1
|
Mohseni R, Mahdavi Sharif P, Behfar M, Modaresi MR, Shirzadi R, Mardani M, Jafari L, Jafari F, Nikfetrat Z, Hamidieh AA. Evaluation of safety and efficacy of allogeneic adipose tissue-derived mesenchymal stem cells in pediatric bronchiolitis obliterans syndrome (BoS) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Stem Cell Res Ther 2023; 14:256. [PMID: 37726865 PMCID: PMC10510238 DOI: 10.1186/s13287-023-03498-y] [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] [Received: 08/06/2023] [Accepted: 09/13/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Allo-HSCT is a definite approach for the management of a wide variety of lethal and debilitating malignant and non-malignant disorders. However, its two main complications, acute and chronic graft-versus-host disease (GVHD), exert significant morbidities and mortalities. BoS, as a manifestation of chronic lung GVHD, is a gruesome complication of allo-HSCT, and for those with steroid-refractory disease, no approved second-line therapies exist. Mesenchymal stem cells (MSCs) exert anti-inflammatory and growth-promoting effects, and their administration against a wide range of inflammatory and neurologic disorders, as well as GVHD, has been associated with promising outcomes. However, literature on the safety and effectiveness of MSC therapy for BoS and pediatric cGVHD is scarce. METHODS We designed a single-arm trial to administer adipose tissue (AT)-derived MSCs to pediatric patients with refractory BoS after allo-HSCT. AT-MSCs from obese, otherwise healthy donors were cultured in an ISO class 1 clean room and injected into the antecubital vein of eligible patients with a dose of 1 × 106/kg. The primary endpoints included a complete or partial response to therapy [in terms of increased forced expiratory volume in one second (FEV1) values and steroid dose reduction] and its safety profile. RESULTS Four eligible patients with a median age of 6.5 years were enrolled in the study. Steroid-induced osteoporosis and myopathy were present in three cases. A partial response was evident in three cases after a single injection of AT-MSCs. The treatment was safe and tolerable, and no treatment-related adverse events were noted. Two patients developed manageable COVID-19 infections one and 4 months after AT-MSC injection. After a median follow-up duration of 19 months, all cases are still alive and have had no indications for lung transplantation. CONCLUSIONS AT-MSCs could be safely administered to our pediatric cases with BoS post-allo-HSCT. Considering their advanced stage of disease, their sub-optimal functional capacity due to steroid-induced complications, and COVID-19 infection post-treatment, we believe that AT-MSC therapy can have possible efficacy in the management of pediatric BoS. The conduction of further studies with larger sample sizes and more frequent injections is prudent for further optimization of AT-MSC therapy against BoS. Trial registration Iranian Registry of Clinical Trials (IRCT), IRCT20201202049568N2. Registered 22 February 2021, https://en.irct.ir/trial/53143 .
Collapse
Affiliation(s)
- Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Pouya Mahdavi Sharif
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Mohammad Reza Modaresi
- Pediatric Respiratory and Sleep Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Rohola Shirzadi
- Pediatric Pulmonary Disease and Sleep Medicine Research Center, Pediatric Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahta Mardani
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Leila Jafari
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Fahimeh Jafari
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Zeynab Nikfetrat
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran.
| |
Collapse
|
2
|
Wuttisarnwattana P, Eck BL, Gargesha M, Wilson DL. Optimal slice thickness for improved accuracy of quantitative analysis of fluorescent cell and microsphere distribution in cryo-images. Sci Rep 2023; 13:10907. [PMID: 37407807 DOI: 10.1038/s41598-023-37927-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 06/29/2023] [Indexed: 07/07/2023] Open
Abstract
Cryo-imaging has been effectively used to study the biodistribution of fluorescent cells or microspheres in animal models. Sequential slice-by-slice fluorescent imaging enables detection of fluorescent cells or microspheres for corresponding quantification of their distribution in tissue. However, if slices are too thin, there will be data overload and excessive scan times. If slices are too thick, then cells can be missed. In this study, we developed a model for detection of fluorescent cells or microspheres to aid optimal slice thickness determination. Key factors include: section thickness (X), fluorescent cell intensity (Ifluo), effective tissue attenuation coefficient (μT), and a detection threshold (T). The model suggests an optimal slice thickness value that provides near-ideal sensitivity while minimizing scan time. The model also suggests a correction method to compensate for missed cells in the case that image data were acquired with overly large slice thickness. This approach allows cryo-imaging operators to use larger slice thickness to expedite the scan time without significant loss of cell count. We validated the model using real data from two independent studies: fluorescent microspheres in a pig heart and fluorescently labeled stem cells in a mouse model. Results show that slice thickness and detection sensitivity relationships from simulations and real data were well-matched with 99% correlation and 2% root-mean-square (RMS) error. We also discussed the detection characteristics in situations where key assumptions of the model were not met such as fluorescence intensity variation and spatial distribution. Finally, we show that with proper settings, cryo-imaging can provide accurate quantification of the fluorescent cell biodistribution with remarkably high recovery ratios (number of detections/delivery). As cryo-imaging technology has been used in many biological applications, our optimal slice thickness determination and data correction methods can play a crucial role in further advancing its usability and reliability.
Collapse
Affiliation(s)
- Patiwet Wuttisarnwattana
- Biomedical Engineering Institute, Department of Computer Engineering, Excellence Center in Infrastructure Technology and Transportation Engineering, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Brendan L Eck
- Imaging Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | | | - David L Wilson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
3
|
Klein OR, Ktena YP, Pierce E, Fu HH, Haile A, Liu C, Cooke KR. Defibrotide modulates pulmonary endothelial cell activation and protects against lung inflammation in pre-clinical models of LPS-induced lung injury and idiopathic pneumonia syndrome. Front Immunol 2023; 14:1186422. [PMID: 37441074 PMCID: PMC10335747 DOI: 10.3389/fimmu.2023.1186422] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction A multiple organ dysfunction syndrome (MODS) workshop convened by the National Institute of Child Health and Human Development in 2015 identified acute respiratory distress syndrome (ARDS) and complications of allogeneic blood and marrow transplantation (allo-BMT) as contributors to MODS in pediatric patients. Pulmonary dysfunction also remains a significant complication of allo-BMT. Idiopathic pneumonia syndrome (IPS) defines non-infectious, acute, lung injury that occurs post-transplant. Injury and activation to endothelial cells (ECs) contribute to each form of lung inflammation. Methods Two murine models were employed. In an ARDS model, naïve B6 mice receive an intravenous (i.v.) injection of lipopolysaccharide (LPS). In the established model of IPS, naïve B6D2F1 mice receive lethal total body irradiation followed by BMT from either allogeneic (B6) or syngeneic (B6D2F1) donors. Lung inflammation was subsequently assessed in each scenario. Results Intravenous injection of LPS to B6 mice resulted in enhanced mRNA expression of TNFα, IL-6, Ang-2, E-, and P-selectin in whole lung homogenates. The expression of Ang-2 in this context is regulated in part by TNFα. Additionally, EC activation was associated with increased total protein and cellularity in broncho-alveolar lavage fluid (BALF). Similar findings were noted during the development of experimental IPS. We hypothesized that interventions maintaining EC integrity would reduce the severity of ARDS and IPS. Defibrotide (DF) is FDA approved for the treatment of BMT patients with sinusoidal obstruction syndrome and renal or pulmonary dysfunction. DF stabilizes activated ECs and protect them from further injury. Intravenous administration of DF before and after LPS injection significantly reduced mRNA expression of TNFα, IL6, Ang-2, E-, and P-selectin compared to controls. BALF showed decreased cellularity, reflecting less EC damage and leak. Allogeneic BMT mice were treated from day -1 through day 14 with DF intraperitoneally, and lungs were harvested at 3 weeks. Compared to controls, DF treatment reduced mRNA expression of TNFα, IL6, Ang-2, E-, and P- selectin, BALF cellularity, and lung histopathology. Conclusion The administration of DF modulates EC injury in models of ARDS and IPS. Cytokine inhibition in combination with agents that stabilize EC integrity may be an attractive strategy for patients in each setting.
Collapse
Affiliation(s)
- Orly R. Klein
- Department of Oncology, Pediatric Blood and Marrow Transplant Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Yiouli P. Ktena
- Department of Oncology, Pediatric Blood and Marrow Transplant Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Elizabeth Pierce
- Department of Pediatrics, Pediatric Blood and Marrow Transplant Program, Case Western Reserve University, School of Medicine, Cleveland, OH, United States
| | - Han-Hsuan Fu
- Department of Oncology, Pediatric Blood and Marrow Transplant Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Azeb Haile
- Department of Oncology, Pediatric Blood and Marrow Transplant Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States
| | - Kenneth R. Cooke
- Department of Oncology, Pediatric Blood and Marrow Transplant Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| |
Collapse
|
4
|
Liu Y, Huang W, Wang H, Lu W, Guo J, Yu L, Wang L. Influence of SPIO labelling on the function of BMSCs in chemokine receptors expression and chemotaxis. PeerJ 2023; 11:e15388. [PMID: 37283891 PMCID: PMC10241165 DOI: 10.7717/peerj.15388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/19/2023] [Indexed: 06/08/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) are increasingly being used in bone marrow transplantation (BMT) to enable homing of the allogeneic hematopoietic stem cells and suppress acute graft versus host disease (aGVHD). The aim of this study was to optimize the labelling of BMSCs with superparamagnetic iron oxide particles (SPIOs), and evaluate the impact of the SPIOs on the biological characteristics, gene expression profile and chemotaxis function of the BMSCs. The viability and proliferation rates of the SPIO-labeled BMSCs were analyzed by trypan blue staining and CCK-8 assay respectively, and the chemotaxis function was evaluated by the transwell assay. The expression levels of chemokine receptors were measured by RT-PCR and flow cytometry. The SPIOs had no effect on the viability of the BMSCs regardless of the labelling concentration and culture duration. The labelling rate of the cells was higher when cultured for 48 h with the SPIOs. Furthermore, cells labeled with 25 µg/ml SPIOs for 48 h had the highest proliferation rates, along with increased expression of chemokine receptor genes and proteins. However, there was no significant difference between the chemotaxis function of the labeled and unlabeled BMSCs. To summarize, labelling BMSCs with 25 µg/ml SPIOs for 48h did not affect their biological characteristics and chemotaxis function, which can be of significance for in vivo applications.
Collapse
Affiliation(s)
- Yuanchun Liu
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wanyi Huang
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huiyang Wang
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wei Lu
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiayu Guo
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Li Yu
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lina Wang
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
5
|
Wuttisarnwattana P, Auephanwiriyakul S. Spleen Tissue Segmentation Algorithm for Cryo-Imaging Data. J Digit Imaging 2023; 36:588-602. [PMID: 36441277 PMCID: PMC10039202 DOI: 10.1007/s10278-022-00736-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/29/2022] Open
Abstract
Spleen tissue segmentation is an essential process for analyzing various immunological diseases as observed in the cryo-imaging data. Because manual labeling of the spleen tissue by human experts is not efficient, an automatic segmentation algorithm is needed. In this study, we developed a novel algorithm for automatically segmenting spleen substructures including white pulp and red pulp for the first time. The algorithm is designed for datasets created by a cryo-imaging system. This unique technology can effectively enable cellular tracking anywhere in the whole mouse with single-cell sensitivity. The proposed algorithm consists of four components: initial spleen mask creation, feature extraction, Supervised Patch-based Fuzzy c-Mean (spFCM) classification, and post-processing. The algorithm accurately and efficiently labeled spleen tissues in all experiment settings. The algorithm also improved the spleen segmentation throughput by 90 folds as compared to the manual segmentation. Moreover, we show that our novel spFCM algorithm outperformed traditional fast-learning classifiers as well as the U-Net deep-learning model in many aspects. Two major contributions of this paper are (1) an explainable algorithm for segmenting spleen tissues in cryo-images for the first time and (2) an spFCM algorithm as a new classifier. We also discussed that our work can be beneficial to researchers who work not only in the fields of graft-versus-host disease (GVHD) mouse models, but also in that of other immunological disease models where spleen analysis is essential. Future work building upon our research may lay the foundations for biomedical studies that utilize cryo-imaging technology.
Collapse
Affiliation(s)
- Patiwet Wuttisarnwattana
- Department of Computer Engineering, Faculty of Engineering, Chiang Mai University, Chiang Mai, 50300, Thailand.
- Optimization Theory and Applications for Engineering Systems Research Group (OASYS), Chiang Mai University, Chiang Mai, 50300, Thailand.
- Excellence Center in Infrastructure Technology and Transportation Engineering (ExCITE), Chiang Mai University, Chiang Mai, 50300, Thailand.
