1
|
Shi H, Ding Y, Sun P, Lv Z, Wang C, Ma H, Lu J, Yu B, Li W, Wang C. Chemical approaches targeting the hurdles of hepatocyte transplantation: mechanisms, applications, and advances. Front Cell Dev Biol 2024; 12:1480226. [PMID: 39544361 PMCID: PMC11560891 DOI: 10.3389/fcell.2024.1480226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Hepatocyte transplantation (HTx) has been a novel cell-based therapy for severe liver diseases, as the donor livers for orthotopic liver transplantation are of great shortage. However, HTx has been confronted with two main hurdles: limited high-quality hepatocyte sources and low cell engraftment and repopulation rate. To cope with, researchers have investigated on various strategies, including small molecule drugs with unique advantages. Small molecules are promising chemical tools to modulate cell fate and function for generating high quality hepatocyte sources. In addition, endothelial barrier, immune responses, and low proliferative efficiency of donor hepatocytes mainly contributes to low cell engraftment and repopulation rate. Interfering these biological processes with small molecules is beneficial for improving cell engraftment and repopulation. In this review, we will discuss the applications and advances of small molecules in modulating cell differentiation and reprogramming for hepatocyte resources and in improving cell engraftment and repopulation as well as its underlying mechanisms.
Collapse
Affiliation(s)
- Huanxiao Shi
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Yi Ding
- Experimental Teaching Center, Naval Medical University, Shanghai, China
| | - Pingxin Sun
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Zhuman Lv
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Chunyan Wang
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Haoxin Ma
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Junyu Lu
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Bing Yu
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Wenlin Li
- Department of Cell Biology, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai, China
| | - Chao Wang
- Department of Cell Biology, Naval Medical University, Shanghai, China
| |
Collapse
|
2
|
Karpf S, Glöckner Burmeister N, Dubreil L, Ghosh S, Hollandi R, Pichon J, Leroux I, Henkel A, Lutz V, Jurkevičius J, Latshaw A, Kilin V, Kutscher T, Wiggert M, Saavedra-Villanueva O, Vogel A, Huber RA, Horvath P, Rouger K, Bonacina L. Harmonic Imaging of Stem Cells in Whole Blood at GHz Pixel Rate. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401472. [PMID: 38863131 DOI: 10.1002/smll.202401472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/21/2024] [Indexed: 06/13/2024]
Abstract
The pre-clinical validation of cell therapies requires monitoring the biodistribution of transplanted cells in tissues of host organisms. Real-time detection of these cells in the circulatory system and identification of their aggregation state is a crucial piece of information, but necessitates deep penetration and fast imaging with high selectivity, subcellular resolution, and high throughput. In this study, multiphoton-based in-flow detection of human stem cells in whole, unfiltered blood is demonstrated in a microfluidic channel. The approach relies on a multiphoton microscope with diffractive scanning in the direction perpendicular to the flow via a rapidly wavelength-swept laser. Stem cells are labeled with metal oxide harmonic nanoparticles. Thanks to their strong and quasi-instantaneous second harmonic generation (SHG), an imaging rate in excess of 10 000 frames per second is achieved with pixel dwell times of 1 ns, a duration shorter than typical fluorescence lifetimes yet compatible with SHG. Through automated cell identification and segmentation, morphological features of each individual detected event are extracted and cell aggregates are distinguished from isolated cells. This combination of high-speed multiphoton microscopy and high-sensitivity SHG nanoparticle labeling in turbid media promises the detection of rare cells in the bloodstream for assessing novel cell-based therapies.
Collapse
Affiliation(s)
- Sebastian Karpf
- Institute of Biomedical Optics (BMO), University Of Luebeck, 23562, Luebeck, Germany
| | | | | | - Shayantani Ghosh
- Department of Applied Physics, Université de Genève, Rue de l'Ecole-de-Médecine, 20, Geneva, 1205, Switzerland
| | - Reka Hollandi
- Synthetic and Systems Biology Unit, Biological Research Centre (BRC), Szeged, H-6726, Hungary
| | | | | | - Alessandra Henkel
- Institute of Biomedical Optics (BMO), University Of Luebeck, 23562, Luebeck, Germany
| | - Valerie Lutz
- Institute of Biomedical Optics (BMO), University Of Luebeck, 23562, Luebeck, Germany
| | - Jonas Jurkevičius
- Institute of Biomedical Optics (BMO), University Of Luebeck, 23562, Luebeck, Germany
| | - Alexandra Latshaw
- Department of Applied Physics, Université de Genève, Rue de l'Ecole-de-Médecine, 20, Geneva, 1205, Switzerland
| | - Vasyl Kilin
- Department of Applied Physics, Université de Genève, Rue de l'Ecole-de-Médecine, 20, Geneva, 1205, Switzerland
| | - Tonio Kutscher
- Institute of Biomedical Optics (BMO), University Of Luebeck, 23562, Luebeck, Germany
| | - Moritz Wiggert
- Department of Applied Physics, Université de Genève, Rue de l'Ecole-de-Médecine, 20, Geneva, 1205, Switzerland
| | | | - Alfred Vogel
- Institute of Biomedical Optics (BMO), University Of Luebeck, 23562, Luebeck, Germany
| | - Robert A Huber
- Institute of Biomedical Optics (BMO), University Of Luebeck, 23562, Luebeck, Germany
| | - Peter Horvath
- Synthetic and Systems Biology Unit, Biological Research Centre (BRC), Szeged, H-6726, Hungary
| | - Karl Rouger
- Oniris, INRAE, PAnther, Nantes, F-44307, France
| | - Luigi Bonacina
- Department of Applied Physics, Université de Genève, Rue de l'Ecole-de-Médecine, 20, Geneva, 1205, Switzerland
| |
Collapse
|
3
|
Kwon J, Kim MY, Lee S, Lee J, Yoon HY. Pulmonary passage of canine adipose tissue-derived mesenchymal stem cells through intravenous transplantation in mouse model. J Vet Sci 2024; 25:e36. [PMID: 38834506 PMCID: PMC11156597 DOI: 10.4142/jvs.23300] [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: 12/06/2023] [Revised: 01/16/2024] [Accepted: 03/29/2024] [Indexed: 06/06/2024] Open
Abstract
