1
|
Kandekar S, Punatar S, Khattry N, Gokarn A, Jindal N, Mirgh S, Chichra A, Tembhare P, Rane P, Gawde J, Mathew L, Patil A, Chiplunkar S, Kode J. Low levels of CD26 on certain cellular subtypes of donor harvest is associated with better clinical outcomes post allogeneic stem cell transplantation through regulation of NF-κB pathway and pro-inflammatory cytokines. Int Immunopharmacol 2023; 125:111054. [PMID: 37890379 DOI: 10.1016/j.intimp.2023.111054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND We had previously reported significant association of immunoectoenzyme CD26 expression on donor harvest with acute Graft-versus-Host-Disease (aGVHD) in allogeneic stem cell transplantation (ASCT) patients. The current study was aimed at analysing CD26 signaling pathway molecules and understanding their impact on immune reconstitution and clinical outcomes post-ASCT. SUBJECTS AND METHODOLOGY The study cohort included 26 transplant donors/patients who underwent reduced intensity (n = 21), myeloablative (n = 4) and non-myeloablative (n = 1) ASCT for hematological malignancies. Donors were matched related donors (n = 19) and haploidentical donors (n = 7). Surface expression of CD26, CD73 and ADA, and various immune cell subtypes were assessed by multicolour-flow cytometry. Soluble CD26 (sCD26) and cytokine levels were measured in plasma samples by ELISA and Multiplex Luminex assay, respectively. Immune cells from healthy individuals were stimulated with phytohemagglutinin (PHA) in the presence or absence of CD26 inhibitor. Effect of CD26 inhibition on NF-κB localization in PHA stimulated cells was analysed by immunofluorescence and confocal microscopy. Pro-inflammatory cytokines from the culture supernatants were detected with Cytometric bead array flow cytometry. Association of all measured markers with clinical outcomes was evaluated using appropriate statistical tests. RESULTS CD26 surface expression on PBSC donor harvest cells showed increased risk of chronic GVHD (cGVHD, p = 0.055). Amongst the various immune cell subtypes, decreased B cells in harvest showed significant association with aGVHD (p = 0.022) whereas increased myeloid dendritic cells and CD3+T cells at Day100 in peripheral blood of transplant recipients correlated with cGVHD (p = 0.046) and aGVHD (p = 0.035), respectively. Further, high sCD26 in transplant recipients at Day100 exhibited association with reduced event-free survival (EFS) (p = 0.011). Higher CD26 expression on more & less mature NK cells, naïve & post-switched memory B cells and Treg cells in the donor harvest (p < 0.05) led to lower EFS in transplant recipients. Mechanistically, CD26 inhibitor caused dose-dependent reduction in CD26 enzyme activity and in pro-inflammatory cytokine production in post mitogen-stimulated T cell cultures. CONCLUSION Our study has implicated that lower CD26 expression on immune cell subtypes of the donor stem cell harvest is associated with reduced risk of GVHD and better survival. The underlying mechanism was found to be through NF-κB pathway and pro-inflammatory cytokines. Based on these observations, chemically designed or natural resources-based CD26 inhibitors can be explored further in clinical trials for improving ASCT outcomes.
Collapse
Affiliation(s)
- Shruti Kandekar
- Kode Lab, Tumor Immunology & Immunotherapy Group, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Sachin Punatar
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Navin Khattry
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Anant Gokarn
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Nishant Jindal
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Sumeet Mirgh
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Akanksha Chichra
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Prashant Tembhare
- Hematopathology Lab, Clinical Research Centre, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Pallavi Rane
- Clinical Research Secretariat, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India
| | - Jitendra Gawde
- Clinical Research Secretariat, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India
| | - Libin Mathew
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India
| | - Anand Patil
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Shubhada Chiplunkar
- Chiplunkar Lab, Tumor Immunology & Immunotherapy Group, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Jyoti Kode
- Kode Lab, Tumor Immunology & Immunotherapy Group, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India.
| |
Collapse
|
2
|
Guo B, Huang X, Chen Y, Broxmeyer HE. Ex Vivo Expansion and Homing of Human Cord Blood Hematopoietic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1442:85-104. [PMID: 38228960 DOI: 10.1007/978-981-99-7471-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Cord blood (CB) has been proven to be an alternative source of haematopoietic stem cells (HSCs) for clinical transplantation and has multiple advantages, including but not limited to greater HLA compatibility, lower incidence of graft-versus-host disease (GvHD), higher survival rates and lower relapse rates among patients with minimal residual disease. However, the limited number of HSCs in a single CB unit limits the wider use of CB in clinical treatment. Many efforts have been made to enhance the efficacy of CB HSC transplantation, particularly by ex vivo expansion or enhancing the homing efficiency of HSCs. In this chapter, we will document the major advances regarding human HSC ex vivo expansion and homing and will also discuss the possibility of clinical translation of such laboratory work.
Collapse
Affiliation(s)
- Bin Guo
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xinxin Huang
- Xuhui Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Yandan Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, School of Medicine, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Cooper SH, Capitano ML, Broxmeyer HE. Experimental Models of Mouse and Human Hematopoietic Stem Cell Transplantation. Methods Mol Biol 2023; 2567:205-232. [PMID: 36255704 DOI: 10.1007/978-1-0716-2679-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Experimental hematopoietic stem cell transplantation (HSCT) is an invaluable tool in determining the function and characteristics of hematopoietic stem cells (HSC) from experimental mouse and human donor groups. These groups could include, but are not limited to, genetically altered populations (gene knockout/knockin models), ex vivo manipulated cell populations, or in vivo modulated cell populations. The basic fundamentals of this process involve taking cells from a mouse/human donor source and putting them into another mouse (recipient) after preconditioning of the recipient with either total body irradiation (TBI) for mouse donor cells or into sublethally irradiated immune-deficient mice for human donor cells. Then, at pre-determined time points post-transplant, sampling a small amount of peripheral blood (PB) and at the termination of the evalaution, bone marrow (BM) to determine donor contribution and function by phenotypic analysis. Exploiting the congenic mouse strains of C57BL/6 (CD45.1- CD45.2+), BoyJ (CD45.1+ CD45.2-), and their F1-crossed hybrid C57BL/6 × BoyJ (CD45.1+ CD45.2+), we are able to quantify donor, competitor, and recipient mouse cell contributions to the engraftment state. Human donor cell engraftment (e.g., from the cord blood [CB], mobilized PB, or BM) is assessed by human cell phenotyping in sublethally irradiated immune-deficient mouse recipients (e.g., NOD scid gamma mice that are deficient in B cells, T cells, and natural killer cells and have defective dendritic cells and macrophages). Engraftment of cells from primary mouse recipients into secondary mice allows for an estimation of the self-renewal capacity of the original donor HSC. This chapter outlines concepts, methods, and techniques for mouse and human cell models of HSCT and for assessment of donor cells collected and processed in hypoxia versus ambient air.
