategies aimed at promoting self-renewal of human HSCs.
Collapse
Affiliation(s)
- Nicholas Chadwick
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | |
Collapse
|
2220
|
Trowbridge JJ, Scott MP, Bhatia M. Hedgehog modulates cell cycle regulators in stem cells to control hematopoietic regeneration. Proc Natl Acad Sci U S A 2006; 103:14134-9. [PMID: 16968775 PMCID: PMC1599924 DOI: 10.1073/pnas.0604568103] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The signals that control the regenerative ability of hematopoietic stem cells (HSCs) in response to damage are unknown. Here, we demonstrate that downstream activation of the Hedgehog (Hh) signaling pathway induces cycling and expansion of primitive bone marrow hematopoietic cells under homeostatic conditions and during acute regeneration. However, this effect is at the expense of HSC function, because continued Hh activation during regeneration represses expression of specific cell cycle regulators, leading to HSC exhaustion. In vivo treatment with an inhibitor of the Hh pathway rescues these transcriptional and functional defects in HSCs. Our study establishes Hh signaling as a regulator of the HSC cell cycle machinery that balances hematopoietic homeostasis and regeneration in vivo.
Collapse
Affiliation(s)
- Jennifer J. Trowbridge
- *Department of Microbiology and Immunology, University of Western Ontario, 1151 Richmond Street, Suite 2, London, ON, Canada N6A 5B8
| | - Matthew P. Scott
- Departments of Biology, Genetics, and Bioengineering, Stanford University School of Medicine, Howard Hughes Medical Institute, Stanford, CA 94305-5439
| | - Mickie Bhatia
- *Department of Microbiology and Immunology, University of Western Ontario, 1151 Richmond Street, Suite 2, London, ON, Canada N6A 5B8
- McMaster Stem Cell and Cancer Research Institute, The Michael G. DeGroote School of Medicine, Hamilton, ON, Canada L9G 4L6; and
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, ON, Canada L8N 3Z5
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
2221
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells are responsible for generating all types of blood cells. As such they are under a high degree of regulation, both internal and external. With the identification of the hematopoietic stem cell niche, there has been increased investigation into extrinsic regulation of hematopoietic stem cells with emphasis on developmental signaling pathways. The purpose of this review is to discuss recent advances and findings in how these different pathways interact to achieve a balanced control of these stem cells. RECENT FINDINGS Studies indicating the importance of pathways such as Wnt, Notch, bone morphogenic protein, Sonic hedgehog and fibroblast growth factor in controlling the fate of hematopoietic stem cells are the most significant recent findings. These pathways have been implicated to affect various aspects of hematopoietic stem cells, including self-renewal, proliferation and lineage determination. Equally important are studies showing, by inactivation of various pathway components, the complexity of signal integration at the stem cell level in vivo. Additionally, some recent reports have provided evidence for direct interaction or cross-talk between different signaling pathways in this regulation. SUMMARY We review highlights of the recent advances made toward resolving the mechanisms of external regulation of hematopoietic stem cells. Understanding the interaction of different signaling pathways in the context of the hematopoietic stem cell niche is essential for increasing their therapeutic potential.
Collapse
Affiliation(s)
- Jason Ross
- Department of Pathology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | |
Collapse
|
2222
|
Abstract
The mucosal lining (endometrium) of the human uterus undergoes cyclical processes of regeneration, differentiation and shedding as part of the menstrual cycle. Endometrial regeneration also follows parturition, almost complete resection and in post-menopausal women taking estrogen replacement therapy. In non-menstruating species, there are cycles of endometrial growth and apoptosis rather than physical shedding. The concept that endometrial stem/progenitor cells are responsible for the remarkable regenerative capacity of endometrium was proposed many years ago. However, attempts to isolate, characterize and locate endometrial stem cells have only been undertaken in the last few years as experimental approaches to identify adult stem/progenitor cells in other tissues have been developed. Adult stem cells are defined by their functional properties rather than by marker expression. Evidence for the existence of adult stem/progenitor cells in human and mouse endometrium is now emerging because functional stem cell assays are being applied to uterine cells and tissues. These fundamental studies on endometrial stem/progenitor cells will provide new insights into the pathophysiology of various gynaecological disorders associated with abnormal endometrial proliferation, including endometrial cancer, endometrial hyperplasia, endometriosis and adenomyosis.
Collapse
Affiliation(s)
- C E Gargett
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
2223
|
Suda T, Arai F, Hirao A. Hematopoietic stem cells and their niche. Trends Immunol 2006; 26:426-33. [PMID: 15979407 DOI: 10.1016/j.it.2005.06.006] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Revised: 04/22/2005] [Accepted: 06/14/2005] [Indexed: 11/19/2022]
Abstract
Recent evidence indicates that osteoblasts are crucial components of the particular microenvironments, or niches, for hematopoietic stem cells (HSCs) in adult bone marrow (BM). Stem cells persist in an immature state within the BM. The quiescence of HSCs is controlled dynamically by the signaling of receptors-ligands and cell-adhesion molecules. In this review, the characteristics of HSCs in the niche are discussed. The understanding of the relationship between normal and cancer stem cells and their niches should lead to the development of new strategies directed toward regeneration medicine and cancer therapeutics.
Collapse
Affiliation(s)
- Toshio Suda
- Department of Cell Differentiation, The Sakaguchi Laboratory of Developmental Biology, School of Medicine, Keio University, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | | | | |
Collapse
|
2224
|
Oguro H, Iwama A, Morita Y, Kamijo T, van Lohuizen M, Nakauchi H. Differential impact of Ink4a and Arf on hematopoietic stem cells and their bone marrow microenvironment in Bmi1-deficient mice. ACTA ACUST UNITED AC 2006; 203:2247-53. [PMID: 16954369 PMCID: PMC2118102 DOI: 10.1084/jem.20052477] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The polycomb group (PcG) protein Bmi1 plays an essential role in the self-renewal of hematopoietic and neural stem cells. Derepression of the Ink4a/Arf gene locus has been largely attributed to Bmi1-deficient phenotypes in the nervous system. However, its role in hematopoietic stem cell (HSC) self-renewal remained undetermined. In this study, we show that derepressed p16Ink4a and p19Arf in Bmi1-deficient mice were tightly associated with a loss of self-renewing HSCs. The deletion of both Ink4a and Arf genes substantially restored the self-renewal capacity of Bmi1−/− HSCs. Thus, Bmi1 regulates HSCs by acting as a critical failsafe against the p16Ink4a- and p19Arf-dependent premature loss of HSCs. We further identified a novel role for Bmi1 in the organization of a functional bone marrow (BM) microenvironment. The BM microenvironment in Bmi1−/− mice appeared severely defective in supporting hematopoiesis. The deletion of both Ink4a and Arf genes did not considerably restore the impaired BM microenvironment, leading to a sustained postnatal HSC depletion in Bmi1−/−Ink4a-Arf−/− mice. Our findings unveil a differential role of derepressed Ink4a and Arf on HSCs and their BM microenvironment in Bmi1-deficient mice. Collectively, Bmi1 regulates self-renewing HSCs in both cell-autonomous and nonautonomous manners.
