1
|
Sim HJ, Bhattarai G, Kim MH, So HS, Poudel SB, Cho ES, Kook SH, Lee JC. Local and Systemic Overexpression of COMP-Ang1 Induces Ang1/Tie2-Related Thrombocytopenia and SDF-1/CXCR4-Dependent Anemia. Stem Cells 2022; 41:93-104. [PMID: 36368017 PMCID: PMC9887089 DOI: 10.1093/stmcls/sxac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/03/2022] [Indexed: 11/13/2022]
Abstract
While supplemental angiopoietin-1 (Ang1) improves hematopoiesis, excessive Ang1 induces bone marrow (BM) impairment, hematopoietic stem cell (HSC) senescence, and erythropoietic defect. Here, we examined how excessive Ang1 disturbs hematopoiesis and explored whether hematopoietic defects were related to its level using K14-Cre;c-Ang1 and Col2.3-Cre;c-Ang1 transgenic mice that systemically and locally overexpress cartilage oligomeric matrix protein-Ang1, respectively. We also investigated the impacts of Tie2 inhibitor and AMD3100 on hematopoietic development. Transgenic mice exhibited excessive angiogenic phenotypes, but K14-Cre;c-Ang1 mice showed more severe defects in growth and life span with higher presence of Ang1 compared with Col2.3-Cre;c-Ang1 mice. Dissimilar to K14-Cre;c-Ang1 mice, Col2.3-Cre;c-Ang1 mice did not show impaired BM retention or senescence of HSCs, erythropoietic defect, or disruption of the stromal cell-derived factor 1 (SDF-1)/CXCR4 axis. However, these mice exhibited a defect in platelet production depending on the expression of Tie2 and globin transcription factor 1 (GATA-1), but not GATA-2, in megakaryocyte progenitor (MP) cells. Treatment with Tie2 inhibitor recovered GATA-1 expression in MP cells and platelet production without changes in circulating RBC in transgenic mice. Consecutive AMD3100 administration not only induced irrecoverable senescence of HSCs but also suppressed formation of RBC, but not platelets, via correlated decreases in number of erythroblasts and their GATA-1 expression in B6 mice. Our results indicate that genetic overexpression of Ang1 impairs hematopoietic development depending on its level, in which megakaryopoiesis is preferentially impaired via activation of Ang1/Tie2 signaling, whereas erythropoietic defect is orchestrated by HSC senescence, inflammation, and disruption of the SDF-1/CXCR4 axis.
Collapse
Affiliation(s)
- Hyun-Jaung Sim
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea,Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Govinda Bhattarai
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Min-Hye Kim
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Han-Sol So
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Sher Bahadur Poudel
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Sung-Ho Kook
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea,Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Jeong-Chae Lee
- Corresponding author: Jeong-Chae Lee, PhD, School of Dentistry, Jeonbuk National University, Jeonju 54896, South Korea. Tel: +82 63 270 4049; Fax: +82 63 270 4004; ; or, Sung-Ho Kook, PhD, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, South Korea. Tel: +82 63 270 3327; Fax: +82 63 270 4312; E-mail: ; or, Eui-Sic Cho, PhD, DDS, School of Dentistry, Jeonbuk National University, Jeonju 54896, South Korea. Tel: +82 63 270 4045; Fax: +82 63 270 4004;
| |
Collapse
|
2
|
Kang SM, Park JH. Pleiotropic Benefits of DPP-4 Inhibitors Beyond Glycemic Control. Clin Med Insights Endocrinol Diabetes 2021; 14:11795514211051698. [PMID: 34733107 PMCID: PMC8558587 DOI: 10.1177/11795514211051698] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
Dipeptidyl peptidase (DPP)-4 inhibitors are oral anti-diabetic medications that block the activity of the ubiquitous enzyme DPP-4. Inhibition of this enzyme increases the level of circulating active glucagon-like peptide (GLP)-1 secreted from L-cells in the small intestine. GLP-1 increases the glucose level, dependent on insulin secretion from pancreatic β-cells; it also decreases the abnormally increased level of glucagon, eventually decreasing the blood glucose level in patients with type 2 diabetes. DPP-4 is involved in many physiological processes other than the degradation of GLP-1. Therefore, the inhibition of DPP-4 may have numerous effects beyond glucose control. In this article, we review the pleiotropic effects of DPP-4 inhibitors beyond glucose control, including their strong beneficial effects on the stress induced accelerated senescence of vascular cells, and the possible clinical implications of these effects.
Collapse
Affiliation(s)
- Seon Mee Kang
- Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Jeong Hyun Park
- Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| |
Collapse
|
3
|
Luo C, Wang L, Wu G, Huang X, Zhang Y, Ma Y, Xie M, Sun Y, Huang Y, Huang Z, Song Q, Li H, Hou Y, Li X, Xu S, Chen J. Comparison of the efficacy of hematopoietic stem cell mobilization regimens: a systematic review and network meta-analysis of preclinical studies. Stem Cell Res Ther 2021; 12:310. [PMID: 34051862 PMCID: PMC8164253 DOI: 10.1186/s13287-021-02379-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mobilization failure may occur when the conventional hematopoietic stem cells (HSCs) mobilization agent granulocyte colony-stimulating factor (G-CSF) is used alone, new regimens were developed to improve mobilization efficacy. Multiple studies have been performed to investigate the efficacy of these regimens via animal models, but the results are inconsistent. We aim to compare the efficacy of different HSC mobilization regimens and identify new promising regimens with a network meta-analysis of preclinical studies. METHODS We searched Medline and Embase databases for the eligible animal studies that compared the efficacy of different HSC mobilization regimens. Primary outcome is the number of total colony-forming cells (CFCs) in per milliliter of peripheral blood (/ml PB), and the secondary outcome is the number of Lin- Sca1+ Kit+ (LSK) cells/ml PB. Bayesian network meta-analyses were performed following the guidelines of the National Institute for Health and Care Excellence Decision Support Unit (NICE DSU) with WinBUGS version 1.4.3. G-CSF-based regimens were classified into the SD (standard dose, 200-250 μg/kg/day) group and the LD (low dose, 100-150 μg/kg/day) group based on doses, and were classified into the short-term (2-3 days) group and the long-term (4-5 days) group based on administration duration. Long-term SD G-CSF was chosen as the reference treatment. Results are presented as the mean differences (MD) with the associated 95% credibility interval (95% CrI) for each regimen. RESULTS We included 95 eligible studies and reviewed the efficacy of 94 mobilization agents. Then 21 studies using the poor mobilizer mice model (C57BL/6 mice) to investigate the efficacy of different mobilization regimens were included for network meta-analysis. Network meta-analyses indicated that compared with long-term SD G-CSF alone, 14 regimens including long-term SD G-CSF + Me6, long-term SD G-CSF + AMD3100 + EP80031, long-term SD G-CSF + AMD3100 + FG-4497, long-term SD G-CSF + ML141, long-term SD G-CSF + desipramine, AMD3100 + meloxicam, long-term SD G-CSF + reboxetine, AMD3100 + VPC01091, long-term SD G-CSF + FG-4497, Me6, long-term SD G-CSF + EP80031, POL5551, long-term SD G-CSF + AMD3100, AMD1300 + EP80031 and long-term LD G-CSF + meloxicam significantly increased the collections of total CFCs. G-CSF + Me6 ranked first among these regimens in consideration of the number of harvested CFCs/ml PB (MD 2168.0, 95% CrI 2062.0-2272.0). In addition, 7 regimens including long-term SD G-CSF + AMD3100, AMD3100 + EP80031, long-term SD G-CSF + EP80031, short-term SD G-CSF + AMD3100 + IL-33, long-term SD G-CSF + ML141, short-term LD G-CSF + ARL67156, and long-term LD G-CSF + meloxicam significantly increased the collections of LSK cells compared with G-CSF alone. Long-term SD G-CSF + AMD3100 ranked first among these regimens in consideration of the number of harvested LSK cells/ml PB (MD 2577.0, 95% CrI 2422.0-2733.0). CONCLUSIONS Considering the number of CFC and LSK cells in PB as outcomes, G-CSF plus AMD3100, Me6, EP80031, ML141, FG-4497, IL-33, ARL67156, meloxicam, desipramine, and reboxetine are all promising mobilizing regimens for future investigation.
Collapse
Affiliation(s)
- Chengxin Luo
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guixian Wu
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Xiangtao Huang
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Yali Zhang
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Yanni Ma
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Mingling Xie
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Yanni Sun
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Yarui Huang
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Zhen Huang
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Qiuyue Song
- Department of Health Statistics, Third Military Medical University, Chongqing, China
| | - Hui Li
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Yu Hou
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China
| | - Xi Li
- Institute of Infectious Disease, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
| | - Shuangnian Xu
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China.
| | - Jieping Chen
- Center for Hematology, Southwest Hospital, Third Military Medical University, #30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Key Laboratory of Cancer Immunotherapy of Chongqing, Chongqing, China.
| |
Collapse
|
4
|
Rajendiran S, Smith-Berdan S, Kunz L, Risolino M, Selleri L, Schroeder T, Forsberg EC. Ubiquitous overexpression of CXCL12 confers radiation protection and enhances mobilization of hematopoietic stem and progenitor cells. Stem Cells 2020; 38:1159-1174. [PMID: 32442338 DOI: 10.1002/stem.3205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
C-X-C motif chemokine ligand 12 (CXCL12; aka SDF1α) is a major regulator of a number of cellular systems, including hematopoiesis, where it influences hematopoietic cell trafficking, proliferation, and survival during homeostasis and upon stress and disease. A variety of constitutive, temporal, ubiquitous, and cell-specific loss-of-function models have documented the functional consequences on hematopoiesis upon deletion of Cxcl12. Here, in contrast to loss-of-function experiments, we implemented a gain-of-function approach by generating a doxycycline-inducible transgenic mouse model that enables spatial and temporal overexpression of Cxcl12. We demonstrated that ubiquitous CXCL12 overexpression led to an increase in multipotent progenitors in the bone marrow and spleen. The CXCL12+ mice displayed reduced reconstitution potential as either donors or recipients in transplantation experiments. Additionally, we discovered that Cxcl12 overexpression improved hematopoietic stem and progenitor cell mobilization into the blood, and conferred radioprotection by promoting quiescence. Thus, this new CXCL12+ mouse model provided new insights into major facets of hematopoiesis and serves as a versatile resource for studying CXCL12 function in a variety of contexts.