IMPORTANCE The intravenous administration of adipose tissue-derived mesenchymal stem cells (AdMSCs) in veterinary medicine is an attractive treatment option. On the other hand, it can result in severe complications, including pulmonary thromboembolism (PTE). OBJECTIVE The present study assessed the occurrence of PTE after the intravenous infusion of canine AdMSCs (cAdMSCs) into experimental animals. METHODS Five-week-old male BALB/c hairless mice were categorized into groups labeled A to G. In the control group (A), fluorescently stained 2 × 106 cAdMSCs were diluted in 200 μL of suspension and injected into the tail vein as a single bolus. The remaining groups included the following: group B with 5 × 106 cells, group C with 3 × 106 cells, group D with 1 × 106 cells, group E with 1 × 106 cells injected twice with a one-day interval, group F with 2 × 106 cells in 100 μL of suspension, and group G with 2 × 106 cells in 300 μL of suspension. RESULTS Group D achieved a 100% survival rate, while none of the subjects in groups B and C survived (p = 0.002). Blood tests revealed a tendency for the D-dimer levels to increase as the cell dose increased (p = 0.006). The platelet count was higher in the low cell concentration groups and lower in the high cell concentration groups (p = 0.028). A histological examination revealed PTE in most deceased subjects (96.30%). CONCLUSIONS AND RELEVANCE PTE was verified, and various variables were identified as potential contributing factors, including the cell dose, injection frequency, and suspension volume.
Collapse
Affiliation(s)
- Jaeyeon Kwon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Mu-Young Kim
- Department of Small Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32611, USA
| | - Soojung Lee
- Department of Companion Animal Health, Yeonsung University, Anyang 14011, Korea
| | - Jeongik Lee
- Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, School of Medicine, Konkuk University, Seoul 05029, Korea
| | - Hun-Young Yoon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- KU Center for Animal Blood Medical Science, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
4
|
Cao JK, Hong XY, Feng ZC, Li QP. Mesenchymal stem cells-based therapies for severe ARDS with ECMO: a review. Intensive Care Med Exp 2024; 12:12. [PMID: 38332384 PMCID: PMC10853094 DOI: 10.1186/s40635-024-00596-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is the primary cause of respiratory failure in critically ill patients. Despite remarkable therapeutic advances in recent years, ARDS remains a life-threatening clinical complication with high morbidity and mortality, especially during the global spread of the coronavirus disease 2019 (COVID-19) pandemic. Previous studies have demonstrated that mesenchymal stem cell (MSC)-based therapy is a potential alternative strategy for the treatment of refractory respiratory diseases including ARDS, while extracorporeal membrane oxygenation (ECMO) as the last resort treatment to sustain life can help improve the survival of ARDS patients. In recent years, several studies have explored the effects of ECMO combined with MSC-based therapies in the treatment of ARDS, and some of them have demonstrated that this combination can provide better therapeutic effects, while others have argued that some critical issues need to be solved before it can be applied to clinical practice. This review presents an overview of the current status, clinical challenges and future prospects of ECMO combined with MSCs in the treatment of ARDS.
Collapse
Affiliation(s)
- Jing-Ke Cao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Yang Hong
- Department of Pediatric Intensive Care Unit, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, NO.5 Nanmencang, Dongcheng District, 100700, Beijing, China
| | - Zhi-Chun Feng
- Department of Neonatology, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, NO. 5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Qiu-Ping Li
- Department of Neonatology, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, NO. 5 Nanmencang, Dongcheng District, Beijing, 100700, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Bivalirudin exerts antiviral activity against respiratory syncytial virus-induced lung infections in neonatal mice. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2022; 72:415-425. [PMID: 36651544 DOI: 10.2478/acph-2022-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 01/26/2023]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of small airways inflammation in the lungs (bronchiolitis) in neonates and immunocompromised adults. The deregulation of cellular and plasma components leads to increased morbidity and mortality. The activation of the clotting cascade plays a key role in the progression of disease severity during viral infection. The current investigation studied the effect of bivalirudin (BR) on the progression and cellular effects of RSV-induced infection in the neonatal mice model. Mice (5-7 days old) were inoculated intranasally with RSV with or without BR administration (2 mg kg-1 day-1, i.v.) for 2 weeks. Tissue histopathology, inflammatory signalling genes such as TLR, and cytokines were analyzed. The results showed pneumocytes exhibiting nuclear pyknosis, cellular infiltration in lung tissue and increased lung titers in RSV-infected mice compared to the control. Furthermore, RSV-infected mice demonstrated altered clotting parameters such as D-dimer, soluble thrombomodulin, and increased inflammatory cytokines IL-5, 6, IFN-γ, IL-13, and CXCL1. Additionally, the mRNA expression analysis displayed increased levels of IL-33, TLR3, and TLR7 genes in RSV-infected lung tissue. Further, to delineate the role of micro RNAs, the qRT-PCR analysis was done, and the results displayed an increase in miR-136, miR-30b, and let-7i. At the same time, the down-regulated expression of miR-221 in RSV-infected mice compared to the control. BR treatment reduced the cellular infiltration with reduced inflammatory cytokines and normalized clotting indices. Thus, the study shows that RSV infection induces specific changes in lung tissue and the clotting related signalling mechanism. Additionally, BR treatment significantly reduces bronchiolitis and prevents the severity of the infections suggesting that BR can possibly be used to reduce the viral-mediated infections in neonates.