Collapse
Affiliation(s)
- Hideyuki Oguro
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, Institute of Medical Sciences, University of Tokyo, Tokyo 108-8679, Japan
| | | | | | | | | | | |
Collapse
|
2225
|
Weber JM, Forsythe SR, Christianson CA, Frisch BJ, Gigliotti BJ, Jordan CT, Milner LA, Guzman ML, Calvi LM. Parathyroid hormone stimulates expression of the Notch ligand Jagged1 in osteoblastic cells. Bone 2006; 39:485-93. [PMID: 16647886 DOI: 10.1016/j.bone.2006.03.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 03/06/2006] [Indexed: 12/11/2022]
Abstract
We previously demonstrated that activation of the Parathyroid Hormone Receptor (PTH1R) in osteoblastic cells increases the Notch ligand Jagged1 and expands hematopoietic stem cells (HSC) through Notch signaling. However, regulation of Jagged1 by PTH in osteoblasts is poorly understood. The present study demonstrates that PTH treatment increases Jagged1 levels in a subpopulation of osteoblastic cells in vivo and in UMR106 osteoblastic cells in vitro. Since PTH(1-34) activates both Adenylate Cyclase/Protein Kinase A (AC/PKA) and Protein Kinase C (PKC) downstream of the PTH1R in osteoblastic cells, we independently determined the effect of either pathway on Jagged1. Activation of AC with Forskolin or PKA with PTH(1-31) or cell-permeable cAMP analogues increased osteoblastic Jagged1. This PTH-dependent Jagged1 increase was blocked by H89 and PKI, specific PKA inhibitors. In contrast, PKC activation with phorbol ester (PMA) or PTH(13-34) did not stimulate Jagged1 expression, and PTH-dependent Jagged1 stimulation was not blocked by Gö6976, a conventional PKC inhibitor. Therefore, PTH treatment stimulates osteoblastic Jagged1 mainly through the AC/PKA signaling pathway downstream of the PTH1R. Since Jagged1/Notch signaling has been implicated not only in stromal-HSC interactions but also in osteoblastic differentiation, Jagged1 may play a critical role in mediating the PTH-dependent expansion of HSC, as well as the anabolic effect of PTH in bone.
Collapse
Affiliation(s)
- Jonathan M Weber
- Endocrine Division, Department of Medicine, University of Rochester School of Medicine, 601 Elmwood Avenue Box 693 Rochester, NY 14642, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
2226
|
Bryder D, Rossi DJ, Weissman IL. Hematopoietic stem cells: the paradigmatic tissue-specific stem cell. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:338-46. [PMID: 16877336 PMCID: PMC1698791 DOI: 10.2353/ajpath.2006.060312] [Citation(s) in RCA: 433] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The recent prospective isolation of a wide variety of somatically derived stem cells has affirmed the notion that homeostatic maintenance of most tissues and organs is mediated by tissue-specific stem and progenitor cells and fueled enthusiasm for the use of such cells in strategies aimed at repairing or replacing damaged, diseased, or genetically deficient tissues and organs. Hematopoietic stem cells (HSCs) are arguably the most well-characterized tissue-specific stem cell, with decades of basic research and clinical application providing not only a profound understanding of the principles of stem cell biology, but also of its potential pitfalls. It is our belief that emerging stem cell fields can benefit greatly from an understanding of the lessons learned from the study of HSCs. In this review we discuss some general concepts regarding stem cell biology learned from the study of HSCs with a highlight on recent work pertaining to emerging topics of interest for stem cell biology.
Collapse
Affiliation(s)
- David Bryder
- Stanford University School of Medicine, B257 Beckman Center, Stanford, CA 94305-5323, USA
| | | | | |
Collapse
|
2227
|
Dar A, Kollet O, Lapidot T. Mutual, reciprocal SDF-1/CXCR4 interactions between hematopoietic and bone marrow stromal cells regulate human stem cell migration and development in NOD/SCID chimeric mice. Exp Hematol 2006; 34:967-75. [PMID: 16863903 DOI: 10.1016/j.exphem.2006.04.002] [Citation(s) in RCA: 262] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2006] [Indexed: 12/16/2022]
Abstract
The chemokine SDF-1 (CXCL12) and its receptor CXCR4 are involved in regulation of migration, survival, and development of multiple cell types, including human hematopoietic CD34+/CD38-/low and stromal STRO-1+ stem cells. During steady-state homeostasis, CXCR4 is expressed by hematopoietic cells and also by stromal cells, which are the main source for SDF-1 in the bone marrow (BM). Stress-induced modulations in SDF-1 and CXCR4 levels participate in recruitment of immature and maturing leukocytes from the BM reservoir to damaged organs as part of host defense and repair mechanism. In addition, trafficking of SDF-1 is mediated by CXCR4, expressed by endothelial and various stromal cell types in the BM, spleen, and other organs, but not by hematopoietic cells. Transcytosis of functional SDF-1 to the BM takes place also in the stem cell-rich endothelium and endosteum regions, regulating hematopoietic and stromal interactions in the stem cell niche. Dynamic levels of SDF-1 and CXCR4 expression induce proliferation of hematopoietic and mesenchymal progenitors, recruitment of bone-resorbing osteoclasts, osteoblasts, neutrophils, and other myeloid cells, leading to leukocyte mobilization. These studies will be reviewed together with the mechanisms that regulate SDF-1 and CXCR4 physiologic function, inactivation, presentation, and availability. Moreover, the role and the dynamic modulations of this ligand and its receptor in alarm and pathologic conditions will be discussed as well.