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai, 50300, Thailand.
| | - Sansanee Auephanwiriyakul
- Department of Computer Engineering, Faculty of Engineering, Chiang Mai University, Chiang Mai, 50300, Thailand.
- Excellence Center in Infrastructure Technology and Transportation Engineering (ExCITE), Chiang Mai University, Chiang Mai, 50300, Thailand.
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai, 50300, Thailand.
| |
Collapse
|
6
|
Li H, Dai H, Li J. Immunomodulatory properties of mesenchymal stromal/stem cells: The link with metabolism. J Adv Res 2023; 45:15-29. [PMID: 35659923 PMCID: PMC10006530 DOI: 10.1016/j.jare.2022.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/17/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) are the most promising stem cells for the treatment of multiple inflammatory and immune diseases due to their easy acquisition and potent immuno-regulatory capacities. These immune functions mainly depend on the MSC secretion of soluble factors. Recent studies have shown that the metabolism of MSCs plays critical roles in immunomodulation, which not only provides energy and building blocks for macromolecule synthesis but is also involved in the signaling pathway regulation. AIM OF REVIEW A thorough understanding of metabolic regulation in MSC immunomodulatory properties can provide new sights to the enhancement of MSC-based therapy. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC immune regulation can be affected by cellular metabolism (glucose, adenosine triphosphate, lipid and amino acid metabolism), which further mediates MSC therapy efficiency in inflammatory and immune diseases. The enhancement of glycolysis of MSCs, such as signaling molecule activation, inflammatory cytokines priming, or environmental control can promote MSC immune functions and therapeutic potential. Besides glucose metabolism, inflammatory stimuli also alter the lipid molecular profile of MSCs, but the direct link with immunomodulatory properties remains to be further explored. Arginine metabolism, glutamine-glutamate metabolism and tryptophan-kynurenine via indoleamine 2,3-dioxygenase (IDO) metabolism all contribute to the immune regulation of MSCs. In addition to the metabolism dictating the MSC immune functions, MSCs also influence the metabolism of immune cells and thus determine their behaviors. However, more direct evidence of the metabolism in MSC immune abilities as well as the underlying mechanism requires to be uncovered.
Collapse
Affiliation(s)
- Hanyue Li
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Hongwei Dai
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| |
Collapse
|
7
|
Wuttisarnwattana P, Eid S, Wilson DL, Cooke KR. Assessment of therapeutic role of mesenchymal stromal cells in mouse models of graft-versus-host disease using cryo-imaging. Sci Rep 2023; 13:1698. [PMID: 36717650 PMCID: PMC9886911 DOI: 10.1038/s41598-023-28478-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 01/19/2023] [Indexed: 02/01/2023] Open
Abstract
Insights regarding the biodistribution and homing of mesenchymal stromal cells (MSCs), as well as their interaction with alloreactive T-cells are critical for understanding how MSCs can regulate graft-versus-host disease (GVHD) following allogeneic (allo) bone marrow transplantation (BMT). We developed novel assays based on 3D, microscopic, cryo-imaging of whole-mouse-sized volumes to assess the therapeutic potential of human MSCs using an established mouse GVHD model. Following infusion, we quantitatively tracked fluorescently labeled, donor-derived, T-cells and third party MSCs in BMT recipients using multispectral cryo-imaging. Specific MSC homing sites were identified in the marginal zones in the spleen and the lymph nodes, where we believe MSC immunomodulation takes place. The number of MSCs found in spleen of the allo BMT recipients was about 200% more than that observed in the syngeneic group. To more carefully define the effects MSCs had on T cell activation and expansion, we developed novel T-cell proliferation assays including secondary lymphoid organ (SLO) enlargement and Carboxyfluoescein succinimidyl ester (CFSE) dilution. As anticipated, significant SLO volume enlargement and CFSE dilution was observed in allo but not syn BMT recipients due to rapid proliferation and expansion of labeled T-cells. MSC treatment markedly attenuated CFSE dilution and volume enlargement of SLO. These assays confirm evidence of potent, in vivo, immunomodulatory properties of MSC following allo BMT. Our innovative platform includes novel methods for tracking cells of interest as well as assessing therapeutic function of MSCs during GVHD induction. Our results support the use of MSCs treatment or prevention of GVHD and illuminate the wider adoption of MSCs as a standard medicinal cell therapy.
Collapse
Affiliation(s)
- Patiwet Wuttisarnwattana
- Optimization Theory and Applications for Engineering Systems Research Group, Department of Computer Engineering, Excellence Center in Infrastructure Technology and Transportation Engineering, Biomedical Engineering Institute, Chiang Mai University, Chiang Mai, Thailand.
| | - Saada Eid
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - David L Wilson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Kenneth R Cooke
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Hospital, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
8
|
Boland L, Bitterlich LM, Hogan AE, Ankrum JA, English K. Translating MSC Therapy in the Age of Obesity. Front Immunol 2022; 13:943333. [PMID: 35860241 PMCID: PMC9289617 DOI: 10.3389/fimmu.2022.943333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/10/2022] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stromal cell (MSC) therapy has seen increased attention as a possible option to treat a number of inflammatory conditions including COVID-19 acute respiratory distress syndrome (ARDS). As rates of obesity and metabolic disease continue to rise worldwide, increasing proportions of patients treated with MSC therapy will be living with obesity. The obese environment poses critical challenges for immunomodulatory therapies that should be accounted for during development and testing of MSCs. In this review, we look to cancer immunotherapy as a model for the challenges MSCs may face in obese environments. We then outline current evidence that obesity alters MSC immunomodulatory function, drastically modifies the host immune system, and therefore reshapes interactions between MSCs and immune cells. Finally, we argue that obese environments may alter essential features of allogeneic MSCs and offer potential strategies for licensing of MSCs to enhance their efficacy in the obese microenvironment. Our aim is to combine insights from basic research in MSC biology and clinical trials to inform new strategies to ensure MSC therapy is effective for a broad range of patients.
Collapse
Affiliation(s)
- Lauren Boland
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| | - Laura Melanie Bitterlich
- Biology Department, Maynooth University, Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth, Ireland
| | - Andrew E. Hogan
- Biology Department, Maynooth University, Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth, Ireland
| | - James A. Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- *Correspondence: James A. Ankrum, ; Karen English,
| | - Karen English
- Biology Department, Maynooth University, Maynooth, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth, Ireland
- *Correspondence: James A. Ankrum, ; Karen English,
| |
Collapse
|
9
|
Ktena YP, Koldobskiy MA, Barbato MI, Fu HH, Luznik L, Llosa NJ, Haile A, Klein OR, Liu C, Gamper CJ, Cooke KR. Donor T cell DNMT3a regulates alloreactivity in mouse models of hematopoietic stem cell transplantation. J Clin Invest 2022; 132:e158047. [PMID: 35608905 PMCID: PMC9246380 DOI: 10.1172/jci158047] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
DNA methyltransferase 3a (DNMT3a) is an important part of the epigenetic machinery that stabilizes patterns of activated T cell responses. We hypothesized that donor T cell DNMT3a regulates alloreactivity after allogeneic blood and marrow transplantation (allo-BMT). T cell conditional Dnmt3a KO mice were used as donors in allo-BMT models. Mice receiving allo-BMT from KO donors developed severe acute graft-versus-host disease (aGVHD), with increases in inflammatory cytokine levels and organ histopathology scores. KO T cells migrated and proliferated in secondary lymphoid organs earlier and demonstrated an advantage in trafficking to the small intestine. Donor T cell subsets were purified after BMT for whole-genome bisulfite sequencing (WGBS) and RNA-Seq. KO T cells had global methylation similar to that of WT cells, with distinct, localized areas of hypomethylation. Using a highly sensitive computational method, we produced a comprehensive profile of the altered epigenome landscape. Hypomethylation corresponded with changes in gene expression in several pathways of T cell signaling and differentiation. Additionally, Dnmt3a-KO T cells resulted in superior graft-versus-tumor activity. Our findings demonstrate a critical role for DNMT3a in regulating T cell alloreactivity and reveal pathways that control T cell tolerance. These results also provide a platform for deciphering clinical data that associate donor DNMT3a mutations with increased GVHD, decreased relapse, and improved survival.
Collapse
Affiliation(s)
- Yiouli P. Ktena
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Michael A. Koldobskiy
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Michael I. Barbato
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Han-Hsuan Fu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Leo Luznik
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Nicolas J. Llosa
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Azeb Haile
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Orly R. Klein
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christopher J. Gamper
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Kenneth R. Cooke
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Keshavarz Shahbaz S, Mansourabadi AH, Jafari D. Genetically engineered mesenchymal stromal cells as a new trend for treatment of severe acute graft-versus-host disease. Clin Exp Immunol 2022; 208:12-24. [PMID: 35274673 PMCID: PMC9113247 DOI: 10.1093/cei/uxac016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/25/2021] [Accepted: 02/07/2022] [Indexed: 01/12/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a population of non-hematopoietic and self-renewing cells characterized by the potential to differentiate into different cell subtypes. MSCs have interesting features which have attracted a lot of attention in various clinical investigations. Some basic features of MSCs are including the weak immunogenicity (absence of MHC-II and costimulatory ligands accompanied by the low expression of MHC-I) and the potential of plasticity and multi-organ homing via expressing related surface molecules. MSCs by immunomodulatory effects could also ameliorate several immune-pathological conditions like graft-versus-host diseases (GVHD). The efficacy and potency of MSCs are the main objections of MSCs therapeutic applications. It suggested that improving the MSC immunosuppressive characteristic via genetic engineering to produce therapeutic molecules consider as one of the best options for this purpose. In this review, we explain the functions, immunologic properties, and clinical applications of MSCs to discuss the beneficial application of genetically modified MSCs in GVHD.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Disease, Qazvin University of Medical Science, Qazvin, Iran
| | - Amir Hossein Mansourabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Davood Jafari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Zanjan, Iran
| |
Collapse
|
11
|
A Novel 3D Culture System Using a Chitin-Based Polysaccharide Material Produces High-Quality Allogeneic Human UCMSCs with Dispersed Sphere Morphology. Cells 2022; 11:cells11060995. [PMID: 35326446 PMCID: PMC8947357 DOI: 10.3390/cells11060995] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/07/2022] [Accepted: 03/13/2022] [Indexed: 11/29/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation, in particular allogeneic transplantation, is a promising therapy for a variety of diseases. However, before performing allograft treatment it is necessary to find suitable donors, establish culture methods that maintain cell quality, and reduce cell production costs. Here, we present a new method of producing allogeneic MSCs combining human umbilical cord-derived mesenchymal stem cells (UCMSCs) and chitin-based polysaccharide fibers (Cellhesion® MS). UCMSC numbers significantly increased, and cells grew as dispersed spheres on Cellhesion® MS. Subsequent biological analyses showed that the expression levels of stemness-related and migration-related genes were significantly upregulated, including octamer-binding transcription factor 4 (OCT4), Nanog homeobox (NANOG), and C-X-C chemokine receptor type 4 (CXCR4). The secretion levels of paracrine factors such as prostaglandin E2 (PGE2), TNFα-stimulating gene (TSG)-6, fibroblast growth factor 2 (bFGF), and Angiogenin (Ang) from UCMSCs using Cellhesion® MS were significantly higher than with microcarrier and U-bottom plate culture. In addition, culture supernatant from UCMSCs with Cellhesion® MS had better angiogenic potential than that from monolayer cultured UCMSCs. Furthermore, we succeeded in a scaled-up culture of UCMSCs with Cellhesion® MS using a closed culture bag. Therefore, Cellhesion® MS is a key material for producing high-quality UCMSCs in a three-dimensional (3D) culture system.
Collapse
|
12
|
Zhang C, Delawary M, Huang P, Korchak JA, Suda K, Zubair AC. IL-10 mRNA Engineered MSCs Demonstrate Enhanced Anti-Inflammation in an Acute GvHD Model. Cells 2021; 10:3101. [PMID: 34831324 PMCID: PMC8621791 DOI: 10.3390/cells10113101] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are used in various studies to induce immunomodulatory effects in clinical conditions associated with immune dysregulation such as graft versus host disease (GvHD). However, most of these clinical trials failed to go beyond early phase 2 studies because of limited efficacy. Various methods have been assessed to increase the potency of MSCs. IL-10 is an anti-inflammatory cytokine that is known to modulate immune responses in GvHD. In this study, we evaluated the feasibility of transfecting IL-10 mRNA to enhance MSC therapeutic potential. IL-10 mRNA engineered MSCs (eMSCs-IL10) maintained high levels of IL-10 expression even after freezing and thawing. IL-10 mRNA transfection did not appear to alter MSC intrinsic characteristics. eMSCs-IL10 significantly suppressed T cell proliferation relative to naïve MSCs in vitro. In a mouse model for GvHD, eMSCs-IL10 induced a decrease in plasma level of potent pro-inflammatory cytokines and inhibited CD4+ and CD8+ T cell proliferation in the spleen. In summary, our studies demonstrate the feasibility of potentiating MSCs to enhance their immunomodulatory effects by IL-10 mRNA transfection. The use of non-viral transfection may generate a safe and potent MSC product for treatment of clinical conditions associated with immune dysregulation such as GvHD.