Collapse
Affiliation(s)
- Smrithi Rajendiran
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| | - Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| | - Leo Kunz
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| | - Maurizio Risolino
- Program in Craniofacial Biology, Institute of Human Genetics, Eli and Edyth Broad Center of Regeneration Medicine and Stem Cell Research, Departments of Orofacial Sciences and Anatomy, University of California, San Francisco, California, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Institute of Human Genetics, Eli and Edyth Broad Center of Regeneration Medicine and Stem Cell Research, Departments of Orofacial Sciences and Anatomy, University of California, San Francisco, California, USA
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| |
Collapse
|
5
|
Albakri M, Tashkandi H, Zhou L. A Review of Advances in Hematopoietic Stem Cell Mobilization and the Potential Role of Notch2 Blockade. Cell Transplant 2020; 29:963689720947146. [PMID: 32749152 PMCID: PMC7563033 DOI: 10.1177/0963689720947146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation can be a potential cure for hematological malignancies and some nonhematologic diseases. Hematopoietic stem and progenitor cells (HSPCs) collected from peripheral blood after mobilization are the primary source to provide HSC transplantation. In most of the cases, mobilization by the cytokine granulocyte colony-stimulating factor with chemotherapy, and in some settings, with the CXC chemokine receptor type 4 antagonist plerixafor, can achieve high yield of hematopoietic progenitor cells (HPCs). However, adequate mobilization is not always successful in a significant portion of donors. Research is going on to find new agents or strategies to increase HSC mobilization. Here, we briefly review the history of HSC transplantation, current mobilization regimens, some of the novel agents that are under investigation for clinical practice, and our recent findings from animal studies regarding Notch and ligand interaction as potential targets for HSPC mobilization.
Collapse
Affiliation(s)
- Marwah Albakri
- Department of Pathology, Case Western Reserve University, Cleveland,
OH, USA
| | - Hammad Tashkandi
- Department of Pathology, University of Pittsburgh Medical Center,
PA, USA
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University, Cleveland,
OH, USA
| |
Collapse
|
6
|
Valent P, Sadovnik I, Eisenwort G, Herrmann H, Bauer K, Mueller N, Sperr WR, Wicklein D, Schumacher U. Redistribution, homing and organ-invasion of neoplastic stem cells in myeloid neoplasms. Semin Cancer Biol 2019; 60:191-201. [PMID: 31408723 DOI: 10.1016/j.semcancer.2019.07.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023]
Abstract
The development of a myeloid neoplasm is a step-wise process that originates from leukemic stem cells (LSC) and includes pre-leukemic stages, overt leukemia and a drug-resistant terminal phase. Organ-invasion may occur in any stage, but is usually associated with advanced disease and a poor prognosis. Sometimes, extra-medullary organ invasion shows a metastasis-like or even sarcoma-like destructive growth of neoplastic cells in local tissue sites. Examples are myeloid sarcoma, mast cell sarcoma and localized blast phase of chronic myeloid leukemia. So far, little is known about mechanisms underlying re-distribution and extramedullary dissemination of LSC in myeloid neoplasms. In this article, we discuss mechanisms through which LSC can mobilize out of the bone marrow niche, can transmigrate from the blood stream into extramedullary organs, can invade local tissue sites and can potentially create or support the formation of local stem cell niches. In addition, we discuss strategies to interfere with LSC expansion and organ invasion by targeted drug therapies.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria.
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria
| | - Gregor Eisenwort
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria
| | - Harald Herrmann
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria; Department of Radiotherapy, Medical University of Vienna, Department of Medicine III, Austria
| | - Karin Bauer
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria
| | - Niklas Mueller
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Department of Internal Medicine III, Division of Hematology and Oncology, Hospital of the Ludwig-Maximilians-University Munich, Germany
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, Austria
| | - Daniel Wicklein
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
7
|
Quickly attainable and highly engrafting hematopoietic stem cells. BLOOD SCIENCE 2019; 1:113-115. [PMID: 35402793 PMCID: PMC8975002 DOI: 10.1097/bs9.0000000000000003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 11/26/2022] Open
|
8
|
Long-Acting IL-33 Mobilizes High-Quality Hematopoietic Stem and Progenitor Cells More Efficiently Than Granulocyte Colony-Stimulating Factor or AMD3100. Biol Blood Marrow Transplant 2019; 25:1475-1485. [PMID: 31163266 DOI: 10.1016/j.bbmt.2019.05.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/15/2019] [Accepted: 05/27/2019] [Indexed: 02/08/2023]
Abstract
Mobilization of hematopoietic stem and progenitor cells (HSPCs) has become increasingly important for hematopoietic cell transplantation. Current mobilization approaches are insufficient because they fail to mobilize sufficient numbers of cells in a significant fraction of patients and are biased toward myeloid immune reconstitution. A novel, single drug mobilization agent that allows a more balanced (myeloid and lymphoid) reconstitution would therefore be highly favorable to improve transplantation outcome. In this present study, we tested commercially available IL-33 molecules and engineered novel variants of IL-33. These molecules were tested in cell-based assays in vitro and in mobilization models in vivo. We observed for the first time that IL-33 treatment in mice mobilized HSPCs and common myeloid progenitors more efficiently than clinical mobilizing agents granulocyte colony-stimulating factor (G-CSF) or AMD3100. We engineered several oxidation-resistant IL-33 variants with equal or better in vitro activity. In vivo, these variants mobilized HSPCs and, interestingly, also hematopoietic stem cells, common lymphoid progenitor cells, and endothelial progenitor cells more efficiently than wild-type IL-33 or G-CSF. We then engineered an IL-33-Fc fusion molecule, a single dose of which was sufficient to significantly increase the mobilization of HSPCs after 4 days. In conclusion, our findings suggest that long-acting, oxidation-resistant IL-33 may be a novel approach for HSPC transplantation. IL-33-mobilized HSPCs differ from cells mobilized with G-CSF and AMD3100, and it is possible that these differences may result in better transplantation outcomes.
Collapse
|
9
|
Avogaro A, Fadini GP. The pleiotropic cardiovascular effects of dipeptidyl peptidase-4 inhibitors. Br J Clin Pharmacol 2018; 84:1686-1695. [PMID: 29667232 DOI: 10.1111/bcp.13611] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022] Open
Abstract
Patients with Type 2 diabetes have an excess risk for cardiovascular disease. One of the several approaches, included in the guidelines for the management of Type 2 diabetes, is based on dipeptidyl peptidase 4 (DPP-4; also termed CD26) inhibitors, also called gliptins. Gliptins inhibit the degradation of glucagon-like peptide-1 (GLP-1): this effect is associated with increased circulating insulin-to-glucagon ratio, and a consequent reduction of HbA1c. In addition to incretin hormones, there are several proteins that may be affected by DPP-4 and its inhibition: among these some are relevant for the cardiovascular system homeostasis such as SDF-1α and its receptor CXCR4, brain natriuretic peptides, neuropeptide Y and peptide YY. In this review, we will discuss the pathophysiological relevance of gliptin pleiotropism and its translational potential.
Collapse
Affiliation(s)
- Angelo Avogaro
- Department of Medicine, Section of Diabetes and Metabolic Diseases, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, Section of Diabetes and Metabolic Diseases, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Lapostolle V, Chevaleyre J, Duchez P, Rodriguez L, Vlaski-Lafarge M, Sandvig I, Brunet de la Grange P, Ivanovic Z. Repopulating hematopoietic stem cells from steady-state blood before and after ex vivo culture are enriched in the CD34 +CD133 +CXCR4 low fraction. Haematologica 2018; 103:1604-1615. [PMID: 29858385 PMCID: PMC6165804 DOI: 10.3324/haematol.2017.183962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
The feasibility of ex vivo expansion allows us to consider the steady-state peripheral blood as an alternative source of hematopoietic stem progenitor cells for transplantation when growth factor-induced cell mobilization is contraindicated or inapplicable. Ex vivo expansion dramatically enhances the in vivo reconstituting cell population from steady-state blood. In order to investigate phenotype and the expression of homing molecules, the expression of CD34, CD133, CD90, CD45RA, CD26 and CD9 was determined on sorted CD34+ cells according to CXCR4 (“neg”, “low” “bright”) and CD133 expression before and after ex vivo expansion. Hematopoietic stem cell activity was determined in vivo on the basis of hematopoietic repopulation of primary and secondary recipients - NSG immuno-deficient mice. In vivo reconstituting cells in the steady-state blood CD34+ cell fraction before expansion belong to the CD133+ population and are CXCR4low or, to a lesser extent, CXCR4neg, while after ex vivo expansion they are contained only in the CD133+CXCR4low cells. The failure of the CXCR4bright population to engraft is probably due to the exclusive expression of CD26 by these cells. The limiting-dilution analysis showed that both repopulating cell number and individual proliferative capacity were enhanced by ex vivo expansion. Thus, steady-state peripheral blood cells exhibit a different phenotype compared to mobilized and cord blood cells, as well as to those issued from the bone marrow. These data represent the first phenotypic characterization of steady-state blood cells exhibiting short- and long-term hematopoietic reconstituting potential, which can be expanded ex vivo, a sine qua non for their subsequent use for transplantation.