Collapse
Affiliation(s)
- Ayelet Dar
- Department of Immunology, Weizmann Institute, Rehovot, Israel
| | | | | |
Collapse
|
2228
|
Blank U, Karlsson G, Moody JL, Utsugisawa T, Magnusson M, Singbrant S, Larsson J, Karlsson S. Smad7 promotes self-renewal of hematopoietic stem cells. Blood 2006; 108:4246-54. [PMID: 16917010 DOI: 10.1182/blood-2006-02-005611] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Smad-signaling pathway downstream of the transforming growth factor-beta superfamily of ligands is an evolutionarily conserved signaling circuitry with critical functions in a wide variety of biologic processes. To investigate the role of this pathway in the regulation of hematopoietic stem cells (HSCs), we have blocked Smad signaling by retroviral gene transfer of the inhibitory Smad7 to murine HSCs. We report here that the self-renewal capacity of HSCs is promoted in vivo upon blocking of the entire Smad pathway, as shown by both primary and secondary bone marrow (BM) transplantations. Importantly, HSCs overexpressing Smad7 have an unperturbed differentiation capacity as evidenced by normal contribution to both lymphoid and myeloid cell lineages, suggesting that the Smad pathway regulates self-renewal independently of differentiation. Moreover, phosphorylation of Smads was inhibited in response to ligand stimulation in BM cells, thus verifying impairment of the Smad-signaling cascade in Smad7-overexpressing cells. Taken together, these data reveal an important and previously unappreciated role for the Smad-signaling pathway in the regulation of self-renewal of HSCs in vivo.
Collapse
Affiliation(s)
- Ulrika Blank
- Molecular Medicine and Gene Therapy, Institute of Laboratory Medicine, Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
2229
|
Akala OO, Clarke MF. Hematopoietic stem cell self-renewal. Curr Opin Genet Dev 2006; 16:496-501. [PMID: 16919448 DOI: 10.1016/j.gde.2006.08.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Accepted: 08/04/2006] [Indexed: 01/03/2023]
Abstract
Recent studies have begun to elucidate the mechanisms controlling hematopoietic stem cell (HSC) self-renewal. Self-renewal requires the integration of survival signals and proliferation controls with the maintenance of an undifferentiated state. This demands a complex crosstalk between extrinsic signals from the microenvironment and the cell-intrinsic regulators of self-renewal. The Polycomb protein Bmi1 is absolutely required for the maintenance of both adult HSCs and neural stem cells. Evidence from studies in murine and human embryonic stem cells indicates that Polycomb group proteins play a dynamic role in concert with master transcriptional regulators in actively maintaining an undifferentiated state, suggesting that this mechanism applies to multiple types of stem cell. Recently, various new players that regulate HSC maintenance (e.g. Mcl1, Tel/Etv6, Gfi1, Pten and Stat5) have been identified. In order to better understand HSC self-renewal, we need to understand how these pathways are coordinated.
Collapse
Affiliation(s)
- Omobolaji O Akala
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, 1050 Arastradero Road, Palo Alto, CA 94304-1334, USA
| | | |
Collapse
|
2230
|
Illa-Bochaca I, Montuenga LM. The regenerative nidi of the locust midgut as a model to study epithelial cell differentiation from stem cells. ACTA ACUST UNITED AC 2006; 209:2215-23. [PMID: 16709922 DOI: 10.1242/jeb.02249] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A better knowledge of the regulatory mechanisms involved in stem cell proliferation and/or differentiation could reveal new methods for the treatment of some diseases. Most previous studies in the field of stem cell biology have been carried out on cultured isolated cells. In the case of adult tissue stem cells, mesenchymal bone marrow derived cells have been most widely studied, while the undifferentiated stem cells present in the epithelial tissues are less known. In order to advance further our understanding of epithelial tissue stem cells, new in vivo models are required. The present study focuses on the dynamics of a new and simple model of intestinal epithelial regeneration found in the midgut of the migratory locust, Locusta migratoria (Linnaeus 1758). The locust midgut consists of three cell types: columnar cells, endocrine cells and undifferentiated regenerative clustered cells. The undifferentiated epithelial midgut cells give rise to two other cell types and are located in a nest of regenerative cells known as regenerative niche. We have performed single and continuous bromodeoxyuridine (BrdU) administration experiments to study regeneration niches and their cellular dynamics. Immunocytochemistry and immunofluorescence techniques were used to detect the incorporation of BrdU into regenerative niches and the presence of FMRFamide-like immunoreactivity, as a marker for endocrine cell differentiation. Some isolated label retaining cells (LRC) were observed at the niche base 10-15 days after the final BrdU administration. We propose that these cells are the stem cells of this epithelial tissue. We also calculated the length of the cell cycle phases for a subpopulation of transit amplifying cells within the regenerative niche: G1, 2.5+/-0.5 h; S, 5.5+/-0.5 h; G2, 0.75+/-0.25 h; M, 2.5+/-0.5 h. These amplifying cells will give rise to the columnar epithelial non-endocrine lineage. The differentiation of an endocrine cell from a niche stem cell occurs less frequently and thus leads to a lower proportion of endocrine cells as compared with epithelial columnar digestive cells (up to three endocrine cells per niche). Endocrine cell commitment seems to occur very early in the differentiation process within the niche, as double-labelled BrdU and FMRF endocrine cells have never been found. The only exception is the endocrine cells located in the ampullar region of the midgut, some of which show double immunostaining after long-term chronic BrdU injection. In summary, we have characterized a new and simple animal model of epithelial stem cell regeneration that may be useful for understanding the complex biological process that drives tissue renewal from undifferentiated and uncommitted progenitor cells.
Collapse
Affiliation(s)
- Irineu Illa-Bochaca
- Department of Histology and Pathology, Schools of Sciences and Medicine, University of Navarra, E-31080 Pamplona, Spain
| | | |
Collapse
|
2231
|
Papaccio G, Graziano A, d'Aquino R, Graziano MF, Pirozzi G, Menditti D, De Rosa A, Carinci F, Laino G. Long-term cryopreservation of dental pulp stem cells (SBP-DPSCs) and their differentiated osteoblasts: a cell source for tissue repair. J Cell Physiol 2006; 208:319-25. [PMID: 16622855 DOI: 10.1002/jcp.20667] [Citation(s) in RCA: 174] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
It is not known whether cells derived from stem cells retain their differentiation and morpho-functional properties after long-term cryopreservation. This information is of importance to evaluate their potential for long-term storage with a view to subsequent use in therapy. Here, we describe the morpho-functional properties of dental pulp stem cells (SBP-DPSCs), and of their differentiated osteoblasts, recovered after long-term cryopreservation. After storage for 2 years, we found that stem cells are still capable of differentiation, and that their differentiated cytotypes proliferate and produce woven bone tissue. In addition, cells still express all their respective surface antigens, confirming cellular integrity. In particular, SBP-DPSCs differentiated into pre-osteoblasts, showing diffuse positivity for ALP, BAP, RUNX-2, and calcein. Recovered osteoblasts expressed bone-specific markers and were easily recognizable ultrastructurally, with no alterations observed at this level. In addition, after in vivo transplantation, woven bone converted into a 3D lamellar bone type. Therefore, dental pulp stem cells and their osteoblast-derived cells can be long-term cryopreserved and may prove to be attractive for clinical applications.
