1
|
Chang HH, Liou YS, Sun DS. Unraveling the interplay between inflammation and stem cell mobilization or homing: Implications for tissue repair and therapeutics. Tzu Chi Med J 2024; 36:349-359. [PMID: 39421490 PMCID: PMC11483098 DOI: 10.4103/tcmj.tcmj_100_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 04/29/2024] [Accepted: 06/14/2024] [Indexed: 10/19/2024] Open
Abstract
Inflammation and stem cell mobilization or homing play pivotal roles in tissue repair and regeneration. This review explores their intricate interplay, elucidating their collaborative role in maintaining tissue homeostasis and responding to injury or disease. While examining the fundamentals of stem cells, we detail the mechanisms underlying inflammation, including immune cell recruitment and inflammatory mediator release, highlighting their self-renewal and differentiation capabilities. Central to our exploration is the modulation of hematopoietic stem cell behavior by inflammatory cues, driving their mobilization from the bone marrow niche into circulation. Key cytokines, chemokines, growth factors, and autophagy, an intracellular catabolic mechanism involved in this process, are discussed alongside their clinical relevance. Furthermore, mesenchymal stem cell homing in response to inflammation contributes to tissue repair processes. In addition, we discuss stem cell resilience in the face of inflammatory challenges. Moreover, we examine the reciprocal influence of stem cells on the inflammatory milieu, shaping immune responses and tissue repair. We underscore the potential of targeting inflammation-induced stem cell mobilization for regenerative therapies through extensive literature analysis and clinical insights. By unraveling the complex interplay between inflammation and stem cells, this review advances our understanding of tissue repair mechanisms and offers promising avenues for clinical translation in regenerative medicine.
Collapse
Affiliation(s)
- Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Yu-Shan Liou
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
2
|
Alsaati N, Grier A, Ochfeld E, McClory S, Heimall J. Hematopoietic stem cell transplantation for primary immunodeficiency. Allergy Asthma Proc 2024; 45:371-383. [PMID: 39294909 DOI: 10.2500/aap.2024.45.240069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Primary immunodeficiencies, also commonly called inborn errors of immunity (IEI), are commonly due to developmental or functional defects in peripheral blood cells derived from hematopoietic stem cells. In light of this, for the past 50 years, hematopoietic stem cell transplantation (HSCT) has been used as a definitive therapy for IEI. The fields of both clinical immunology and transplantation medicine have had significant advances. This, in turn, has allowed for both an increasing ability to determine a monogenic etiology for many IEIs and an increasing ability to successfully treat these patients with HSCT. Therefore, it has become more common for the practicing allergist/immunologist to diagnose and manage a broad range of patients with IEI before and after HSCT. This review aims to provide practical guidance for the clinical allergist/immunologist on the basics of HSCT and known outcomes in selected forms of IEI, the importance of pre-HSCT supportive care, and the critical importance of and guidance for life-long immunologic and medical monitoring of these patients.
Collapse
Affiliation(s)
- Nouf Alsaati
- From the Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania; and
| | - Alexandra Grier
- From the Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania; and
| | - Elisa Ochfeld
- From the Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania; and
| | - Susan McClory
- Cell Therapy and Transplant Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania
| | - Jennifer Heimall
- From the Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania; and
| |
Collapse
|
3
|
Hamerschlak N, Gómez-Almaguer D, McLornan DP. Editorial: 50 years of BMT: risk stratification, donor matching and stem cell collection for transplantation. Front Oncol 2023; 13:1321334. [PMID: 38169637 PMCID: PMC10758429 DOI: 10.3389/fonc.2023.1321334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/13/2023] [Indexed: 01/05/2024] Open
Affiliation(s)
- Nelson Hamerschlak
- Hospital Israelita Albert Einstein, Department of Bone Marrow Transplant, São Paulo, SP, Brazil
| | - David Gómez-Almaguer
- Servicio de Hematología, Hospital Universitario, Universidad Autánoma de Nuevo León, Monterrey, Mexico
| | - Donal P. McLornan
- Department of Haematology and Stem Cell Transplantation, University College London Hospitals NHS Trust, London, United Kingdom
| |
Collapse
|
4
|
Chang HH, Liou YS, Sun DS. Hematopoietic stem cell mobilization. Tzu Chi Med J 2022; 34:270-275. [PMID: 35912054 PMCID: PMC9333105 DOI: 10.4103/tcmj.tcmj_98_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 06/25/2021] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation has been used to treat hematopoietic diseases for over 50 years. HSCs can be isolated from bone marrow (BM), umbilical cord blood, or peripheral blood. Because of lower costs, shorter hospitalization, and faster engraftment, peripheral blood has become the predominant source of HSCs for transplantation. The major factors determining the rate of successful HSC transplantation include the degree of human leukocyte antigen matching between the donor and recipient and the number of HSCs for transplantation. Administration of granulocyte colony-stimulating factor (G-CSF) alone or combined with plerixafor (AMD3100) are clinical used methods to promote HSC mobilization from BM to the peripheral blood for HSC transplantations. However, a significant portion of healthy donors or patients may be poor mobilizers of G-CSF, resulting in an insufficient number of HSCs for the transplantation and necessitating alternative strategies to increase the apheresis yield. The detailed mechanisms underlying G-CSF-mediated HSC mobilization remain to be elucidated. This review summarizes the current research on deciphering the mechanism of HSC mobilization.
Collapse
Affiliation(s)
- Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Yu-Shan Liou
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
5
|
Inflammatory monocytes promote pre-engraftment syndrome and tocilizumab can therapeutically limit pathology in patients. Nat Commun 2021; 12:4137. [PMID: 34230468 PMCID: PMC8260612 DOI: 10.1038/s41467-021-24412-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 06/15/2021] [Indexed: 12/01/2022] Open
Abstract
Unrelated cord blood transplantation (UCBT) is an effective treatment for hematopoietic disorders. However, this attractive approach is frequently accompanied by pre-engraftment syndrome (PES), severe cases of PES are associated with enhanced mortality and morbidity, but the pathogenesis of PES remains unclear. Here we show that GM-CSF produced by cord blood-derived inflammatory monocytes drives PES pathology, and that monocytes are the main source of IL-6 during PES. Further, we report the outcome of a single arm, single-center clinical study of tocilizumab in the treatment of steroid-refractory severe PES patients (www.chictr.org.cn ChiCTR1800015472). The study met the primary outcome measure since none of the patients was nonrelapse death during the 100 days follow-up. The study also met key secondary outcomes measures of neutrophil engraftment and hematopoiesis. These findings offer a therapeutic strategy with which to tackle PES and improve nonrelapse mortality. Pre-engraftment syndrome is a major consideration during clinical application of unrelated cord blood transfusion and monocytes represent a critical cell type in immune-pathogenesis. Here the authors further establish the role of monocytes and GM-CSF in pre-engraftment syndrome and show clinical administration of tocilizumab limits pathology in pre-engraftment syndrome pathology in patients.
Collapse
|
6
|
Xu ZL, Huang XJ. Optimizing outcomes for haploidentical hematopoietic stem cell transplantation in severe aplastic anemia with intensive GVHD prophylaxis: a review of current findings. Expert Rev Hematol 2021; 14:449-455. [PMID: 33945370 DOI: 10.1080/17474086.2021.1923475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has resulted in increased levels of disease-free survival in severe aplastic anemia (SAA). Haploidentical transplantation (haplo-SCT) was previously not recommended due to unacceptable incidences of graft-versus-host disease (GvHD) and graft failures. With the advent of intensive GvHD prophylaxis strategies, the outcomes obtained with haplo-SCT for SAA have gradually improved.Areas covered: A comprehensive search considered PubMed reported articles before 1 February 2021, presented abstracts, and clinical trials pertaining to haplo-HSCT for SAA. This manuscript covers modern approaches with intensive GvHD prophylaxis in haplo-SCT for SAA. The representative methods consist of granulocyte colony stimulating factor (G-CSF) plus ATG-based and posttransplantation cyclophosphamide (PT-Cy)-based protocols.Expert opinion: Currently, haplo-SCT has become a feasible option for treating SAA. The G-CSF/ATG-based protocol included the largest sample size and reported comparable survival rates with identical siblings. The PT-Cy protocol resulted in a relatively lower incidence of GvHD and seemingly poorer but continuously improved engraftment with augmented conditioning. The optimized outcomes are constantly updated with the modification of the conditioning regimen, donor selection, graft source and GvHD prophylaxis. In the future, we should pay more attention to quality of life in addition to survival, and personalized haplo-SCT may improve outcomes.