Collapse
Affiliation(s)
- Scott H Cooper
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Maegan L Capitano
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hal E Broxmeyer
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
4
|
Felker S, Shrestha A, Bailey J, Pillis DM, Siniard D, Malik P. Differential CXCR4 expression on hematopoietic progenitor cells versus stem cells directs homing and engraftment. JCI Insight 2022; 7:151847. [PMID: 35531956 PMCID: PMC9090236 DOI: 10.1172/jci.insight.151847] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Gene therapy involves a substantial loss of hematopoietic stem and progenitor cells (HSPC) during processing and homing. Intra-BM (i.b.m.) transplantation can reduce homing losses, but prior studies have not yielded promising results. We studied the mechanisms involved in homing and engraftment of i.b.m. transplanted and i.v. transplanted genetically modified (GM) human HSPC. We found that i.b.m. HSPC transplantation improved engraftment of hematopoietic progenitor cells (HPC) but not of long-term repopulating hematopoietic stem cells (HSC). Mechanistically, HPC expressed higher functional levels of CXCR4 than HSC, conferring them a retention and homing advantage when transplanted i.b.m. Removing HPC and transplanting an HSC-enriched population i.b.m. significantly increased long-term engraftment over i.v. transplantation. Transient upregulation of CXCR4 on GM HSC-enriched cells, using a noncytotoxic portion of viral protein R (VPR) fused to CXCR4 delivered as a protein in lentiviral particles, resulted in higher homing and long-term engraftment of GM HSC transplanted either i.v. or i.b.m. compared with standard i.v. transplants. Overall, we show a mechanism for why i.b.m. transplants do not significantly improve long-term engraftment over i.v. transplants. I.b.m. transplantation becomes relevant when an HSC-enriched population is delivered. Alternatively, CXCR4 expression on HSC, when transiently increased using a protein delivery method, improves homing and engraftment specifically of GM HSC.
Collapse
Affiliation(s)
- Sydney Felker
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center (CCHMC) and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Experimental Hematology and Cancer Biology and
| | | | - Jeff Bailey
- Division of Experimental Hematology and Cancer Biology and
| | - Devin M Pillis
- Division of Experimental Hematology and Cancer Biology and
| | - Dylan Siniard
- Division of Experimental Hematology and Cancer Biology and
| | - Punam Malik
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center (CCHMC) and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Experimental Hematology and Cancer Biology and
- Division of Hematology, CCHMC, Cincinnati, Ohio, USA
| |
Collapse
|
5
|
Punatar S, Kandekar S, Khattry N, Gokarn A, Prabhash K, Bakshi A, Rane P, Mathew L, Chiplunkar S, Kode J. CD26 expression on donor harvest as a risk predictive biomarker for developing graft-versus-host disease post-allogeneic hematopoietic stem cell transplantation: A ten-year follow-up study. Cancer Biomark 2022; 33:17-28. [PMID: 34334382 DOI: 10.3233/cbm-210137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (ASCT) is the preferred treatment option for patients with several hematologic disorders and immunodeficiency syndromes. Graft-versus-host disease (GVHD) is an immune mediated post-transplant complication which has a major impact on long-term transplant outcomes. OBJECTIVE Current efforts are focused on identification of new markers that serve as potential predictors of GVHD and other post-transplant clinical outcomes. METHODS This study includes donor harvests collected from twenty-three allogeneic donors during period 2008-2009 and respective transplant recipients followed for clinical outcomes till March 2019. Percent CD26+ and CD34+ cells in donor harvest were analyzed using flow cytometry. Percent expression and infused dose of CD26+ and CD34+ cells were evaluated for association with various clinical outcomes. RESULTS Total 23 healthy donors with median age of 28 years (13 males), and transplant recipients with median age of 24 years (17 males) formed the study cohort. The diagnosis included malignant (n= 13) and non-malignant (n= 10) hematological disorders. Median CD34brCD45lo HSC expression was 0.57% (IQR 0.24-1.03) while median CD26 expression was 19.64% (IQR 8.96-33.56) of all nucleated cells. CD26 expression was associated with donor age (P= 0.037). CD26 percent expression correlated with WBC engraftment (P= 0.015) and with acute GVHD (P= 0.023) whereas infused CD26 cell dose correlated with WBC engraftment (P= 0.004) and risk of CMV reactivation (P= 0.020). There was no statistically significant correlation of either CD26 expression or cell dose with chronic GVHD, EFS or OS. CONCLUSIONS Our findings suggest a role of CD26 expression on human donor harvest as a potential predictor of acute GVHD. This association warrants further exploration.