Collapse
Affiliation(s)
- Cuiping Zhang
- Center for Regenerative Medicine and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL 32224, USA; (C.Z.); (P.H.); (J.A.K.)
| | - Mina Delawary
- Cell Therapy Research Laboratories, Daiichi Sankyo, Co., Ltd., Tokyo 1408710, Japan; (M.D.); (K.S.)
| | - Peng Huang
- Center for Regenerative Medicine and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL 32224, USA; (C.Z.); (P.H.); (J.A.K.)
| | - Jennifer A. Korchak
- Center for Regenerative Medicine and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL 32224, USA; (C.Z.); (P.H.); (J.A.K.)
| | - Koji Suda
- Cell Therapy Research Laboratories, Daiichi Sankyo, Co., Ltd., Tokyo 1408710, Japan; (M.D.); (K.S.)
| | - Abba C. Zubair
- Center for Regenerative Medicine and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL 32224, USA; (C.Z.); (P.H.); (J.A.K.)
| |
Collapse
|
13
|
Kasikis S, Baez J, Gandhi I, Grupp S, Kitko CL, Kowalyk S, Merli P, Morales G, Pulsipher MA, Qayed M, Wölfl M, Yanik G, See F, Hayes J, Grossman F, Burke E, Young R, Levine JE, Ferrara JLM. Mesenchymal stromal cell therapy induces high responses and survival in children with steroid refractory GVHD and poor risk biomarkers. Bone Marrow Transplant 2021; 56:2869-2870. [PMID: 34471240 PMCID: PMC9840529 DOI: 10.1038/s41409-021-01442-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/29/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Stelios Kasikis
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, NY
| | - Janna Baez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, NY
| | - Isha Gandhi
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, NY
| | - Stephan Grupp
- Children’s Hospital of Philadelphia and Perelman School of Medicine
| | | | - Steven Kowalyk
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, NY
| | - Pietro Merli
- Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Pediatrico Bambino Gesuù, Rome, Italy
| | - George Morales
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, NY
| | - Michael A. Pulsipher
- Section of Transplantation and Cellular Therapy, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Emory University and Children’s Healthcare of Atlanta, Atlanta, GA
| | - Matthias Wölfl
- Children’s Hospital, University of Würzburg, Würzburg Germany
| | | | | | | | | | | | - Rachel Young
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, NY
| | - John E. Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, NY
| | | |
Collapse
|
14
|
Metheny L, Eid S, Wuttisarnwattana P, Auletta JJ, Liu C, Van Dervort A, Paez C, Lee Z, Wilson D, Lazarus HM, Deans R, Vant Hof W, Ktena Y, Cooke KR. Human multipotent adult progenitor cells effectively reduce graft-vs-host disease while preserving graft-vs-leukemia activity. STEM CELLS (DAYTON, OHIO) 2021; 39:1506-1519. [PMID: 34255899 PMCID: PMC8596993 DOI: 10.1002/stem.3434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/24/2021] [Indexed: 11/13/2022]
Abstract
Graft‐vs‐host disease (GvHD) limits successful outcomes following allogeneic blood and marrow transplantation (allo‐BMT). We examined whether the administration of human, bone marrow‐derived, multipotent adult progenitor cells (MAPCs™) could regulate experimental GvHD. The immunoregulatory capacity of MAPC cells was evaluated in vivo using established murine GvHD models. Injection of MAPC cells on day +1 (D1) and +4 (D4) significantly reduced T‐cell expansion and the numbers of donor‐derived, Tumor Necrosis Factor Alpha (TNFα) and Interferon Gamma (IFNγ)‐producing, CD4+ and CD8+ cells by D10 compared with untreated controls. These findings were associated with reductions in serum levels of TNFα and IFNγ, intestinal and hepatic inflammation and systemic GvHD as measured by survival and clinical score. Biodistribution studies showed that MAPC cells tracked from the lung and to the liver, spleen, and mesenteric nodes within 24 hours after injection. MAPC cells inhibited mouse T‐cell proliferation in vitro and this effect was associated with reduced T‐cell activation and inflammatory cytokine secretion and robust increases in the concentrations of Prostaglandin E2 (PGE2) and Transforming Growth Factor Beta (TGFβ). Indomethacin and E‐prostanoid 2 (EP2) receptor antagonism both reversed while EP2 agonism restored MAPC cell‐mediated in vitro T‐cell suppression, confirming the role for PGE2. Furthermore, cyclo‐oxygenase inhibition following allo‐BMT abrogated the protective effects of MAPC cells. Importantly, MAPC cells had no effect on the generation cytotoxic T lymphocyte activity in vitro, and the administration of MAPC cells in the setting of leukemic challenge resulted in superior leukemia‐free survival. Collectively, these data provide valuable information regarding the biodistribution and regulatory capacity of MAPC cells, which may inform future clinical trial design.
Collapse
Affiliation(s)
- Leland Metheny
- University Hospitals Seidman Cancer CenterClevelandOhioUSA
- Case Comprehensive Cancer CenterClevelandOhioUSA
| | - Saada Eid
- Department of PediatricsCase Western Reserve UniversityClevelandOhioUSA
| | - Patiwet Wuttisarnwattana
- Department of Computer EngineeringChiang Mai UniversityChiang MaiThailand
- Department of Biomedical Engineering CenterChiang Mai UniversityChiang MaiThailand
| | - Jeffery J. Auletta
- Host Defense Program, Hematology, Oncology, and Infectious DiseasesNationwide Children's HospitalColumbusOhioUSA
| | - Chen Liu
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
| | - Alana Van Dervort
- Department of PediatricsCase Western Reserve UniversityClevelandOhioUSA
| | - Conner Paez
- Department of PediatricsCase Western Reserve UniversityClevelandOhioUSA
| | - ZhengHong Lee
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - David Wilson
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | | | | | | | - Yiouli Ktena
- Department of OncologyJohns Hopkins Sidney Kimmel Comprehensive Cancer CenterBaltimoreMarylandUSA
| | - Kenneth R. Cooke
- Department of OncologyJohns Hopkins Sidney Kimmel Comprehensive Cancer CenterBaltimoreMarylandUSA
| |
Collapse
|
15
|
Mesenchymal stromal cells in hematopoietic cell transplantation. Blood Adv 2021; 4:5877-5887. [PMID: 33232479 DOI: 10.1182/bloodadvances.2020002646] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are widely recognized to possess potent immunomodulatory activity, as well as to stimulate repair and regeneration of diseased or damaged tissue. These fundamental properties suggest important applications in hematopoietic cell transplantation. Although the mechanisms of therapeutic activity in vivo are yet to be fully elucidated, MSCs seem to suppress lymphocytes by paracrine mechanisms, including secreted mediators and metabolic modulators. Most recently, host macrophage engulfment of apoptotic MSCs has emerged as an important contributor to the immune suppressive microenvironment. Although bone marrow-derived MSCs are the most commonly studied, the tissue source of MSCs may be a critical determinant of immunomodulatory function. The key application of MSC therapy in hematopoietic cell transplantation is to prevent or treat graft-versus-host disease (GVHD). The pathogenesis of GVHD reveals multiple potential targets. Moreover, the recently proposed concept of tissue tolerance suggests a new possible mechanism of MSC therapy for GVHD. Beyond GVHD, MSCs may facilitate hematopoietic stem cell engraftment, which could gain greater importance with increasing use of haploidentical transplantation. Despite many challenges and much doubt, commercial MSC products for pediatric steroid-refractory GVHD have been licensed in Japan, conditionally licensed in Canada and New Zealand, and have been recommended for approval by an FDA Advisory Committee in the United States. Here, we review key historical data in the context of the most salient recent findings to present the current state of MSCs as adjunct cell therapy in hematopoietic cell transplantation.
Collapse
|
16
|
van Heeckeren AM, Sutton MT, Fletcher DR, Hodges CA, Caplan AI, Bonfield TL. Enhancing Cystic Fibrosis Immune Regulation. Front Pharmacol 2021; 12:573065. [PMID: 34054509 PMCID: PMC8155373 DOI: 10.3389/fphar.2021.573065] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/29/2021] [Indexed: 01/08/2023] Open
Abstract
In cystic fibrosis (CF), sustained infection and exuberant inflammation results in debilitating and often fatal lung disease. Advancement in CF therapeutics has provided successful treatment regimens for a variety of clinical consequences in CF; however effective means to treat the pulmonary infection and inflammation continues to be problematic. Even with the successful development of small molecule cystic fibrosis transmembrane conductance regulator (CFTR) correctors and potentiators, there is only a modest effect on established infection and inflammation in CF patients. In the pursuit of therapeutics to treat inflammation, the conundrum to address is how to overcome the inflammatory response without jeopardizing the required immunity to manage pathogens and prevent infection. The key therapeutic would have the capacity to dull the inflammatory response, while sustaining the ability to manage infections. Advances in cell-based therapy have opened up the avenue for dynamic and versatile immune interventions that may support this requirement. Cell based therapy has the capacity to augment the patient’s own ability to manage their inflammatory status while at the same time sustaining anti-pathogen immunity. The studies highlighted in this manuscript outline the potential use of cell-based therapy for CF. The data demonstrate that 1) total bone marrow aspirates containing Cftr sufficient hematopoietic and mesenchymal stem cells (hMSCs) provide Cftr deficient mice >50% improvement in survival and improved management of infection and inflammation; 2) myeloid cells can provide sufficient Cftr to provide pre-clinical anti-inflammatory and antimicrobial benefit; 3) hMSCs provide significant improvement in survival and management of infection and inflammation in CF; 4) the combined interaction between macrophages and hMSCs can potentially enhance anti-inflammatory and antimicrobial support through manipulating PPARγ. These data support the development of optimized cell-based therapeutics to enhance CF patient’s own immune repertoire and capacity to maintain the balance between inflammation and pathogen management.
Collapse
Affiliation(s)
- Anna M van Heeckeren
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Morgan T Sutton
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,St. Jude Children's Research Hospital Graduate School of Biomedical Sciences, Memphis, TN, United States
| | - David R Fletcher
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Craig A Hodges
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Arnold I Caplan
- Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Tracey L Bonfield
- Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Skeletal Research Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,National Center for Regenerative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States.,Departments of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
17
|
Eckard AR, Borow KM, Mack EH, Burke E, Atz AM. Remestemcel-L Therapy for COVID-19-Associated Multisystem Inflammatory Syndrome in Children. Pediatrics 2021; 147:peds.2020-046573. [PMID: 33579813 DOI: 10.1542/peds.2020-046573] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2021] [Indexed: 11/24/2022] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a serious postinfectious immune dysregulation associated with coronavirus disease 2019 that may present with severe and life-threatening cardiovascular dysfunction, hemodynamic instability, shock, and multisystem organ failure. Optimal treatment is unknown. Current standard of care consists of nonspecific anti-inflammatory and antithrombotic therapies. Interventions that target MIS-C's distinctive clinical features and immunophenotype are indicated. Remestemcel-L, an investigational mesenchymal stromal cell therapy, is a promising candidate for treatment of MIS-C because of its beneficial anti-inflammatory, immunomodulatory, endothelial function and vascular stabilizing effects, which align well with the pathophysiology of MIS-C. Here, we present the first two patients with life-threatening MIS-C ever treated with remestemcel-L under an expanded access program. Both were previously healthy children without any indication of previous coronavirus disease 2019 infection or exposure. They presented with severe clinical illness including myocardial dysfunction, hemodynamic instability, hypotension, acute kidney injury, and shock. At the time of hospital admission, both had negative polymerase chain reaction (PCR) test results and positive serology results for severe acute respiratory syndrome coronavirus 2. Both children received standard of care MIS-C treatment. Although the patients showed some clinical improvement, left ventricular ejection fraction remained reduced and inflammatory biomarkers remained significantly elevated. When treated with two intravenous doses of remestemcel-L separated by 48 hours, rapid normalization of left ventricular ejection fraction, notable reductions in biomarkers of systemic and cardiac inflammation, and improved clinical status occurred. Neither child experienced adverse effects associated with remestemcel-L administration. This treatment appears promising as a novel immunomodulatory cellular therapy for children with clinically significant cardiovascular manifestations of MIS-C.