Collapse
Affiliation(s)
- Zheng-Li Xu
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| | - Xiao-Jun Huang
- National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China
| |
Collapse
|
7
|
Barnhouse V, Petrikas N, Crosby C, Zoldan J, Harley B. Perivascular Secretome Influences Hematopoietic Stem Cell Maintenance in a Gelatin Hydrogel. Ann Biomed Eng 2021; 49:780-792. [PMID: 32939609 PMCID: PMC7854499 DOI: 10.1007/s10439-020-02602-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022]
Abstract
Adult hematopoietic stem cells (HSCs) produce the body's full complement of blood and immune cells. They reside in specialized microenvironments, or niches, within the bone marrow. The perivascular niche near blood vessels is believed to help maintain primitive HSCs in an undifferentiated state but demonstration of this effect is difficult. In vivo studies make it challenging to determine the direct effect of the endosteal and perivascular niches as they can be in close proximity, and two-dimensional in vitro cultures often lack an instructive extracellular matrix environment. We describe a tissue engineering approach to develop and characterize a three-dimensional perivascular tissue model to investigate the influence of the perivascular secretome on HSC behavior. We generate 3D endothelial networks in methacrylamide-functionalized gelatin hydrogels using human umbilical vein endothelial cells (HUVECs) and mesenchymal stromal cells (MSCs). We identify a subset of secreted factors important for HSC function, and examine the response of primary murine HSCs in hydrogels to the perivascular secretome. Within 4 days of culture, perivascular conditioned media promoted maintenance of a greater fraction of hematopoietic stem and progenitor cells. This work represents an important first-generation perivascular model to investigate the role of niche secreted factors on the maintenance of primary HSCs.
Collapse
Affiliation(s)
- Victoria Barnhouse
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Nathan Petrikas
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Cody Crosby
- Department of Biomedical Engineering, University of Texas at Austin, Austin, USA
| | - Janet Zoldan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, USA
| | - Brendan Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
8
|
Aladag E, Goker H, Demiroglu H, Aksu S, Sayinalp N, Haznedaroglu IC, Ozcebe OI, Buyukasik Y. Long-term results of allogeneic peripheral blood hematopoietic stem cell transplantation for severe aplastic anemia. Transfus Apher Sci 2021; 60:103050. [PMID: 33446450 DOI: 10.1016/j.transci.2020.103050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/21/2020] [Accepted: 12/30/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Aplastic anemia (AA) is a life-threatening disorder and may be associated with significant morbidity and mortality Currently, the first treatment option is allogeneic hematopoietic stem cell transplant (allo-HSCT) for patients younger than 40 years. Bone marrow is recommended as the stem cell source due to less graft versus host disease (GVHD) risk and better outcomes than peripheral blood (PB)-derived stem cell. The aim of this study is to share the data of AA patients who have underwent PB-derived allo-HSCT in our bone marrow transplantation center. METHODS Twenty-seven patients who underwent PB-derived allo-HSCT from human leukocyte antigen matched sibling donors were analyzed retrospectively. RESULTS The median follow-up time was 95.2 months (range, 4.8-235 months). The 10-year survival was 89 %. The median neutrophil and platelet engraftment time was 11 days (range, 9-16 days) and 13 days (range, 11-29 days), respectively. Primary platelet engraftment failure was observed in 1 patient (3.7 %). Acute and chronic GVHD observed in 2 (7.4 %) and 3 (11.1 %) patients, respectively. Neutropenic fever was observed in 13 (44.8 %) of patients until the engraftment after allo-HSCT. One patient died due to CMV infections, two died due to septic shock secondary to fungal infection. CONCLUSION Although there is no prospective data directly comparing BM with PB as stem cell source in AA, observational studies indicates better OS with BM. PB can be used in certain situations such as higher risk for graft failure and donor preference. This study demonstrated that PB-derived stem cell seems to be a reasonable alternative to BM.
Collapse
Affiliation(s)
- Elifcan Aladag
- Hacettepe University Faculty of Medicine, Department of Hematology, 06100, Ankara, Turkey
| | - Hakan Goker
- Hacettepe University Faculty of Medicine, Department of Hematology, 06100, Ankara, Turkey.
| | - Haluk Demiroglu
- Hacettepe University Faculty of Medicine, Department of Hematology, 06100, Ankara, Turkey
| | - Salih Aksu
- Hacettepe University Faculty of Medicine, Department of Hematology, 06100, Ankara, Turkey
| | - Nilgun Sayinalp
- Hacettepe University Faculty of Medicine, Department of Hematology, 06100, Ankara, Turkey
| | | | - Osman Ilhami Ozcebe
- Hacettepe University Faculty of Medicine, Department of Hematology, 06100, Ankara, Turkey
| | - Yahya Buyukasik
- Hacettepe University Faculty of Medicine, Department of Hematology, 06100, Ankara, Turkey
| |
Collapse
|
9
|
Jawdat D, Almashaqbeh W, Sumaily A, Albaloushi N, Jammah S, Alsultan A. Screening for pre-leukemia TEL-AML1 chromosomal translocation in banked cord blood units: cord blood bank perspective. Cell Tissue Bank 2020; 21:625-630. [PMID: 32812094 DOI: 10.1007/s10561-020-09855-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 08/01/2020] [Indexed: 01/03/2023]
Abstract
Acute lymphocytic leukemia is the most common leukemia in children. Many studies suggest the existence of two subsequent hits in order for the disease to occur. TEL-AML1 (ETV6-RUNX1) is considered an initial genetic hit that occurs prenatally and generates a pre-leukemia clone. In cord blood (CB) stem cell transplantation, donor cell leukemia (DCL) is one of the complications associated with the presence of the pre-leukemic clone. The aim of this study was to identify the prevalence of ETV6-RUNX1 translocation in CB units and the feasibility in implementing such a screening test, to ensure the safety of the CB units. A total of 424 CB samples were tested from the CB units banked at KAIMRC-CBB. RNA was extracted and cDNA synthesis was performed on 1 ug input RNA using Reverse Transcriptase RT-PCR methodology. Chromosomal translocation ETV6-RUNX1 was tested using real time quantitative PCR methodology. Our study showed undetectable levels of ETV6-RUNX1 in all tested CB samples. The samples were analyzed for the chromosomal translocation ETV6-RUNX1 under controlled conditions, using control and fusion genes with known concentrations. The result of this study does not rule out the importance of this screening test in predicting and/or preventing DCL. Moreover, the outcome strengthens the adopted system in our CBB for mother medical history screening prior to donation. We propose adding this test during the verification testing stage, prior to the release of CB units selected for transplantation rather than at the banking stage.