Collapse
Affiliation(s)
- Sachin Punatar
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Shruti Kandekar
- Tumor Immunology and Immunotherapy Group, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Navin Khattry
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Anant Gokarn
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Kumar Prabhash
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Ashish Bakshi
- Department of Bone Marrow Transplantation, Department of Medical Oncology, Hiranandani Hospital, Powai, Mumbai, India
| | - Pallavi Rane
- Epidemiology and Clinical Trials Unit, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Libin Mathew
- Stem Cell Transplant Unit, Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Shubhada Chiplunkar
- Tumor Immunology and Immunotherapy Group, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Jyoti Kode
- Tumor Immunology and Immunotherapy Group, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| |
Collapse
|
6
|
Tay J, Barbier V, Helwani FM, Price GR, Levesque JP, Winkler IG. Prostacyclin is an endosteal bone marrow niche component and its clinical analog iloprost protects hematopoietic stem cell potential during stress. Stem Cells 2021; 39:1532-1545. [PMID: 34260805 DOI: 10.1002/stem.3438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 06/18/2021] [Indexed: 06/13/2023]
Abstract
Hematopoietic stem cells (HSCs) with superior reconstitution potential are reported to be enriched in the endosteal compared to central bone marrow (BM) region. To investigate whether specific factors at the endosteum may contribute to HSC potency, we screened for candidate HSC niche factors enriched in the endosteal compared to central BM regions. Together with key known HSC supporting factors Kitl and Cxcl12, we report that prostacyclin/prostaglandin I2 (PGI2 ) synthase (Ptgis) was one of the most highly enriched mRNAs (>10-fold) in endosteal compared to central BM. As PGI2 signals through receptors distinct from prostaglandin E2 (PGE2 ), we investigated functional roles for PGI2 at the endosteal niche using therapeutic PGI2 analogs, iloprost, and cicaprost. We found PGI2 analogs strongly reduced HSC differentiation in vitro. Ex vivo iloprost pulse treatment also significantly boosted long-term competitive repopulation (LT-CR) potential of HSCs upon transplantation. This was associated with increased tyrosine-phosphorylation of transducer and activator of transcription-3 (STAT3) signaling in HSCs but not altered cell cycling. In vivo, iloprost administration protected BM HSC potential from radiation or granulocyte colony-stimulating factor-induced exhaustion, and restored HSC homing potential with increased Kitl and Cxcl12 transcription in the BM. In conclusion, we propose that PGI2 is a novel HSC regulator enriched in the endosteum that promotes HSC regenerative potential following stress.
Collapse
Affiliation(s)
- Joshua Tay
- Stem Cell and Cancer Group, Blood and Bone Diseases Program, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Valerie Barbier
- Stem Cell and Cancer Group, Blood and Bone Diseases Program, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Falak M Helwani
- Stem Cell Biology Group, Blood and Bone Diseases Program, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Gareth R Price
- Stem Cell and Cancer Group, Blood and Bone Diseases Program, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Jean-Pierre Levesque
- Stem Cell Biology Group, Blood and Bone Diseases Program, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Ingrid G Winkler
- Stem Cell and Cancer Group, Blood and Bone Diseases Program, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
7
|
Ropa J, Broxmeyer HE. An expanded role for dipeptidyl peptidase 4 in cell regulation. Curr Opin Hematol 2021; 27:215-224. [PMID: 32487805 DOI: 10.1097/moh.0000000000000590] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Dipeptidyl peptidase 4 (DPP4) is a serine protease with diverse regulatory functions in healthy and diseased cells. Much remains unknown about the mechanisms and targets of DPP4. Here we discuss new studies exploring DPP4-mediated cellular regulation, provide an updated list of potential targets of DPP4, and discuss clinical implications of each. RECENT FINDINGS Recent studies have sought enhanced efficacy of targeting DPP4's role in regulating hematopoietic stem and progenitor cells for improved clinical application. Further studies have identified DPP4 functions in different cellular compartments and have proposed ways to target this protein in malignancy. These findings, together with an expanded list of putative extracellular, cell surface, and intracellular DPP4 targets, provide insight into new DPP4-mediated cell regulation. SUMMARY DPP4 posttranslationally modifies proteins and peptides with essential roles in hematopoietic cell regulation, stem cell transplantation, and malignancy. Targets include secreted signaling factors and may include membrane proteins and transcription factors critical for different hematopoietic functions. Knowing these targets and functions can provide insight into new regulatory roles for DPP4 that may be targeted to enhance transplantation, treat disease, and better understand different regulatory pathways of hematopoiesis.
Collapse
Affiliation(s)
- James Ropa
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | |
Collapse
|
8
|
Progress towards improving homing and engraftment of hematopoietic stem cells for clinical transplantation. Curr Opin Hematol 2020; 26:266-272. [PMID: 31045644 DOI: 10.1097/moh.0000000000000510] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Hematopoietic cell transplantation (HCT) is a life-saving treatment for a variety of hematological and nonhematological disorders. Successful clinical outcomes after transplantation rely on adequate hematopoietic stem cell (HSC) numbers, and the homing and subsequent short-term and long-term engraftment of these cells in the bone marrow. Enhancing the homing capability of HSCs has the potential for high impact on improving HCT and patient survival. RECENT FINDINGS There are a number of ways to enhance HSC engraftment. Neutralizing negative epigenetic regulation by histone deacetylase 5 (HDAC5) increases surface CXCR4 expression and promotes human HSC homing and engraftment in immune-deficient NSG (NOD.Cg-Prkdc IL2rgt/Sz) mice. Short-term treatment of cells with glucocorticoids, pharmacological stabilization of hypoxia-inducible factor (HIF)-1α, increasing membrane lipid raft aggregation, and inhibition of dipeptidyl peptidase 4 (DPP4) facilitates HSC homing and engraftment. Added to these procedures, modulating the mitochondria permeability transition pore (MPTP) to mitigate ambient air-induced extra physiological oxygen stress/shock (EPHOSS) by hypoxic harvest and processing, or using cyclosporine A during air collection increases functional HSC numbers and improves HSC engraftment. SUMMARY A better understanding of the regulation of human HSC homing mediated by various signaling pathways will facilitate development of more efficient means to enhance HCT efficacy.