Collapse
Affiliation(s)
- Véronique Lapostolle
- Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,U1035 INSERM/Bordeaux University, France
| | - Jean Chevaleyre
- Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,U1035 INSERM/Bordeaux University, France
| | - Pascale Duchez
- Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,U1035 INSERM/Bordeaux University, France
| | - Laura Rodriguez
- Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,U1035 INSERM/Bordeaux University, France
| | - Marija Vlaski-Lafarge
- Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France.,U1035 INSERM/Bordeaux University, France
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | | | - Zoran Ivanovic
- Etablissement Français du Sang Nouvelle Aquitaine, Bordeaux, France .,U1035 INSERM/Bordeaux University, France
| |
Collapse
|
11
|
Quickly Attainable and Highly Engrafting Hematopoietic Stem Cells. BLOOD SCIENCE 2018. [DOI: 10.2478/bls-2018-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
12
|
Wang W, Yu S, Myers J, Wang Y, Xin WW, Albakri M, Xin AW, Li M, Huang AY, Xin W, Siebel CW, Lazarus HM, Zhou L. Notch2 blockade enhances hematopoietic stem cell mobilization and homing. Haematologica 2017; 102:1785-1795. [PMID: 28729299 PMCID: PMC5622863 DOI: 10.3324/haematol.2017.168674] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/13/2017] [Indexed: 12/21/2022] Open
Abstract
Despite use of newer approaches, some patients being considered for autologous hematopoietic cell transplantation (HCT) may only mobilize limited numbers of hematopoietic progenitor cells (HPCs) into blood, precluding use of the procedure, or being placed at increased risk of complications due to slow hematopoietic reconstitution. Developing more efficacious HPC mobilization regimens and strategies may enhance the mobilization process and improve patient outcome. Although Notch signaling is not essential for homeostasis of adult hematopoietic stem cells (HSCs), Notch-ligand adhesive interaction maintains HSC quiescence and niche retention. Using Notch receptor blocking antibodies, we report that Notch2 blockade, but not Notch1 blockade, sensitizes hematopoietic stem cells and progenitors (HSPCs) to mobilization stimuli and leads to enhanced egress from marrow to the periphery. Notch2 blockade leads to transient myeloid progenitor expansion without affecting HSC homeostasis and self-renewal. We show that transient Notch2 blockade or Notch2-loss in mice lacking Notch2 receptor lead to decreased CXCR4 expression by HSC but increased cell cycling with CXCR4 transcription being directly regulated by the Notch transcriptional protein RBPJ. In addition, we found that Notch2-blocked or Notch2-deficient marrow HSPCs show an increased homing to the marrow, while mobilized Notch2-blocked, but not Notch2-deficient stem cells and progenitors, displayed a competitive repopulating advantage and enhanced hematopoietic reconstitution. These findings suggest that blocking Notch2 combined with the current clinical regimen may further enhance HPC mobilization and improve engraftment during HCT.
Collapse
Affiliation(s)
- Weihuan Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Shuiliang Yu
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Jay Myers
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Yiwei Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - William W Xin
- School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Marwah Albakri
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | | | - Ming Li
- Biostatistics and Bioinformatics Core Facility, Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alex Y Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA .,Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Wei Xin
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Christian W Siebel
- Department of Molecular Biology Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
13
|
Ben Nasr M, Bassi R, Usuelli V, Valderrama-Vasquez A, Tezza S, D'Addio F, Fiorina P. The use of hematopoietic stem cells in autoimmune diseases. Regen Med 2016; 11:395-405. [PMID: 27165670 DOI: 10.2217/rme-2015-0057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Hematopoietic stem cells (HSCs) have been shown recently to hold much promise in curing autoimmune diseases. Newly diagnosed Type 1 diabetes individuals have been successfully reverted to normoglycemia by administration of autologous HSCs in association with a nonmyeloablative regimen (antithymocyte globulin + cyclophasmide). Furthermore, recent trials reported positive results by using HSCs in treatment of systemic sclerosis, multiple sclerosis and rheumatoid arthritis as well. Early data suggested that HSCs possess immunological properties that may be harnessed to alleviate the symptoms of individuals with autoimmune disorders and possibly induce remission of autoimmune diseases. Mechanistically, HSCs may facilitate the generation of regulatory T cells, may inhibit the function of autoreactive T-cell function and may reshape the immune system.
Collapse
Affiliation(s)
- Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Transplant Medicine Division, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Roberto Bassi
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Transplant Medicine Division, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Vera Usuelli
- Transplant Medicine Division, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Sara Tezza
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francesca D'Addio
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Transplant Medicine Division, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Transplant Medicine Division, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
14
|
Kopinska A, Krawczyk-Kulis M, Dziaczkowska-Suszek J, Bieszczad K, Jagoda K, Kyrcz-Krzemien S. The importance of the number of transplanted cells with dipeptidyl peptidase-4 expression on the haematopoietic recovery and lymphocyte reconstitution in patients with multiple myeloma after autologous haematopoietic stem-cell transplantation. Hematol Oncol 2015. [DOI: 10.1002/hon.2267] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Anna Kopinska
- Department of Hematology and Bone Marrow Transplantation; Silesian Medical University; Katowice Poland
| | - Małgorzata Krawczyk-Kulis
- Department of Hematology and Bone Marrow Transplantation; Silesian Medical University; Katowice Poland
| | | | - Katarzyna Bieszczad
- Department of Hematology and Bone Marrow Transplantation; Silesian Medical University; Katowice Poland
| | - Krystyna Jagoda
- Department of Hematology and Bone Marrow Transplantation; Silesian Medical University; Katowice Poland
| | - Slawomira Kyrcz-Krzemien
- Department of Hematology and Bone Marrow Transplantation; Silesian Medical University; Katowice Poland
| |
Collapse
|
15
|
Zhong J, Rajagopalan S. Dipeptidyl Peptidase-4 Regulation of SDF-1/CXCR4 Axis: Implications for Cardiovascular Disease. Front Immunol 2015; 6:477. [PMID: 26441982 PMCID: PMC4585326 DOI: 10.3389/fimmu.2015.00477] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/03/2015] [Indexed: 12/19/2022] Open
Abstract
Dipeptidyl peptidase-4 (DPP4) is a ubiquitously expressed protease that regulates diverse number of physiological functions. As a dipeptidase, it exerts its catalytic effects on proteins/peptides with proline, alanine, or serine in the penultimate (P1) amino acid residue from the amino terminus. The evidence to date supports an important effect of DPP4 in catalytic cleavage of incretin peptides and this perhaps represents the main mechanism by which DPP4 inhibition improves glycemic control. DPP4 also plays an important role in the degradation of multiple chemokines of which stromal cell-derived factor-1 (SDF-1, also known as CXCL12) is perhaps an increasingly recognized target, given its importance in processes, such as hematopoiesis, angiogenesis, and stem cell homing. In the current review, we will summarize the importance of DPP4-mediated enzymatic processing of cytokines/chemokines with an emphasis on SDF-1 and resultant implications for cardiovascular physiology and disease.
Collapse
Affiliation(s)
- Jixin Zhong
- Division of Cardiovascular Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| |
Collapse
|
16
|
Frederich R, Alexander JH, Fiedorek FT, Donovan M, Berglind N, Harris S, Chen R, Wolf R, Mahaffey KW. A Systematic Assessment of Cardiovascular Outcomes in the Saxagliptin Drug Development Program for Type 2 Diabetes. Postgrad Med 2015; 122:16-27. [DOI: 10.3810/pgm.2010.05.2138] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
17
|
Hoggatt J, Speth JM, Pelus LM. Concise review: Sowing the seeds of a fruitful harvest: hematopoietic stem cell mobilization. Stem Cells 2015; 31:2599-606. [PMID: 24123398 DOI: 10.1002/stem.1574] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 05/06/2013] [Indexed: 02/06/2023]
Abstract
Hematopoietic stem cell transplantation is the only curative option for a number of malignant and nonmalignant diseases. As the use of hematopoietic transplant has expanded, so too has the source of stem and progenitor cells. The predominate source of stem and progenitors today, particularly in settings of autologous transplantation, is mobilized peripheral blood. This review will highlight the historical advances which led to the widespread use of peripheral blood stem cells for transplantation, with a look toward future enhancements to mobilization strategies.