Collapse
Affiliation(s)
- Dunia Jawdat
- Cord Blood Bank, King Abdullah International Medical Research Center/College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences/King Abdulaziz Medical City-Riyadh, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia. .,Saudi Stem Cells Donor Registry and Cord Blood Bank, King Abdullah International Medical Research Center, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Riyadh, 11426, Saudi Arabia.
| | - Walid Almashaqbeh
- Cord Blood Bank, King Abdullah International Medical Research Center/College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences/King Abdulaziz Medical City-Riyadh, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,Cell Therapy Laboratory, Department of Pathology and Laboratory Medicine/King Abdullah International Medical Research Center/College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences/King Abdulaziz Medical City-Riyadh, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdulrahman Sumaily
- Cord Blood Bank, King Abdullah International Medical Research Center/College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences/King Abdulaziz Medical City-Riyadh, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Nada Albaloushi
- Cord Blood Bank, King Abdullah International Medical Research Center/College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences/King Abdulaziz Medical City-Riyadh, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Sami Jammah
- Cell Therapy Laboratory, Department of Pathology and Laboratory Medicine/King Abdullah International Medical Research Center/College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences/King Abdulaziz Medical City-Riyadh, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdulrahman Alsultan
- Department of Pediatric, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Budgude P, Kale V, Vaidya A. Mesenchymal stromal cell‐derived extracellular vesicles as cell‐free biologics for the ex vivo expansion of hematopoietic stem cells. Cell Biol Int 2020; 44:1078-1102. [DOI: 10.1002/cbin.11313] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Pallavi Budgude
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
- Symbiosis School of Biological SciencesSymbiosis International (Deemed University) Pune 412115 India
| |
Collapse
|
11
|
Belleghem SMV, Mahadik B, Snodderly KL, Fisher JP. Overview of Tissue Engineering Concepts and Applications. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00081-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
12
|
Combination of Chemical and Neurotrophin Stimulation Modulates Neurotransmitter Receptor Expression and Activity in Transdifferentiating Human Adipose Stromal Cells. Stem Cell Rev Rep 2019; 15:851-863. [PMID: 31529274 DOI: 10.1007/s12015-019-09915-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adipose stromal cells are promising tools for clinical applications in regeneration therapies, due to their ease of isolation from tissue and its high yield; however, their ability to transdifferentiate into neural phenotypes is still a matter of controversy. Here, we show that combined chemical and neurotrophin stimulation resulted in neuron-like morphology and regulated expression and activity of several genes involved in neurogenesis and neurotransmission as well as ion currents mediated by NMDA and GABA receptors. Among them, expression patterns of genes coding for kinin-B1 and B2, α7 nicotinic, M1, M3 and M4 muscarinic acetylcholine, glutamatergic (AMPA2 and mGlu2), purinergic P2Y1 and P2Y4 and GABAergic (GABA-A, β3-subunit) receptors and neuronal nitric oxide synthase were up-regulated compared to levels of undifferentiated cells. Simultaneously, expression levels of P2X1, P2X4, P2X7 and P2Y6 purinergic and M5 muscarinic acetylcholine receptors were down-regulated. Agonist-induced activity levels of the studied receptor classes also augmented during neuronal transdifferentiation. Transdifferentiated cells expressed high levels of neuronal β3-tubulin, NF-H, NeuN and MAP-2 proteins as well as increased ASCL1, MYT1 and POU3F2 gene expression known to drive neuronal fate determination. The presented work contributes to a better understanding of transdifferentiation induced by neurotrophins for a prospective broad spectrum of medical applications.
Collapse
|
13
|
Yi T, Li X, Wang E, Zhang Y, Fu Y, Li J, Jiang T. Activation of the Nuclear Erythroid 2-Related Factor 2 Antioxidant Responsive Element (Nrf2-ARE) Signaling Pathway Alleviates Acute Graft-Versus-Host Disease by Reducing Oxidative Stress and Inhibiting Infiltration of Inflammatory Cells in an Allogeneic Stem Cell Transplantation Mouse Model. Med Sci Monit 2018; 24:5973-5979. [PMID: 30148822 PMCID: PMC6122273 DOI: 10.12659/msm.908130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Background Acute graft-versus-host disease (aGVHD) limits the wider application of hematopoietic stem cell transplantation (HSCT). We explored the relationship between the Nrf2-ARE signaling pathway and aGVHD and identified effective and efficient therapeutic targets for the prevention and management of aGVHD following HSCT. Material/Methods C57BL/6 and BALB/c mice were used to establish the aGVHD model. The bone marrow and spleen mononuclear cells were separated from the donor mice and injected into the caudal vein of recipient mice that had undergone total body irradiation (TBI, 8 Gy). Sulforaphane (SFN) was used to activate the Nrf2-ARE signaling pathway. Results The long-term survival rate of the SFN group was higher than that of the control group (40% vs. 0%, p<0.05, n=10). There were worse pathological changes and a greater infiltration of inflammatory cells in the liver, small intestine, and lung tissues of the control group. Furthermore, the Nrf2, NQO1, and HO-1 mRNA and protein levels were higher in the small intestines of the SFN group than in the control group (p<0.05, n=4). Conclusions The Nrf2-ARE signaling pathway plays a vital role in preventing aGVHD in an HSCT mouse model by regulating the expression of the downstream antioxidant genes NQO1 and HO-1 and by inhibiting the local inflammatory reaction.
Collapse
Affiliation(s)
- Ting Yi
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland).,Department of Hematology, Changsha Central Hospital, Changsha, Hunan, China (mainland)
| | - Xiaogang Li
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Erhua Wang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Yanan Zhang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Yunfeng Fu
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Jieping Li
- Department of Hematology, Changsha Central Hospital, Changsha, Hunan, China (mainland)
| | - Tiebin Jiang
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
14
|
Fuerst D, Frank S, Mueller C, Beelen DW, Schetelig J, Niederwieser D, Finke J, Bunjes D, Kröger N, Neuchel C, Tsamadou C, Schrezenmeier H, Beyersmann J, Mytilineos J. Competing-risk outcomes after hematopoietic stem cell transplantation from the perspective of time-dependent effects. Haematologica 2018; 103:1527-1534. [PMID: 29880610 PMCID: PMC6119156 DOI: 10.3324/haematol.2017.183012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 05/30/2018] [Indexed: 11/14/2022] Open
Abstract
The success of hematopoietic stem cell transplantation is determined by multiple factors. Additional complexity is conferred by covariables showing time-dependent effects. We evaluated the effect of predictors on competing-risk outcomes after hematopoietic stem cell transplantation in a time-dependent manner. We analyzed 14951 outcomes of adult patients with hematologic malignancies who underwent a first allogeneic transplant. We extended the combined endpoints of disease-free and overall survival to competing-risk settings: disease-free survival was split into relapse and non-relapse mortality. Overall survival was divided into transplant-related mortality, death from other causes and death from unknown causes. For time-dependent effects we computed estimators before and after a covariable-specific cut-point. Patients treated with reduced intensity conditioning had a constantly higher risk of relapse compared to patients treated with myeloablative conditioning. For non-relapse mortality, patients treated with reduced intensity conditioning had a reduced mortality risk but this effect was only seen in the first 4 months after transplantation (hazard ratio: 0.76, P<0.001) and not afterwards. Graft source exhibited a time-dependent effect on both transplant-related mortality (in first year: hazard ratio 0.70, P<0.001; after first year: hazard ratio 1.47, P=0.002) and non-relapse mortality (in first 8 months: hazard ratio 0.75, P<0.001; after first 8 months: hazard ratio 1.38, P<0.001). Patients with a poor Karnofsky performance score (<80) had a considerably higher risk of all endpoints in the first 4 months. The competing-risk analysis for overall survival and disease-free survival allows resolution of effects with different vectors early and later after stem cell transplantation, as shown for graft source. This information may be useful in risk assessment of treatment choices and for counseling patients on an individual basis.