Collapse
|
9
|
Zebedin-Brandl E, Themanns M, Kazemi Z, Nasrollahi-Shirazi S, Mussbacher M, Heyes E, Meissl K, Prchal-Murphy M, Strohmaier W, Krumpl G, Freissmuth M. Regimen-dependent synergism and antagonism of treprostinil and vildagliptin in hematopoietic cell transplantation. J Mol Med (Berl) 2019; 98:233-243. [PMID: 31872285 PMCID: PMC7007891 DOI: 10.1007/s00109-019-01869-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/12/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023]
Abstract
The cell dose in umbilical cord blood units is a major determinant for the outcome of hematopoietic cell transplantation. Prostaglandin analogs and dipeptidylpeptidase-4 (DPP4/CD26)-inhibitors enhance the ability of hematopoietic stem cells (HSCs) to reconstitute hematopoiesis. Here we explored the synergism between treprostinil, a stable prostaglandin agonist, and the DPP4/CD26-inhibitor vildagliptin. The combination of treprostinil and forskolin caused a modest but statistically significant increase in the surface levels of DPP4/CD26 on hematopoietic stem and progenitor cells (HSPCs) derived from murine bone and human cord blood. Their migration towards stromal cell-derived factor-1 (SDF-1/CXCL12) was enhanced, if they were pretreated with treprostinil and forskolin, and further augmented by vildagliptin. Administration of vildagliptin rescued 25% of lethally irradiated recipient mice injected with a limiting number of untreated HSPCs, but 90 to 100% of recipients injected with HSPCs preincubated with treprostinil and forskolin. The efficacy of vildagliptin surpassed that of treprostinil (60% rescue). Surprisingly, concomitant administration of vildagliptin and treprostinil resulted in poor survival of recipients indicating mutual antagonism, which was recapitulated when homing of and colony formation by HSPCs were assessed. These observations of regimen-dependent synergism and antagonism of treprostinil and vildagliptin are of translational relevance for the design of clinical trials. KEY MESSAGES: Pretreatment with treprostinil increases surface levels of DPP4/CD26 in HSPCs. Vildagliptin enhances in vitro migration of pretreated HSPCs. Vildagliptin enhances in vivo homing and engraftment of pretreated HSPCs. Unexpected mutual antagonism in vivo by concomitant administration of vildagliptin and treprostinil.
Collapse
Affiliation(s)
- Eva Zebedin-Brandl
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090, Vienna, Austria
| | - Madeleine Themanns
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090, Vienna, Austria
| | - Zahra Kazemi
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090, Vienna, Austria
| | - Shahrooz Nasrollahi-Shirazi
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090, Vienna, Austria
| | - Marion Mussbacher
- Institute of Vascular Biology, Centre of Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Elizabeth Heyes
- Institute for Medical Biochemistry, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Katrin Meissl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine, 1210, Vienna, Austria
| | | | - Guenther Krumpl
- MRN Medical Research GmbH, Postgasse 11, 1010, Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
10
|
Huang X, Guo B, Capitano M, Broxmeyer HE. Past, present, and future efforts to enhance the efficacy of cord blood hematopoietic cell transplantation. F1000Res 2019; 8. [PMID: 31723413 PMCID: PMC6823900 DOI: 10.12688/f1000research.20002.1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2019] [Indexed: 12/22/2022] Open
Abstract
Cord blood (CB) has been used as a viable source of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) in over 35,000 clinical hematopoietic cell transplantation (HCT) efforts to treat the same variety of malignant and non-malignant disorders treated by bone marrow (BM) and mobilized peripheral blood (mPB) using HLA-matched or partially HLA-disparate related or unrelated donor cells for adult and children recipients. This review documents the beginning of this clinical effort that started in the 1980’s, the pros and cons of CB HCT compared to BM and mPB HCT, and recent experimental and clinical efforts to enhance the efficacy of CB HCT. These efforts include means for increasing HSC numbers in single CB collections, expanding functional HSCs
ex vivo, and improving CB HSC homing and engraftment, all with the goal of clinical translation. Concluding remarks highlight the need for phase I/II clinical trials to test the experimental procedures that are described, either alone or in combination.
Collapse
Affiliation(s)
- Xinxin Huang
- Xuhui Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Bin Guo
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maegan Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202-5181, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202-5181, USA
| |
Collapse
|
11
|
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
|
12
|
Loew A, Köhnke T, Rehbeil E, Pietzner A, Weylandt KH. A Role for Lipid Mediators in Acute Myeloid Leukemia. Int J Mol Sci 2019; 20:ijms20102425. [PMID: 31100828 PMCID: PMC6567850 DOI: 10.3390/ijms20102425] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/05/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022] Open
Abstract
In spite of therapeutic improvements in the treatment of different hematologic malignancies, the prognosis of acute myeloid leukemia (AML) treated solely with conventional induction and consolidation chemotherapy remains poor, especially in association with high risk chromosomal or molecular aberrations. Recent discoveries describe the complex interaction of immune effector cells, as well as the role of the bone marrow microenvironment in the development, maintenance and progression of AML. Lipids, and in particular omega-3 as well as omega-6 polyunsaturated fatty acids (PUFAs) have been shown to play a vital role as signaling molecules of immune processes in numerous benign and malignant conditions. While the majority of research in cancer has been focused on the role of lipid mediators in solid tumors, some data are showing their involvement also in hematologic malignancies. There is a considerable amount of evidence that AML cells are targetable by innate and adaptive immune mechanisms, paving the way for immune therapy approaches in AML. In this article we review the current data showing the lipid mediator and lipidome patterns in AML and their potential links to immune mechanisms.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Bone Marrow
- Disease Progression
- Fatty Acids, Omega-3/immunology
- Fatty Acids, Omega-3/therapeutic use
- Fatty Acids, Omega-6/immunology
- Fatty Acids, Omega-6/therapeutic use
- Fatty Acids, Unsaturated
- Hematologic Neoplasms/drug therapy
- Hematopoiesis
- Humans
- Immunity, Innate/drug effects
- Immunotherapy
- Inflammation
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/immunology
- Lipids/immunology
- Lipids/therapeutic use
- Neoplasms/drug therapy
- Prognosis
- Tumor Microenvironment
Collapse
Affiliation(s)
- Andreas Loew
- Department of Medicine B, Ruppin General Hospital, Brandenburg Medical School, 16816 Neuruppin, Germany.
| | - Thomas Köhnke
- Department of Internal Medicine III, University of Munich, 81377 Munich, Germany.
| | - Emma Rehbeil
- Department of Medicine B, Ruppin General Hospital, Brandenburg Medical School, 16816 Neuruppin, Germany.
| | - Anne Pietzner
- Department of Medicine B, Ruppin General Hospital, Brandenburg Medical School, 16816 Neuruppin, Germany.
| | - Karsten-H Weylandt
- Department of Medicine B, Ruppin General Hospital, Brandenburg Medical School, 16816 Neuruppin, Germany.