Collapse
Affiliation(s)
- Gianpaolo Papaccio
- Dipartimento di Medicina Sperimentale, Sezione di Istologia ed Embriologia, Secondo Ateneo di Napoli, Napoli, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
2232
|
Abstract
The Notch signaling pathway is among the most commonly used communication channels in animal cells. Recent studies have demonstrated that this pathway is indispensable for cells in various stages of maturation, including terminal differentiation. One main focus in mammalian studies is the role of Notch in embryonic and postembryonic stem cell systems. In this review, the roles of Notch signaling in various mammalian stem and early progenitor cells are summarized.
Collapse
Affiliation(s)
- Shigeru Chiba
- Department of Cell Therapy and Transplantation Medicine, University of Tokyo, 7-3-1 Hongo, Tokyo 113-8655, Japan.
| |
Collapse
|
2233
|
Winkler IG, Lévesque JP. Mechanisms of hematopoietic stem cell mobilization: When innate immunity assails the cells that make blood and bone. Exp Hematol 2006; 34:996-1009. [PMID: 16863906 DOI: 10.1016/j.exphem.2006.04.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Indexed: 01/13/2023]
Abstract
Mobilization is now used worldwide to collect large numbers of hematopoietic stem and progenitor cells (HSPCs) for transplantation. Although the first mobilizing agents were discovered largely by accident, discovery of more efficient mobilizing agents will require a better understanding of the molecular mechanisms responsible. During the past 5 years, a number of mechanisms have been identified, shedding new light on the dynamics of the hematopoietic system in vivo and on the intricate relationship between hematopoiesis, innate immunity, and bone. After briefly reviewing the mechanisms by which circulating HSPCs home into the bone marrow and what keeps them there, the current knowledge of mechanisms responsible for HSPC mobilization in response to hematopoietic growth factors such as granulocyte colony-stimulating factor, chemotherapy, chemokines, and polyanions will be discussed together with current strategies developed to further increase HSPC mobilization.
Collapse
Affiliation(s)
- Ingrid G Winkler
- Haematopoietic Stem Cell Laboratory, Cancer Biotherapies Program, Mater Medical Research Institute, University of Queensland, South Brisbane, Queensland, Australia
| | | |
Collapse
|
2234
|
Deregowski V, Gazzerro E, Priest L, Rydziel S, Canalis E. Role of the RAM domain and ankyrin repeats on notch signaling and activity in cells of osteoblastic lineage. J Bone Miner Res 2006; 21:1317-26. [PMID: 16869730 DOI: 10.1359/jbmr.060505] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED Notch proteins belong to a family of single pass transmembrane receptors that are activated after interactions with the membrane-bound ligands Delta and Jagged/Serrate. We determined the pathways responsible for the inhibitory effects of Notch on osteoblastogenesis and the contributions of the RAM domain and ankyrin repeats to this process in cells of the osteoblastic lineage. INTRODUCTION Notch receptors play a role in osteoblast differentiation. Activation of Notch results in its cleavage and the release of its intracellular domain (NICD), which interacts with the CBF1/RBP-Jkappa, Suppressor of Hairless, Lag-1 (CSL) family of transcription factors. The interaction is presumably mediated by the RBP-Jkappa-associated module (RAM) of NICD, although the role of the ankyrin repeats is uncertain. MATERIALS AND METHODS To determine the contributions of the RAM domain and ankyrin repeats to the inhibitory effects of Notch on osteoblastogenesis, ST-2 and MC3T3-E1 cells were transfected or transduced with vectors expressing NICD, RAM (NICD DeltaRAM), and ankyrin (NICD DeltaANK) deletion mutants. RESULTS Notch increased the transactivation of transiently transfected 12xCSL-Luc constructs, containing 12 repeats of an RBP-Jkappa/CSL binding site, and of the hairy and E (spl) (HES)-1 promoter. Deletion of the ankyrin repeats resulted in the loss of 12xCSL-Luc and HES-1 promoter transactivation, whereas deletion of the RAM domain caused a partial loss of 12xCSL-Luc and sustained HES-1 promoter transactivation. NICD overexpression inhibited osteocalcin mRNA levels and alkaline phosphatase activity in ST-2 cells, and deletion of the ankyrin repeats, and to a lesser extent of the RAM domain, resulted in loss of the NICD inhibitory effect. NICD inhibited Wnt signaling and deletion of ankyrin repeats or the RAM domain restored Wnt signaling activity. CONCLUSIONS The RAM domain and ankyrin repeats are required for Notch signaling and activity, and the CSL pathway is central to the inhibitory effect of Notch on osteoblastogenesis.
Collapse
Affiliation(s)
- Valerie Deregowski
- Department of Research Saint Francis Hospital and Medical Center, Hartford, Connecticut 06105-1299, USA
| | | | | | | | | |
Collapse
|
2235
|
Mathew S, Davies M, Lund R, Saab G, Hruska KA. Function and effect of bone morphogenetic protein-7 in kidney bone and the bone-vascular links in chronic kidney disease. Eur J Clin Invest 2006; 36 Suppl 2:43-50. [PMID: 16884397 DOI: 10.1111/j.1365-2362.2006.01663.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In two independent and separate studies, we have shown that renal injury and chronic kidney disease (CKD) directly inhibit skeletal anabolism, and that stimulation of bone formation decreased the serum phosphate. In the first study, the serum Ca PO(4), parathyroid hormone (PTH), and calcitriol were maintained normal after renal ablation in mice, and even mild renal injury equivalent to stage 3 CKD decreased bone formation rates. More recently, these observations were rediscovered in low-density lipoprotein receptor null (LDLR-/-) mice fed high-fat/cholesterol diets, a model of the metabolic syndrome (hypertension, obesity, dyslipidemia and insulin resistance). We demonstrated that these mice have vascular calcification (VC) of both the intimal atherosclerotic type and medial calcification. We have also shown that VC is made worse by CKD and ameliorated by bone morphogenetic protein-7 (BMP-7). The finding that high-fat fed LDLR-/- animals with CKD had hyperphosphatemia which was prevented in BMP-7-treated animals lead us to examine the skeletons of these mice. It was found that significant reductions in bone formation rates were associated with high-fat feeding, and superimposing CKD resulted in the adynamic bone disorder (ABD), while VC was made worse. The effect of CKD to decrease skeletal anabolism (decreased bone formation rates and reduced number of bone modelling units) occurred despite secondary hyperparathyroidism. The BMP-7 treatment corrected the ABD and hyperphosphatemia, owing to BMP-7-driven stimulation of skeletal phosphate deposition reducing plasma phosphate and thereby removing a major stimulus to VC. A pathological link between abnormal bone mineralization and VC through the serum phosphorus was demonstrated by the partial effectiveness of directly reducing the serum phosphate by a phosphate binder that had no skeletal action. Thus, in the metabolic syndrome with CKD, a reduction in bone forming potential of osteogenic cells leads to the ABD producing hyperphosphatemia and VC, processes ameliorated by BMP-7, in part through increased bone formation and skeletal deposition of phosphate and in part through direct actions on vascular smooth muscle cells. We have demonstrated that the processes leading to vascular calcification begin with even mild levels of renal injury affecting the skeleton before demonstrable hyperphosphatemia and that they are preventable and treatable. Therefore, early intervention in the skeletal disorder associated with CKD is warranted and may affect mortality of the disease.