Collapse
Affiliation(s)
- Allison Ross Eckard
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina;
| | | | - Elizabeth H Mack
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | | | - Andrew M Atz
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
18
|
Gruhn B, Brodt G, Ernst J. Extended Treatment With Mesenchymal Stromal Cells-Frankfurt am Main in a Pediatric Patient With Steroid-refractory Acute Gastrointestinal Graft-Versus-Host Disease: Case Report and Review of the Literature. J Pediatr Hematol Oncol 2021; 43:e419-e425. [PMID: 32118816 DOI: 10.1097/mph.0000000000001758] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/30/2020] [Indexed: 12/14/2022]
Abstract
In acute graft-versus-host disease (aGVHD) following allogeneic hematopoietic stem cell transplantation, there are various options available after the failure of initial steroid therapy. Since the publication of the first study in 2008, mesenchymal stromal cells (MSCs) have also been used with increasing frequency, including in pediatric patients with steroid-refractory aGVHD, and the manufacturing process has undergone further development. MSC-Frankfurt am Main (MSC-FFM, Obnitix), which is manufactured from pooled mononuclear bone marrow cells from 8 donors using a standardized process, resulted in a response rate of 84% in children with steroid-refractory aGVHD. We report on a 13-year-old female patient with acute myeloid leukemia who received Obnitix as a third-line treatment for gastrointestinal (GI) aGVHD in a life-threatening situation. The patient was initially given a total of 4 Obnitix infusions as per the regulatory approval, with her symptoms improving from day 9 after the first infusion. The second cycle of 4 Obnitix infusions followed due to persistent severe protein-losing enteropathy and resulted in complete remission. A systematic review of the literature on MSC in pediatric patients with steroid-refractory aGVHD confirms that MSC treatment beyond 4 weeks is employed in accordance with treatment protocols or on a case-by-case basis. To summarize, aGVHD activity can be checked endoscopically in patients with persistent GI symptoms and a second Obnitix cycle can then be administered if appropriate, with the goal of achieving complete remission. Future studies should also investigate the potential influence of tissue repair properties as an element in MSCs' efficacy in GI aGVHD.
Collapse
Affiliation(s)
- Bernd Gruhn
- Department of Pediatrics, Jena University Hospital, Jena, Germany
| | | | | |
Collapse
|
19
|
Yoo KH. Strategies to enhance graft performance in cord blood transplantation. PRECISION AND FUTURE MEDICINE 2021. [DOI: 10.23838/pfm.2020.00149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
20
|
Tago Y, Kobayashi C, Ogura M, Wada J, Yamaguchi S, Yamaguchi T, Hayashi M, Nakaishi T, Kubo H, Ueda Y. Human amnion-derived mesenchymal stem cells attenuate xenogeneic graft-versus-host disease by preventing T cell activation and proliferation. Sci Rep 2021; 11:2406. [PMID: 33510297 PMCID: PMC7843654 DOI: 10.1038/s41598-021-81916-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022] Open
Abstract
Acute graft-versus-host disease (GVHD) is characterized by severe tissue damage that is a life-threatening complication of allogeneic hematopoietic stem cell transplantation. Due to their immunosuppressive properties, mesenchymal stem cells (MSC) have been increasingly examined for the treatment of immune-related diseases. We aimed to assess the immunosuppressive effects of human amnion-derived MSC (AMSC) in a xenogeneic GVHD NOD/Shi-scid IL2rγnull mouse model using human peripheral blood mononuclear cells (PBMC). Additionally, we used human bone marrow-derived MSC (BMSC) as comparative controls to determine differences in immunomodulatory functions depending on the MSC origin. Administration of AMSC significantly prolonged survival, and reduced human tumor necrosis factor-α (TNF-α) concentration and percentage of programmed cell death protein-1 receptor (PD-1)+CD8+ T cell populations compared with in GVHD control mice. Furthermore, colonic inflammation score and percentage of human CD8+ T cell populations in AMSC-treated mice were significantly lower than in GVHD control and BMSC-treated mice. Interestingly, gene expression and protein secretion of the PD-1 ligands were higher in AMSC than in BMSC. These findings are the first to demonstrate that AMSC exhibit marked immunosuppression and delay acute GVHD progression by preventing T cell activation and proliferation via the PD-1 pathway.
Collapse
Affiliation(s)
- Yoshiyuki Tago
- Biotechnology Research Laboratories, Kaneka Corporation, 1-8, Miyamae-cho, Takasago-cho, Takasago, Hyogo, 676-8688, Japan. .,Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan.
| | - Chiho Kobayashi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Mineko Ogura
- Biotechnology Research Laboratories, Kaneka Corporation, 1-8, Miyamae-cho, Takasago-cho, Takasago, Hyogo, 676-8688, Japan
| | - Jutaro Wada
- Biotechnology Research Laboratories, Kaneka Corporation, 1-8, Miyamae-cho, Takasago-cho, Takasago, Hyogo, 676-8688, Japan
| | - Sho Yamaguchi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Takashi Yamaguchi
- Biotechnology Research Laboratories, Kaneka Corporation, 1-8, Miyamae-cho, Takasago-cho, Takasago, Hyogo, 676-8688, Japan
| | - Masahiro Hayashi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Tomoyuki Nakaishi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Hiroshi Kubo
- Biotechnology Research Laboratories, Kaneka Corporation, 1-8, Miyamae-cho, Takasago-cho, Takasago, Hyogo, 676-8688, Japan
| | - Yasuyoshi Ueda
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| |
Collapse
|
21
|
Yu Q, Wang H, Zhang L, Wei W. Advances in the treatment of graft-versus-host disease with immunomodulatory cells. Int Immunopharmacol 2021; 92:107349. [PMID: 33486323 DOI: 10.1016/j.intimp.2020.107349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 12/19/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has been widely used to treat hematological malignancies and genetic diseases. Graft-versus-host disease (GVHD) induced by donor immune system is the most common complication, contributing to severe morbidity and mortality after allo-HSCT. Currently, in terms of the prevention and treatment of GVHD, the major first-line therapeutic drugs are corticosteroids. However, most patients with systemic corticosteroid treatment are prone to steroid-refractory and poor prognosis. The use of several immune cells including Tregs, Bregs and mesenchymal stromal cells (MSCs) as an alternative on prevention or therapy of GVHD has been demonstrated to be beneficial. However, there are still many defects to a certain degree. Based on immune cells, it is promising to develop new and better approaches to improve GVHD. In this article, we will review the current advance of immune cells (Tregs, Bregs, MSCs) with negative regulation in the treatment of GVHD and present emerging strategies for the prevention and treatment of GVHD by other immune regulatory cells and chimeric antigen receptor (CAR) Tregs. In addition, these new therapeutic options need to be further evaluated in well-designed prospective multicenter trials to determine the optimal treatment for GVHD patients and improve their prognosis.
Collapse
Affiliation(s)
- Qianqian Yu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei 230032, China
| | - Han Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei 230032, China
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei 230032, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei 230032, China.
| |
Collapse
|
22
|
Li H, Liu Q, Gao X, Zhang D, Mao S, Jia Y. IFN-γ gene loaded human umbilical mesenchymal stromal cells targeting therapy for Graft-versus-host disease. Int J Pharm 2021; 592:120058. [PMID: 33220383 DOI: 10.1016/j.ijpharm.2020.120058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/19/2020] [Accepted: 11/03/2020] [Indexed: 02/05/2023]
Abstract
Graft-versus-host disease (GVHD) is a frequent complication following allogeneic hematopoietic stem cell transplantation (allo-HSCT). The application of mesenchymal stromal cells (MSCs) to treat GVHD patients refractory to initial steroid treatment has led to impressive results. In this study, we explored the potential of human umbilical mesenchymal stem cells (HUMSCs) transfected with the IFN-γ gene of human (h)/mice (m) (HUMSCs + Ad-h/mIFN-γ) carried by a recombinant adenoviral vector in the prevention and treatment of GVHD. We demonstrated that HUMSCs + Ad-h/mIFN-γ efficiently suppressed T lymphocyte proliferation and activation, induced G1 cell cycle arrest and apoptosis in vitro. To assess the in vivo efficacy of HUMSCs + Ad-h/mIFN-γ, Balb/c mice were induced to develop GVHD symptoms by tail vein injection of C57BL/6 splenocytes after irradiation. Weight, hair, survival, hemogram, and chimera condition of GVHD model mice were monitored before and after treatment, respectively. The results showed that HUMSCs + Ad-h/mIFN-γ reduced GVHD's incidence and severity on the model mice and provided a significant survival benefit. In conclusion, this study may provide validated evidence that the introduction of IFN-γ into HUMSCs would help ameliorate GVHD after allo-HSCT.
Collapse
Affiliation(s)
- Hui Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, People's Republic of China; Department of Hematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital & Affiliated Hospital of University of Electronic Science and Technology of China, Chengdu, People's Republic of China.
| | - Qi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, People's Republic of China
| | - Xiaofeng Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, People's Republic of China
| | - Di Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, People's Republic of China
| | - Shengjun Mao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu, People's Republic of China
| | - Yongqian Jia
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
23
|
Analysis of Same Selected Immunomodulatory Properties of Chorionic Mesenchymal Stem Cells. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10249040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells (MSCs) represent a population of adherent cells that can be isolated from multiple adult tissues. MSCs have immunomodulatory capacity and the ability to differentiate into many cell lines. Research study examines the immunomodulatory properties of MSCs isolated from chorion (CMSCs). Following the stimulation process, it was found that MSCs are capable of immunomodulatory action via the release of bioactive molecules as well as through direct contact with the immune cells. Immunomodulatory potential of the CMSCs was analyzed by modifying proliferative capacity of mitogen-activated lymphocytes. CMSCs and lymphocytes were tested in cell-to-cell contact. Lymphocytes were stained with carboxyfluorescein diacetate succinimidyl ester. Inhibition of the proliferation of activated lymphocytes was observed. Following the co-cultivation, the expression of markers involved in the immune response modulation was assessed. Afterwards, an increase in CMSCs expression of IL-10 was detected. Following the co-cultivation with activated lymphocyte, adhesion molecules CD54 and CD44 in the CMSCs increased. An increase of CD54 expression was observed. The properties of CMSCs, adherence and differentiation ability, were confirmed. The phenotype of CMSCs CD105+, CD90+, CD73+, CD44+, CD29+, CD45−, CD34−, CD54+ was characterized. It was demonstrated that chorion-derived MSCs have important immunomodulatory effects.
Collapse
|
24
|
Ciccocioppo R, Comoli P, Astori G, Del Bufalo F, Prapa M, Dominici M, Locatelli F. Developing cell therapies as drug products. Br J Pharmacol 2020; 178:262-279. [PMID: 33140850 DOI: 10.1111/bph.15305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023] Open
Abstract
In the last 20 years, the global regulatory frameworks for drug assessment have been managing the challenges posed by using cellular products as new therapeutic tools. Currently, they are defined as "Advanced Therapy Medicinal Products", comprising a large group of cellular types that either alone or in combination with gene and tissue engineering technology. They have the potential to change the natural course of still lethal or highly debilitating diseases, including cancers, opportunistic infections and chronic inflammatory conditions. Globally, more than 50 cell-based products have obtained market authorization. This overview describes the advantages and unsolved challenges on developing cells as innovative therapeutic vehicles. The main cell therapy players and the legal framework are discussed, starting from chimeric antigen receptor T-cells for leukaemia and solid tumours, dealing then with lymphocytes as potent anti-microbiological tools and then focusing on mesenchymal stem/stromal cells whose role covers regenerative medicine, immunology and anti-tumour therapy.