Collapse
|
6
|
Dosing Limitation for Intra-Renal Arterial Infusion of Mesenchymal Stromal Cells. Int J Mol Sci 2022; 23:ijms23158268. [PMID: 35955404 PMCID: PMC9368110 DOI: 10.3390/ijms23158268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 02/01/2023] Open
Abstract
The immunomodulatory and regenerative properties of mesenchymal stromal cells (MSCs) make MSC therapy a promising therapeutic strategy in kidney disease. A targeted MSC administration via the renal artery offers an efficient delivery method with limited spillover to other organs. Although local administration alleviates safety issues with MSCs in systemic circulation, it introduces new safety concerns in the kidneys. In a porcine model, we employed intra-renal arterial infusion of ten million allogenic adipose tissue-derived MSCs. In order to trigger any potential adverse events, a higher dose (hundred million MSCs) was also included. The kidney function was studied by magnetic resonance imaging after the MSC infusion and again at two weeks post-treatment. The kidneys were assessed by single kidney glomerular filtration rate (skGFR) measurements, histology and inflammation, and fibrosis-related gene expression. None of the measured parameters were affected immediately after the administration of ten million MSCs, but the administration of one hundred million MSCs induced severe adverse events. Renal perfusion was reduced immediately after MSC administration which coincided with the presence of microthrombi in the glomeruli and signs of an instant blood-mediated inflammatory reaction. At two weeks post-treatment, the kidneys that were treated with one hundred million MSCs showed reduced skGFR, signs of tissue inflammation, and glomerular and tubular damage. In conclusions, the intra-renal administration of ten million MSCs is well-tolerated by the porcine kidney. However, higher concentrations (one hundred million MSCs) caused severe kidney damage, implying that very high doses of intra-renally administered MSCs should be undertaken with caution.
Collapse
|
7
|
Moll G, Ankrum JA, Olson SD, Nolta JA. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:2-13. [PMID: 35641163 PMCID: PMC8895495 DOI: 10.1093/stcltm/szab005] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/27/2021] [Indexed: 11/22/2022] Open
Abstract
The number of mesenchymal stromal/stem cell (MSC) therapeutics and types of clinical applications have greatly diversified during the past decade, including rapid growth of poorly regulated “Stem Cell Clinics” offering diverse “Unproven Stem Cell Interventions.” This product diversification necessitates a critical evaluation of the reliance on the 2006 MSC minimal criteria to not only define MSC identity but characterize MSC suitability for intravascular administration. While high-quality MSC therapeutics have been safely administered intravascularly in well-controlled clinical trials, repeated case reports of mild-to-more-severe adverse events have been reported. These are most commonly related to thromboembolic complications upon infusion of highly procoagulant tissue factor (TF/CD142)-expressing MSC products. As TF/CD142 expression varies widely depending on the source and manufacturing process of the MSC product, additional clinical cell product characterization and guidelines are needed to ensure the safe use of MSC products. To minimize risk to patients receiving MSC therapy, we here propose to supplement the minimal criteria used for characterization of MSCs, to include criteria that assess the suitability of MSC products for intravascular use. If cell products are intended for intravascular delivery, which is true for half of all clinical applications involving MSCs, the effects of MSC on coagulation and hemocompatibility should be assessed and expression of TF/CD142 should be included as a phenotypic safety marker. This adjunct criterion will ensure both the identity of the MSCs as well as the safety of the MSCs has been vetted prior to intravascular delivery of MSC products.