Collapse
Affiliation(s)
- S Mathew
- Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
2236
|
Weng AP, Millholland JM, Yashiro-Ohtani Y, Arcangeli ML, Lau A, Wai C, Del Bianco C, Rodriguez CG, Sai H, Tobias J, Li Y, Wolfe MS, Shachaf C, Felsher D, Blacklow SC, Pear WS, Aster JC. c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma. Genes Dev 2006; 20:2096-109. [PMID: 16847353 PMCID: PMC1536060 DOI: 10.1101/gad.1450406] [Citation(s) in RCA: 684] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Accepted: 06/06/2006] [Indexed: 02/06/2023]
Abstract
Human acute T-cell lymphoblastic leukemias and lymphomas (T-ALL) are commonly associated with gain-of-function mutations in Notch1 that contribute to T-ALL induction and maintenance. Starting from an expression-profiling screen, we identified c-myc as a direct target of Notch1 in Notch-dependent T-ALL cell lines, in which Notch accounts for the majority of c-myc expression. In functional assays, inhibitors of c-myc interfere with the progrowth effects of activated Notch1, and enforced expression of c-myc rescues multiple Notch1-dependent T-ALL cell lines from Notch withdrawal. The existence of a Notch1-c-myc signaling axis was bolstered further by experiments using c-myc-dependent murine T-ALL cells, which are rescued from withdrawal of c-myc by retroviral transduction of activated Notch1. This Notch1-mediated rescue is associated with the up-regulation of endogenous murine c-myc and its downstream transcriptional targets, and the acquisition of sensitivity to Notch pathway inhibitors. Additionally, we show that primary murine thymocytes at the DN3 stage of development depend on ligand-induced Notch signaling to maintain c-myc expression. Together, these data implicate c-myc as a developmentally regulated direct downstream target of Notch1 that contributes to the growth of T-ALL cells.
Collapse
Affiliation(s)
- Andrew P Weng
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2237
|
Wang Y, Probin V, Zhou D. Cancer therapy-induced residual bone marrow injury-Mechanisms of induction and implication for therapy. CURRENT CANCER THERAPY REVIEWS 2006; 2:271-279. [PMID: 19936034 PMCID: PMC2779029 DOI: 10.2174/157339406777934717] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone marrow (BM) suppression is the important dose-limiting side effect of chemotherapy and radiotherapy for cancer. Although acute myelosuppression is an immediate concern for patients undergoing cancer therapy, its management has been improved significantly in recent years by the use of various hematopoietic growth factors. However, many patients receiving chemotherapy and/or ionizing radiation (IR) also develop residual (or long-term) BM injury (a sustained decrease in HSC reserves due to an impairment in HSC self-renewal) after the recovery from acute myelosuppression. Unlike acute myelosuppression, residual BM injury is latent and long lasting and shows little tendency for recovery. Following additional hematopoietic stress such as subsequent cycles of consolidation cancer treatment or autologous BM transplantation, residual BM injury can deteriorate to become a hypoplastic or myelodysplastic syndrome. This article review some of the new developments in elucidating the cellular and molecular mechanisms whereby chemotherapy and radiotherapy cause residual BM injury. Particularly, we discuss the role of induction of hematopoietic stem cell (HSC) senescence via the p53-p21(Cip1/Waf1) and/or p16(Ink4a)-RB pathways in the induction of the injury and the therapeutic potential of molecularly targeting these pathways for amelioration of chemotherapy- and radiotherapy-induced long-term BM toxicity.
Collapse
Affiliation(s)
- Yong Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Virginia Probin
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Daohong Zhou
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
2238
|
Abstract
Stem-cell populations are established in 'niches'--specific anatomic locations that regulate how they participate in tissue generation, maintenance and repair. The niche saves stem cells from depletion, while protecting the host from over-exuberant stem-cell proliferation. It constitutes a basic unit of tissue physiology, integrating signals that mediate the balanced response of stem cells to the needs of organisms. Yet the niche may also induce pathologies by imposing aberrant function on stem cells or other targets. The interplay between stem cells and their niche creates the dynamic system necessary for sustaining tissues, and for the ultimate design of stem-cell therapeutics.
Collapse
Affiliation(s)
- David T Scadden
- Massachusetts General Hospital Center for Regenerative Medicine, Harvard Stem Cell Institute, 185 Cambridge Street, CPZN - 4265A, Boston, Massachusetts 02114, USA.
| |
Collapse
|
2239
|
Nagayoshi K, Ohkawa H, Yorozu K, Higuchi M, Higashi S, Kubota N, Fukui H, Imai N, Gojo S, Hata JI, Kobayashi Y, Umezawa A. Increased mobilization of c-kit+ Sca-1+ Lin- (KSL) cells and colony-forming units in spleen (CFU-S) following de novo formation of a stem cell niche depends on dynamic, but not stable, membranous ossification. J Cell Physiol 2006; 208:188-94. [PMID: 16575918 DOI: 10.1002/jcp.20652] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Stem cells are thought to inhabit in a unique microenvironment, known as "niche," in which they undergo asymmetric cell divisions that results in reproducing both stem cells and progenies to maintain various tissues throughout life. The cells of osteoblastic lineage have been identified as a key participant in regulating the number of hematopoietic stem cells (HSCs). HSCs receive their regulatory messages from the microenvironment in the bone marrow. This would account for a reason why the localization of hematopoiesis is usually restricted in the bone marrow. To clarify the above possibility we employed a cell implantation-based strategy with a unique osteoblast cell line (KUSA-A1) derived from a C3H/He mouse. The implantation of KUSA-A 1 cells resulted in the generation of ectopic bones in the subcutaneous tissues of the athymic BALB/c nu/nu mice. Subsequently the mice obtained a greater amount of the bone marrow than normal mice, and they showed an increased number of HSCs. These results indicate that the newly generated osteoblasts-derived ectopic bones are responsible for the increase in the number of the HSC population. Furthermore, the increased number of HSCs directly correlates with both the magnitude of dynamic osteogenic process and the size of the newly generated bone or "niche."