Collapse
Affiliation(s)
- Jonathan Hoggatt
- Harvard University, Department of Stem Cell and Regenerative Biology, Massachusetts General Hospital, Center for Regenerative Medicine, Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | | | | |
Collapse
|
18
|
Bari S, Seah KKH, Poon Z, Cheung AMS, Fan X, Ong SY, Li S, Koh LP, Hwang WYK. Expansion and homing of umbilical cord blood hematopoietic stem and progenitor cells for clinical transplantation. Biol Blood Marrow Transplant 2014; 21:1008-19. [PMID: 25555449 DOI: 10.1016/j.bbmt.2014.12.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 12/22/2014] [Indexed: 12/26/2022]
Abstract
The successful expansion of hematopoietic stem and progenitor cells (HSPCs) from umbilical cord blood (UCB) for transplantation could revolutionize clinical practice by improving transplantation-related outcomes and making available UCB units that have suboptimal cell doses for transplantation. New cytokine combinations appear able to promote HSPC growth with minimal differentiation into mature precursors and new agents, such as insulin-like growth factor-binding protein 2, are being used in clinical trials. Molecules that simulate the HSPC niche, such as Notch ligand, have also shown promise. Further improvements have been made with the use of mesenchymal stromal cells, which have made possible UCB expansion without a potentially deleterious prior CD34/CD133 cell selection step. Chemical molecules, such as copper chelators, nicotinamide, and aryl hydrocarbon antagonists, have shown excellent outcomes in clinical studies. The use of bioreactors could further add to HSPC studies in future. Drugs that could improve HSPC homing also appear to have potential in improving engraftment times in UCB transplantation. Technologies to expand HSPC from UCB and to enhance the homing of these cells appear to have attained the goal of accelerating hematopoietic recovery. Further discoveries and clinical studies are likely to make the goal of true HSPC expansion a reality for many applications in future.
Collapse
Affiliation(s)
- Sudipto Bari
- Department of Hematology, Singapore General Hospital, Singapore; Department of Pharmacy, National University of Singapore, Singapore
| | | | - Zhiyong Poon
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research and Technology, Singapore
| | | | - Xiubo Fan
- Department of Clinical Research, Singapore General Hospital, Singapore
| | - Shin-Yeu Ong
- Department of Hematology, Singapore General Hospital, Singapore
| | - Shang Li
- Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore
| | - Liang Piu Koh
- Department of Hematology-Oncology, National University Cancer Institute, Singapore
| | - William Ying Khee Hwang
- Department of Hematology, Singapore General Hospital, Singapore; Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore; Singapore Cord Blood Bank, Singapore.
| |
Collapse
|
19
|
Solh M. Haploidentical vs cord blood transplantation for adults with acute myelogenous leukemia. World J Stem Cells 2014; 6:371-9. [PMID: 25258659 PMCID: PMC4172666 DOI: 10.4252/wjsc.v6.i4.371] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/12/2014] [Accepted: 07/17/2014] [Indexed: 02/06/2023] Open
Abstract
Hematopoeitic cell transplantation is established as a curative treatment for patients w acute myelogenous leukemia. Haploidentical family donor and umbilical cord blood (UCB) are alternative sources of stem cells for patients lacking a matched sibling or unrelated donor. The early challenges of transplant complications related to poor engraftment and graft-vs-host disease have been overcome with new strategies such as using 2 units and increased cell dose in UCB and T-cell depletion and post transplantation cyclophosphamide in haploidentical transplantation. The outcomes of alternative transplantation for acute leukemia were compared to other traditional graft sources. For patients lacking a matched sibling or unrelated donor, either strategy is a suitable option. The choice should rely mostly on the urgency of the transplantation and the available cell dose as well as the expertise available at the transplant center. This manuscript reviews the options of alternative donor transplantation and highlights recent advances in each of these promising transplantation options.
Collapse
Affiliation(s)
- Melhem Solh
- Melhem Solh, the Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, GA 30342, United States
| |
Collapse
|
20
|
Broxmeyer HE, Farag S. Background and future considerations for human cord blood hematopoietic cell transplantation, including economic concerns. Stem Cells Dev 2014; 22 Suppl 1:103-10. [PMID: 24304086 DOI: 10.1089/scd.2013.0382] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cord blood (CB) has been used since 1988 as a source of hematopoietic stem cells (HSCs) and progenitor cells for hematopoietic cell transplantation (HCT) to treat patients with malignant and nonmalignant disorders. CB has both advantages and disadvantages when compared with other tissue sources of HSCs such as bone marrow and mobilized peripheral blood, which are also being used in the setting of HCT. This short review focuses on some historical information, as well as current efforts that are being assessed to enhance the efficacy of CB HCT. Also of importance are the costs of CB, and the feasibility and economics of using such to be identified, and newly confirmed improvements worldwide for the greatest number of patients. In this context, simple methods that would not necessarily entail the need for selected cell-processing facilities to ex vivo expand or improve the CB graft's functional activity may be of interest, with one such possibility being the use of an orally active inhibitor of the enzyme dipeptidylpeptidase 4, alone or in combination with other new and innovative approaches for improving HSC engraftment and in vivo repopulating capability of CB.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- 1 Department of Microbiology & Immunology, Indiana University School of Medicine , Indianapolis, Indiana
| | | |
Collapse
|
21
|
Kedarisetty CK, Anand L, Khanam A, Kumar A, Rastogi A, Maiwall R, Sarin SK. Growth factors enhance liver regeneration in acute-on-chronic liver failure. Hepatol Int 2014. [PMID: 26201333 DOI: 10.1007/s12072-014-9538-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute-on-chronic liver failure is a distinct syndrome characterized by a rapid progression of liver disease culminating in organ failure and death. The only definitive treatment is liver transplantation. However, there is a possible element of reversibility and hepatic regeneration if the acute insult can be tided over. Exogenously administered growth factors may stimulate hepatocytes, hepatic progenitor cells and bone marrow-derived cells to supplement hepatic regeneration. The proposed review is intended to provide an in-depth analysis of the individual components of hepatic and bone marrow niches and highlight the growing role of various growth factors in liver regeneration in health and in liver failure.
Collapse
Affiliation(s)
| | - Lovkesh Anand
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Arshi Khanam
- Department of Research, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Anupam Kumar
- Department of Research, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Archana Rastogi
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rakhi Maiwall
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India.
| |
Collapse
|
22
|
Rennert RC, Sorkin M, Garg RK, Gurtner GC. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med 2013; 7:833-50. [PMID: 23164083 DOI: 10.2217/rme.12.82] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tissue repair and regeneration are thought to involve resident cell proliferation as well as the selective recruitment of circulating stem and progenitor cell populations through complex signaling cascades. Many of these recruited cells originate from the bone marrow, and specific subpopulations of bone marrow cells have been isolated and used to augment adult tissue regeneration in preclinical models. Clinical studies of cell-based therapies have reported mixed results, however, and a variety of approaches to enhance the regenerative capacity of stem cell therapies are being developed based on emerging insights into the mechanisms of progenitor cell biology and recruitment following injury. This article discusses the function and mechanisms of recruitment of important bone marrow-derived stem and progenitor cell populations following injury, as well as the emerging therapeutic applications targeting these cells.
Collapse
Affiliation(s)
- Robert C Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Hagey Building GK-201, Stanford, CA 94305-5148, USA
| | | | | | | |
Collapse
|
23
|
Farag SS, Srivastava S, Messina-Graham S, Schwartz J, Robertson MJ, Abonour R, Cornetta K, Wood L, Secrest A, Strother RM, Jones DR, Broxmeyer HE. In vivo DPP-4 inhibition to enhance engraftment of single-unit cord blood transplants in adults with hematological malignancies. Stem Cells Dev 2013; 22:1007-15. [PMID: 23270493 DOI: 10.1089/scd.2012.0636] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Delayed engraftment is a significant limitation of umbilical cord blood (UCB) transplantation due to low stem cell numbers. Inhibition of dipeptidyl peptidase (DPP)-4 enhanced engraftment in murine transplants. We evaluated the feasibility of systemic DPP-4 inhibition using sitagliptin to enhance engraftment of single-unit UCB grafts in adults with hematological malignancies. Twenty-four patients (21-58 years) received myeloablative conditioning, followed by sitagliptin 600 mg orally days -1 to +2, and single UCB grafts day 0. Seventeen receiving red cell-depleted (RCD) grafts, matched at 4 (n=10) or 5 (n=7) of 6 human leucocyte antigen (HLA) loci with median nucleated cell dose 3.6 (2.5-5.2)×10(7)/kg, engrafted at median of 21 (range, 13-50) days with cumulative incidence of 94% (95% confidence interval, 84%-100%) at 50 days. Plasma DDP-4 activity was reduced to 23%±7% within 2 h. Area under DPP-4 activity-time curve (AUCA) correlated with engraftment; 9 of 11 with AUCA <6,000 activity·h engrafted within ≤21 days, while all 6 with higher AUCA engrafted later (P=0.002). Seven patients receiving red cell replete grafts had 10-fold lower colony forming units after thawing compared with RCD grafts, with poor engraftment. Systemic DPP-4 inhibition was well tolerated and may enhance engraftment. Optimizing sitagliptin dosing to achieve more sustained DPP-4 inhibition may further improve outcome.
Collapse
Affiliation(s)
- Sherif S Farag
- Division of Hematology-Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Matheeussen V, Jungraithmayr W, De Meester I. Dipeptidyl peptidase 4 as a therapeutic target in ischemia/reperfusion injury. Pharmacol Ther 2012; 136:267-82. [DOI: 10.1016/j.pharmthera.2012.07.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 07/16/2012] [Indexed: 01/21/2023]
|
25
|
Abstract
The use of mobilized peripheral blood stem cells (PBSCs) has largely replaced the use of bone marrow as a source of stem cells for both allogeneic and autologous stem cell transplantation. G-CSF with or without chemotherapy is the most commonly used regimen for stem cell mobilization. Some donors or patients, especially the heavily pretreated patients, fail to mobilize the targeted number of stem cells with this regimen. A better understanding of the mechanisms involved in hematopoietic stem cell (HSC) trafficking could lead to the development of newer mobilizing agents and therapeutic approaches. This review will cover the current methods for stem cell mobilization and recent developments in the understanding of the biology of stem cells and the bone marrow microenvironment.