Collapse
Affiliation(s)
- Daniel Fuerst
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen and University Clinic Ulm, Germany.,Institute of Transfusion Medicine, University of Ulm, Germany
| | - Sandra Frank
- Institute of Statistics, University of Ulm, Germany.,DRST - German Registry for Stem Cell Transplantation, Germany
| | - Carlheinz Mueller
- DRST - German Registry for Stem Cell Transplantation, Germany.,Zentrales Knochenmarkspender-Register Deutschland (ZKRD - German Bone Marrow Donor Registry), Ulm, Germany
| | - Dietrich W Beelen
- DRST - German Registry for Stem Cell Transplantation, Germany.,Department of Bone Marrow Transplantation, University Hospital, University of Duisburg-Essen, Essen
| | - Johannes Schetelig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | | | - Jürgen Finke
- Faculty of Medicine and Medical Center, University of Freiburg, Germany
| | - Donald Bunjes
- Department of Internal Medicine III, University of Ulm, Germany
| | - Nicolaus Kröger
- Department for Stem Cell Transplantation, University Cancer Center Hamburg, Germany
| | - Christine Neuchel
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen and University Clinic Ulm, Germany.,Institute of Transfusion Medicine, University of Ulm, Germany
| | - Chrysanthi Tsamadou
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen and University Clinic Ulm, Germany.,Institute of Transfusion Medicine, University of Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen and University Clinic Ulm, Germany.,Institute of Transfusion Medicine, University of Ulm, Germany
| | | | - Joannis Mytilineos
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen and University Clinic Ulm, Germany .,Institute of Transfusion Medicine, University of Ulm, Germany.,DRST - German Registry for Stem Cell Transplantation, Germany
| |
Collapse
|
15
|
|
16
|
Moalic-Allain V. Medical and ethical considerations on hematopoietic stem cells mobilization for healthy donors. Transfus Clin Biol 2018; 25:136-143. [PMID: 29555414 DOI: 10.1016/j.tracli.2018.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 02/26/2018] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem cell transplantation is a common procedure potentially beneficial to many individuals with cancer, hematological, or inherited disorders, and has highlighted the need of related or unrelated donors to perform allograft. Donation of hematopoietic stem cells, either through bone marrow harvest or peripheral blood stem cell collection, is well-established and widespread. Over the past two decades, the peripheral blood stem cell collection by aphaeresis has become the main source of hematopoietic stem cells for transplantation, due to faster engraftment and practicability and lower risk of relapse for high-risk patients. For peripheral blood stem cell donation, donors require mobilization of hematopoietic stem cells from bone marrow into the blood stream. This is performed by growth factors injections. This article is a review of reported applications of growth factors (original granulocyte colony stimulating factor and its biosimilars), for healthy donors' peripheral blood stem cell mobilization, in terms of toxicity, side effects, efficacy and follow-up. There is still an ethical dilemma for clinicians involved in allograft, because they expose healthy donors to drugs. It is important to dispel some of the critical concerns regarding their use in healthy volunteers, particularly because they receive no personal therapeutic benefit from this procedure.
Collapse
Affiliation(s)
- V Moalic-Allain
- Laboratoire de génétique moléculaire et d'histocompatibilité, pôle de pathologie biologie, CHRU Morvan, bâtiment 5 bis, RDC, 2, avenue Foch, 29609 Brest cedex, France.
| |
Collapse
|
17
|
Cid AR, Montesinos P, Sánchez‐Guiu I, Haya S, Lorenzo JI, Sanz J, Moscardo F, Puig N, Planelles D, Bonanad S, Sanz GF, Vicente V, González‐Manchón C, Lozano ML, Rivera J, Sanz MA. Allogeneic hematopoietic cell transplantation in an adult patient with Glanzmann thrombasthenia. Clin Case Rep 2017; 5:1887-1890. [PMID: 29152293 PMCID: PMC5676254 DOI: 10.1002/ccr3.1206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 07/27/2017] [Accepted: 08/19/2017] [Indexed: 11/17/2022] Open
Abstract
Glanzmann thrombasthenia is a rare bleeding disorder that can present life‐threatening bleeding. Our patients develop antiplatelet antibodies that become refractory to any pharmacological treatment. Allogeneic hematopoietic stem‐cell transplantation is the only currently curative procedure, but has major risks mainly in adult; indeed, our patient died.
Collapse
Affiliation(s)
- Ana R. Cid
- Unidad de Hemostasia y TrombosisServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| | - Pau Montesinos
- Unidad de Trasplante de Células HematopoyéticasServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| | - Isabel Sánchez‐Guiu
- Servicio de Hematología y Oncología MédicaHospital Universitario Morales MeseguerCentro Regional de HemodonaciónUniversidad de Murcia, IMIB‐Arrixaca, CIBERERMurciaSpain
| | - Saturnino Haya
- Unidad de Hemostasia y TrombosisServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| | - Jose I. Lorenzo
- Unidad de Trasplante de Células HematopoyéticasServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| | - Jaime Sanz
- Unidad de Trasplante de Células HematopoyéticasServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| | - Federico Moscardo
- Unidad de Trasplante de Células HematopoyéticasServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| | - Nieves Puig
- Centro de Transfusión de la Comunidad ValencianaValenciaSpain
| | | | - Santiago Bonanad
- Unidad de Hemostasia y TrombosisServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| | - Guillermo F. Sanz
- Unidad de Trasplante de Células HematopoyéticasServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| | - Vicente Vicente
- Servicio de Hematología y Oncología MédicaHospital Universitario Morales MeseguerCentro Regional de HemodonaciónUniversidad de Murcia, IMIB‐Arrixaca, CIBERERMurciaSpain
| | - Consuelo González‐Manchón
- Departament Cellular and Molecular MedicineCentro de Investigaciones Biológicas (C.S.I.C.)MadridSpain
| | - María L. Lozano
- Servicio de Hematología y Oncología MédicaHospital Universitario Morales MeseguerCentro Regional de HemodonaciónUniversidad de Murcia, IMIB‐Arrixaca, CIBERERMurciaSpain
| | - José Rivera
- Servicio de Hematología y Oncología MédicaHospital Universitario Morales MeseguerCentro Regional de HemodonaciónUniversidad de Murcia, IMIB‐Arrixaca, CIBERERMurciaSpain
| | - Miguel A. Sanz
- Unidad de Hemostasia y TrombosisServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
- Unidad de Trasplante de Células HematopoyéticasServicio de HematologíaHospital Universitario y Politécnico La FeValenciaSpain
| |
Collapse
|
18
|
Moore RLL, Worrallo MJ, Mitchell PD, Harriman J, Glen KE, Thomas RJ. Immobilisation of Delta-like 1 ligand for the scalable and controlled manufacture of hematopoietic progenitor cells in a stirred bioreactor. BMC Biotechnol 2017; 17:65. [PMID: 28778182 PMCID: PMC5544980 DOI: 10.1186/s12896-017-0383-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022] Open
Abstract
Background Umbilical cord blood provides a source of hematopoietic stem cells for transplantation with immunological and availability advantages over conventional bone marrow sources. Limited cell numbers and slower engraftment from umbilical cord blood units has led to the clinical development of immobilised Notch ligand Delta-Like 1 to promote ex vivo expansion of a rapidly engrafting cell population. However, current immobilisation methods are not simple to scale in a controlled manner. Results Delta-Like 1 was immobilised onto streptavidin coated magnetic particles via a heterobifunctionalised polyethylene glycol linker molecule to provide an easily manipulated format of surface protein presentation. CD34+ enriched cord blood cells were treated with Delta-Like 1 immobilised particles, and immunophenotypic markers measured to monitor population distributions using cluster identification, characterization, and regression software. The amenability of the approach to scalability was evaluated in a micro-scale stirred tank bioreactor. Surface concentration of Delta-Like 1 was well controlled used differing stoichiometric reagent ratios. Protein immobilisation was a cost effective process and particles were efficiently removed from the final cell product. Immobilised Delta-Like 1 is functional and stimulates qualitatively similar CD34hi, CD38lo, CD90lo, CD133hi, CD135hi progenitor expansion in both static culture and scalable stirred culture platforms. Conclusions Immobilised Delta-Like 1 in this form has the potential to improve the manufacturing efficiency and control of final ex vivo expanded cell product through compatibility with highly controlled and characterised suspension culture systems. Electronic supplementary material The online version of this article (doi:10.1186/s12896-017-0383-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rebecca L L Moore
- Healthcare Engineering Research Group, Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, UK.