Collapse
Affiliation(s)
- Kazunari Nagayoshi
- Department of Reproductive Biology and Pathology, National Research Institute for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2240
|
Chute JP, Muramoto GG, Whitesides J, Colvin M, Safi R, Chao NJ, McDonnell DP. Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proc Natl Acad Sci U S A 2006; 103:11707-12. [PMID: 16857736 PMCID: PMC1544234 DOI: 10.1073/pnas.0603806103] [Citation(s) in RCA: 344] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme that is expressed in the liver and is required for the conversion of retinol (vitamin A) to retinoic acids. ALDH is also highly enriched in hematopoietic stem cells (HSCs) and is considered a selectable marker of human HSCs, although its contribution to stem cell fate remains unknown. In this study, we demonstrate that ALDH is a key regulator of HSC differentiation. Inhibition of ALDH with diethylaminobenzaldehyde (DEAB) delayed the differentiation of human HSCs that otherwise occurred in response to cytokines. Moreover, short-term culture with DEAB caused a 3.4-fold expansion in the most primitive assayable human cells, the nonobese diabetic/severe combined immunodeficiency mouse repopulating cells, compared with day 0 CD34(+)CD38(-)lin(-) cells. The effects of DEAB on HSC differentiation could be reversed by the coadministration of the retinoic acid receptor agonist, all-trans-retinoic acid, suggesting that the ability of ALDH to generate retinoic acids is important in determining HSC fate. DEAB treatment also caused a decrease in retinoic acid receptor-mediated signaling within human HSCs, suggesting directly that inhibition of ALDH promotes HSC self-renewal via reduction of retinoic acid activity. Modulation of ALDH activity and retinoid signaling is a previously unrecognized and effective strategy to amplify human HSCs.
Collapse
Affiliation(s)
- John P Chute
- Division of Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | | | |
Collapse
|
2241
|
Yamazaki S, Iwama A, Takayanagi SI, Morita Y, Eto K, Ema H, Nakauchi H. Cytokine signals modulated via lipid rafts mimic niche signals and induce hibernation in hematopoietic stem cells. EMBO J 2006; 25:3515-23. [PMID: 16858398 PMCID: PMC1538571 DOI: 10.1038/sj.emboj.7601236] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Accepted: 06/21/2006] [Indexed: 11/09/2022] Open
Abstract
Hematopoietic stem cells (HSCs) reside in the bone marrow (BM) niche in a noncycling state and enter the cell cycle at long intervals. However, little is known about inter- and intracellular signaling mechanisms underlying this unique property of HSCs. Here, we show that lipid raft clustering is a key event in the regulation of HSC dormancy. Freshly isolated HSCs from the BM niche lack lipid raft clustering, exhibit repression of the AKT-FOXO signaling pathway, and express abundant p57(Kip2) cyclin-dependent kinase inhibitor. Lipid raft clustering induced by cytokines is essential for HSC re-entry into the cell cycle. Conversely, inhibition of lipid raft clustering caused sustained nuclear accumulation of FOXO transcription factors and induced HSC hibernation ex vivo. These data establish a critical role for lipid rafts in regulating the cell cycle, the survival, and the entry into apoptosis of HSCs and uncover a striking similarity in HSC hibernation and Caenorhabditis elegans dauer formation.
Collapse
Affiliation(s)
- Satoshi Yamazaki
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- ReproCELL Inc., Tokyo, Japan
| | - Atsushi Iwama
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shin-ichiro Takayanagi
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yohei Morita
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- ReproCELL Inc., Tokyo, Japan
| | - Koji Eto
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hideo Ema
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Laboratory of Stem Cell Therapy, Center for Experimental Medicine, The Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan. Tel.: +81 3 5449 5330; Fax: +81 3 5449 5451; E-mail:
| |
Collapse
|
2242
|
Suzuki T, Yokoyama Y, Kumano K, Takanashi M, Kozuma S, Takato T, Nakahata T, Nishikawa M, Sakano S, Kurokawa M, Ogawa S, Chiba S. Highly efficient ex vivo expansion of human hematopoietic stem cells using Delta1-Fc chimeric protein. Stem Cells 2006; 24:2456-65. [PMID: 16857897 DOI: 10.1634/stemcells.2006-0258] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Ex vivo expansion of hematopoietic stem cells (HSCs) has been explored in the fields of stem cell biology, gene therapy, and clinical transplantation. Here, we demonstrate efficient ex vivo expansion of HSCs measured by long-term severe combined immunodeficient (SCID) repopulating cells (SRCs) from human cord blood CD133-sorted cells using a soluble form of Delta1. After a 3-week culture on immobilized Delta1 supplemented with stem cell factor, thrombopoietin, Flt-3 ligand, interleukin (IL)-3, and IL-6/soluble IL-6 receptor chimeric protein (FP6) in a serum- and stromal cell-free condition, we achieved approximately sixfold expansion of SRCs when evaluated by limiting dilution/transplantation assays. The maintenance of full multipotency and self-renewal capacity during culture was confirmed by transplantation to nonobese diabetic/SCID/gammac(null) mice, which showed myeloid, B, T, and natural killer cells as well as CD133(+)CD34(+) cells, and hematopoietic reconstitution in the secondary recipients. Interestingly, the CD133-sorted cells contained approximately 4.5 times more SRCs than the CD34-sorted cells. The present study provides a promising method to expand HSCs and encourages future trials on clinical transplantation.