- Medical Department, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany.
| |
Collapse
|
13
|
Broxmeyer HE. Long-Overdue Guidelines for the Cord Blood Banking Community. Stem Cells Transl Med 2019; 8:320-322. [PMID: 30843664 PMCID: PMC6431605 DOI: 10.1002/sctm.19-0056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Hal E. Broxmeyer
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
14
|
Carter BZ, Mak PY, Wang X, Tao W, Ruvolo V, Mak D, Mu H, Burks JK, Andreeff M. An ARC-Regulated IL1β/Cox-2/PGE2/β-Catenin/ARC Circuit Controls Leukemia-Microenvironment Interactions and Confers Drug Resistance in AML. Cancer Res 2019; 79:1165-1177. [PMID: 30674535 DOI: 10.1158/0008-5472.can-18-0921] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/17/2018] [Accepted: 01/16/2019] [Indexed: 12/29/2022]
Abstract
The apoptosis repressor with caspase recruitment domain (ARC) protein is a strong independent adverse prognostic marker in acute myeloid leukemia (AML). We previously reported that ARC regulates leukemia-microenvironment interactions through the NFκB/IL1β signaling network. Malignant cells have been reported to release IL1β, which induces PGE2 synthesis in mesenchymal stromal cells (MSC), in turn activating β-catenin signaling and inducing the cancer stem cell phenotype. Although Cox-2 and its enzymatic product PGE2 play major roles in inflammation and cancer, the regulation and role of PGE2 in AML are largely unknown. Here, we report that AML-MSC cocultures greatly increase Cox-2 expression in MSC and PGE2 production in an ARC/IL1β-dependent manner. PGE2 induced the expression of β-catenin, which regulated ARC and augmented chemoresistance in AML cells; inhibition of β-catenin decreased ARC and sensitized AML cells to chemotherapy. NOD/SCIDIL2RγNull-3/GM/SF mice transplanted with ARC-knockdown AML cells had significantly lower leukemia burden, lower serum levels of IL1β/PGE2, and lower tissue human ARC and β-catenin levels, prolonged survival, and increased sensitivity to chemotherapy than controls. Collectively, we present a new mechanism of action of antiapoptotic ARC by which ARC regulates PGE2 production in the tumor microenvironment and microenvironment-mediated chemoresistance in AML.Significance: The antiapoptotic protein ARC promotes AML aggressiveness by enabling detrimental cross-talk with bone marrow mesenchymal stromal cells.
Collapse
Affiliation(s)
- Bing Z Carter
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Po Yee Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiangmeng Wang
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wenjing Tao
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivian Ruvolo
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Duncan Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Mu
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jared K Burks
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
15
|
Zubkov EA, Zorkina YA, Orshanskaya EV, Khlebnikova NN, Krupina NA, Chekhonin VP. Changes in Gene Expression Profiles in Adult Rat Brain Structures after Neonatal Action of Dipeptidyl Peptidase-IV Inhibitors. Neuropsychobiology 2018; 76:89-99. [PMID: 29860255 DOI: 10.1159/000488367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/13/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Previous studies have shown the development of emotional and motivational disorders, such as anxiety-depression-like disorders with increased aggression in adolescent and adult Wistar rats, occurs after neonatal exposure to the dipeptidyl peptidase-IV (DPP-IV, EC 3.4.14.5) inhibitors diprotin A and sitagliptin (postnatal days 5-18). METHODS In this study, using real-time PCR, we evaluated changes in the gene expression of serine protease DPP-IV and prolyl endopeptidase (PREP, EC 3.4.21.26; dpp4 and prep genes), monoamine oxidase А (maoA) and B (maoB), and serotonin transporter (SERT; sert) in the brain structures from 3-month-old rats after postnatal action of DPP-IV inhibitors diprotin A and sitagliptin. RESULTS Dpp4, sert, and maoB gene expression increased and maoA gene expression changed with a tendency to increase in the striatum of rats with neonatal sitagliptin action. The increase of maoA gene expression was also shown in the amygdala. An increase in prep gene expression was found in the striatum of rats with the neonatal action of diprotin A, and a decrease in maoB gene expression was observed in the amygdala. We detected a significant downward trend in sert gene expression in the frontal cortex and amygdala, as well as a tendency to increase in maoA gene expression in the hypothalamus. DISCUSSION These findings suggest that changes in the expression of the abovementioned genes are associated with the development of anxiety and depression, with increased aggression caused by the neonatal action of diprotin A and sitagliptin.
Collapse
Affiliation(s)
- Eugene A Zubkov
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psyсhiatry and Narcology, Moscow, Russian Federation
| | - Yana A Zorkina
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psyсhiatry and Narcology, Moscow, Russian Federation
| | - Elena V Orshanskaya
- The Institute of General Pathology and Pathophysiology, Moscow, Russian Federation
| | | | - Natalia A Krupina
- The Institute of General Pathology and Pathophysiology, Moscow, Russian Federation
| | - Vladimir P Chekhonin
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center of Psyсhiatry and Narcology, Moscow, Russian Federation
| |
Collapse
|
16
|
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
|
17
|
Cai Q, Capitano M, Huang X, Guo B, Cooper S, Broxmeyer HE. Combinations of antioxidants and/or of epigenetic enzyme inhibitors allow for enhanced collection of mouse bone marrow hematopoietic stem cells in ambient air. Blood Cells Mol Dis 2018; 71:23-28. [PMID: 29422232 DOI: 10.1016/j.bcmd.2018.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 12/11/2022]
Abstract
Hematopoietic cell transplantation (HCT) is a treatment for malignant and non-malignant disorders. However, sometimes the numbers of donor hematopoietic stem cells (HSC) are limiting, which can compromise the success of HCT. We recently published that collection and processing of mouse bone marrow (BM) and human cord blood cells in a hypoxic atmosphere of 3% O2 or in ambient air (~21% O2) in the presence of cyclosporine A yields increased numbers of HSC. We now show that collection and processing of mouse BM cells in ambient air in the presence of specific combinations of anti-oxidants and/or inhibitors of epigenetic enzymes can also enhance the collection of HSC, information of potential relevance for enhanced efficacy of HCT.