| | - Matthew J Worrallo
- Healthcare Engineering Research Group, Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, UK
| | - Peter D Mitchell
- Healthcare Engineering Research Group, Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, UK
| | - Jon Harriman
- Healthcare Engineering Research Group, Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, UK
| | - Katie E Glen
- Healthcare Engineering Research Group, Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, UK
| | - Robert J Thomas
- Healthcare Engineering Research Group, Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, UK
| |
Collapse
|
19
|
Kimura L, Alvarez G, Li N, Pawlikowska-Haddal A, Moore TB, Casillas J, Lee KW. Temporary resolution of insulin requirement in acquired partial lipodystrophy associated with chronic graft-versus-host disease. Pediatr Blood Cancer 2017; 64. [PMID: 28371314 DOI: 10.1002/pbc.26427] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/07/2016] [Accepted: 11/28/2016] [Indexed: 02/02/2023]
Abstract
This is a case presentation describing a high insulin requirement that suddenly resolved in a patient with acute lymphoblastic leukemia treated with stem cell transplantation complicated by chronic graft-versus-host disease. The patient was diagnosed with acquired partial lipodystrophy that did not require alternative therapies such as leptin or insulin-like growth factor 1.
Collapse
Affiliation(s)
- Leslie Kimura
- Department of Pediatric Endocrinology, UCLA Mattel Children's Hospital, Los Angeles, California
| | - Griselda Alvarez
- Department of Pediatric Endocrinology, Children's Hospital of Los Angeles, Los Angeles, California
| | - Ning Li
- Department of Biomathematics, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Anna Pawlikowska-Haddal
- Department of Pediatric Endocrinology, UCLA Mattel Children's Hospital, Los Angeles, California
| | - Theodore B Moore
- Department of Pediatric Hematology-Oncology, UCLA Mattel Children's Hospital, Los Angeles, California
| | - Jacqueline Casillas
- Department of Pediatric Hematology-Oncology, UCLA Mattel Children's Hospital, Los Angeles, California
| | | |
Collapse
|
20
|
Tang Y, Pan ZY, Zou Y, He Y, Yang PY, Tang QQ, Yin F. A comparative assessment of adipose-derived stem cells from subcutaneous and visceral fat as a potential cell source for knee osteoarthritis treatment. J Cell Mol Med 2017; 21:2153-2162. [PMID: 28374574 PMCID: PMC5571554 DOI: 10.1111/jcmm.13138] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/30/2016] [Indexed: 02/06/2023] Open
Abstract
The intra‐articular injection of adipose‐derived stem cells (ASCs) is a novel potential therapy for patients with osteoarthritis (OA). However, the efficacy of ASCs from different regions of the body remains unknown. This study investigated whether ASCs from subcutaneous or visceral adipose tissue provide the same improvement of OA. Mouse and human subcutaneous and visceral adipose tissue were excised for ASC isolation. Morphology, proliferation, surface markers and adipocyte differentiation of subcutaneous ASCs (S‐ASCs) and visceral ASCs (V‐ASCs) were analysed. A surgically induced rat model of OA was established, and 4 weeks after the operation, S‐ASCs, V‐ASCs or phosphate‐buffered saline (PBS, control) were injected into the articular cavity. Histology, immunohistochemistry and gene expression analyses were performed 6 weeks after ASC injection. The ability of ASCs to differentiate into chondrocytes was assessed by in vitro chondrogenesis, and the immunosuppressive activity of ASCs was evaluated by co‐culturing with macrophages. The proliferation of V‐ASCs was significantly greater than that of S‐ASCs, but S‐ASCs had the greater adipogenic capacity than V‐ASCs. In addition, the infracted cartilage treated with S‐ASCs showed significantly greater improvement than cartilage treated with PBS or V‐ASCs. Moreover, S‐ASCs showed better chondrogenic potential and immunosuppression in vitro. Subcutaneous adipose tissue is an effective cell source for cell therapy of OA as it promotes stem cell differentiation into chondrocytes and inhibits immunological reactions.
Collapse
Affiliation(s)
- Yan Tang
- Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhang-Yi Pan
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Zou
- Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yi He
- Department of Cardiothoracic Surgery, Xinhua Hospital, Shanghai Jiaotong University of Medicine College, Shanghai, China
| | - Peng-Yuan Yang
- Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qi-Qun Tang
- Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Feng Yin
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
21
|
Xu JY, Cai WY, Tian M, Liu D, Huang RC. Stem cell transplantation dose in patients with acute myocardial infarction: A meta-analysis. Chronic Dis Transl Med 2016; 2:92-101. [PMID: 29063029 PMCID: PMC5643746 DOI: 10.1016/j.cdtm.2016.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE To evaluate whether stem cell transplantation improves global left ventricular ejection fraction (LVEF) in patients with acute myocardial infarction (AMI), and to determine the appropriate stem cell therapy dose as well as the effective period after stem cell transplantation for therapy. METHODS A systematic literature search included Pubmed, MEDLINE, China National Knowledge Infrastructure (CNKI), Chinese Biomedical Literature Database (CBM), and Cochrane Evidence-Based Medicine databases. The retrieval time limit ranged from January 1990 to June 2016. We also obtained full texts through manual retrieval, interlibrary loan and document delivery service, or by contacting the authors directly. According to our inclusion and exclusion criteria, data were extracted independently by two evaluators. In case of disagreement, a joint discussion occurred and a third researcher was utilized. Data were analyzed quantitatively using Revman 5.2. Summary results are presented as the weighted mean difference (WMD) with 95% confidence intervals (CIs). We collected individual trial data and conducted a meta-analysis to compare changes in global left ventricular ejection fraction (ΔLVEF) after stem cell therapy. In this study, four subgroups were based on stem cell dose (≤1 × 107 cells, ≤1 × 108 cells, ≤1 × 109 cells, and ≤1 × 1010 cells) and three subgroups were based on follow-up time (<6 months, 6-12 months, and ≥12 months). RESULTS Thirty-four studies, which included 40 randomized controlled trials, were included in this meta-analysis, and 1927 patients were evaluated. Changes in global LVEF were significantly higher in the stem cell transplantation group than in the control group (95% CI: 2.35-4.26%, P < 0.01). We found no significant differences in ΔLVEF between the bone marrow stem cells (BMCs) group and control group when the dose of BMCs was ≤1 × 107 [ΔLVEF 95% CI: 0.12-3.96%, P = 0.04]. The ΔLVEF in the BMCs groups was significantly higher than in the control groups when the dose of BMCs was ≤1 × 108 [ΔLVEF 95% CI: 0.95-4.25%, P = 0.002] and ≤1 × 109 [ΔLVEF 95% CI: 2.31-4.20%, P < 0.01]. In addition, when the dose of BMCs was between 109 and 1010 cells, we did not observe any significant differences [ΔLVEF 95% CI: -0.99-11.82%, P = 0.10]. Our data suggest stem cell therapy improves cardiac function in AMI patients when treated with an appropriate dose of BMCs. CONCLUSION Stem cell transplantation after AMI could improve global LVEF. Stem cells may be effectively administered to patients with AMI doses between 108 and 109 cells.