Collapse
Affiliation(s)
- Ibraheem H Motabi
- Siteman Cancer Center, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110, USA.
| | | |
Collapse
|
26
|
Sahin AO, Buitenhuis M. Molecular mechanisms underlying adhesion and migration of hematopoietic stem cells. Cell Adh Migr 2012; 6:39-48. [PMID: 22647939 DOI: 10.4161/cam.18975] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem cell transplantation is the most powerful treatment modality for a large number of hematopoietic malignancies, including leukemia. Successful hematopoietic recovery after transplantation depends on homing of hematopoietic stem cells to the bone marrow and subsequent lodging of those cells in specific niches in the bone marrow. Migration of hematopoietic stem cells to the bone marrow is a highly regulated process that requires correct regulation of the expression and activity of various molecules including chemoattractants, selectins and integrins. This review will discuss recent studies that have extended our understanding of the molecular mechanisms underlying adhesion, migration and bone marrow homing of hematopoietic stem cells.
Collapse
Affiliation(s)
- Aysegul Ocal Sahin
- Department of Hematology and Erasmus MC Stem Cell Institute for Regenerative Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
27
|
PEG-albumin plasma expansion increases expression of MCP-1 evidencing increased circulatory wall shear stress: an experimental study. PLoS One 2012; 7:e39111. [PMID: 22720043 PMCID: PMC3375248 DOI: 10.1371/journal.pone.0039111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 05/16/2012] [Indexed: 11/19/2022] Open
Abstract
Treatment of blood loss with plasma expanders lowers blood viscosity, increasing cardiac output. However, increased flow velocity by conventional plasma expanders does not compensate for decreased viscosity in maintaining vessel wall shear stress (WSS), decreasing endothelial nitric oxide (NO) production. A new type of plasma expander using polyethylene glycol conjugate albumin (PEG-Alb) causes supra-perfusion when used in extreme hemodilution and is effective in treating hemorrhagic shock, although it is minimally viscogenic. An acute 40% hemodilution/exchange-transfusion protocol was used to compare 4% PEG-Alb to Ringer's lactate, Dextran 70 kDa and 6% Hetastarch (670 kDa) in unanesthetized CD-1 mice. Serum cytokine analysis showed that PEG-Alb elevates monocyte chemotactic protein-1 (MCP-1), a member of a small inducible gene family, as well as expression of MIP-1α, and MIP-2. MCP-1 is specific to increased WSS. Given the direct link between increased WSS and production of NO, the beneficial resuscitation effects due to PEG-Alb plasma expansion appear to be due to increased WSS through increased perfusion and blood flow rather than blood viscosity.
Collapse
|
28
|
Abstract
Stem cell technology holds great promises for the cures of devastating diseases, injuries, aging, and even cancers as it is applied in regenerative medicine. Recent breakthroughs in the development of induced pluripotent stem cell techniques and efficient differentiation strategies have generated tremendous enthusiasm and efforts to explore the therapeutic potential of stem cells. Small molecules, which target specific signaling pathways and/or proteins, have been demonstrated to be particularly valuable for manipulating cell fate, state, and function. Such small molecules not only are useful in generating desired cell types in vitro for various applications but also could be further developed as conventional therapeutics to stimulate patients' endogenous cells to repair and regenerate in vivo. Here, we focus on recent progress in the use of small molecules in stem cell biology and regenerative medicine.
Collapse
Affiliation(s)
- Saiyong Zhu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
29
|
Abstract
Dynamic interactions between hematopoietic cells and their specialized bone marrow microenvironments, namely the vascular and osteoblastic 'niches', regulate hematopoiesis. The vascular niche is conducive for thrombopoiesis and megakaryocytes may, in turn, regulate the vascular niche, especially in supporting vascular and hematopoietic regeneration following irradiation or chemotherapy. A role for platelets in tumor growth and metastasis is well established and, more recently, the vascular niche has also been implicated as an area for preferential homing and engraftment of malignant cells. This article aims to provide an overview of the dynamic interactions between cellular and molecular components of the bone marrow vascular niche and the potential role of megakaryocytes in bone marrow malignancy.
Collapse
Affiliation(s)
- B Psaila
- Department of Haematology, Imperial College School of Medicine, London, UK.
| | | | | |
Collapse
|
30
|
Mendt M, Cardier JE. Stromal-derived factor-1 and its receptor, CXCR4, are constitutively expressed by mouse liver sinusoidal endothelial cells: implications for the regulation of hematopoietic cell migration to the liver during extramedullary hematopoiesis. Stem Cells Dev 2012; 21:2142-51. [PMID: 22121892 DOI: 10.1089/scd.2011.0565] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stromal-derived factor (SDF)-1 is the main regulating factor for trafficking/homing of hematopoietic stem cells (HSC) to the bone marrow (BM). It is possible that this chemokine may also play a fundamental role in regulating the migration of HSC to several organs during extramedullary hematopoiesis. Because liver sinusoidal endothelial cells (LSEC) constitute an extramedullary niche for HSC, it is possible that these cells represent one of the main cellular sources of SDF-1 at the liver. Here, we show that LSEC express SDF-1 at the mRNA and protein level. Biological assays showed that conditioned medium from LSEC (LSEC-CM) stimulated the migration of BM progenitor lineage-negative (BM/Lin⁻) cells. This effect was significantly reduced by AMD3100, indicating that the SDF-1/CXCR4 axis is involved in the stimulatory migrating effect induced by LSEC-CM. Early localization of HSC in SDF-1-expressing LSEC microenvironment together with increased levels of this chemokine in hepatic homogenates was found in an experimental model of liver extramedullary hematopoiesis. Flow cytometry studies showed that LSEC express the CXCR4 receptor. Functional assays showed that activation of this receptor by SDF-1 stimulated the migration of LSEC and increased the expression of PECAM-1. Our findings suggest that LSEC through the production of SDF-1 may constitute a fundamental niche for regulation of HSC migration to the liver. To our knowledge, this is the first report showing that LSEC not only express and secrete SDF-1, but also its receptor CXCR4.
Collapse
Affiliation(s)
- Mayela Mendt
- Unidad de Terapia Celular-Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas-IVIC, Apartado, Caracas, Venezuela
| | | |
Collapse
|
31
|
Wise JK, Sumner DR, Virdi AS. Modulation of stromal cell-derived factor-1/CXC chemokine receptor 4 axis enhances rhBMP-2-induced ectopic bone formation. Tissue Eng Part A 2012; 18:860-9. [PMID: 22035136 DOI: 10.1089/ten.tea.2011.0187] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Enhancement of in vivo mobilization and homing of endogenous mesenchymal stem cells (MSCs) to an injury site is an innovative strategy for improvement of bone tissue engineering and repair. The present study was designed to determine whether mobilization by AMD3100 and/or local homing by delivery of stromal cell-derived factor-1 (SDF-1) enhances recombinant human bone morphogenetic protein-2 (rhBMP-2) induced ectopic bone formation in an established rat model. Rats received an injection of either saline or AMD3100 treatment 1 h before harvesting of bone marrow for in vitro colony-forming unit-fibroblasts (CFU-F) culture or the in vivo subcutaneous implantation of absorbable collagen sponges (ACSs) loaded with saline, recombinant human bone morphogenetic protein-2 (rhBMP-2), SDF-1, or the combination of SDF-1 and rhBMP-2. AMD3100 treatment resulted in a significant decrease in CFU-F number, compared with saline, which confirmed that a single systemic AMD3100 treatment rapidly mobilized MSCs from the bone marrow. At 28 and 56 days, bone formation in the explanted ACS was assessed by microcomputed tomography (μCT) and histology. At 28 days, AMD3100 and/or SDF-1 had no statistically significant effect on bone volume (BV) or bone mineral content (BMC), but histology revealed more active bone formation with treatment of AMD3100, loading of SDF-1, or the combination of both AMD3100 and SDF-1, compared with saline-treated rhBMP-2 loaded ACS. At 56 days, the addition of AMD3100 treatment, loading of SDF-1, or the combination of both resulted in a statistically significant stimulatory effect on BV and BMC, compared with the saline-treated rhBMP-2 loaded ACS. Histology of the 56-day ACS were consistent with the μCT analysis, exhibiting more mature and mineralized bone formation with AMD3100 treatment, SDF-1 loading, or the combination of both, compared with the saline-treated rhBMP-2 loaded ACS. The present study is the first that provides evidence of the efficacy of AMD3100 and SDF-1 treatment to stimulate trafficking of MSCs to an ectopic implant site, in order to ultimately enhance rhBMP-2 induced long-term bone formation.
Collapse
Affiliation(s)
- Joel K Wise
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | |
Collapse
|
32
|
Abstract
Hematopoietic stem and progenitor mobilization has revolutionized the field of hematopoietic transplantation. Currently, hematopoietic grafts acquired from the peripheral blood of patients or donors treated with granulocyte-colony stimulating factor (G-CSF) are the preferred source for transplantation. G-CSF mobilization regimens, however, are associated with known morbidities and a significant number of normal donors and patient populations fail to mobilize sufficient numbers of hematopoietic stem and progenitor cells for transplantation, necessitating the need for non-G-CSF mobilization strategies. Mechanistic studies evaluating hematopoietic bone marrow niche interactions have uncovered novel agents with the capacity for hematopoietic mobilization. This chapter provides a comprehensive overview of mobilizing agents, other than G-CSF, and experimental procedures and technical aspects important to evaluate and define their hematopoietic mobilizing activities alone and in combination.