Collapse
Affiliation(s)
- Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy
| | - Patrizia Comoli
- Cell Factory and Paediatric Haematology/Oncology Unit, Fondazione I.R.C.C.S. Policlinico San Matteo, Pavia, Italy
| | - Giuseppe Astori
- Laboratory of Advanced Cellular Therapies, Haematology Unit, San Bortolo Hospital, A.U.L.S.S. 8 "Berica", Vicenza, Italy
| | - Francesca Del Bufalo
- Department of Paediatric Haematology and Oncology and Cell and Gene Therapy, I.R.C.C.S. Bambino Gesù Children's Hospital, Rome, Italy
| | - Malvina Prapa
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Franco Locatelli
- Department of Paediatric Haematology and Oncology and Cell and Gene Therapy, I.R.C.C.S. Bambino Gesù Children's Hospital, Rome, Italy.,Department of Paediatrics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Burnham AJ, Foppiani EM, Horwitz EM. Key Metabolic Pathways in MSC-Mediated Immunomodulation: Implications for the Prophylaxis and Treatment of Graft Versus Host Disease. Front Immunol 2020; 11:609277. [PMID: 33365034 PMCID: PMC7750397 DOI: 10.3389/fimmu.2020.609277] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/10/2020] [Indexed: 01/18/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are spindle-shaped, plastic-adherent cells in vitro with potent immunosuppressive activity both in vitro and in vivo. MSCs have been employed as a cellular immunotherapy in diverse preclinical models and clinical trials, but most commonly as agents for the prophylaxis or therapy of graft versus host disease after hematopoietic cell transplantation. In addition to the oft studied secreted cytokines, several metabolic pathways intrinsic to MSCs, notably indoleamine 2,3-dioxygenase, prostaglandin E2, hypoxia-inducible factor 1 α, heme oxygenase-1, as well as energy-generating metabolism, have been shown to play roles in the immunomodulatory activity of MSCs. In this review, we discuss these key metabolic pathways in MSCs which have been reported to contribute to MSC therapeutic effects in the setting of hematopoietic cell transplantation and graft versus host disease. Understanding the contribution of MSC metabolism to immunomodulatory activity may substantially inform the development of future clinical applications of MSCs.
Collapse
Affiliation(s)
- Andre J Burnham
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Elisabetta Manuela Foppiani
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Edwin M Horwitz
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
26
|
Cimino M, Parreira P, Bidarra SJ, Gonçalves RM, Barrias CC, Martins MCL. Effect of surface chemistry on hMSC growth under xeno-free conditions. Colloids Surf B Biointerfaces 2020; 189:110836. [DOI: 10.1016/j.colsurfb.2020.110836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 01/17/2020] [Accepted: 01/29/2020] [Indexed: 01/05/2023]
|
27
|
Wuttisarnwattana P, Eid S, Gargesha M, Cooke KR, Wilson DL. Cryo-imaging of Stem Cell Biodistribution in Mouse Model of Graft-Versus-Host-Disease. Ann Biomed Eng 2020; 48:1702-1711. [PMID: 32103369 DOI: 10.1007/s10439-020-02487-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/21/2020] [Indexed: 12/13/2022]
Abstract
We demonstrated the use of multispectral cryo-imaging and software to analyze human mesenchymal stromal cells (hMSCs) biodistribution in mouse models of graft-versus-host-disease (GVHD) following allogeneic bone marrow transplantation (BMT). We injected quantum dot labeled MSCs via tail vein to mice receiving BMT and analyzed hMSC biodistribution in major organs (e.g. lung, liver, spleen, kidneys and bone marrow). We compared the biodistribution of hMSCs in mice following allogeneic BMT recipients (with GVHD) to the biodistribution following syngeneic BMT (without GVHD). Cryo-imaging system revealed cellular biodistribution and redistribution patterns in the animal model. We initially found clusters of cells in the lung that eventually dissociated to single cells and redistributed to other organs within 72 h. The in vivo half-life of the exogenous MSCs was about 21 h. We found that the biodistribution of stromal cells was not related to blood flow, rather cells preferentially homed to specific organs. In conclusion, cryo-imaging was suitable for analyzing the cellular biodistribution. It could provide capabilities of visualizing cells anywhere in the mouse model with single cell sensitivity. By characterizing the biodistribution and anatomical specificity of a therapeutic cellular product, we believe that cryo-imaging can play an important role in the advancement of stem and stromal cell therapies and regenerative medicine.
Collapse
Affiliation(s)
- Patiwet Wuttisarnwattana
- Department of Computer Engineering, Faculty of Engineering, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Biomedical Engineering Institute, Chiang Mai University, Chiang Mai, Thailand.
| | - Saada Eid
- Department of Pediatric Hematology and Oncology, Case Western Reserve University, Cleveland, OH, USA
| | | | - Kenneth R Cooke
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David L Wilson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
28
|
Dal Collo G, Adamo A, Gatti A, Tamellini E, Bazzoni R, Takam Kamga P, Tecchio C, Quaglia FM, Krampera M. Functional dosing of mesenchymal stromal cell-derived extracellular vesicles for the prevention of acute graft-versus-host-disease. Stem Cells 2020; 38:698-711. [PMID: 32064745 DOI: 10.1002/stem.3160] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023]
Abstract
Graft-vs-host-disease (GvHD) is currently the main complication of allogeneic hematopoietic stem cell transplantation. Mortality and morbidity rates are particularly high, especially in steroid-refractory acute GvHD (aGvHD). Immune regulatory human bone marrow mesenchymal stromal cells (hMB-MSCs) represent a therapeutic approach to address this issue. Unfortunately, their effect is hardly predictable in vivo due to several variables, that is, MSC tissue origin, concentration, dose number, administration route and timing, and inflammatory status of the recipient. Interestingly, human bone marrow MSC-derived extracellular vesicles (hBM-MSC-EVs) display many of the hBM-MSC immunoregulatory properties due to their content in paracrine factors that greatly varies according to the collection method. In this study, we focused on the immunological characterization of hBM-MSC-EVs on their capability of inducing regulatory T-cells (T-regs) both in vitro and in a xenograft mouse model of aGvHD. We correlated these data with the aGvHD incidence and degree following hBM-MSC-EV intravenous administration. Thus, we first quantified the EV immunomodulation in vitro in terms of EV immunomodulatory functional unit (EV-IFU), that is, the lowest concentration of EVs leading in vitro to at least threefold increase of the T-regs compared with controls. Second, we established the EV therapeutic dose in vivo (EV-TD) corresponding to 10-fold the in vitro EV-IFU. According to this approach, we observed a significant improvement of both mouse survival and control of aGvHD onset and progression. This study confirms that EVs may represent an alternative to whole MSCs for aGvHD prevention, once the effective dose is reproducibly identified according to EV-IFU and EV-TD definition.
Collapse
Affiliation(s)
- Giada Dal Collo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Annalisa Adamo
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Alessandro Gatti
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Edoardo Tamellini
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy.,EA4340-BCOH, Biomarker in Cancerology and Onco-Haematology, UVSQ, Université Paris Saclay, Boulogne-Billancourt, France
| | - Cristina Tecchio
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Francesca Maria Quaglia
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
29
|
A Phase 3, Single-Arm, Prospective Study of Remestemcel-L, Ex Vivo Culture-Expanded Adult Human Mesenchymal Stromal Cells for the Treatment of Pediatric Patients Who Failed to Respond to Steroid Treatment for Acute Graft-versus-Host Disease. Biol Blood Marrow Transplant 2020; 26:845-854. [PMID: 32018062 PMCID: PMC8322819 DOI: 10.1016/j.bbmt.2020.01.018] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/17/2020] [Accepted: 01/26/2020] [Indexed: 12/17/2022]
Abstract
Steroid-refractory acute graft-versus-host disease (SR-aGVHD) following hematopoietic cell transplantation (HSCT) is associated with poor clinical outcomes. Currently, there are no safe and effective therapies approved for use in the pediatric population under the age of 12 years. Accordingly, there is an urgent need for new treatments that are safe, well tolerated, and effective in managing this debilitating and potentially fatal complication of HSCT. In early phase clinical trials, mesenchymal stromal cells (MSCs) have demonstrated efficacy in the treatment of acute GVHD (aGVHD) in pediatric patients. We now report the results of a phase 3, prospective, single-arm, multicenter study (NCT02336230) in 54 children with primary SR-aGVHD who were naive to other immunosuppressant therapies for aGVHD treated with MSC product (remestemcel-L) dosed at 2 × 106 cells/kg twice weekly for 4 weeks. Remestemcel-L therapy significantly improved day 28 overall response rate (OR) compared with the prespecified control OR value of 45% (70.4% versus 45%, P = .0003). The statistically significant OR (70.4%) was sustained through day 100, including an increase in complete response from 29.6% at day 28 to 44.4% at day 100. Overall survival was 74.1% at day 100 and 68.5% at day 180. Overall response in all participants at day 28 was highly predictive of improved survival through 180 days, and survival was significantly greater in day 28 responders compared with nonresponders through day 100 (86.8% versus 47.1% for responders and nonresponders, respectively, P = .0001) and through day 180 (78.9% versus 43.8%, P = .003). Remestemcel-L was well tolerated with no identified infusion-related toxicities or other safety concerns. This study provides robust, prospective evidence of the safety, tolerability, and efficacy of remestemcel-L as first-line therapy after initial steroid failure in pediatric SR-aGVHD.
Collapse
|
30
|
García JR, Quirós M, Han WM, O'Leary MN, Cox GN, Nusrat A, García AJ. IFN-γ-tethered hydrogels enhance mesenchymal stem cell-based immunomodulation and promote tissue repair. Biomaterials 2019; 220:119403. [PMID: 31401468 DOI: 10.1016/j.biomaterials.2019.119403] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/17/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022]
Abstract
Because of their immunomodulatory activities, human mesenchymal stem cells (hMSCs) are being explored to treat a variety of chronic conditions such as inflammatory bowel disorders and graft-vs-host disease. Treating hMSCs with IFN-γ prior to administration augments these immunomodulatory properties; however, this ex vivo treatment limits the broad applicability of this therapy due to technical and regulatory issues. In this study, we engineered an injectable synthetic hydrogel with tethered recombinant IFN-γ that activates encapsulated hMSCs to increase their immunomodulatory functions and avoids the need for ex vivo manipulation. Tethering IFN-γ to the hydrogel increases retention of IFN-γ within the biomaterial while preserving its biological activity. hMSCs encapsulated within hydrogels with tethered IFN-γ exhibited significant differences in cytokine secretion and showed a potent ability to halt activated T-cell proliferation and monocyte-derived dendritic cell differentiation compared to hMSCs that were pre-treated with IFN-γ and untreated hMSCs. Importantly, hMSCs encapsulated within hydrogels with tethered IFN-γ accelerated healing of colonic mucosal wounds in both immunocompromised and immunocompetent mice. This novel approach for licensing hMSCs with IFN-γ may enhance the clinical translation and efficacy of hMSC-based therapies.
Collapse
Affiliation(s)
- José R García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Miguel Quirós
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Woojin M Han
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | | | | | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
31
|
Geiger S, Ozay EI, Geumann U, Hereth MK, Magnusson T, Shanthalingam S, Hirsch D, Kälin S, Günther C, Osborne BA, Tew GN, Hermann FG, Minter LM. Alpha-1 Antitrypsin-Expressing Mesenchymal Stromal Cells Confer a Long-Term Survival Benefit in a Mouse Model of Lethal GvHD. Mol Ther 2019; 27:1436-1451. [PMID: 31138510 DOI: 10.1016/j.ymthe.2019.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/02/2019] [Accepted: 05/04/2019] [Indexed: 12/29/2022] Open
Abstract
Acute graft-versus-host disease is a frequent complication associated with allogeneic hematopoietic stem cell transplantation. Patients that become refractory to initial steroid treatment have a poor prognosis. apceth-201 consists of human allogeneic mesenchymal stromal cells, engineered by lentiviral transduction to express the protease inhibitor alpha-1 antitrypsin, to augment the anti-inflammatory potential of the mesenchymal stromal cells. We show that apceth-201 mesenchymal stromal cells efficiently suppress T cell proliferation and polarize macrophages to an anti-inflammatory M2 type, in vitro. To assess the in vivo efficacy of apceth-201, it was tested in two different mouse models of acute graft-versus-host disease. Control animals in a humanized model succumbed quickly to disease, whereas median survival was doubled in apceth-201-treated animals. The product was also tested in a graft-versus-host disease model system that closely mimics haploidentical hematopoietic stem cell transplantation, an approach that is now being evaluated for use in the clinic. Control animals succumbed quickly to disease, whereas treatment with apceth-201 resulted in long-term survival of 57% of the animals. Within 25 days after the second injection, clinical scores returned to baseline in responding animals, indicating complete resolution of graft-versus-host disease. These promising data have led to planning of a phase I study using apceth-201.