Collapse
Affiliation(s)
- Jia-Ying Xu
- The Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, China
| | - Wen-Yuan Cai
- The Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, China
| | - Ming Tian
- The Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, China
| | - Dai Liu
- The Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, China
| | - Rong-Chong Huang
- The Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, China
| |
Collapse
|
22
|
|
23
|
Wagner JE, Brunstein CG, Boitano AE, DeFor TE, McKenna D, Sumstad D, Blazar BR, Tolar J, Le C, Jones J, Cooke MP, Bleul CC. Phase I/II Trial of StemRegenin-1 Expanded Umbilical Cord Blood Hematopoietic Stem Cells Supports Testing as a Stand-Alone Graft. Cell Stem Cell 2015; 18:144-55. [PMID: 26669897 DOI: 10.1016/j.stem.2015.10.004] [Citation(s) in RCA: 244] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/12/2015] [Accepted: 10/19/2015] [Indexed: 01/17/2023]
Abstract
Clinical application of umbilical cord blood (UCB) as a source of hematopoietic stem cells for transplantation is limited by low CD34+ cell dose, increased risk of graft failure, and slow hematopoietic recovery. While the cell dose limitation is partially mitigated by using two UCB units, larger-dosed single units would be preferable. We have evaluated the feasibility and safety of StemRegenin-1 (SR-1), an aryl hydrocarbon receptor antagonist that expands CD34+ cells, by placing one of the two units in expansion culture. SR-1 produced a 330-fold increase in CD34+ cells and led to engraftment in 17/17 patients at a median of 15 days for neutrophils and 49 days for platelets, significantly faster than in patients treated with unmanipulated UCB. Taken together, the marked expansion, absence of graft failure, and enhanced hematopoietic recovery support testing of SR-1 expansion as a stand-alone graft and suggest it may ameliorate a limitation of UCB transplant.
Collapse
Affiliation(s)
- John E Wagner
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN 55455, USA; Molecular and Cellular Therapeutics Facility, University of Minnesota, Minneapolis, MN 55108, USA.
| | - Claudio G Brunstein
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anthony E Boitano
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Todd E DeFor
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - David McKenna
- Molecular and Cellular Therapeutics Facility, University of Minnesota, Minneapolis, MN 55108, USA
| | - Darin Sumstad
- Molecular and Cellular Therapeutics Facility, University of Minnesota, Minneapolis, MN 55108, USA
| | - Bruce R Blazar
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jakub Tolar
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chap Le
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Julie Jones
- Novartis Institutes for BioMedical Research, Basel 4000, Switzerland
| | - Michael P Cooke
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Conrad C Bleul
- Novartis Institutes for BioMedical Research, Basel 4000, Switzerland
| |
Collapse
|
24
|
Choi JS, Mahadik BP, Harley BAC. Engineering the hematopoietic stem cell niche: Frontiers in biomaterial science. Biotechnol J 2015; 10:1529-45. [PMID: 26356030 PMCID: PMC4724421 DOI: 10.1002/biot.201400758] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/15/2015] [Accepted: 07/16/2015] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSCs) play a crucial role in the generation of the body's blood and immune cells. This process takes place primarily in the bone marrow in specialized 'niche' microenvironments, which provide signals responsible for maintaining a balance between HSC quiescence, self-renewal, and lineage specification required for life-long hematopoiesis. While our understanding of these signaling mechanisms continues to improve, our ability to engineer them in vitro for the expansion of clinically relevant HSC populations is still lacking. In this review, we focus on development of biomaterials-based culture platforms for in vitro study of interactions between HSCs and their local microenvironment. The tools and techniques used for both examining HSC-niche interactions as well as applying these findings towards controlled HSC expansion or directed differentiation in 2D and 3D platforms are discussed. These novel techniques hold the potential to push the existing boundaries of HSC cultures towards high-throughput, real-time, and single-cell level biomimetic approaches that enable a more nuanced understanding of HSC regulation and function. Their application in conjunction with innovative biomaterial platforms can pave the way for engineering artificial bone marrow niches for clinical applications as well as elucidating the pathology of blood-related cancers and disorders.
Collapse
Affiliation(s)
- Ji Sun Choi
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bhushan P Mahadik
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
25
|
Huang W, Yu L, Cao T, Li Y, Liu Z, Li H, Bo J, Zhao Y, Jing Y, Wang S, Zhu H, Dou L, Wang Q, Gao C. The efficacy and safety of rabbit anti-thymocyte globulin vs rabbit anti-T-lymphocyte globulin in peripheral blood stem cell transplantation from unrelated donors. Leuk Lymphoma 2015; 57:355-363. [PMID: 26118935 DOI: 10.3109/10428194.2015.1045901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The comparative efficacy and safety of antithymocyte globulin (ATG) at fixed doses in patients undergoing allogeneic peripheral blood stem cell transplantation from unrelated donors (UR-PBSCT) has not been evaluated. In this study, the records of 56 patients and 54 patients who received pre-transplant ATG-Thymoglobulin (ATG-T) at a total dose of 10 mg/kg and ATG- Fresenius (ATG-F) at a total dose of 20 mg/kg, respectively, were retrospectively analyzed. ATG-F patients had a significantly lower probability of developing chronic graft-vs-host disease (cGVHD) than those treated with ATG-T (p = 0.04). ATG-F was associated with a non-significant trend towards lower relapse rates and higher survival at 3- and 5-years of follow-up compared with ATG-T. A significantly greater proportion of ATG-T patients experienced chills and high fever than ATG-F patients (p < 0.01). The current findings suggest that ATG-F may more effectively and safely prevent cGVHD without increasing relapse rates in patients undergoing UR-PBSCT.
Collapse
Affiliation(s)
- Wenrong Huang
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China.,b Department of Hematology , Hainan Branch of Chinese PLA General Hospital , Hainan province , PR China
| | - Li Yu
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Tingting Cao
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Yanfen Li
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Zhanxiang Liu
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Honghua Li
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Jian Bo
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Yu Zhao
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Yu Jing
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Shuhong Wang
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Haiyan Zhu
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Liping Dou
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Qunshun Wang
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Chunji Gao
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| |
Collapse
|
26
|
Kuchma MD, Kyryk VM, Svitina HM, Shablii YM, Lukash LL, Lobyntseva GS, Shablii VA. Comparative Analysis of the Hematopoietic Progenitor Cells from Placenta, Cord Blood, and Fetal Liver, Based on Their Immunophenotype. BIOMED RESEARCH INTERNATIONAL 2015; 2015:418752. [PMID: 26347038 PMCID: PMC4540977 DOI: 10.1155/2015/418752] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/03/2015] [Accepted: 07/08/2015] [Indexed: 11/18/2022]
Abstract
We have investigated the characteristics of human hematopoietic progenitor cells (HPCs) with the CD34(+)CD45(low)SSC(low) phenotype from full-term placental tissue (FTPT) as compared to cord blood (CB) and fetal liver (FL) cells. We demonstrated the presence of cell subpopulations at various stages of the differentiation with such immunophenotypes as CD34(+/low)CD45(low/-), CD34(++)CD45(low/-), CD34(+++)CD45(low/-), CD34(+/low)CD45(hi), and CD34(++)CD45(hi) in both first trimester placental tissue (FiTPT) and FTPT which implies their higher phenotypic heterogeneity compared to CB. HPCs of the FTPT origin expressed the CD90 antigen at a higher level compared to its expression by the CB HPCs and the CD133 antigen expression being at the same level in both cases. The HPCs compartment of FTPT versus CB contained higher number of myeloid and erythroid committed cells but lower number of myeloid and lymphoid ones compared to FL HPCs. HPCs of the FTPT and CB origin possess similar potentials for the multilineage differentiation in vitro and similar ratios of myeloid and erythroid progenitors among the committed cells. This observation suggests that the active hematopoiesis occurs in the FTPT. We obtained viable HPCs from cryopreserved placental tissue fragments allowing us to develop procedures for banking and testing of placenta-derived HPCs for clinical use.