Collapse
|
33
|
Abstract
Current approaches aiming to cure type 1 diabetes (T1D) have made a negligible number of patients insulin-independent. In this review, we revisit the role of stem cell (SC)-based applications in curing T1D. The optimal therapeutic approach for T1D should ideally preserve the remaining β-cells, restore β-cell function, and protect the replaced insulin-producing cells from autoimmunity. SCs possess immunological and regenerative properties that could be harnessed to improve the treatment of T1D; indeed, SCs may reestablish peripheral tolerance toward β-cells through reshaping of the immune response and inhibition of autoreactive T-cell function. Furthermore, SC-derived insulin-producing cells are capable of engrafting and reversing hyperglycemia in mice. Bone marrow mesenchymal SCs display a hypoimmunogenic phenotype as well as a broad range of immunomodulatory capabilities, they have been shown to cure newly diabetic nonobese diabetic (NOD) mice, and they are currently undergoing evaluation in two clinical trials. Cord blood SCs have been shown to facilitate the generation of regulatory T cells, thereby reverting hyperglycemia in NOD mice. T1D patients treated with cord blood SCs also did not show any adverse reaction in the absence of major effects on glycometabolic control. Although hematopoietic SCs rarely revert hyperglycemia in NOD mice, they exhibit profound immunomodulatory properties in humans; newly hyperglycemic T1D patients have been successfully reverted to normoglycemia with autologous nonmyeloablative hematopoietic SC transplantation. Finally, embryonic SCs also offer exciting prospects because they are able to generate glucose-responsive insulin-producing cells. Easy enthusiasm should be mitigated mainly because of the potential oncogenicity of SCs.
Collapse
Affiliation(s)
- Paolo Fiorina
- Transplantation Research Center, Division of Nephrology, Children's Hospital/Harvard Medical School, 221 Longwood Avenue, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
34
|
Schwaiger E, Klaus C, Matheeussen V, Baranyi U, Pilat N, Ramsey H, Korom S, De Meester I, Wekerle T. Dipeptidyl peptidase IV (DPPIV/CD26) inhibition does not improve engraftment of unfractionated syngeneic or allogeneic bone marrow after nonmyeloablative conditioning. Exp Hematol 2011; 40:97-106. [PMID: 22085453 PMCID: PMC3265670 DOI: 10.1016/j.exphem.2011.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/26/2011] [Accepted: 10/31/2011] [Indexed: 12/30/2022]
Abstract
In order to develop minimally toxic bone marrow transplantation (BMT) protocols suitable for use in a wider range of indications, it is important to identify ways to enhance BM engraftment at a given level of recipient conditioning. CXCL12/stromal cell-derived factor-1α plays a crucial physiological role in homing of hematopoietic stem cells to BM. It is regulated by the ectopeptidase dipeptidyl peptidase IV (DPPIV; DPP4) known as CD26, which cleaves dipeptides from the N-terminus of polypeptide chains. Blocking DPPIV enzymatic activity had a beneficial effect on hematopoietic stem cell engraftment in various but very specific experimental settings. Here we investigated whether inhibition of DPPIV enzymatic activity through Diprotin A or sitagliptin (Januvia) improves BM engraftment in nonmyeloablative murine models of syngeneic (i.e., CD45-congenic) and allogeneic (i.e., Balb/c to B6) BMT (1 Gy total body irradiation, 10–15 × 106 unseparated BM cells/mouse). Neither Diprotin A administered in vivo at the time of BMT and/or used for in vitro pretreatment of BM nor sitagliptin administered in vivo had a detectable effect on the level of multilineage chimerism (follow-up >20 weeks). Similarly, sitagliptin did not enhance chimerism after allogeneic BMT, even though DPPIV enzymatic activity measured in serum was profoundly inhibited (>98% inhibition at peak exposure). Our results provide evidence that DPPIV inhibition via Diprotin A or sitagliptin does not improve engraftment of unseparated BM in a nonmyeloablative BMT setting.
Collapse
Affiliation(s)
- Elisabeth Schwaiger
- Division of Transplantation, Department of Surgery, Vienna General Hospital, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
La mobilisation des progéniteurs hématopoïétiques : nouvelles cibles et nouvelles modalités thérapeutiques. Bull Cancer 2011; 98:951-61. [DOI: 10.1684/bdc.2011.1405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
36
|
Tong Y, Xu W, Han H, Chen Y, Yang J, Qiao H, Hong D, Wu Y, Zhou C. Tanshinone IIA increases recruitment of bone marrow mesenchymal stem cells to infarct region via up-regulating stromal cell-derived factor-1/CXC chemokine receptor 4 axis in a myocardial ischemia model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2011; 18:443-450. [PMID: 21146968 DOI: 10.1016/j.phymed.2010.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Revised: 09/07/2010] [Accepted: 10/12/2010] [Indexed: 05/30/2023]
Abstract
Systemic administration with bone marrow mesenchymal stem cells (BMSCs) is a promising approach to cure myocardial ischemia (MI), while the efficacy of cell transplantation is limited by the low engraftment of BMSCs. Tanshinone IIA (Tan IIA) has been reported many times for the treatment of MI. Therefore, the present study was performed to investigate whether Tan IIA could increase the migration of BMSCs to ischemic region and its potential mechanisms. In our study, we found that combination treatment with Tan IIA and BMSCs significantly alleviated the infarct size when compared with control group (31.46 ± 3.00% vs. 46.95 ± 6.51%, p<0.05). Results of real-time PCR showed that Tanshinone IIA (Tan IIA) did increase the migration of BMSCs to ischemic region in vivo, which was correlated with cardiac function recovery after MI. Furthermore, 2 μM Tan IIA could enhance the migration capability of BMSCs in vitro (3.69-fold of control), and this enhancement could be blocked by AMD3100 (a CXC chemokine receptor 4 blocker). CXCR4, together with its specific receptor, stromal cell-derived factor-1 (SDF-1) plays a critical role in the stem cell recruitment. Our experiment indicated that Tan IIA could promote SDF-1α expression in the infarct area and enhance the CXCR4 expression of BMSCs in vitro. Therefore, we postulated that Tan IIA could increase the BMSCs migration via up-regulating SDF1/CXCR4 axis.
Collapse
Affiliation(s)
- Yinghui Tong
- College of Pharmaceutical Sciences, Zhejiang University, No. 358 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Once considered biological waste, umbilical cord blood (UCB) has become an accepted source of haematopoietic stem cells (HSCs). With initial success in the pediatric setting, UCB transplantation continues to gain favor in the adult patient population. Novel approaches to UCB transplantation include use of two units and a variety of graft manipulations. Additional uses for UCB are currently being explored and include applications in regenerative medicine and immunotherapy.
Collapse
Affiliation(s)
- D H McKenna
- Department of Laboratory Medicine and Pathology, Division of Transfusion Medicine, University of Minnesota, Saint Paul, MN 55108, USA.
| | | |
Collapse
|
38
|
Hoggatt J, Pelus LM. Mobilization of hematopoietic stem cells from the bone marrow niche to the blood compartment. Stem Cell Res Ther 2011; 2:13. [PMID: 21418553 PMCID: PMC3226284 DOI: 10.1186/scrt54] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The vast majority of hematopoietic stem cells (HSCs) reside in specialized niches within the bone marrow during steady state, maintaining lifelong blood cell production. A small number of HSCs normally traffic throughout the body; however, exogenous stimuli can enhance their release from the niche and entry into the peripheral circulation. This process, termed mobilization, has become the primary means to acquire a stem cell graft for hematopoietic transplant at most transplant centers. Currently, the preferred method of HSC mobilization for subsequent transplantation is treatment of the donor with granulocyte colony-stimulating factor. The mobilizing effect of granulocyte colony-stimulating factor is not completely understood, but recent studies suggest that its capacity to mobilize HSCs, at least in part, is a consequence of alterations to the hematopoietic niche. The present article reviews some of the key mechanisms mediating HSC mobilization, highlighting recent advances and controversies in the field.
Collapse
Affiliation(s)
- Jonathan Hoggatt
- Department of Microbiology & Immunology, Indiana University School of Medicine, 950 West Walnut Street, R2-302, Indianapolis, IN 46202, USA
| | | |
Collapse
|
39
|
Christopher MJ, Rao M, Liu F, Woloszynek JR, Link DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. ACTA ACUST UNITED AC 2011; 208:251-60. [PMID: 21282380 PMCID: PMC3039862 DOI: 10.1084/jem.20101700] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF), the prototypical mobilizing cytokine, induces hematopoietic stem and progenitor cell (HSPC) mobilization from the bone marrow in a cell-nonautonomous fashion. This process is mediated, in part, through suppression of osteoblasts and disruption of CXCR4/CXCL12 signaling. The cellular targets of G-CSF that initiate the mobilization cascade have not been identified. We use mixed G-CSF receptor (G-CSFR)-deficient bone marrow chimeras to show that G-CSF-induced mobilization of HSPCs correlates poorly with the number of wild-type neutrophils. We generated transgenic mice in which expression of the G-CSFR is restricted to cells of the monocytic lineage. G-CSF-induced HSPC mobilization, osteoblast suppression, and inhibition of CXCL12 expression in the bone marrow of these transgenic mice are intact, demonstrating that G-CSFR signals in monocytic cells are sufficient to induce HSPC mobilization. Moreover, G-CSF treatment of wild-type mice is associated with marked loss of monocytic cells in the bone marrow. Finally, we show that bone marrow macrophages produce factors that support the growth and/or survival of osteoblasts in vitro. Together, these data suggest a model in which G-CSFR signals in bone marrow monocytic cells inhibit the production of trophic factors required for osteoblast lineage cell maintenance, ultimately leading to HSPC mobilization.