Collapse
Affiliation(s)
- Qingchun Cai
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maegan Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xinxin Huang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bin Guo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Scott Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
18
|
Farag SS, Nelson R, Cairo MS, O'Leary HA, Zhang S, Huntley C, Delgado D, Schwartz J, Zaid MA, Abonour R, Robertson M, Broxmeyer H. High-dose sitagliptin for systemic inhibition of dipeptidylpeptidase-4 to enhance engraftment of single cord umbilical cord blood transplantation. Oncotarget 2017; 8:110350-110357. [PMID: 29299152 PMCID: PMC5746387 DOI: 10.18632/oncotarget.22739] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/30/2017] [Indexed: 01/31/2023] Open
Abstract
Delayed engraftment remains a limitation of umbilical cord blood (UCB) transplantation. We previously showed that inhibition of dipeptidylpeptidase (DPP)-4 using sitagliptin 600 mg daily was safe with encouraging results on engraftment, but inhibition was not sustained. We evaluated the efficacy and feasibility of higher doses of sitagliptin to enhance engraftment of UCB in patients with hematological cancers. Fifteen patients, median age 41 (range, 18-59) years, received single UCB grafts matched at 4 (n=11) or 5 (n=4) of 6 HLA loci with median nucleated cell dose of 3.5 (range, 2.57-4.57) x107/kg. Sitagliptin 600 mg every 12 hours was administered days -1 to +2. All patients engrafted by day 30, with 12 (80%) engrafting by day 21. The median time to neutrophil engraftment was 19 (range, 12-30) days. Plasma DPP-4 activity was better inhibited with a mean residual trough DPP-4 activity of 70%±19%. Compared to patients previously treated with 600 mg/day, sitagliptin 600 mg every 12 hours appeared to improve engraftment, supporting the hypothesis that more sustained DPP-4 inhibition is required. In-vivo inhibition of DPP-4 using high-dose sitagliptin compares favorably with other approaches to enhance UCB engraftment with greater simplicity, and may show synergy in combination with other strategies.
Collapse
Affiliation(s)
- Sherif S Farag
- Division of Hematology and Oncology, Department of Medicine, Indianapolis, Indiana, USA.,Indiana University Simon Cancer Center, Indianapolis, Indiana, USA
| | - Robert Nelson
- Division of Hematology and Oncology, Department of Medicine, Indianapolis, Indiana, USA.,Indiana University Simon Cancer Center, Indianapolis, Indiana, USA
| | - Mitchell S Cairo
- Children and Adolescent Cancer and Blood Diseases Center and Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Heather A O'Leary
- Division of Hematology and Oncology, Department of Medicine, Indianapolis, Indiana, USA.,Indiana University Simon Cancer Center, Indianapolis, Indiana, USA
| | - Shuhong Zhang
- Division of Hematology and Oncology, Department of Medicine, Indianapolis, Indiana, USA
| | - Carol Huntley
- Indiana University Simon Cancer Center, Indianapolis, Indiana, USA
| | - David Delgado
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jennifer Schwartz
- Division of Hematology and Oncology, Department of Medicine, Indianapolis, Indiana, USA
| | - Mohammad Abu Zaid
- Division of Hematology and Oncology, Department of Medicine, Indianapolis, Indiana, USA
| | - Rafat Abonour
- Division of Hematology and Oncology, Department of Medicine, Indianapolis, Indiana, USA
| | - Michael Robertson
- Division of Hematology and Oncology, Department of Medicine, Indianapolis, Indiana, USA.,Indiana University Simon Cancer Center, Indianapolis, Indiana, USA
| | - Hal Broxmeyer
- Department of Microbiology and Immunology, Indianapolis, Indiana, USA.,Indiana University Simon Cancer Center, Indianapolis, Indiana, USA
| |
Collapse
|
19
|
Blood on the tracks: hematopoietic stem cell-endothelial cell interactions in homing and engraftment. J Mol Med (Berl) 2017; 95:809-819. [PMID: 28702683 DOI: 10.1007/s00109-017-1559-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/24/2017] [Accepted: 06/08/2017] [Indexed: 01/13/2023]
Abstract
Cells of the hematopoietic system undergo rapid turnover. Each day, humans require the production of about one hundred billion new blood cells for proper function. Hematopoietic stem cells (HSCs) are rare cells that reside in specialized niches and are required throughout life to produce specific progenitor cells that will replenish all blood lineages. There is, however, an incomplete understanding of the molecular and physical properties that regulate HSC migration, homing, engraftment, and maintenance in the niche. Endothelial cells (ECs) are intimately associated with HSCs throughout the life of the stem cell, from the specialized endothelial cells that give rise to HSCs, to the perivascular niche endothelial cells that regulate HSC homeostasis. Recent studies have dissected the unique molecular and physical properties of the endothelial cells in the HSC vascular niche and their role in HSC biology, which may be manipulated to enhance hematopoietic stem cell transplantation therapies.