Collapse
Affiliation(s)
- Maria D. Kuchma
- Institute of Cell Therapy, Komarova Avenue 3, Kyiv 03680, Ukraine
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Zabolotnogo Street 150, Kyiv 03680, Ukraine
| | - Vitaliy M. Kyryk
- State Institute of Genetics and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, Vyshgorodska Street 67, Kyiv 04114, Ukraine
| | - Hanna M. Svitina
- Institute of Cell Therapy, Komarova Avenue 3, Kyiv 03680, Ukraine
| | - Yulia M. Shablii
- Institute of Cell Therapy, Komarova Avenue 3, Kyiv 03680, Ukraine
| | - Lubov L. Lukash
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Zabolotnogo Street 150, Kyiv 03680, Ukraine
| | | | | |
Collapse
|
27
|
Wang B, Hu Y, Liu L, Hu K, Tie R, He Y, Fu S, Zhu N, Luo Y, Yu X, Huang H. Phenotypical and functional characterization of bone marrow mesenchymal stem cells in patients with chronic graft-versus-host disease. Biol Blood Marrow Transplant 2015; 21:1020-8. [PMID: 25708216 DOI: 10.1016/j.bbmt.2015.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 02/10/2015] [Indexed: 12/13/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is a critical complication after allogeneic hematopoietic stem cell transplantation. The conditioning therapy has been involved in the impairment of bone marrow (BM) mesenchymal stem/stromal cells (MSCs). However, the potential implication of MSCs in the pathophysiology of cGVHD has not been investigated. We analyzed expanded MSCs from patients with cGVHD and compared them with those from transplantation patients without cGVHD. The MSCs from both groups were of host origin and their reserves were comparable. They showed similar morphology, immunophenotype, population doubling times, self-renewal capacity, differentiation, and migration potential. The immunomodulatory potential of the 2 groups was also identical, they were both capable of inhibiting phytohemagglutinin-activated peripheral blood mononuclear cells (PBMCs) proliferation and inducing regulatory T cells after coculturing with CD4(+) T cells, and the immunosuppressive factors were secreted similarly in both MSCs whether in normal culture or coculture with PBMCs. No significant differences were observed in the cellular senescence and apoptosis between 2 groups. In addition, MSCs from patients with cGVHD displayed normal phenotype and function compared with their counterparts from healthy donors, although reduced frequency in BM mononuclear cell fraction was observed in these patients. Taken together, our results suggest that MSCs do not seem to contribute to the pathogenesis of cGVHD and indicate the feasibility of autologous cell therapy in patients who are not completely responding to standard immunosuppressive therapy for cGVHD.
Collapse
Affiliation(s)
- Binsheng Wang
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lizhen Liu
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kaimin Hu
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying He
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shan Fu
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ni Zhu
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Luo
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohong Yu
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
28
|
Cord blood collection and banking from a population with highly diverse geographic origins increase HLA diversity in the registry and do not lower the proportion of validated cord blood units: experience of the Marseille Cord Blood Bank. Bone Marrow Transplant 2015; 50:531-5. [PMID: 25621799 DOI: 10.1038/bmt.2014.314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/09/2014] [Accepted: 11/29/2014] [Indexed: 12/11/2022]
Abstract
Several Cord Blood (CB) Bank studies suggested that ethnicity impaired CB unit (CBU) qualification. The Bone Marrow Donors Worldwide registries present an over-representation of unrelated donors (UD) from Northwestern European descent. This raises the question of equality of access to hematopoietic stem cells transplant, especially in the Mediterranean zone, which has taken in many waves of immigration. The aim of our study is to address whether, in the Marseille CB Bank, CBU qualification rate is impaired by geographic origin. The study compared biological characteristics of 106 CBU disqualified for total nucleated cell (TNC) count (dCBU) and 136 qualified CBU in relation to registry enrichment and haplotype origin. A high proportion (>80%) of both dCBU and CBU had at least one non-European haplotype and enrich CB and UD registries to a higher extent than those with two European haplotypes (P<0.001). No difference was observed between TNC count and volume according to geographic origin. Our study shows that diverse Mediterranean origins do not have an impact on the CBU qualification rate. Partnership with Mediterranean birth clinics with highly trained staff is a reasonable option to increase the HLA diversity of CB Bank inventories and to improve the representation of minorities.
Collapse
|
29
|
Outcomes of peripheral blood stem cell transplantation patients from HLA-mismatched unrelated donor with antithymocyte globulin (ATG)-Thymoglobulin versus ATG-Fresenius: a single-center study. Med Oncol 2015; 32:465. [PMID: 25588925 DOI: 10.1007/s12032-014-0465-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 12/16/2014] [Indexed: 10/24/2022]
Abstract
Although antithymocyte globulin (ATG) had been widely used in hematopoietic stem cell transplantation from unrelated donor due to its ability to prevent acute and chronic graft-versus-host disease (GVHD), the comparative efficacy and safety of ATG-Thymoglobulin (ATG-T) and ATG-Fresenius (ATG-F) in patients undergoing HLA-mismatched allogeneic peripheral blood stem cell transplantation from unrelated donors (UR-PBSCT) has not been evaluated. Retrospective analysis of patients who underwent HLA-mismatched UR-PBSCT between January 2003 and December 2013 and received pre-transplant ATG-T at a total dose of 10 mg/kg or ATG-F at a total dose of 20 mg/kg was performed. Patients who received ATG-T (n = 23) or ATG-F (n = 28) had similar baseline demographic, disease, and transplant characteristics. There were no significant between-groups differences in the probability of acute GVHD (P = 0.721) and chronic GVHD (P = 0.439). ATG-F was associated with nonsignificant trends toward higher disease-free survival at 3-year follow-up compared with ATG-T (45.7 ± 11.1 vs 61.3 ± 9.7 %, respectively, P = 0.07). A significantly greater proportion of ATG-T patients experienced high fever than ATG-F patients (P < 0.01) during ATG infusion. There was no difference in the rate of infection between the two treatment groups. There were less adverse effects comparing ATG-F with ATG-T. ATG-T at a total dose of 10 mg/kg and ATG-F at a total dose of 20 mg/kg had a similar clinical outcome in the setting of HLA-mismatched UR-PBSCT.
Collapse
|
30
|
Wagner JE, Eapen M, Carter S, Wang Y, Schultz KR, Wall DA, Bunin N, Delaney C, Haut P, Margolis D, Peres E, Verneris MR, Walters M, Horowitz MM, Kurtzberg J. One-unit versus two-unit cord-blood transplantation for hematologic cancers. N Engl J Med 2014; 371:1685-94. [PMID: 25354103 PMCID: PMC4257059 DOI: 10.1056/nejmoa1405584] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Umbilical-cord blood has been used as the source of hematopoietic stem cells in an estimated 30,000 transplants. The limited number of hematopoietic cells in a single cord-blood unit prevents its use in recipients with larger body mass and results in delayed hematopoietic recovery and higher mortality. Therefore, we hypothesized that the greater numbers of hematopoietic cells in two units of cord blood would be associated with improved outcomes after transplantation. METHODS Between December 1, 2006, and February 24, 2012, a total of 224 patients 1 to 21 years of age with hematologic cancer were randomly assigned to undergo double-unit (111 patients) or single-unit (113 patients) cord-blood transplantation after a uniform myeloablative conditioning regimen and immunoprophylaxis for graft-versus-host disease (GVHD). The primary end point was 1-year overall survival. RESULTS Treatment groups were matched for age, sex, self-reported race (white vs. nonwhite), performance status, degree of donor-recipient HLA matching, and disease type and status at transplantation. The 1-year overall survival rate was 65% (95% confidence interval [CI], 56 to 74) and 73% (95% CI, 63 to 80) among recipients of double and single cord-blood units, respectively (P=0.17). Similar outcomes in the two groups were also observed with respect to the rates of disease-free survival, neutrophil recovery, transplantation-related death, relapse, infections, immunologic reconstitution, and grade II-IV acute GVHD. However, improved platelet recovery and lower incidences of grade III and IV acute and extensive chronic GVHD were observed among recipients of a single cord-blood unit. CONCLUSIONS We found that among children and adolescents with hematologic cancer, survival rates were similar after single-unit and double-unit cord-blood transplantation; however, a single-unit cord-blood transplant was associated with better platelet recovery and a lower risk of GVHD. (Funded by the National Heart, Lung, and Blood Institute and the National Cancer Institute; ClinicalTrials.gov number, NCT00412360.).