Collapse
Affiliation(s)
- Matthew J Christopher
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
40
|
Abstract
The regulated migration of stem cells is critical for organogenesis during development and for tissue -homeostasis and repair during adulthood. Human bone marrow (BM) represents an accessible reservoir containing regenerative cell types from hematopoietic, endothelial, and mesenchymal-stromal lineages that together coordinate hematopoiesis and promote the repair of damaged vasculature and tissues throughout the body. Thus, a detailed understanding of lineage-specific stem cell mobilization, homing, and subsequent engraftment in areas of injury or disease is of critical importance to the rational development of novel cell-mediated regenerative therapies. Stem cell trafficking via the circulation from site of origin to peripheral tissues requires fundamental molecular pathways governing (1) niche-specific deadhesion of progenitor cells; (2) chemoattraction to guide progenitor cell homing; and (3) interstitial navigation and adhesion/retention of recruited progenitor cells. This overview chapter summarizes the diversity of migratory strategies employed by hematopoietic, endothelial, and mesenchymal-stromal progenitor cells during repair and regeneration after tissue damage. Further elucidation of stem cell homing and migration pathways will allow greater application of stem cells for targeted cell therapy and/or drug delivery for tissue repair. Strikingly similar migratory mechanisms appear to govern the in vivo migration of recently characterized cancer stem cells (CSC) in leukemias and solid tumors, indicating that conserved principles of stem cell migration and niche specificity will provide new information to target CSC in anticancer therapy.
Collapse
|
41
|
Fiorina P, Pietramaggiori G, Scherer SS, Jurewicz M, Mathews JC, Vergani A, Thomas G, Orsenigo E, Staudacher C, La Rosa S, Capella C, Carothers A, Zerwes HG, Luzi L, Abdi R, Orgill DP. The Mobilization and Effect of Endogenous Bone Marrow Progenitor Cells in Diabetic Wound Healing. Cell Transplant 2010; 19:1369-81. [DOI: 10.3727/096368910x514288] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Diabetic patients suffer from impaired wound healing, characterized by only modest angiogenesis and cell proliferation. Stem cells may stimulate healing, but little is known about the kinetics of mobilization and function of bone marrow progenitor cells (BM-PCs) during diabetic wound repair. The objective of this study was to investigate the kinetics of BM-PC mobilization and their role during early diabetic wound repair in diabetic db/db mice. After wounding, circulating hematopoietic stem cells (Lin-c-Kit+Sca-1+) stably increased in the periphery and lymphoid tissue of db/db mice compared to unwounded controls. Peripheral endothelial progenitor cells (CD34+VEGFR+) were 2.5- and 3.5-fold increased on days 6 and 10 after wounding, respectively. Targeting the CXCR4—CXCL12 axis induced an increased release and engraftment of endogenous BM-PCs that was paralleled by an increased expression of CXCL12/SDF-1α in the wounds. Increased levels of peripheral and engrafted BM-PCs corresponded to stimulated angiogenesis and cell proliferation, while the addition of an agonist (GM-CSF) or an antagonist (ACK2) did not further modulate wound healing. Macroscopic histological correlations showed that increased levels of stem cells corresponded to higher levels of wound reepithelialization. After wounding, a natural release of endogenous BM-PCs was shown in diabetic mice, but only low levels of these cells homed in the healing tissue. Higher levels of CXCL12/SDF-1α and circulating stem cells were required to enhance their engraftment and biological effects. Despite controversial data about the functional impairment of diabetic BM-PCs, in this model our data showed a residual capacity of these cells to trigger angiogenesis and cell proliferation.
Collapse
Affiliation(s)
- Paolo Fiorina
- Transplantation Research Center, Division of Nephrology, Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine & Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Giorgio Pietramaggiori
- Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Surgery, University of Geneva, Geneva, Switzerland
| | - Saja S. Scherer
- Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Surgery, University of Geneva, Geneva, Switzerland
| | - Mollie Jurewicz
- Transplantation Research Center, Division of Nephrology, Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmine C. Mathews
- Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea Vergani
- Transplantation Research Center, Division of Nephrology, Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine & Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Gebhard Thomas
- Autoimmunity, Transplantation and Inflammation and Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Elena Orsenigo
- Department of Medicine & Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Carlo Staudacher
- Department of Medicine & Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Stefano La Rosa
- Department of Pathology, Ospedale di Circolo and Department of Human Morphology, University of Insubria, Varese, Italy
| | - Carlo Capella
- Department of Pathology, Ospedale di Circolo and Department of Human Morphology, University of Insubria, Varese, Italy
| | - Adelaide Carothers
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hans-Gunter Zerwes
- Autoimmunity, Transplantation and Inflammation and Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Livio Luzi
- Facolta di Scienze Motorie, Università di Milano, Milan, Italy
| | - Reza Abdi
- Transplantation Research Center, Division of Nephrology, Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dennis P. Orgill
- Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Kang Y, Chen BJ, DeOliveira D, Mito J, Chao NJ. Selective enhancement of donor hematopoietic cell engraftment by the CXCR4 antagonist AMD3100 in a mouse transplantation model. PLoS One 2010; 5:e11316. [PMID: 20596257 PMCID: PMC2893128 DOI: 10.1371/journal.pone.0011316] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 05/27/2010] [Indexed: 11/19/2022] Open
Abstract
The interaction between stromal cell-derived factor-1 (SDF-1) with CXCR4 chemokine receptors plays an important role in hematopoiesis following hematopoietic stem cell transplantation. We examined the efficacy of post transplant administration of a specific CXCR4 antagonist (AMD3100) in improving animal survival and in enhancing donor hematopoietic cell engraftment using a congeneic mouse transplantation model. AMD3100 was administered subcutaneously at 5 mg/kg body weight 3 times a week beginning at day +2 post-transplant. Post-transplant administration of AMD3100 significantly improves animal survival. AMD3100 reduces pro-inflammatory cytokine/chemokine production. Furthermore, post transplant administration of AMD3100 selectively enhances donor cell engraftment and promotes recovery of all donor cell lineages (myeloid cells, T and B lymphocytes, erythrocytes and platelets). This enhancement results from a combined effect of increased marrow niche availability and greater cell division induced by AMD3100. Our studies shed new lights into the biological roles of SDF-1/CXCR4 interaction in hematopoietic stem cell engraftment following transplantation and in transplant-related mortality. Our results indicate that AMD3100 provides a novel approach for enhancing hematological recovery following transplantation, and will likely benefit patients undergoing transplantation.
Collapse
Affiliation(s)
- Yubin Kang
- Divisions of Hematology, Oncology and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Benny J. Chen
- Division of Cellular Therapy/Adult Bone Marrow Transplantation, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Divino DeOliveira
- Division of Cellular Therapy/Adult Bone Marrow Transplantation, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Jeffrey Mito
- Division of Cellular Therapy/Adult Bone Marrow Transplantation, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Nelson J. Chao
- Division of Cellular Therapy/Adult Bone Marrow Transplantation, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
43
|
Brunstein CG, Laughlin MJ. Extending cord blood transplant to adults: dealing with problems and results overall. Semin Hematol 2010; 47:86-96. [PMID: 20109616 DOI: 10.1053/j.seminhematol.2009.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The development of newer strategies to overcome, in particular, the cell dose limitation, has increased the availability of umbilical cord blood (UCB) as a source of hematopoietic stem cells (HSC) for transplantation of adults. Among these strategies is the development of the double UCB, ex vivo, and reduced-intensity transplantation platforms. Several ongoing registry-based and single-institution and multicenter clinical trials are investigating ways to make UCB transplantation safer and to improve the outcomes of adults after UCB transplantation. We review the background data and promising newer strategies that will further expand the utilization of UCB for the treatment of adults.
Collapse
Affiliation(s)
- Claudio G Brunstein
- Blood and Marrow Transplant Program, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
44
|
Ramirez PA, Wagner JE, Brunstein CG. Going straight to the point: intra-BM injection of hematopoietic progenitors. Bone Marrow Transplant 2010; 45:1127-33. [DOI: 10.1038/bmt.2010.39] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
45
|
Microarray analysis of retinal endothelial tip cells identifies CXCR4 as a mediator of tip cell morphology and branching. Blood 2010; 115:5102-10. [PMID: 20154215 DOI: 10.1182/blood-2009-07-230284] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The development of the vertebrate vascular system is mediated by both genetic patterning of vessels and by angiogenic sprouting in response to hypoxia. Both of these processes depend on the detection of environmental guidance cues by endothelial cells. A specialized subtype of endothelial cell known as the tip cell is thought to be involved in the detection and response to these cues, but the molecular signaling pathways used by tip cells to mediate tissue vascularization remain largely uncharacterized. To identify genes critical to tip cell function, we have developed a method to isolate them using laser capture microdissection, permitting comparison of RNA extracted from endothelial tip cells with that of endothelial stalk cells using microarray analysis. Genes enriched in tip cells include ESM-1, angiopoietin-2, and SLP-76. CXCR4, a receptor for the chemokine stromal-cell derived factor-1, was also identified as a tip cell-enriched gene, and we provide evidence for a novel role for this receptor in mediating tip cell morphology and vascular patterning in the neonatal retina.