Collapse
Affiliation(s)
| | - Emrah I Ozay
- Program in Molecular & Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA; Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ulf Geumann
- apceth Biopharma GmbH, 81377 Munich, Germany
| | | | | | - Sudarvili Shanthalingam
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | | | | | | | - Barbara A Osborne
- Program in Molecular & Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA; Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Gregory N Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | | | - Lisa M Minter
- Program in Molecular & Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA; Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
32
|
Kundrotas G, Karabanovas V, Pleckaitis M, Juraleviciute M, Steponkiene S, Gudleviciene Z, Rotomskis R. Uptake and distribution of carboxylated quantum dots in human mesenchymal stem cells: cell growing density matters. J Nanobiotechnology 2019; 17:39. [PMID: 30866960 PMCID: PMC6417192 DOI: 10.1186/s12951-019-0470-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/26/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Human mesenchymal stem cells (MSCs) have drawn much attention in the field of regenerative medicine for their immunomodulatory and anti-inflammatory effects. MSCs possess specific tumor-oriented migration and incorporation highlighting the potential for MSCs to be used as an ideal carrier for anticancer agents. Bone marrow is the main source of MSCs for clinical applications. MSCs tracking in vivo is a critical component of the safety and efficacy evaluation of therapeutic cell products; therefore, cells must be labeled with contrast agents to enable visualization of the MSCs migration in vivo. Due to their unique properties, quantum dots (QDs) are emerging as optimal tools in long-term MSC optical imaging applications. The aim of this study was to investigate the uptake dynamics, cytotoxity, subcellular and extracellular distribution of non-targeted carboxylated quantum dots in human bone marrow MSCs at different cell growing densities. RESULTS QDs had no negative impact on MSC viability throughout the experiment and accumulated in all observed cells efficiently; however, in some MSCs QDs induced formation of lipid droplets. At low cell growing densities QDs distribute within MSCs cytoplasm already after 1 h of incubation reaching saturation after 6 h. After 24 h QDs localize mainly in the perinuclear region of the cells in endosomes. Interestingly, in more confluent culture QDs localize mostly outside MSCs. QDs abundantly mark MSC long filopodia-like structures attaching neighboring cells. At high cell density cultivation, we for the first time demonstrated that carboxylated QDs localize in human bone marrow MSC extracellular matrix. Moreover, we observed that average photoluminescence lifetime of QDs distributed in extracellular matrix are longer than lifetimes of QDs entrapped in endocytic vesicles; thus, for the first time showing the possibility to identify and distinguish localization of QDs in various extracellular and intracellular structures using fluorescence-lifetime imaging microscopy without additional staining assays. CONCLUSION Carboxylated QDs can be used as nonspecific and effective dye for staining of human bone marrow MSCs and their specific extracellular structures. These results are promising in fundamental stem cell biology as well as in cellular therapy, anticancer drug delivery and tissue engineering.
Collapse
Affiliation(s)
- Gabrielis Kundrotas
- Biobank, National Cancer Institute, Baublio Str. 3b, 08406, Vilnius, Lithuania
- Laboratory of Immunology, National Cancer Institute, Baublio Str. 3b, 08406, Vilnius, Lithuania
| | - Vitalijus Karabanovas
- Biomedical Physics Laboratory, National Cancer Institute, Baublio Str. 3b, 08406, Vilnius, Lithuania
- Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Sauletekis Ave. 11, 10223, Vilnius, Lithuania
| | - Marijus Pleckaitis
- Biomedical Physics Laboratory, National Cancer Institute, Baublio Str. 3b, 08406, Vilnius, Lithuania
| | - Marina Juraleviciute
- Biomedical Physics Laboratory, National Cancer Institute, Baublio Str. 3b, 08406, Vilnius, Lithuania
| | - Simona Steponkiene
- Biomedical Physics Laboratory, National Cancer Institute, Baublio Str. 3b, 08406, Vilnius, Lithuania
| | - Zivile Gudleviciene
- Biobank, National Cancer Institute, Baublio Str. 3b, 08406, Vilnius, Lithuania
| | - Ricardas Rotomskis
- Biomedical Physics Laboratory, National Cancer Institute, Baublio Str. 3b, 08406, Vilnius, Lithuania.
- Biophotonics Group of Laser Research Center, Faculty of Physics, Vilnius University, Sauletekis Ave. 9, 10222, Vilnius, Lithuania.
| |
Collapse
|
33
|
Beljanski V, Grinnemo KH, Österholm C. Pleiotropic roles of autophagy in stem cell-based therapies. Cytotherapy 2019; 21:380-392. [PMID: 30876741 DOI: 10.1016/j.jcyt.2019.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/25/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
Abstract
Stem cells (SCs) have been proven to possess regenerative and immunomodulatory properties and can be used to treat diseases that involve loss of cells due to tissue damage or inflammation. For this approach to succeed, SCs or their derivatives should be able to engraft in the target tissue at least for a short period of time. Unfortunately, once injected, therapeutic SCs will encounter a hostile environment, including hypoxia, lack of nutrients and stromal support, and cells may also be targeted and rejected by the immune system. Therefore, SC's stress-response mechanisms likely play a significant role in survival of injected cells and possibly contribute to their therapeutic efficacy. Autphagy, a stress-response pathway, is involved in many different cellular processes, such as survival during hypoxia and nutrient deprivation, cellular differentiation and de-differentiation, and it can also contribute to their immunovisibility by regulating antigen presentation and cytokine secretion. Autophagy machinery interacts with many proteins and signaling pathways that regulate SC properties, including PI3K/Akt, mammalian target of rapamycin (mTOR), Wnt, Hedgehog and Notch, and it is also involved in regulating intracellular reactive oxygen species (ROS) levels. In this review, we contend that autophagy is an important therapeutic target that can be used to improve the outcome of SC-based tissue repair and regeneration. Further research should reveal whether inhibition or stimulation of autophagy increases the therapeutic utility of SCs and it should also identify appropriate therapeutic regimens that can be applied in the clinic.
Collapse
Affiliation(s)
- Vladimir Beljanski
- NSU Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, Florida, USA.
| | - Karl-Henrik Grinnemo
- Department of Molecular Medicine and Surgery, Division of Cardiothoracic Surgery and Anesthesiology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Department of Surgical Sciences, Division of Cardiothoracic Surgery and Anesthesiology, Uppsala University, Akademiska University Hospital, Uppsala, Sweden
| | - Cecilia Österholm
- Department of Molecular Medicine and Surgery, Division of Cardiothoracic Surgery and Anesthesiology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
34
|
Najar M, Fayyad-Kazan M, Merimi M, Meuleman N, Bron D, Fayyad-Kazan H, Lagneaux L. Reciprocal immuno-biological alterations occur during the co-culture of natural killer cells and adipose tissue-derived mesenchymal stromal cells. Cytotechnology 2019; 71:375-388. [PMID: 30632032 DOI: 10.1007/s10616-019-00294-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023] Open
Abstract
Due to their immune-therapeutic value, adipose tissue-derived mesenchymal stromal cells (AT-MSCs) require a better characterization of their interplay with natural killer (NK) cells known to contribute to the graft-versus-leukemia effects. When cultivated together, AT-MSCs showed cellular cytotoxicity and were therefore killed by NK cells in an activating-cytokine dependent manner. In the presence of AT-MSCs, both ligands and receptors known to drive NK cell interactions were significantly altered. During this co-culture, the proliferation of NK cells was slightly reduced, while their IFN-γ and TNF-α secretion was significantly increased. NK cells displayed sustained degranulation accompanied by increased discharge of their cytolytic granules (perforin, granzymes A and B). On the other hand, activated NK cells reduced the expression of serpins C1 and B9 in AT-MSCs. Collectively, reciprocal immuno-biological alterations occur during the co-culture of NK cells and AT-MSCs. Understanding these changes will increase the safety and efficacy of cell-based immuno-oncotherapy.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada
| | - Mohammad Fayyad-Kazan
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121 Boulevard de Waterloo, 1000, Brussels, Belgium.
| | - Makram Merimi
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121 Boulevard de Waterloo, 1000, Brussels, Belgium.,Laboratory of Physiology, Genetics and Ethnopharmacology, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121 Boulevard de Waterloo, 1000, Brussels, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121 Boulevard de Waterloo, 1000, Brussels, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| |
Collapse
|
35
|
Chang YJ, Zhao XY, Huang XJ. Strategies for Enhancing and Preserving Anti-leukemia Effects Without Aggravating Graft-Versus-Host Disease. Front Immunol 2018; 9:3041. [PMID: 30619371 PMCID: PMC6308132 DOI: 10.3389/fimmu.2018.03041] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/10/2018] [Indexed: 12/29/2022] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) is a curable method for the treatment of hematological malignancies. In the past two decades, the establishment of haploidentical transplant modalities make “everyone has a donor” become a reality. However, graft-versus-host disease (GVHD) and relapse remain the major two causes of death either in the human leukocyte antigen (HLA)-matched transplant or haploidentical transplant settings, both of which restrict the improvement of transplant outcomes. Preclinical mice model showed that both donor-derived T cells and natural killer (NK) cells play important role in the pathogenesis of GVHD and the effects of graft-versus-leukemia (GVL). Hence, understanding the immune mechanisms of GVHD and GVL would provide potential strategies for the control of leukemia relapse without aggravating GVHD. The purpose of the current review is to summarize the biology of GVHD and GVL responses in preclinical models and to discuss potential novel therapeutic strategies to reduce the relapse rate after allo-SCT. We will also review the approaches, including optimal donor selection and, conditioning regimens, donor lymphocyte infusion, BCR/ABL-specific CTL, and chimeric antigen receptor-modified T cells, which have been successfully used in the clinic to enhance and preserve anti-leukemia activity, especially GVL effects, without aggravating GVHD or alleviate GVHD.
Collapse
Affiliation(s)
- Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
36
|
Najar M, Ouhaddi Y, Bouhtit F, Melki R, Afif H, Boukhatem N, Merimi M, Fahmi H. Empowering the immune fate of bone marrow mesenchymal stromal cells: gene and protein changes. Inflamm Res 2018; 68:167-176. [PMID: 30426152 DOI: 10.1007/s00011-018-1198-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/23/2018] [Accepted: 11/07/2018] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE AND DESIGN Bone marrow mesenchymal stromal cells (BM-MSCs) are referred as a promising immunotherapeutic cell product. New approaches using empowered MSCs should be developed as for the treatment or prevention of different immunological diseases. Such preconditioning by new licensing stimuli will empower the immune fate of BM-MSCs and, therefore, promote a better and more efficient biological. Here, our main goal was to establish the immunological profile of BM-MSCs following inflammatory priming and in particular their capacity to adjust their immune-related proteome and transcriptome. MATERIAL AND METHODS To run this study, we have used BM-MSC cell cultures, a pro-inflammatory cytokine cocktail priming, flow cytometry analysis, qPCR and ELISA techniques. RESULTS Different expression levels of several immunological mediators such as COX-1, COX-2, LIF, HGF, Gal-1, HO-1, IL-11, IL-8, IL-6 and TGF-β were constitutively observed in BM-MSCs. Inflammation priming substantially but differentially modulated the gene and protein expression profiles of these mediators. Thus, expressions of COX-2, LIF, HGF, IL-11, IL-8 and IL-6 were highly increased/induced and those of COX-1, Gal-1, and TGF-β were reduced. CONCLUSIONS Collectively, we demonstrated that BM-MSCs are endowed with a specific and modular regulatory machinery which is potentially involved in immunomodulation. Moreover, BM-MSCs are highly sensitive to inflammation and respond to such signal by properly adjusting their gene and protein expression of regulatory factors. Using such preconditioning may empower the immune fate of MSCs and, therefore, enhance their value for cell-based immunotherapy.
Collapse
Affiliation(s)
- Mehdi Najar
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada.
| | - Yassine Ouhaddi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada
| | - Fatima Bouhtit
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Rahma Melki
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Hassan Afif
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada
| | - Noureddine Boukhatem
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Makram Merimi
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco.,Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, 900 Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada.
| |
Collapse
|
37
|
Hilger N, Mueller C, Stahl L, Mueller AM, Zoennchen B, Dluczek S, Halbich C, Wickenhauser C, Gerloff D, Wurm AA, Behre G, Kretschmer A, Fricke S. Incubation of Immune Cell Grafts With MAX.16H5 IgG1 Anti-Human CD4 Antibody Prolonged Survival After Hematopoietic Stem Cell Transplantation in a Mouse Model for Fms Like Tyrosine Kinase 3 Positive Acute Myeloid Leukemia. Front Immunol 2018; 9:2408. [PMID: 30405611 PMCID: PMC6204383 DOI: 10.3389/fimmu.2018.02408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/28/2018] [Indexed: 12/24/2022] Open
Abstract
Despite the constant development of innovative therapeutic options for hematological malignancies, the gold-standard therapy regimen for curative treatment often includes allogeneic hematopoietic stem cell transplantation (HSCT). The graft-vs.-leukemia effect (GVL) is one of the main therapeutic goals that arises from HSCT. On the other hand, graft-vs.-host disease (GVHD) is still one of the main and most serious complications following allogeneic HSCT. In acute myeloid leukemia (AML), HSCT together with high-dose chemotherapy is used as a treatment option. An aggressive progression of the disease, a decreased response to treatment, and a poor prognosis are connected to internal tandem duplication (ITD) mutations in the Fms like tyrosine kinase 3 (FLT3) gene, which affects around 30% of AML patients. In this study, C3H/HeN mice received an allogeneic graft together with 32D-FLT3ITD AML cells to induce acute GVHD and GVL. It was examined if pre-incubation of the graft with the anti-human cluster of differentiation (CD) 4 antibody MAX.16H5 IgG1 prevented the development of GVHD and whether the graft function was impaired. Animals receiving grafts pre-incubated with the antibody together with FLT3ITD AML cells survived significantly longer than mice receiving untreated grafts. The observed prolonged survival due to MAX.16H5 incubation of immune cell grafts prior to transplantation may allow an extended application of additional targeted strategies in the treatment of AML.