Collapse
Affiliation(s)
- John E Wagner
- From the University of Minnesota Medical School, Minneapolis (J.E.W., M.R.V.); Medical College of Wisconsin, Milwaukee (M.E., D.M., M.M.H.); EMMES Corporation, Rockville, MD (S.C., Y.W.); BC Children's Hospital, Vancouver (K.R.S.), and University of Manitoba, Winnipeg (D.A.W.) - both in Canada; Children's Hospital of Philadelphia, Philadelphia (N.B.); Fred Hutchinson Cancer Research Center, Seattle (C.D.); Indiana University, Indianapolis (P.H.); University of Michigan, Ann Arbor (E.P.); University of California San Francisco Benioff Children's Hospital Oakland, Oakland (M.W.); and Duke University, Durham, NC (J.K.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hematopoietic stem cell niche maintenance during homeostasis and regeneration. Nat Med 2014; 20:833-46. [PMID: 25100529 DOI: 10.1038/nm.3647] [Citation(s) in RCA: 584] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 07/03/2014] [Indexed: 02/08/2023]
Abstract
The bone marrow niche has mystified scientists for many years, leading to widespread investigation to shed light into its molecular and cellular composition. Considerable efforts have been devoted toward uncovering the regulatory mechanisms of hematopoietic stem cell (HSC) niche maintenance. Recent advances in imaging and genetic manipulation of mouse models have allowed the identification of distinct vascular niches that have been shown to orchestrate the balance between quiescence, proliferation and regeneration of the bone marrow after injury. Here we highlight the recently discovered intrinsic mechanisms, microenvironmental interactions and communication with surrounding cells involved in HSC regulation, during homeostasis and in regeneration after injury and discuss their implications for regenerative therapy.
Collapse
|
32
|
Mohty M, Malard F, Savani BN. High-dose total body irradiation and myeloablative conditioning before allogeneic hematopoietic cell transplantation: time to rethink? Biol Blood Marrow Transplant 2014; 21:620-4. [PMID: 25246296 DOI: 10.1016/j.bbmt.2014.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/15/2014] [Indexed: 10/24/2022]
Abstract
Over the last decade, the care of patients undergoing allogeneic hematopoietic cell transplantation (allo-HCT) has significantly improved, leading to a decrease in deaths related to allo-HCT as well as improved long-term survival. However, for many patients, long-term survivorship is associated with a substantial burden of chronic morbidities. Indeed, malignant and nonmalignant late complications after allo-HCT are numerous and usually multifactorial, with all organs and tissues a potential target. In many cases, these long-term side effects are associated with the use of high-dose total body irradiation, myeloablative conditioning regimens, and the onset of chronic graft-versus-host disease. It appears to be essential to change the natural history of these late effects. This requires the introduction of improved conditioning regimens and the development of lifelong monitoring controls, patient counseling, and preventative treatment measures. This approach will allow us to pursue our efforts to improve patient outcome.
Collapse
Affiliation(s)
- Mohamad Mohty
- Department of Haematology, Saint Antoine Hospital, Paris, France; INSERM UMR 938, Paris, France; Université Pierre et Marie Curie, Paris, France.
| | - Florent Malard
- Department of Haematology, Saint Antoine Hospital, Paris, France; INSERM UMR 938, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Bipin N Savani
- Hematology and Stem Cell Transplantation Section, Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center and Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
33
|
Sousa H, Boutolleau D, Ribeiro J, Teixeira AL, Pinho Vaz C, Campilho F, Branca R, Campos A, Baldaque I, Medeiros R. Cytomegalovirus infection in patients who underwent allogeneic hematopoietic stem cell transplantation in Portugal: a five-year retrospective review. Biol Blood Marrow Transplant 2014; 20:1958-67. [PMID: 25139217 DOI: 10.1016/j.bbmt.2014.08.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/12/2014] [Indexed: 02/03/2023]
Abstract
Cytomegalovirus (CMV) infection is 1 of the leading causes of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (aHSCT), mainly within the first 100 days after transplantation. We aimed to characterize CMV infection in a cohort of 305 patients with different malignancies undergoing aHSCT at the Portuguese Institute of Oncology of Porto between January 2008 and December 2012. In total, 184 patients (60.3%) developed CMV infection, mainly viral reactivations rather than primary infections (96.2% versus 3.8%, respectively). The majority of patients (166 of 184) developed CMV infection ≤100 days after transplantation, with median time to infection of 29 days (range, 0 to 1285) and median duration of infection of 10 days (range, 2 to 372). Multivariate analysis revealed that CMV infection was increased in donor (D)-/recipient (R)+ and D+/R+ (odds ratio [OR], 10.5; 95% confidence interval [CI], 4.35 to 25.4; P < .001) and in patients with mismatched or unrelated donors (OR, 2.54; 95% CI, 1.34 to 4.80; P = .004). Cox regression model showed that the risk of death was significantly increased in patients >38 years old (OR, 1.89; 95% CI, 1.14 to 3.12; P = .0137), who underwent transplantation with peripheral blood (OR, 3.02; 95% CI, 1.33 to 6.86; P = .008), with mismatched or unrelated donor (OR, 2.16; 95% CI, 1.48 to 3.13; P < .001), and who developed CMV infection (OR, 1.76; 95% CI, 1.07 to 2.90; P = .025). Moreover, patients who developed CMV infection had a significantly reduced median post-transplantation survival (16 versus 36 months; P = .002).
Collapse
Affiliation(s)
- Hugo Sousa
- Virology Service, Portuguese Institute of Oncology of Porto, Porto, Portugal; Molecular Oncology Group, Portuguese Institute of Oncology of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal.
| | - David Boutolleau
- Sorbonne Universités, UPMC Université Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France; INSERM, U1135, CIMI-Paris, Paris, France; AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière - Charles Foix, Service de Virologie, Paris, France
| | - Joana Ribeiro
- Virology Service, Portuguese Institute of Oncology of Porto, Porto, Portugal; Molecular Oncology Group, Portuguese Institute of Oncology of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ana L Teixeira
- Virology Service, Portuguese Institute of Oncology of Porto, Porto, Portugal; Molecular Oncology Group, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Carlos Pinho Vaz
- Bone Marrow Transplantation Unit, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Fernando Campilho
- Bone Marrow Transplantation Unit, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Rosa Branca
- Bone Marrow Transplantation Unit, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - António Campos
- Bone Marrow Transplantation Unit, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Inês Baldaque
- Virology Service, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Rui Medeiros
- Virology Service, Portuguese Institute of Oncology of Porto, Porto, Portugal; Molecular Oncology Group, Portuguese Institute of Oncology of Porto, Porto, Portugal; Research Department, Portuguese League Against Cancer (LPCC-NRNorte), Porto, Portugal
| |
Collapse
|
34
|
Domingo-Gonzalez R, Moore BB. Innate Immunity Post-Hematopoietic Stem Cell Transplantation: Focus on Epigenetics. ACTA ACUST UNITED AC 2014; 5:189-197. [PMID: 26709355 DOI: 10.3233/nib-140079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epigenetic regulation of gene expression is important for normal biological processes like immune cell development, immune responses, and differentiation from hematopoietic stem cells. Furthermore, it is well understood that epigenetic mechanisms can include methylation, histone modification, and more recently, microRNAs. Interestingly, aberrant epigenetic modification can also promote pathology in many diseases like cancer. The effects of methylation on gene expression and its resulting phenotype have been extensively studied. In this review, we discuss the inhibition of innate immunity that occurs in humans and animal models post-stem cell transplant. In addition, we highlight the changes methylation and microRNA profiles have on regulating pulmonary innate immune responses in the context of hematopoietic stem cell transplantation in experimental animal models.
Collapse
Affiliation(s)
| | - Bethany B Moore
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA ; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|