Collapse
Affiliation(s)
- Guido Moll
- BIH Center for Regenerative Therapies (BCRT) and Berlin Brandenburg School of Regenerative Therapies (BSRT), Berlin Institute of Health (BIH) at the Charité—Universitätsmedizin Berlin, corporate member of Freie Universität zu Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Corresponding author: Guido Moll, PhD, BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering and Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Jan A Nolta
- Director of the Stem Cell Program, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
8
|
Laner-Plamberger S, Oeller M, Rohde E, Schallmoser K, Strunk D. Heparin and Derivatives for Advanced Cell Therapies. Int J Mol Sci 2021; 22:12041. [PMID: 34769471 PMCID: PMC8584295 DOI: 10.3390/ijms222112041] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/27/2022] Open
Abstract
Heparin and its derivatives are saving thousands of human lives annually, by successfully preventing and treating thromboembolic events. Although the mode of action during anticoagulation is well studied, their influence on cell behavior is not fully understood as is the risk of bleeding and other side effects. New applications in regenerative medicine have evolved supporting production of cell-based therapeutics or as a substrate for creating functionalized matrices in biotechnology. The currently resurgent interest in heparins is related to the expected combined anti-inflammatory, anti-thrombotic and anti-viral action against COVID-19. Based on a concise summary of key biochemical and clinical data, this review summarizes the impact for manufacturing and application of cell therapeutics and highlights the need for discriminating the different heparins.
Collapse
Affiliation(s)
- Sandra Laner-Plamberger
- Department of Transfusion Medicine, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (S.L.-P.); (M.O.); (E.R.)
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria;
| | - Michaela Oeller
- Department of Transfusion Medicine, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (S.L.-P.); (M.O.); (E.R.)
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria;
| | - Eva Rohde
- Department of Transfusion Medicine, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (S.L.-P.); (M.O.); (E.R.)
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria;
| | - Katharina Schallmoser
- Department of Transfusion Medicine, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (S.L.-P.); (M.O.); (E.R.)
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria;
| | - Dirk Strunk
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria;
- Cell Therapy Institute, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
9
|
Riedl J, Popp C, Eide C, Ebens C, Tolar J. Mesenchymal stromal cells in wound healing applications: role of the secretome, targeted delivery and impact on recessive dystrophic epidermolysis bullosa treatment. Cytotherapy 2021; 23:961-973. [PMID: 34376336 PMCID: PMC8569889 DOI: 10.1016/j.jcyt.2021.06.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/25/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multi-potent stromal-derived cells capable of self-renewal that possess several advantageous properties for wound healing, making them of interest to the field of dermatology. Research has focused on characterizing the unique properties of MSCs, which broadly revolve around their regenerative and more recently discovered immunomodulatory capacities. Because of ease of harvesting and expansion, differentiation potential and low immunogenicity, MSCs have been leading candidates for tissue engineering and regenerative medicine applications for wound healing, yet results from clinical studies have been variable, and promising pre-clinical work has been difficult to reproduce. Therefore, the specific mechanisms of how MSCs influence the local microenvironment in distinct wound etiologies warrant further research. Of specific interest in MSC-mediated healing is harnessing the secretome, which is composed of components known to positively influence wound healing. Molecules released by the MSC secretome can promote re-epithelialization and angiogenesis while inhibiting fibrosis and microbial invasion. This review focuses on the therapeutic interest in MSCs with regard to wound healing applications, including burns and diabetic ulcers, with specific attention to the genetic skin disease recessive dystrophic epidermolysis bullosa. This review also compares various delivery methods to support skin regeneration in the hopes of combating the poor engraftment of MSCs after delivery, which is one of the major pitfalls in clinical studies utilizing MSCs.
Collapse
Affiliation(s)
- Julia Riedl
- Medical Scientist Training Program (MD/PhD), University of Minnesota, Minneapolis, Minnesota, USA; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Courtney Popp
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cindy Eide
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christen Ebens
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
10
|
Nevens F, Gustot T, Laterre PF, Lasser LL, Haralampiev LE, Vargas V, Lyubomirova D, Albillos A, Najimi M, Michel S, Stoykov I, Gordillo N, Vainilovich Y, Barthel V, Clerget-Chossat N, Sokal EM. A phase II study of human allogeneic liver-derived progenitor cell therapy for acute-on-chronic liver failure and acute decompensation. JHEP Rep 2021; 3:100291. [PMID: 34169246 PMCID: PMC8207211 DOI: 10.1016/j.jhepr.2021.100291] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/16/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND & AIMS Human allogeneic liver-derived progenitor cells (HALPC, HepaStem®; Promethera Biosciences, Mont-Saint-Guibert, Belgium) are an advanced therapy medicinal product that could potentially alleviate systemic inflammation and ameliorate liver function in patients with acute-on-chronic liver failure (ACLF) or acute decompensation of cirrhosis (AD). METHODS This open-label phase II study was conducted in 9 centres in Belgium, Spain, and Bulgaria between 2016 and 2019. The primary objective was to assess the safety of HALPC therapy up to Day 28 and the secondary objectives were to assess its safety and preliminary efficacy up to Month 3. RESULTS The 24 treated patients (mean age: 51 years) were mostly male with an alcoholic cirrhosis. On pre-infusion Day 1, 15 patients had ACLF and 9 patients had AD. Two of the 3 initial patients treated with high HALPC doses (∼5×106 cells/kg body weight [BW]) had severe adverse bleeding events attributed to treatment. In 21 patients subsequently treated with lower HALPC doses (0.6 or 1.2×106 cells/kg BW, 1 or 2 times 7 days apart), no serious adverse events were related to treatment, and the other adverse events were in line with those expected in patients with ACLF and AD. Overall, markers of systemic inflammation and altered liver function decreased gradually for the surviving patients. The Day-28 and Month-3 survival rates were 83% (20/24) and 71% (17/24), and at Month 3, no patient had ACLF. CONCLUSIONS The treatment of patients with ACLF or AD with up to 2 doses of 1.2×106 HALPC/kg BW appeared safe. The results of this study support the initiation of a proof-of-concept study in a larger cohort of patients with ACLF to further confirm the safety and evaluate the efficacy of HALPC therapy. CLINICAL TRIALS REGISTRATION EudraCT 2016-001177-32. LAY SUMMARY Patients with liver cirrhosis may suffer from the rapid onset of organ failure or multiple organ failure associated with a high risk of death in the short term. This clinical study of 24 patients suggests that an advanced therapy based on the intravenous infusion of low doses of human allogeneic liver-derived progenitor cells is safe and supports the next phase of clinical development of this type of therapy.