Collapse
Affiliation(s)
- Nadja Hilger
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.,Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Claudia Mueller
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Lilly Stahl
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Anne M Mueller
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Bianca Zoennchen
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Sarah Dluczek
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Christoph Halbich
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | | | - Dennis Gerloff
- Department of Dermatology and Venereology, University Hospital Halle, Halle, Germany
| | - Alexander A Wurm
- Division of Hematology and Medical Oncology, Leipzig University Hospital, Leipzig, Germany
| | - Gerhard Behre
- Division of Hematology and Medical Oncology, Leipzig University Hospital, Leipzig, Germany
| | - Anna Kretschmer
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Stephan Fricke
- Immune Tolerance, Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
38
|
Tian T, Yu Z, Zhang N, Chang Y, Zhang Y, Zhang L, Zhou S, Zhang C, Feng G, Huang F. Modified acellular nerve-delivering PMSCs improve functional recovery in rats after complete spinal cord transection. Biomater Sci 2018; 5:2480-2492. [PMID: 29106428 DOI: 10.1039/c7bm00485k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Due to the poor regeneration capacity of neurons and the inhibitory microenvironment, spontaneous regeneration in spinal cord injury (SCI) remains challenging. Tissue engineering is considered a promising approach for enhancing the regeneration of SCI by reconstructing the inherent structure and improving the microenvironment. In this study, the possibility of engineering a nerve complex, which is constructed by acellular nerve delivering placenta mesenchymal stem cells (PMSCs), was assessed for the recovery of a transected spinal cord. Modified acellular nerve grafts were developed, and PMSCs labeled with green fluorescent protein (GFP) were seeded on the graft to construct the engineered nerve complex. Then, the engineered nerve complex was implanted into a 2 mm-length transected gap of the spinal cord. Four weeks after the transplantation, numerous surviving PMSCs were observed in the lesion cavity by immunofluorescence staining. Moreover, co-localization between GFP and neurofilament-200 (NF200) and Neuronal Class III β-Tubulin (Tuj1) was observed at the bridge interface. The PMSCs-graft group exhibited significant function improvement as evaluated by the Basso, Beattie and Bresnahan (BBB) locomotion score and footprint analysis. Eight weeks after surgery, the evoked response was restored in the PMSCs-graft group and numerous thick myelin sheathes were observed compared to that in the control groups. Collectively, our findings suggest that the nerve complex prepared by acellular nerve delivering PMSCs enhanced the structure and function regeneration of the spinal cord after SCI.
Collapse
Affiliation(s)
- Ting Tian
- Institute of Human Anatomy and Histology and Embryology, Otology & Neuroscience Center, Binzhou Medical University, 346 Guanhai Road, Laishan District, Shandong Province 264003, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Immune regulatory cell infusion for graft-versus-host disease prevention and therapy. Blood 2018; 131:2651-2660. [PMID: 29728401 DOI: 10.1182/blood-2017-11-785865] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022] Open
Abstract
Current approaches to prevent and treat graft-versus-host disease (GVHD) after stem cell transplantation rely principally on pharmacological immune suppression. Such approaches are limited by drug toxicity, nonspecific immune suppression, and a requirement for long-term therapy. Our increased understanding of the regulatory cells and molecular pathways involved in limiting pathogenic immune responses opens the opportunity for the use of these cell subsets to prevent and/or GVHD. The theoretical advantages of this approach is permanency of effect, potential for facilitating tissue repair, and induction of tolerance that obviates a need for ongoing drug therapy. To date, a number of potential cell subsets have been identified, including FoxP3+ regulatory T (Treg) and FoxP3negIL-10+ (FoxP3-negative) regulatory T (Tr1), natural killer (NK) and natural killer T (NKT) cells, innate lymphoid cells, and various myeloid suppressor populations of hematopoietic (eg, myeloid derived suppressor cells) and stromal origin (eg, mesenchymal stem cells). Despite initial technical challenges relating to large-scale selection and expansion, these regulatory lineages are now undergoing early phase clinical testing. To date, Treg therapies have shown promising results in preventing clinical GVHD when infused early after transplant. Results from ongoing studies over the next 5 years will delineate the most appropriate cell lineage, source (donor, host, third party), timing, and potential exogenous cytokine support needed to achieve the goal of clinical transplant tolerance.
Collapse
|
40
|
Carty F, Corbett JM, Cunha JPMCM, Reading JL, Tree TIM, Ting AE, Stubblefield SR, English K. Multipotent Adult Progenitor Cells Suppress T Cell Activation in In Vivo Models of Homeostatic Proliferation in a Prostaglandin E2-Dependent Manner. Front Immunol 2018; 9:645. [PMID: 29740426 PMCID: PMC5925221 DOI: 10.3389/fimmu.2018.00645] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/14/2018] [Indexed: 12/14/2022] Open
Abstract
Lymphodepletion strategies are used in the setting of transplantation (including bone marrow, hematopoietic cell, and solid organ) to create space or to prevent allograft rejection and graft versus host disease. Following lymphodepletion, there is an excess of IL-7 available, and T cells that escape depletion respond to this cytokine undergoing accelerated proliferation. Moreover, this environment promotes the skew of T cells to a Th1 pro-inflammatory phenotype. Existing immunosuppressive regimens fail to control this homeostatic proliferative (HP) response, and thus the development of strategies to successfully control HP while sparing T cell reconstitution (providing a functioning immune system) represents a significant unmet need in patients requiring lymphodepletion. Multipotent adult progenitor cells (MAPC®) have the capacity to control T cell proliferation and Th1 cytokine production. Herein, this study shows that MAPC cells suppressed anti-thymocyte globulin-induced cytokine production but spared T cell reconstitution in a pre-clinical model of lymphodepletion. Importantly, MAPC cells administered intraperitoneally were efficacious in suppressing interferon-γ production and in promoting the expansion of regulatory T cells in the lymph nodes. MAPC cells administered intraperitoneally accumulated in the omentum but were not present in the spleen suggesting a role for soluble factors. MAPC cells suppressed lymphopenia-induced cytokine production in a prostaglandin E2-dependent manner. This study suggests that MAPC cell therapy may be useful as a novel strategy to target lymphopenia-induced pathogenic T cell responses in lymphodepleted patients.
Collapse
Affiliation(s)
- Fiona Carty
- Department of Biology, Institute of Immunology, Maynooth University, Maynooth, Ireland
| | - Jennifer M Corbett
- Department of Biology, Institute of Immunology, Maynooth University, Maynooth, Ireland
| | | | - James L Reading
- Department of Immunobiology, King's College London, London, United Kingdom
| | - Timothy I M Tree
- Department of Immunobiology, King's College London, London, United Kingdom
| | | | | | - Karen English
- Department of Biology, Institute of Immunology, Maynooth University, Maynooth, Ireland
| |
Collapse
|
41
|
Zhang B, Yeo RWY, Lai RC, Sim EWK, Chin KC, Lim SK. Mesenchymal stromal cell exosome-enhanced regulatory T-cell production through an antigen-presenting cell-mediated pathway. Cytotherapy 2018; 20:687-696. [PMID: 29622483 DOI: 10.1016/j.jcyt.2018.02.372] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/07/2018] [Accepted: 02/27/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS The immunomodulatory property of mesenchymal stromal cell (MSC) exosomes is well documented. On the basis of our previous report that MSC exosomes increased regulatory T-cell (Treg) production in mice with allogenic skin graft but not in ungrafted mice, we hypothesize that an activated immune system is key to exosome-mediated Treg production. METHODS To test our hypothesis, MSC exosomes were incubated with mouse spleen CD4+ T cells that were activated with either anti-CD3/CD28 mAbs or allogenic antigen-presenting cell (APC)-enriched spleen CD11c+ cells to determine whether production of mouse CD4+CD25+ T cells or CD4+CD25+Foxp3+ Tregs could be induced. MSC exosomes were also administered to the lethal chimeric human-SCID mouse model of graft-versus-host disease (GVHD) in which human peripheral blood mononuclear cells were infused into irradiated NSG mice to induce GVHD. RESULTS We report here that MSC exosome-induced production of CD4+CD25+ T cells or CD4+CD25+Foxp3+ Tregs from CD4+ T cells activated by allogeneic APC-enriched CD11C+ cells but not those activated by anti-CD3/CD28 mAbs. This induction was exosome- and APC dose-dependent. In the mouse GVHD model in which GVHD was induced by transplanted human APC-stimulated human anti-mouse CD4+ T cell effectors, MSC exosome alleviated GVHD symptoms and increased survival. Surviving exosome-treated mice had a significantly higher level of human CD4+CD25+CD127low/- Tregs than surviving mice treated with Etanercept, a tumor necrosis factor inhibitor. CONCLUSIONS MSC exosome enhanced Treg production in vitro and in vivo through an APC-mediated pathway.
Collapse
Affiliation(s)
- Bin Zhang
- Institute of Medical Biology, A*STAR, Singapore
| | | | | | | | - Keh Chuang Chin
- Institute of Molecular and Cell Biology, A*STAR, Singapore; Department of Physiology, YLL School of Medicine, NUS, Singapore
| | - Sai Kiang Lim
- Institute of Medical Biology, A*STAR, Singapore; Department of Surgery, YLL School of Medicine, NUS, Singapore.
| |
Collapse
|
42
|
Wang L, Zhang H, Guan L, Zhao S, Gu Z, Wei H, Gao Z, Wang F, Yang N, Luo L, Li Y, Wang L, Liu D, Gao C. Mesenchymal stem cells provide prophylaxis against acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation: A meta-analysis of animal models. Oncotarget 2018; 7:61764-61774. [PMID: 27528221 PMCID: PMC5308689 DOI: 10.18632/oncotarget.11238] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 07/28/2016] [Indexed: 02/06/2023] Open
Abstract
A meta-analysis of animal models was conducted to evaluate the prophylactic effects of mesenchymal stem cells (MSCs) on acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation. A total of 50 studies involving 1848 animals were included. The pooled results showed that MSCs significantly reduced aGVHD-associated mortality (risk ratio = 0.70, 95% confidence interval 0.62 to 0.79, P = 2.73×10−9) and clinical scores (standardized mean difference = −3.60, 95% confidence interval −4.43 to −2.76, P = 3.61×10−17). In addition, MSCs conferred robust favorable prophylactic effects on aGVHD across recipient species, MSC doses, and administration times, but not MSC sources. Our meta-analysis showed that MSCs significantly prevented mortality and alleviated the clinical manifestations of aGVHD in animal models. These data support further clinical trials aimed at evaluating the efficacy of using MSCs to prevent aGVHD.