Collapse
|
46
|
Brunstein CG, Weisdorf DJ. Future of cord blood for oncology uses. Bone Marrow Transplant 2009; 44:699-707. [DOI: 10.1038/bmt.2009.286] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
47
|
Zhang Z, Zhong W, Hall MJ, Kurre P, Spencer D, Skinner A, O'Neill S, Xia Z, Rosenbaum JT. CXCR4 but not CXCR7 is mainly implicated in ocular leukocyte trafficking during ovalbumin-induced acute uveitis. Exp Eye Res 2009; 89:522-31. [PMID: 19524567 PMCID: PMC2745349 DOI: 10.1016/j.exer.2009.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 05/04/2009] [Accepted: 05/20/2009] [Indexed: 01/13/2023]
Abstract
Uveitis is an inflammatory ocular disease characterized by the infiltration of T lymphocytes and other leukocytes into the eye. The recruitment of these inflammatory cells from systemic vasculature to ocular tissue is a well-coordinated multistep process including rolling, firm adhesion and transmigration. CXCL12 (SDF-1alpha) is an endothelial cell-derived cytokine interacting with CXCR4 and CXCR7, two chemokine receptors mainly expressed in T cells, neutrophils and monocytes. Recent studies have shown that CXCR4, CXCR7 and their ligand, CXCL12, are important for the regulation of leukocyte mobilization and trafficking. However, it is unclear whether these two chemokine receptors are implicated in the pathogenesis of uveitis. In this study, we used DO11.10 mice, whose CD4+ T cells are genetically engineered to react with ovalbumin (OVA), to investigate the role of CXCR4 and CXCR7 in an animal model of uveitis. Intravital microscopy revealed that intravitreal OVA challenge of DO11.10 mice caused the infiltration of both T cells and neutrophils. The invasion of these inflammatory cells coincided with the detection of transcriptional up-regulation of CXCR4 and CXCR7 in the eye. In addition, both real-time-PCR and immunohistochemistry revealed an enhanced expression of endothelial CXCL12. Furthermore, intraperitoneal injection of AMD3100 (a specific CXCR4 antagonist) significantly attenuated OVA-induced uveitis and CXCL12-mediated transwell migration. In contrast, intraperitoneal administration of CXCR7 neutralizing antibody did not significantly alter ocular infiltration of inflammatory cells caused by OVA challenge. Our data suggest that CXCR4 but not CXCR7 plays a critical role in antigen-induced ocular inflammation by facilitating leukocyte infiltration. This study not only enhances our knowledge of the immunopathological mechanism of uveitis but also provides a novel rationale to target CXCR4 as an anti-inflammatory strategy to treat uveitis.
Collapse
Affiliation(s)
- Zili Zhang
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Wenwei Zhong
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Mark J. Hall
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Peter Kurre
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Doran Spencer
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Amy Skinner
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Stacey O'Neill
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Zhenwei Xia
- Department of Pediatrics, RuiJin Hospital, Shanghai, China
| | - James T. Rosenbaum
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
48
|
Xu Q, Yuan X, Xu M, McLafferty F, Hu J, Lee BS, Liu G, Zeng Z, Black KL, Yu JS. Chemokine CXC receptor 4--mediated glioma tumor tracking by bone marrow--derived neural progenitor/stem cells. Mol Cancer Ther 2009; 8:2746-53. [PMID: 19723878 DOI: 10.1158/1535-7163.mct-09-0273] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Malignant gliomas manifest frequent tumor recurrence after surgical resection and/or other treatment because of their nature of invasiveness and dissemination. The recognized brain tumor-tracking property of neural progenitor/stem cells opened the possibility of targeting malignant brain tumors using neural progenitor/stem cells. We and others have previously shown that fetal neural progenitor/stem cells can be used to deliver therapeutic molecules to brain tumors. Our recent work has further shown that gene delivery by bone marrow-derived neural progenitor/stem cells achieves therapeutic effects in a glioma model. In this study, we isolate and characterize bone marrow-derived neural progenitor/stem cells, which also express the chemokine receptor chemokine CXC receptor 4 (CXCR4). We show that CXCR4 is required for their chemotaxis and extracellular matrix invasion against a gradient of glioma soluble factors. Furthermore, beta-galactosidase-labeled bone marrow-derived neural progenitor/stem cells implanted in the contralateral side of the brain were shown to track gliomas as early as day 1 and increased through days 3 and 7. Intracranial glioma tracking by bone marrow-derived neural progenitor/stem cells is significantly inhibited by preincubation of bone marrow-derived neural progenitor/stem cells with a blocking anti-CXCR4 antibody, suggesting a CXCR4-dependent tracking mechanism. Glioma tracking bone marrow-derived neural progenitor/stem cells were found to express progenitor/stem cell markers, as well as CXCR4. Although bromodeoxyuridine incorporation assays and proliferating antigen staining indicated that tumor tracking bone marrow-derived neural progenitor/stem cells were mostly nonproliferating, these cells survive in the local tumor environment with little apoptosis. Elucidating the molecular mechanism of brain tumor tracking by adult source stem cells may provide basis for the development of future targeted therapy for malignant brain tumors.
Collapse
Affiliation(s)
- Qijin Xu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Frerker N, Raber K, Bode F, Skripuletz T, Nave H, Klemann C, Pabst R, Stephan M, Schade J, Brabant G, Wedekind D, Jacobs R, Jörns A, Forssmann U, Straub RH, Johannes S, Hoffmann T, Wagner L, Demuth HU, von Hörsten S. Phenotyping of congenic dipeptidyl peptidase 4 (DP4) deficient Dark Agouti (DA) rats suggests involvement of DP4 in neuro-, endocrine, and immune functions. Clin Chem Lab Med 2009; 47:275-87. [PMID: 19327106 DOI: 10.1515/cclm.2009.064] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Treatment of diabetes type 2 using chronic pharmacological inhibition of dipeptidyl peptidase 4 (DP4) still requires an in-depth analysis of models for chronic DP4 deficiency, because adverse reactions induced by some DP4 inhibitors have been described. METHODS In the present study, a novel congenic rat model of DP4 deficiency on a "DP4-high" DA rat genetic background was generated (DA.F344-Dpp4(m)/ SvH rats) and comprehensively phenotyped. RESULTS Similar to chronic pharmacological inhibition of DP4, DP4 deficient rats exhibited a phenotype involving reduced diet-induced body weight gain and improved glucose tolerance associated with increased levels of glucagon-like peptide-1 (GLP-1) and bound leptin as well as decreased aminotransferases and triglycerides. Additionally, DA.F344-Dpp4(m)/SvH rats showed anxiolytic-like and reduced stress-like responses, a phenomenon presently not targeted by DP4 inhibitors. However, several immune alterations, such as differential leukocyte subset composition at baseline, blunted natural killer cell and T-cell functions, and altered cytokine levels were observed. CONCLUSIONS While this animal model confirms a critical role of DP4 in GLP-1-dependent glucose regulation, genetically induced chronic DP4 deficiency apparently also affects stress-regulatory and immuneregulatory systems, indicating that the use of chronic DP4 inhibitors might have the potential to interfere with central nervous system and immune functions in vivo.
Collapse
Affiliation(s)
- Nadine Frerker
- Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Wyss BK, Donnelly AFW, Zhou D, Sinn AL, Pollok KE, Goebel WS. Enhanced homing and engraftment of fresh but not ex vivo cultured murine marrow cells in submyeloablated hosts following CD26 inhibition by Diprotin A. Exp Hematol 2009; 37:814-23. [PMID: 19540435 PMCID: PMC2700780 DOI: 10.1016/j.exphem.2009.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 03/13/2009] [Accepted: 03/16/2009] [Indexed: 11/17/2022]
Abstract
OBJECTIVE We recently reported that murine marrow cultured ex vivo for gamma-retrovirus transduction engrafts approximately 10-fold less well than fresh marrow upon transplantation into submyeloablated hosts. Here, we evaluated homing efficiency as a potential mechanism for this engraftment disparity, and whether CD26 inhibition with the tripeptide Diprotin A (DipA) would enhance engraftment of ex vivo cultured cells in submyeloablated hosts. MATERIALS AND METHODS Homing and engraftment of fresh and ex vivo cultured lineage-negative (lin(-)) marrow cells in submyeloablated congenic hosts with and without DipA treatment was evaluated. Expression of CXCR4 and CD26 on fresh and cultured lin(-) marrow cells was compared. RESULTS Homing of lin(-) cells cultured for gamma-retrovirus transduction was at least threefold less than that of fresh lin(-) cells 20 hours after transplantation into submyeloablated hosts. DipA treatment of fresh lin(-) cells resulted in at least twofold increased homing and engraftment in submyeloablated hosts. DipA treatment, however, did not significantly improve homing or engraftment of cells undergoing a 3-day culture protocol for gamma-retrovirus transduction in submyeloablated hosts. CXCR4 expression on lin(-) cells was significantly decreased following 3 days of culture; CXCR4 expression was not significantly altered following overnight culture. CONCLUSIONS Ex vivo culture of lin(-) cells for gamma-retroviral transduction downregulates CXCR4 expression and markedly impairs homing and engraftment of murine lin(-) marrow in submyeloablated hosts. While inhibition of CD26 activity with DipA increases homing and engraftment of fresh lin(-) cells, DipA treatment does not improve homing and engraftment of cultured lin(-) marrow cells in submyeloablated congenic hosts.
Collapse
Affiliation(s)
- Brandon K. Wyss
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abigail F. W. Donnelly
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dan Zhou
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anthony L. Sinn
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Karen E. Pollok
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Section of Pediatric Hematology/Oncology, James Whitcomb Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - W. Scott Goebel
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Section of Pediatric Hematology/Oncology, James Whitcomb Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|