Collapse
Key Words
- ACLF, acute-on-chronic liver failure
- AD, acute decompensation of liver cirrhosis
- AE, adverse event
- AESI, AE of special interest
- ATMP, advanced therapy medicinal product
- Alcoholic liver disease
- BW, body weight
- CRP, C-reactive protein
- EASL-CLIF, European Association for the Study of Chronic Liver Failure
- HALPC, human allogeneic liver-derived progenitor cells
- INR, international normalised ratio
- Liver regenerative medicine
- MELD, model for end-stage liver disease
- MSC, mesenchymal stem cells
- SAE, serious AE
- SAS, safety analysis set
- SUSAR, suspected unexpected serious adverse reaction
- Stem cell
- TEG, thromboelastography
- TGT, thrombin generation test
- i.v., intravenous
Collapse
Affiliation(s)
- Frederik Nevens
- Department of Gastroenterology and Hepatology, University Hospitals, KU Leuven, Belgium
| | - Thierry Gustot
- Department of Gastroenterology and Hepato-Pancreatology, C.U.B. Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Luc L. Lasser
- Gastroenterology Clinic, CHU Brugmann, Brussels, Belgium
- Department of Hepatogastroenterology, CHU Brugmann, Brussels, Belgium
| | - Lyudmil E. Haralampiev
- Department of Internal Diseases, Multiprofile Hospital for Active Treatment (MEDICA), Ruse, Bulgaria
| | - Victor Vargas
- Liver Unit, Hospital Vall d'Hebron, Universitat Autònoma Barcelona, CIBERehd, Barcelona, Spain
| | - Desislava Lyubomirova
- Department of Clinical Gastroenterology with Hepatology, Gastroenterology Clinic, University Multiprofile Hospital for Active Treatment “Georgi Stranski”, Pleven, Bulgaria
| | - Agustin Albillos
- Gastroenterology and Hepatology, University Hospital Ramón y Cajal, Madrid, Spain
| | - Mustapha Najimi
- UCLouvain, Institute of Experimental and Clinical Research (IREC), Laboratory of Pediatric Hepatology and Cell Therapy (PEDI), Brussels, Belgium
| | | | | | | | | | | | | | - Etienne M. Sokal
- UCLouvain, Institute of Experimental and Clinical Research (IREC), Laboratory of Pediatric Hepatology and Cell Therapy (PEDI), Brussels, Belgium
- Promethera Biosciences, Mont-Saint-Guibert, Belgium
- Cliniques Universitaires Saint-Luc, UCLouvain, Pediatric Hepatology & Gastroenterology Unit, Brussels, Belgium
| |
Collapse
|
11
|
Dollet PE, Hsu MJ, Ambroise J, Rozzi M, Ravau J, André F, Evraerts J, Najimi M, Sokal E, Lombard C. Evaluation of Strategies Aimed at Improving Liver Progenitor Cell Rolling and Subsequent Adhesion to the Endothelium. Cell Transplant 2021; 29:963689720912707. [PMID: 32425073 PMCID: PMC7444224 DOI: 10.1177/0963689720912707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Adult-derived human liver stem/progenitor cells (ADHLSCs) are a promising
alternative to orthotopic liver transplantation in the treatment of inborn
errors of metabolism. However, as is the case with many mesenchymal stromal
cells, ADHLSCs have shown a low level of engraftment, which could be explained
by the fact that they lack expression of selectin ligand and LFA-1 and only
slightly express VLA- 4, molecules that have been shown to be involved in cell
adhesion to the endothelium. In this paper, we have investigated strategies to
increase their rolling and adhesion during the homing process by (1) adding a
selectin ligand (Sialyl Lewis X) to their surface using
biotinyl-N-hydroxy-succinimide–streptavidin bridges, and
(2) protecting the adhesion proteins from trypsinization-induced damage using a
thermosensitive polymer for cell culture and a nonenzymatic cell dissociation
solution (CDS) for harvest. Despite increasing adhesion of ADHLSCs to E-selectin
during an adhesion test in vitro performed under shear stress,
the addition of Sialyl Lewis X did not increase adhesion to endothelial cells
under the same conditions. Cultivating cells on a thermosensitive polymer and
harvesting them with CDS increased their adhesion to endothelial cells under
noninflammatory conditions, compared to the use of trypsin. However, we were not
able to demonstrate any improvement in cell adhesion to the endothelium
following culture on polymer and harvest with CDS, suggesting that alternative
methods of improving engraftment still need to be evaluated.