Collapse
MESH Headings
- AC133 Antigen
- ADP-ribosyl Cyclase 1/analysis
- Animals
- Antigens, CD/analysis
- Antigens, CD34/analysis
- Cell Differentiation/drug effects
- Cell Lineage
- Cell Proliferation/drug effects
- Cells, Cultured
- Cytokine Receptor gp130/metabolism
- Fetal Blood/cytology
- Fetal Blood/immunology
- Fetal Blood/metabolism
- Glycoproteins/analysis
- Hematopoietic Stem Cell Transplantation
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/metabolism
- Humans
- Immunomagnetic Separation
- Interleukin-3/metabolism
- Interleukin-6/metabolism
- Intracellular Signaling Peptides and Proteins
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Peptides/analysis
- Receptors, Fc/genetics
- Receptors, Fc/metabolism
- Receptors, Interleukin/metabolism
- Receptors, Interleukin-6
- Receptors, Notch/metabolism
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Takahiro Suzuki
- Department of Regeneration Medicine for Hematopoiesis, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2243
|
Chua KN, Chai C, Lee PC, Tang YN, Ramakrishna S, Leong KW, Mao HQ. Surface-aminated electrospun nanofibers enhance adhesion and expansion of human umbilical cord blood hematopoietic stem/progenitor cells. Biomaterials 2006; 27:6043-51. [PMID: 16854459 DOI: 10.1016/j.biomaterials.2006.06.017] [Citation(s) in RCA: 212] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Accepted: 06/23/2006] [Indexed: 12/13/2022]
Abstract
Interaction between hematopoietic stem/progenitor cells (HSPCs) and their extra cellular matrix components is an integral part of the signaling control for HSPC survival, proliferation and differentiation. We hypothesized that both substrate topographical cues and biochemical cues could act synergistically with cytokine supplementation to improve ex vivo expansion of HSPCs. In this study, we compared the ex vivo expansion of human umbilical cord blood CD34(+) cells on unmodified, hydroxylated, carboxylated and aminated nanofibers and films. Results from 10-day expansion cultures showed that aminated nanofiber mesh and film were most efficient in supporting the expansion of the CD34(+)CD45(+) cells (195-fold and 178-fold, respectively), as compared to tissue culture polystyrene (50-fold, p<0.05). In particular, aminated nanofiber meshes supported a higher degree of cell adhesion and percentage of HSPCs, as compared to aminated films. SEM imaging revealed the discrete colonies of cells proliferating and interacting with the aminated nanofibers. This study highlights the potential of a biomaterials approach to influence the proliferation and differentiation of HSPCs ex vivo.
Collapse
Affiliation(s)
- Kian-Ngiap Chua
- Division of Bioengineering and NUSNNI, National University of Singapore, Singapore 117576, Singapore
| | | | | | | | | | | | | |
Collapse
|
2244
|
Yaniv I, Stein J, Farkas DL, Askenasy N. The tale of early hematopoietic cell seeding in the bone marrow niche. Stem Cells Dev 2006; 15:4-16. [PMID: 16522158 DOI: 10.1089/scd.2006.15.4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Since introduction of the notion of a "niche" that hosts engraftment and activity of hematopoietic cells, there is a massive effort to discover its structure and decipher its function. Our understanding of the niche is continuously changing with reinterpretation of traditional concepts and apprehension of new insights into the biology of hematopoietic cell homing, seeding, and engraftment. Here we discuss some of the early events in hematopoietic stem cell seeding and engraftment and propose a perspective based on visualization of labeled bone marrow cells in real time in vivo. Primary seeding of hematopoietic cells in the bone marrow niches evolves as a complex and dynamic process; however, it follows discrete topological and chronological patterns. Initial seeding occurs on the endosteal surface of the marrow, which includes heterogeneous niches for primary seeding. Several days after transplantation the endosteal niches become more restrictive, hosting primarily mitotically quiescent cells, and gradual centripetal migration is accompanied by engagement in proliferation and differentiation. The hematopoietic niches evolve as heterogeneous three-dimensional microenvironments that are continuously changing over time.
Collapse
Affiliation(s)
- Isaac Yaniv
- Frankel Laboratory of Experimental Bone Marrow Transplantation, Center for Stem Cell Research, Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel 49202
| | | | | | | |
Collapse
|
2245
|
Lotem J, Sachs L. Epigenetics and the plasticity of differentiation in normal and cancer stem cells. Oncogene 2006; 25:7663-72. [PMID: 16847453 DOI: 10.1038/sj.onc.1209816] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Embryonic stem cells are characterized by their differentiation to all cell types during embryogenesis. In adult life, different tissues also have somatic stem cells, called adult stem cells, which in specific niches can undergo multipotent differentiation. The use of these adult stem cells has considerable therapeutic potential for the regeneration of damaged tissues. In both embryonic and adult stem cells, differentiation is controlled by epigenetic mechanisms, and the plasticity of differentiation in these cells is associated with transcription accessibility for genes expressed in different normal tissues. Abnormalities in genetic and/or epigenetic controls can lead to development of cancer, which is maintained by self-renewing cancer stem cells. Although the genetic abnormalities produce defects in growth and differentiation in cancer stem cells, these cells have not always lost the ability to undergo differentiation through epigenetic changes that by-pass the genomic abnormalities, thus creating the basis for differentiation therapy. Like normal stem cells, cancer stem cells can show plasticity for differentiation. This plasticity of cancer stem cells is also associated with transcription accessibility for genes that are normally expressed in different tissues, including tissues other than those from which the cancers originated. This broad transcription accessibility can also contribute to the behavior of cancer cells by overexpressing genes that promote cell viability, growth and metastasis.
Collapse
Affiliation(s)
- J Lotem
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
2246
|
Abstract
Scientists have tried for decades to understand cancer development in the context of therapeutic strategies. The realization that cancers may rely on "cancer stem cells" that share the self-renewal feature of normal stem cells has changed the perspective with regard to new approaches for treating the disease. In this review, we propose that one of the differences between normal stem cells and cancer stem cells is their degree of dependence on the stem cell niche, a specialized microenvironment in which stem cells reside. The stem cell niche in adult somatic tissues plays an essential role in maintaining stem cells or preventing tumorigenesis by providing primarily inhibitory signals for both proliferation and differentiation. However, the niche also provides transient signals for stem cell division to support ongoing tissue regeneration. The balance between proliferation-inhibiting and proliferation-promoting signals is the key to homeostatic regulation of stem cell maintenance versus tissue regeneration. Loss of the niche can lead to loss of stem cells, indicating the reliance of stem cells on niche signals. Therefore, cancer stem cells may arise from an intrinsic mutation, leading to self-sufficient cell proliferation, and/or may also involve deregulation or alteration of the niche by dominant proliferation-promoting signals. Furthermore, the molecular machinery used by normal stem cells for homing to or mobilizing from the niche may be "hijacked" by cancer stem cells for invasion and metastasis. We hope this examination of the interaction between stem cells and their niche will enhance understanding of the process of cancer development, invasiveness, and metastasis and reveal possible targets for cancer treatment.