Collapse
Affiliation(s)
- Li Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Hematology and Oncology, Laoshan Branch, No. 401 Hospital of Chinese PLA, Qingdao, China
| | - Haiyan Zhang
- Department of Hematology, Linyi People's Hospital, Linyi, China
| | - Lixun Guan
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Shasha Zhao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhenyang Gu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Huaping Wei
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhe Gao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Feiyan Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Nan Yang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lan Luo
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yonghui Li
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lili Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Daihong Liu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Chunji Gao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
43
|
Chen Q, Chen R, Yuan H, Liu P, Hu A, Wu L, Liu J. Immunosuppressive mechanism of Hypoderma lineatum secreted serine esterase, a potential modulatory method used to inhibit transplant rejection. ELECTRON J BIOTECHN 2018. [DOI: 10.1016/j.ejbt.2018.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
44
|
Najar M, Fayyad-Kazan M, Meuleman N, Bron D, Fayyad-Kazan H, Lagneaux L. Immunological impact of Wharton's Jelly mesenchymal stromal cells and natural killer cell co-culture. Mol Cell Biochem 2018; 447:111-124. [PMID: 29380244 DOI: 10.1007/s11010-018-3297-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/23/2018] [Indexed: 12/20/2022]
Abstract
Due to their easier isolation, multilineage potential, and immunomodulatory capacity, Wharton's Jelly-derived mesenchymal stromal cells (WJ-MSCs) exhibit promising efficacy in the field of regenerative medicine and immunotherapy. Characterization of WJ-MSCs-natural killer (NK) cells crosstalk is required for ameliorating the medicinal value of WJ-MSCs. Here, we revealed that the outcome of WJ-MSCs-NK cells crosstalk varied according to the type of cytokines (IL-2, IL-12, IL-15 and IL-21) utilized to activate NK cells. Differently activated NK cells exerted distinct cytotoxicities against WJ-MSCs causing their probable death. Cell surface ligands (CD112, CD155, ULPB-3) and receptors (LAIR, CD226, CD314, CD335, CD336 and CD337) governing the interaction between NK cells and their targets, exhibited altered expression profiles following the co-culture with WJ-MSCs. Although partly inhibited NK cell proliferation, WJ-MSCs enhanced activated NK-cell-mediated secretion of IFN-γ and TNF-α. Moreover, WJ-MSCs reinforced NK cells' degranulation as well as secretion of perforin and granzymes. On the other hand, WJ-MSCs displayed only slight increase in ROS generation but significant decrease in A1 and C1 serpins expression following co-culture with activated NK cells. Altogether, our results highlight that WJ-MSCs-NK cells interaction may affect both cell type features and, therefore, their therapeutic properties.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Mohammad Fayyad-Kazan
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium.
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 121, Boulevard de Waterloo, 1000, Bruxelles, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| |
Collapse
|
45
|
Najar M, Fayyad-Kazan M, Meuleman N, Bron D, Fayyad-Kazan H, Lagneaux L. Immunomodulatory effects of foreskin mesenchymal stromal cells on natural killer cells. J Cell Physiol 2018; 233:5243-5254. [PMID: 29194614 DOI: 10.1002/jcp.26305] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022]
Abstract
Foreskin-mesenchymal stromal cells (FSK-MSCs) are immune-privileged thus making them valuable immunotherapeutic cell product. Characterization of the relationship between FSK-MSCs and natural killer (NK) cells is essential to improve cell-based therapy. In the present study, we studied for the first time FSK-MSCs-NK interaction and showed that the result of such cross talk was robustly dependent on the type of cytokines (IL-2, IL-12, IL-15, and IL-21) employed to activate NK cells. Distinctly activated-NK cells showed uneven cytotoxicity against FSK-MSCs, triggering their death in fine. The expression of different cell-surface ligands (CD112, CD155, ULPB-3) and receptors (LAIR, KIRs) ensuring such interaction was altered following co-culture of both populations. Despite their partial negative effect on NK cell proliferation, FSK-MSCs boosted the capacity of activated NK-cells to secrete IFN-γ and TNF-α. Moreover, FSK-MSCs enhanced degranulation of NK cells, reinforced secretion of perforin and granzymes, while only modestly increased ROS production. On the other hand, FSK-MSCs-mediated expression of C1 and B9 serpins was significantly lowered in the presence of activated NK cells. Altogether, our results highlight major immunological changes following FSK-MSCs-NK interaction. Understanding these outcomes will therefore enhance the value of the therapeutic strategy.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Mohammad Fayyad-Kazan
- Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, Gosselies, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo, Bruxelles, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium.,Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo, Bruxelles, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| |
Collapse
|
46
|
Cimino M, Gonçalves RM, Bauman E, Barroso-Vilares M, Logarinho E, Barrias CC, Martins MCL. Optimization of the use of a pharmaceutical grade xeno-free medium for in vitro expansion of human mesenchymal stem/stromal cells. J Tissue Eng Regen Med 2017; 12:e1785-e1795. [PMID: 29024519 DOI: 10.1002/term.2588] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 09/23/2017] [Accepted: 10/03/2017] [Indexed: 01/04/2023]
Abstract
Human bone marrow-derived mesenchymal stem/stromal cells (hMSCs) are considered promising therapeutic agents in the field of cell therapy and regenerative medicine, mainly due to their relative facility to be isolated, multi-differentiation potential, and immunomodulatory role. However, their application in clinics requires a crucial step of in vitro expansion. Most of the protocols for hMSCs in vitro culture use foetal bovine serum as medium supplement that, being from animal origin, presents several safety concerns and may initiate xenogeneic immune responses after cells transplantation. This work reports the optimization of a pharmaceutical-grade xeno-free strategy for hMSCs in vitro expansion based on the supplementation of basal medium with a pharmaceutical-grade human plasma-derived supplement for cell culture (SCC) and 2 human growth factors (bFGF and TGFβ1), plus a coating of human plasma fibronectin (Fn). After 4 weeks in culture, this strategy improves hMSCs expansion yield about 4.3-fold in comparison with foetal bovine serum supplementation and 4.5-fold compared with a commercially available xeno-free medium. hMSCs expanded in SCC-based formulation maintained their phenotype and differentiation capacity into osteogenic, adipogenic, and chondrogenic lineages, without alterations in cell karyotype. Overall, the SCC-based medium appears to be an excellent alternative for the xeno-free expansion of hMSCs as therapeutic agents for clinical applications.
Collapse
Affiliation(s)
- M Cimino
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal
| | - R M Gonçalves
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto (UP), Porto, Portugal
| | - E Bauman
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal
| | - M Barroso-Vilares
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, University of Porto (UP), Porto, Portugal
| | - E Logarinho
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, University of Porto (UP), Porto, Portugal
| | - C C Barrias
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto (UP), Porto, Portugal
| | - M C L Martins
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto (UP), Porto, Portugal
| |
Collapse
|
47
|
Qian L, Dima D, Berce C, Liu Y, Rus I, Raduly LZ, Liu Y, Petrushev B, Berindan-Neagoe I, Irimie A, Tanase A, Jurj A, Shen J, Tomuleasa C. Protein dysregulation in graft versus host disease. Oncotarget 2017; 9:1483-1491. [PMID: 29416707 PMCID: PMC5787452 DOI: 10.18632/oncotarget.23276] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/05/2017] [Indexed: 12/15/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is a well-established treatment for many malignant and non-malignant hematological disorders. As a frequent complication in up to 50% of all patients, graft-versus-host disease is still the main cause for morbidity and non-relapse mortality. Diagnosis is usually done clinically, even though confirmation by pathology is often used to support the clinical findings. Effective treatment requires intensified immunosuppression as early as possible. Although several promising biomarkers have been proposed for an early diagnosis, no internationally-recognized consensus has yet been established. Protein-based biomarkers represent an interesting tool since they have been recently reported to be an important regulator of various cells, including immune cells such as T cells. Therefore, we assume that protein dysregulation is important in the pathogenesis of acute graft versus host disease and their detection might be an possibility in the early diagnosis and monitoring. In this review, we aim to summarize the previous reports of protein biomarkers, focusing on the pathogenesis of the disease and possible implications in diagnostic approaches.
Collapse
Affiliation(s)
- Liren Qian
- Department of Hematology, Navy General Hospital, Beijing, PR China
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Cristian Berce
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Yu Liu
- Department of Hematology, Navy General Hospital, Beijing, PR China
| | - Ioana Rus
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Lajos-Zsolt Raduly
- Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Yi Liu
- Department of Hematology, Navy General Hospital, Beijing, PR China
| | - Bobe Petrushev
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | | | - Alexandru Irimie
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Ancuta Jurj
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Jianliang Shen
- Department of Hematology, Navy General Hospital, Beijing, PR China
| | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania.,Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| |
Collapse
|
48
|
Sekiya S, Shimizu T. Introduction of vasculature in engineered three-dimensional tissue. Inflamm Regen 2017; 37:25. [PMID: 29259724 PMCID: PMC5725988 DOI: 10.1186/s41232-017-0055-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 10/05/2017] [Indexed: 12/20/2022] Open
Abstract
Background With recent developments in tissue engineering technology, various three-dimensional tissues can be generated now. However, as the tissue thickness increases due to three-dimensionalization, it is difficult to increase the tissue scale without introduction of blood vessels. Main text Many methods for vasculature induction have been reported recently. In this review, we introduced several methods which are adjustable vascularization in three-dimensional tissues according to three steps. First, "selection" provides potents for engineered tissues with vascularization ability. Second, "assembly technology" is used to fabricate tissues as three-dimensional structures and simultaneously inner neo-vasculature. Third, a "perfusion" technique is used for maturation of blood vessels in three-dimensional tissues. In "selection", selection of cells and materials gives the ability to promote angiogenesis in three-dimensional tissues. During the cell assembly step, cell sheet engineering, nanofilm coating technology, and three-dimensional printing technology could be used to produce vascularized three-dimensional tissues. Perfusion techniques to perfuse blood or cell culture medium throughout three-dimensional tissues with a unified inlet and outlet could induce functional blood vessels within retransplantable three-dimensional tissues. Combination of each step technology allows simulation of perivascular microenvironments in target tissues and drive vascularization in three-dimensional tissues. Conclusion The biomimetic microenvironment of target tissues will induce adequate cell-cell interaction, distance, cell morphology, and function within tissues. It could be accelerated for vascularization within three-dimensional tissues and give us the functional tissues. Since vascularized three-dimensional tissues are highly functional, they are expected to contribute to the development of regenerative medicine and drug safety tests for drug discovery in the future.
Collapse
Affiliation(s)
- Sachiko Sekiya
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| |
Collapse
|
49
|
Xeno-Free Strategies for Safe Human Mesenchymal Stem/Stromal Cell Expansion: Supplements and Coatings. Stem Cells Int 2017; 2017:6597815. [PMID: 29158740 PMCID: PMC5660800 DOI: 10.1155/2017/6597815] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022] Open
Abstract
Human mesenchymal stem/stromal cells (hMSCs) have generated great interest in regenerative medicine mainly due to their multidifferentiation potential and immunomodulatory role. Although hMSC can be obtained from different tissues, the number of available cells is always low for clinical applications, thus requiring in vitro expansion. Most of the current protocols for hMSC expansion make use of fetal bovine serum (FBS) as a nutrient-rich supplement. However, regulatory guidelines encourage novel xeno-free alternatives to define safer and standardized protocols for hMSC expansion that preserve their intrinsic therapeutic potential. Since hMSCs are adherent cells, the attachment surface and cell-adhesive components also play a crucial role on their successful expansion. This review focuses on the advantages/disadvantages of FBS-free media and surfaces/coatings that avoid the use of animal serum, overcoming ethical issues and improving the expansion of hMSC for clinical applications in a safe and reproducible way.
Collapse
|
50
|
CTLA4-CD28 chimera gene modification of T cells enhances the therapeutic efficacy of donor lymphocyte infusion for hematological malignancy. Exp Mol Med 2017; 49:e360. [PMID: 28751785 PMCID: PMC5565951 DOI: 10.1038/emm.2017.104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 02/06/2023] Open
Abstract
Donor lymphocyte infusion (DLI) followed by hematopoietic stem cell transplantation has served as an effective prevention/treatment modality against the relapse of some hematologic tumors, such as chronic myeloid leukemia (CML). However, the therapeutic efficacies of DLI for other types of leukemia, including acute lymphocytic leukemia (ALL), have been limited thus far. Therefore, we examined whether increasing the reactivity of donor T cells by gene modification could enhance the therapeutic efficacy of DLI in a murine model of ALL. When a CTLA4-CD28 chimera gene (CTC28) in which the intracellular signaling domain of CTLA4 was replaced with the CD28 signaling domain was introduced into CD4 and CD8 T cells in DLI, the graft-versus-tumor (GVT) effect was significantly increased. This effect was correlated with an increased expansion of donor CD8 T cells in vivo, and the depletion of CD8 T cells abolished this effect. The CD8 T cell expansion and the enhanced GVT effect were dependent on the transduction of both CD4 and CD8 T cells with CTC28, which emphasizes the role of dual modification in this therapeutic effect. The CTC28-transduced T cells that expanded in vivo also exhibited enhanced functionality. Although the potentiation of the GVT effect mediated by the CTC28 gene modification of T cells was accompanied by an increase of graft-versus-host disease (GVHD), the GVHD was not lethal and was mitigated by treatment with IL-10 gene-modified third-party mesenchymal stem cells. Thus, the combined genetic modification of CD4 and CD8 donor T cells with CTC28 could be a promising strategy for enhancing the therapeutic efficacy of DLI.
Collapse
|