Collapse
Affiliation(s)
- Pierre Edouard Dollet
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Mei Ju Hsu
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Jérôme Ambroise
- Centre de Technologies Moléculaires Appliquées (CTMA), Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Milena Rozzi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Joachim Ravau
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Floriane André
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Jonathan Evraerts
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Etienne Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Catherine Lombard
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| |
Collapse
|
12
|
Byrnes D, Masterson CH, Artigas A, Laffey JG. Mesenchymal Stem/Stromal Cells Therapy for Sepsis and Acute Respiratory Distress Syndrome. Semin Respir Crit Care Med 2020; 42:20-39. [PMID: 32767301 DOI: 10.1055/s-0040-1713422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sepsis and acute respiratory distress syndrome (ARDS) constitute devastating conditions with high morbidity and mortality. Sepsis results from abnormal host immune response, with evidence for both pro- and anti-inflammatory activation present from the earliest phases. The "proinflammatory" response predominates initially causing host injury, with later-phase sepsis characterized by immune cell hypofunction and opportunistic superinfection. ARDS is characterized by inflammation and disruption of the alveolar-capillary membrane leading to injury and lung dysfunction. Sepsis is the most common cause of ARDS. Approximately 20% of deaths worldwide in 2017 were due to sepsis, while ARDS occurs in over 10% of all intensive care unit patients and results in a mortality of 30 to 45%. Given the fact that sepsis and ARDS share some-but not all-underlying pathophysiologic injury mechanisms, the lack of specific therapies, and their frequent coexistence in the critically ill, it makes sense to consider therapies for both conditions together. In this article, we will focus on the therapeutic potential of mesenchymal stem/stromal cells (MSCs). MSCs are available from several tissues, including bone marrow, umbilical cord, and adipose tissue. Allogeneic administration is feasible, an important advantage for acute conditions like sepsis or ARDS. They possess diverse mechanisms of action of relevance to sepsis and ARDS, including direct and indirect antibacterial actions, potent effects on the innate and adaptive response, and pro-reparative effects. MSCs can be preactivated thereby potentiating their effects, while the use of their extracellular vesicles can avoid whole cell administration. While early-phase clinical trials suggest safety, considerable challenges exist in moving forward to phase III efficacy studies, and to implementation as a therapy should they prove effective.
Collapse
Affiliation(s)
- Declan Byrnes
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Claire H Masterson
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Antonio Artigas
- Critical Care Center, Corporació Sanitaria Parc Tauli, CIBER Enfermedades Respiratorias, Autonomous University of Barcelona, Sabadell, Spain
| | - John G Laffey
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.,Department of Anaesthesia, SAOLTA University Health Group, Galway University Hospitals, Galway, Ireland
| |
Collapse
|
13
|
Phase I/II Trial of Liver-derived Mesenchymal Stem Cells in Pediatric Liver-based Metabolic Disorders: A Prospective, Open Label, Multicenter, Partially Randomized, Safety Study of One Cycle of Heterologous Human Adult Liver-derived Progenitor Cells (HepaStem) in Urea Cycle Disorders and Crigler-Najjar Syndrome Patients. Transplantation 2020; 103:1903-1915. [PMID: 30801523 DOI: 10.1097/tp.0000000000002605] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Regenerative medicine using stem cell technology is an emerging field that is currently tested for inborn and acquired liver diseases. OBJECTIVE This phase I/II prospective, open label, multicenter, randomized trial aimed primarily at evaluating the safety of Heterologous Human Adult Liver-derived Progenitor Cells (HepaStem) in pediatric patients with urea cycle disorders (UCDs) or Crigler-Najjar (CN) syndrome 6 months posttransplantation. The secondary objective included the assessment of safety up to 12 months postinfusion and of preliminary efficacy. METHODS Fourteen patients with UCDs and 6 with CN syndrome were divided into 3 cohorts by body weight and intraportally infused with 3 doses of HepaStem. Clinical status, portal vein hemodynamics, morphology of the liver, de novo detection of circulating anti-human leukocyte antigen antibodies, and clinically significant adverse events (AEs) and serious adverse events to infusion were evaluated by using an intent-to-treat analysis. RESULTS The overall safety of HepaStem was confirmed. For the entire study period, patient-month incidence rate was 1.76 for the AEs and 0.21 for the serious adverse events, of which 38% occurred within 1 month postinfusion. There was a trend of higher events in UCD as compared with CN patients. Segmental left portal vein thrombosis occurred in 1 patient and intraluminal local transient thrombus in a second patient. The other AEs were in line with expectations for catheter placement, cell infusion, concomitant medications, age, and underlying diseases. CONCLUSIONS This study led to European clinical trial authorization for a phase II study in a homogeneous patient cohort, with repeated infusions and intermediate doses.