Collapse
Affiliation(s)
- Linheng Li
- Stowers Institute for Medical Research, Kansas City, Missouri, USA.
| | | |
Collapse
|
2247
|
Rawlins EL, Hogan BLM. Epithelial stem cells of the lung: privileged few or opportunities for many? Development 2006; 133:2455-65. [PMID: 16735479 DOI: 10.1242/dev.02407] [Citation(s) in RCA: 238] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Most reviews of adult stem cells focus on the relatively undifferentiated cells dedicated to the renewal of rapidly proliferating tissues, such as the skin, gut and blood. By contrast, there is mounting evidence that organs and tissues such as the liver and pancreatic islets, which turn over more slowly,use alternative strategies, including the self-renewal of differentiated cells. The response of these organs to injury may also reveal the potential of differentiated cells to act as stem cells. The lung shows both slow turnover and rapid repair. New experimental approaches, including those based on studies of embryonic development, are needed to identify putative lung stem cells and strategies of lung homeostasis and repair.
Collapse
Affiliation(s)
- Emma L Rawlins
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
2248
|
Schroeder T, Meier-Stiegen F, Schwanbeck R, Eilken H, Nishikawa S, Häsler R, Schreiber S, Bornkamm GW, Nishikawa SI, Just U. Activated Notch1 alters differentiation of embryonic stem cells into mesodermal cell lineages at multiple stages of development. Mech Dev 2006; 123:570-9. [PMID: 16822655 DOI: 10.1016/j.mod.2006.05.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2005] [Revised: 05/14/2006] [Accepted: 05/17/2006] [Indexed: 02/06/2023]
Abstract
Signals of Notch transmembrane receptors function to regulate a wide variety of developmental cell fates. Here we investigate the role of Notch signaling in the development of mesodermal cell types by expressing a tamoxifen-inducible, activated form of Notch1 in embryonic stem cells (ESC). For differentiation of ESC into first mesodermal progenitor cells and then endothelial, mural, cardiac muscle and hematopoietic cells, the OP9 stroma co-culture system was used. Timed activation of Notch signaling by the addition of tamoxifen at various stages during differentiation of ESC into mesodermal cell lineages results in profound alterations in the generation of all of these cells. Differentiation of ESC into Flk1(+) mesodermal cells is inhibited by activated Notch. When Notch signaling is activated in mesodermal cells, generation of cardiac muscle, endothelial and hematopoietic cells is inhibited, favoring the generation of mural cells. Activation of Notch signaling in hematopoietic cells reduces colony formation and maintenance of hematopoiesis. These data suggest that Notch signaling plays a regulatory role in mesodermal development, cardiomyogenesis, the balanced generation of endothelial versus mural cells of blood vessels and hematopoietic development.
Collapse
Affiliation(s)
- Timm Schroeder
- Institute for Clinical Molecular Biology and Tumor Genetics, GSF - National Research Center for Environment and Health, Marchioninistr. 25, 81377 München, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2249
|
Guo Z, Li H, Li X, Yu X, Wang H, Tang P, Mao N. In vitro characteristics and in vivo immunosuppressive activity of compact bone-derived murine mesenchymal progenitor cells. Stem Cells 2006; 24:992-1000. [PMID: 16644925 DOI: 10.1634/stemcells.2005-0224] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In contrast to the considerable amount of data that documents the biological properties of mesenchymal progenitor cells from human and other species, there is still paucity of information about mouse counterparts, as their purification and culture expansion procedures remain rudimentary. In the present study, murine mesenchymal progenitor cell (muMPC) culture was developed by explant culture of collagenase-digested bone fragments after removal of the released cells. During cultivation, fibroblastoid cells sprouted and migrated from the fragments, followed by adherent monolayer development. The cells exhibited homogenous surface antigen profile and presented in vitro multipotential differentiation along osteocyte, chondrocyte, and adipocyte lineages, as evaluated by matched cell or matrix staining and reverse transcription polymerase chain reaction techniques. Also, the surface antigenic epitope changed and potential of proliferation and multidifferentiation decreased with successive subculturing. Functional investigations demonstrated that these cells supported in vitro hematopoiesis and suppressed lymphocyte cell proliferation triggered by ConA or allogeneic splenocytes. Furthermore, muMPCs prolonged the mean survival time of skin grafts across the major histocompatibility barrier (H2(b) --> H2(d)), suggestive of the immunosuppressive effects in vivo. The findings demonstrate that muMPCs obtained with this simple protocol are similar in property to their marrow counterparts, and thus, the protocol described here could be used for further investigations in mouse physiological and pathological models.
Collapse
Affiliation(s)
- Zikuan Guo
- Department of Cell Biology, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | | | | | | | | | | | | |
Collapse
|
2250
|
Song L, Young NJ, Webb NE, Tuan RS. Origin and characterization of multipotential mesenchymal stem cells derived from adult human trabecular bone. Stem Cells Dev 2006; 14:712-21. [PMID: 16433626 DOI: 10.1089/scd.2005.14.712] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Much of the knowledge regarding the regulatory pathways for adult stem cell self-renewal and differentiation has been obtained from the results of in vitro cultures. However, it is unclear if adult stem cells are controlled in the same way under physiological conditions. We examined this issue with respect to the migration of stem cells to tissue injury and how switch from a migratory state to one of proliferation wherein they participate in development. Building on our previous identification of multipotent stem cells in trabecular bone, we have examined the in vitro behavior of these cells within the bone milieu. We found that cell proliferation is inhibited within the trabecular bone niche as cells migrate out of the trabecular bone prior to proliferation. Additionally, multiple cell types were detected in adult trabecular bone, including osteoblasts, osteoclasts, endothelial cells, and Stro-1-positive mesenchymal stem cells. Furthermore, we demonstrated that Stro-1-positive cells migrated out of their native bone niche to generate multipotential stem and progenitor cells during in vitro culture. We conclude that self-renewal and differentiation of adult stem cells in connective tissues are tightly controlled and separately orchestrated processes. A regulatory network of extrinsic factors and intrinsic signals acts to stimulate the exit of stem cells from their niche so that they can localize to sites of wound healing, where they participate in development after functional differentiation.
Collapse
Affiliation(s)
- Lin Song
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|