Collapse
|
20
|
Kazemi Z, Bergmayr C, Prchal-Murphy M, Javaheri T, Themanns M, Pham HTT, Strohmaier W, Sexl V, Freissmuth M, Zebedin-Brandl E. Repurposing Treprostinil for Enhancing Hematopoietic Progenitor Cell Transplantation. Mol Pharmacol 2016; 89:630-44. [PMID: 26989084 PMCID: PMC4885501 DOI: 10.1124/mol.116.103267] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/14/2016] [Indexed: 12/23/2022] Open
Abstract
Activation of Gs-coupled receptors enhances engraftment of hematopoietic stem and progenitor cells (HSPCs). We tested the hypothesis that treprostinil, a prostacyclin analog approved for the treatment of pulmonary hypertension, can be repurposed to improve hematopoietic stem cell transplantation. Murine and human HSPCs were isolated from bone marrow and umbilical cord blood, respectively. Prostanoid receptor agonists and the combination thereof with forskolin were tested for their capacity to stimulate [(3)H]cAMP accumulation in HSPCs. Three independent approaches were employed to verify the ability of agonist-activated HSPCs to reconstitute the bone marrow in lethally irradiated recipient mice. The underlying mechanism was explored in cellular migration assays and by blocking C-X-C motif chemokine receptor 4 (CXCR4). Among several prostanoid agonists tested in combination with forskolin, treprostinil was most efficacious in raising intracellular cAMP levels in murine and human HPSCs. Injection of murine and human HSPCs, which had been pretreated with treprostinil and forskolin, enhanced survival of lethally irradiated recipient mice. Survival was further improved if recipient mice were subcutaneously administered treprostinil (0.15 mg kg(-1) 8 h(-1)) for 10 days. This regimen also reduced the number of HSPCs required to rescue lethally irradiated mice. Enhanced survival of recipient mice was causally related to treprostinil-enhanced CXCR4-dependent migration of HSPCs. Treprostinil stimulates the engraftment of human and murine hematopoietic stem cells without impairing their capacity for self-renewal. The investigated dose range corresponds to the dose approved for human use. Hence, these findings may be readily translated into a clinical application.
Collapse
Affiliation(s)
- Zahra Kazemi
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Christian Bergmayr
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Michaela Prchal-Murphy
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Tahereh Javaheri
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Madeleine Themanns
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Ha T T Pham
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Wolfgang Strohmaier
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Veronika Sexl
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Michael Freissmuth
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| | - Eva Zebedin-Brandl
- Institute of Pharmacology, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (Z.K., C.B., M.T., M.F., E.Z.-B.); Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria (M.P.-M., V.S.); Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria (T.J., H.T.T.P.); SciPharm SàRL, L-2540 Luxembourg (W.S.)
| |
Collapse
|
21
|
Abstract
Clinical cord blood (CB) hematopoietic cell transplantation (HCT) has progressed well since the initial successful CB HCT that saved the life of a young boy with Fanconi anemia. The recipient is alive and well now 28 years out since that first transplant with CB cells from his HLA-matched sister. CB HCT has now been used to treat over 35,000 patients with various malignant and non-malignant disorders mainly using HLA-matched or partially HLA-disparate allogeneic CB cells. There are advantages and disadvantages to using CB for HCT compared to other sources of transplantable hematopoietic stem (HSC) and progenitor (HPC) cells. One disadvantage of the use of CB as a source of transplantable HSC and HPC is the limited number of these cells in a single CB collected, and slower time to neutrophil, platelet and immune cell recovery. This review describes current attempts to: increase the collection of HSC/HPC from CB, enhance the homing of the infused cells, ex-vivo expand numbers of collected HSC/HPC and increase production of the infused CB cells that reach the marrow. The ultimate goal is to manipulate efficiency and efficacy for safe and economical use of single unit CB HCT.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
22
|
Psatha N, Karponi G, Yannaki E. Optimizing autologous cell grafts to improve stem cell gene therapy. Exp Hematol 2016; 44:528-39. [PMID: 27106799 DOI: 10.1016/j.exphem.2016.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 10/21/2022]
Abstract
Over the past decade, stem cell gene therapy has achieved unprecedented curative outcomes for several genetic disorders. Despite the unequivocal success, clinical gene therapy still faces challenges. Genetically engineered hematopoietic stem cells are particularly vulnerable to attenuation of their repopulating capacity once exposed to culture conditions, ultimately leading to low engraftment levels posttransplant. This becomes of particular importance when transduction rates are low or/and competitive transplant conditions are generated by reduced-intensity conditioning in the absence of a selective advantage of the transduced over the unmodified cells. These limitations could partially be overcome by introducing megadoses of genetically modified CD34(+) cells into conditioned patients or by transplanting hematopoietic stem cells hematopoietic stem cells with high engrafting and repopulating potential. On the basis of the lessons gained from cord blood transplantation, we summarize the most promising approaches to date of increasing either the numbers of hematopoietic stem cells for transplantation or/and their engraftability, as a platform toward the optimization of engineered stem cell grafts.
Collapse
Affiliation(s)
- Nikoletta Psatha
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Medicine, University of Washington, Seattle, WA
| | - Garyfalia Karponi
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Evangelia Yannaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Medicine, University of Washington, Seattle, WA.
| |
Collapse
|
23
|
Berezin A. Metabolic memory phenomenon in diabetes mellitus: Achieving and perspectives. Diabetes Metab Syndr 2016; 10:S176-S183. [PMID: 27025794 DOI: 10.1016/j.dsx.2016.03.016] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/05/2016] [Indexed: 12/20/2022]
Abstract
Diabetes mellitus (DM) exhibits raised prevalence worldwide. There is a large body of evidence regarding the incidence of DM closely associates with cardiovascular (CV) complications. In this context, hyperglycaemia, oxidant stress, and inflammation are key factors that contribute in CV events and disease in type1 and type 2 DM, even when metabolic control was optimal and/or intensive glycemic control was implemented. It has been suggested that the effect of poor metabolic control or even transient episodes of hyperglycemia in DM associates in particularly with worsening ability of endogenous vasoreparative systems that are mediated epigenetic changes in several cells (progenitor cells, stem cells, mononuclears, immune cells), and thereby lead to so called "vascular glycemic memory" or "metabolic memory". Both terms are emphasized the fact that prior glucose control has sustained effects that persist even after return to more usual glycemic control. The mechanisms underlying the cellular "metabolic memory" induced by high glucose remain unclear. The review is discussed pathophysiology and clinical relevance of "metabolic" memory phenomenon in DM. The role of oxidative stress, inflammation, and epigenetics in DM and its vascular complications are highlighted. The effects of several therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Alexander Berezin
- Internal Medicine Department, State Medical University of Zaporozhye, 26, Mayakovsky Av., Zaporozhye 69035, Ukraine.