Collapse
|
14
|
Coppin LCF, Smets F, Ambroise J, Sokal EEM, Stéphenne X. Infusion-related thrombogenesis by liver-derived mesenchymal stem cells controlled by anticoagulant drugs in 11 patients with liver-based metabolic disorders. Stem Cell Res Ther 2020; 11:51. [PMID: 32028991 PMCID: PMC7006410 DOI: 10.1186/s13287-020-1572-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/17/2019] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation is a fast-developing therapy in regenerative medicine. However, some concerns have been raised regarding their safety and the infusion-related pro-coagulant activity. The aim of this study is to analyze the induced thrombogenic risk and the safety of adding anticoagulants during intraportal infusions of liver-derived MSCs (HepaStem), in patients with Crigler-Najjar (CN) and urea cycle disorders (UCD). METHODS Eleven patients (6 CN and 5 UCD patients) were included in this partially randomized phase 1/2 study. Three cell doses of HepaStem were investigated: low (12.5 × 106 cells/kg), intermediate (50 × 106 cells/kg), and high doses (200 × 106 cells/kg). A combination of anticoagulants, heparin (10 I.U./5 × 106cells), and bivalirudin (1.75 mg/kg/h) were added during cell infusions. The infusion-related thrombogenic risk and anticoagulation were evaluated by clinical monitoring, blood sampling (platelet and D-dimer levels, activated clotting time, etc.) and liver Doppler ultrasound. Mixed effects linear regression models were used to assess statistically significant differences. RESULTS One patient presented a thrombogenic event such as a partial portal vein thrombus after 6 infusions. Minor adverse effects such as petechiae, epistaxis, and cutaneous hemorrhage at the site of catheter placement were observed in four patients. A significant decrease in platelet and increase in D-dimer levels were observed at the end of the infusion cycle, normalizing spontaneously after 7 days. No significant and clinically relevant increase in portal vein pressure could be observed once the infusion cycle was completed. CONCLUSIONS The safety- and the infusion-related pro-coagulant activity remains a concern in MSC transplantation. In our study, a combination of heparin and bivalirudin was added to prevent the thrombogenic risk induced by HepaStem infusions in 11 patients. A significant decrease in platelet and increase in D-dimer levels were observed, suggesting the activation of coagulation in these patients; however, this was spontaneously reversible in time. We can conclude that adding this combination of anticoagulants is safe and limits infusion-related thrombogenesis to subclinical signs in most of the patients. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT01765283-January 10, 2013.
Collapse
Affiliation(s)
- Louise C F Coppin
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Av Hippocrate 10, B-1200, Brussels, Belgium.
| | - Françoise Smets
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Av Hippocrate 10, B-1200, Brussels, Belgium
| | - Jérome Ambroise
- Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Etienne E M Sokal
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Av Hippocrate 10, B-1200, Brussels, Belgium
| | - Xavier Stéphenne
- Service de Gastro-Entérologie et Hépatologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Av Hippocrate 10, B-1200, Brussels, Belgium
| |
Collapse
|
15
|
Tsuchida T, Murata S, Matsuki K, Mori A, Matsuo M, Mikami S, Okamoto S, Ueno Y, Tadokoro T, Zheng YW, Taniguchi H. The Regenerative Effect of Portal Vein Injection of Liver Organoids by Retrorsine/Partial Hepatectomy in Rats. Int J Mol Sci 2019; 21:178. [PMID: 31887985 PMCID: PMC6981799 DOI: 10.3390/ijms21010178] [Citation(s) in RCA: 27] [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: 10/29/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022] Open
Abstract
In this study, we reveal that liver organoid transplantation through the portal vein is a safe and effective method for the treatment of chronic liver damage. The liver organoids significantly reconstituted the hepatocytes; hence, the liver was significantly enlarged in this group, compared to the monolayer cell transplantation group in the retrorsine/partial hepatectomy (RS/PH) model. In the liver organoid transplantation group, the bile ducts were located in the donor area and connected to the recipient bile ducts. Thus, the rate of bile reconstruction in the liver was significantly higher compared to that in the monolayer group. By transplanting liver organoids, we saw a level of 70% replacement of the damaged liver. Consequently, in the transplantation group, diminished ductular reaction and a decrease of placental glutathione S-transferase (GST-p) precancerous lesions were observed. After trans-portal injection, the human induced pluripotent stem cell (hiPSC)-derived liver organoids revealed no translocation outside the liver; in contrast, the monolayer cells had spread to the lungs. The hiPSC-derived liver organoids were attached to the liver in the immunodeficient RS/PH rats. This study clearly demonstrates that liver organoid transplantation through the portal vein is a safe and effective method for the treatment of chronic liver damage in rats.
Collapse
Affiliation(s)
- Tomonori Tsuchida
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Koichiro Matsuki
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Akihiro Mori
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Megumi Matsuo
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Satoshi Mikami
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Yasuharu Ueno
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Tomomi Tadokoro
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
| | - Yun-Wen Zheng
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Department of Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.T.); (K.M.); (A.M.); (M.M.); (S.M.); (S.O.); (Y.U.); (T.T.); (Y.-W.Z.)
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
16
|
Coppin L, Sokal E, Stéphenne X. Thrombogenic Risk Induced by Intravascular Mesenchymal Stem Cell Therapy: Current Status and Future Perspectives. Cells 2019; 8:cells8101160. [PMID: 31569696 PMCID: PMC6829440 DOI: 10.3390/cells8101160] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are currently studied and used in numerous clinical trials. Nevertheless, some concerns have been raised regarding the safety of these infusions and the thrombogenic risk they induce. MSCs express procoagulant activity (PCA) linked to the expression of tissue factor (TF) that, when in contact with blood, initiates coagulation. Some even describe a dual activation of both the coagulation and the complement pathway, called Instant Blood-Mediated Inflammatory Reaction (IBMIR), explaining the disappointing results and low engraftment rates in clinical trials. However, nowadays, different approaches to modulate the PCA of MSCs and thus control the thrombogenic risk after cell infusion are being studied. This review summarizes both in vitro and in vivo studies on the PCA of MSC of various origins. It further emphasizes the crucial role of TF linked to the PCA of MSCs. Furthermore, optimization of MSC therapy protocols using different methods to control the PCA of MSCs are described.