| |
Collapse
|
24
|
Krupina NA, Khlebnikova NN. Neonatal Exposure to the Dipeptidyl Peptidase-IV Inhibitors Diprotin A and Sitagliptin Induces Depression-Like Behavior, Anxiety, and Latent Aggression in Adolescent and Adult Rats. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/jbbs.2016.64018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
25
|
Ponce DM, Hilden P, Devlin SM, Maloy M, Lubin M, Castro-Malaspina H, Dahi P, Hsu K, Jakubowski AA, Kernan NA, Koehne G, O'Reilly RJ, Papadopoulos EB, Perales MA, Sauter C, Scaradavou A, Tamari R, van den Brink MRM, Young JW, Giralt S, Barker JN. High Disease-Free Survival with Enhanced Protection against Relapse after Double-Unit Cord Blood Transplantation When Compared with T Cell-Depleted Unrelated Donor Transplantation in Patients with Acute Leukemia and Chronic Myelogenous Leukemia. Biol Blood Marrow Transplant 2015; 21:1985-93. [PMID: 26238810 PMCID: PMC4768474 DOI: 10.1016/j.bbmt.2015.07.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/28/2015] [Indexed: 01/30/2023]
Abstract
Double-unit cord blood (DCB) grafts are a rapidly available stem cell source for adults with high-risk leukemias. However, how disease-free survival (DFS) after DCB transplantation (DCBT) compares to that of unrelated donor transplantation (URDT) is not fully established. We analyzed 166 allograft recipients (66 8/8 HLA-matched URDT, 45 7/8 HLA-matched URDT, and 55 DCBT) ages 16 to 60 years with high-risk acute leukemia or chronic myelogenous leukemia (CML). URDT and DCBT recipients were similar except DCBT recipients were more likely to have lower weight and non-European ancestry and to receive intermediate-intensity conditioning. All URDT recipients received a CD34(+) cell-selected (T cell-depleted) graft. Overall, differences between the 3-year transplantation-related mortality were not significant (8/8 URDT, 18%; 7/8 URDT, 39%; and DCBT, 24%; P = .108), whereas the 3-year relapse risk was decreased after DCBT (8/8 URDT, 23%; 7/8 URDT, 20%; and DCBT 9%, P = .037). Three-year DFS was 57% in 8/8 URDT, 41% in 7/8 URDT, and 68% in DCBT recipients (P = .068), and the 3-year DFS in DCBT recipients was higher than that of 7/8 URDT recipients (P = .021). In multivariate analysis in acute leukemia patients, factors adversely associated with DFS were female gender (hazard ratio [HR], 1.68; P = .031), diagnosis of acute lymphoblastic leukemia (HR, 2.09; P = .004), and 7/8 T cell-depleted URDT (HR, 1.91; P = .037). High DFS can be achieved in adults with acute leukemia and CML with low relapse rates after DCBT. Our findings support performing DCBT in adults in preference to HLA-mismatched T cell-depleted URDT and suggest DCBT is a readily available alternative to T cell-depleted 8/8 URDT, especially in patients requiring urgent transplantation.
Collapse
MESH Headings
- Adolescent
- Adult
- Cord Blood Stem Cell Transplantation/methods
- Female
- Graft Survival
- Hematopoietic Stem Cell Transplantation
- Histocompatibility Testing
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Lymphocyte Depletion
- Male
- Middle Aged
- Myeloablative Agonists/therapeutic use
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Recurrence
- Retrospective Studies
- Sex Factors
- Survival Analysis
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- Transplantation Conditioning
- Transplantation, Homologous
- Unrelated Donors
Collapse
Affiliation(s)
- Doris M Ponce
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Patrick Hilden
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Molly Maloy
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marissa Lubin
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hugo Castro-Malaspina
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Parastoo Dahi
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Katharine Hsu
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ann A Jakubowski
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Nancy A Kernan
- Bone Marrow Transplantation Service, Department of Pediatrics; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Guenther Koehne
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Richard J O'Reilly
- Bone Marrow Transplantation Service, Department of Pediatrics; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Esperanza B Papadopoulos
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Craig Sauter
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Andromachi Scaradavou
- Bone Marrow Transplantation Service, Department of Pediatrics; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roni Tamari
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - James W Young
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sergio Giralt
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Juliet N Barker
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
26
|
Abstract
The use of umbilical cord blood (UCB) as an alternative haematopoietic cell source in lieu of bone marrow for haematopoietic reconstitution is increasingly becoming a mainstay treatment for both malignant and nonmalignant diseases, as most individuals will have at least one available, suitably HLA-matched unit of blood. The principal limitation of UCB is the low and finite number of haematopoietic stem and progenitor cells (HSPC) relative to the number found in a typical bone marrow or mobilized peripheral blood allograft, which leads to prolonged engraftment times. In an attempt to overcome this obstacle, strategies that are often based on native processes occurring in the bone marrow microenvironment or 'niche' have been developed with the goal of accelerating UCB engraftment. In broad terms, the two main approaches have been either to expand UCB HSPC ex vivo before transplantation, or to modulate HSPC functionality to increase the efficiency of HSPC homing to the bone marrow niche after transplant both of which enhance the biological activities of the engrafted HSPC. Several early phase clinical trials of these approaches have reported promising results.
Collapse
|
27
|
Abstract
Successful clinical outcomes from transplantation of hematopoietic stem cells (HSCs) depend upon efficient HSC homing to bone marrow (BM), subsequent engraftment, and, finally, BM repopulation. Homing of intravenously administered HSCs from peripheral blood (PB) through the circulation to the BM stem cell niches, which is the first critical step that precedes their engraftment, is enforced by chemotactic factors released in the BM microenvironment that chemoattract HSCs. These chemotactic factors include α-chemokine stromal-derived factor 1 (SDF-1), the bioactive phosphosphingolipids sphingosine-1-phosphate (S1P) and ceramid-1-phosphate (C1P), and the extracellular nucleotides ATP and UTP. Stem cells may also respond to a Ca2+ or H+ gradient by employing calcium- or proton-sensing receptors, respectively. In this review, we will present emerging strategies based on ex vivo manipulation of graft HSCs that are aimed at enhancing the responsiveness of HSCs to BM-secreted chemoattractants and/or promoting HSC adhesion and seeding efficiency in the BM microenvironment.
Collapse
|