Collapse
Affiliation(s)
- Louise Coppin
- Laboratoire d'Hépatologie Pédiatrique et Thérapie Cellulaire, Unité PEDI, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Etienne Sokal
- Laboratoire d'Hépatologie Pédiatrique et Thérapie Cellulaire, Unité PEDI, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| | - Xavier Stéphenne
- Laboratoire d'Hépatologie Pédiatrique et Thérapie Cellulaire, Unité PEDI, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| |
Collapse
|
17
|
Perlee D, de Vos AF, Scicluna BP, Maag A, Mancheño P, de la Rosa O, Dalemans W, Florquin S, Van't Veer C, Lombardo E, van der Poll T. Role of tissue factor in the procoagulant and antibacterial effects of human adipose-derived mesenchymal stem cells during pneumosepsis in mice. Stem Cell Res Ther 2019; 10:286. [PMID: 31547876 PMCID: PMC6757441 DOI: 10.1186/s13287-019-1391-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/03/2019] [Accepted: 08/19/2019] [Indexed: 02/06/2023] Open
Abstract
Background Adult mesenchymal stem cells (MSCs) improve the host response during experimental sepsis in animals. MSCs from various sources express a procoagulant activity that has been linked to the expression of tissue factor. This study sought to determine the role of tissue factor associated with adipose-derived MSCs (ASCs) in their procoagulant and antibacterial effects during pneumonia-derived sepsis. Methods Mice were infused intravenously with ASCs or vehicle after infection with the common human pathogen Klebsiella pneumoniae via the airways. Results Infusion of freshly cultured or cryopreserved ASCs induced the expression of many genes associated with tissue factor signaling and coagulation activation in the lungs. Freshly cultured and cryopreserved ASCs, as well as ASC lysates, exerted procoagulant activity in vitro as determined by a fibrin generation assay, which was almost completely inhibited by an anti-tissue factor antibody. Infusion of cryopreserved ASCs was associated with a rise in plasma thrombin-antithrombin complexes (indicative of coagulation activation) and formation of multiple thrombi in the lungs 4 h post-infusion. Preincubation of ASCs with anti-tissue factor antibody prior to infusion prevented the rise in plasma thrombin-antithrombin complex concentrations but did not influence thrombus formation in the lungs. ASCs reduced bacterial loads in the lungs and liver at 48 h after infection, which was not influenced by preincubation with anti-tissue factor antibody. At this late time point, microthrombi in the lungs were not detected anymore. Conclusion These data indicate that ASC-associated tissue factor is responsible for systemic activation of coagulation after infusion of ASCs but not for the formation of microthrombi in the lungs or antibacterial effects. Electronic supplementary material The online version of this article (10.1186/s13287-019-1391-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Desirée Perlee
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
| | - Alex F de Vos
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Brendon P Scicluna
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Anja Maag
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | - Sandrine Florquin
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis Van't Veer
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Tom van der Poll
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Room G2-130, 1105AZ, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Mesenchymal Stem Cells in the Adult Human Liver: Hype or Hope? Cells 2019; 8:cells8101127. [PMID: 31546729 PMCID: PMC6830330 DOI: 10.3390/cells8101127] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/18/2019] [Accepted: 09/21/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic liver diseases constitute a significant economic, social, and biomedical burden. Among commonly adopted approaches, only organ transplantation can radically help patients with end-stage liver pathologies. Cell therapy with hepatocytes as a treatment for chronic liver disease has demonstrated promising results. However, quality human hepatocytes are in short supply. Stem/progenitor cells capable of differentiating into functionally active hepatocytes provide an attractive alternative approach to cell therapy for liver diseases, as well as to liver-tissue engineering, drug screening, and basic research. The application of methods generally used to isolate mesenchymal stem cells (MSCs) and maintain them in culture to human liver tissue provides cells, designated here as liver MSCs. They have much in common with MSCs from other tissues, but differ in two aspects-expression of a range of hepatocyte-specific genes and, possibly, inherent commitment to hepatogenic differentiation. The aim of this review is to analyze data regarding liver MSCs, probably another type of liver stem/progenitor cells different from hepatic stellate cells or so-called hepatic progenitor cells. The review presents an analysis of the phenotypic characteristics of liver MSCs, their differentiation and therapeutic potential, methods for isolating these cells from human liver, and discusses issues of their origin and heterogeneity. Human liver MSCs are a fascinating object of fundamental research with a potential for important practical applications.
Collapse
|
19
|
Clinical Protocol to Prevent Thrombogenic Effect of Liver-Derived Mesenchymal Cells for Cell-Based Therapies. Cells 2019; 8:cells8080846. [PMID: 31394759 PMCID: PMC6721739 DOI: 10.3390/cells8080846] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/03/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022] Open
Abstract
The efficacy of mesenchymal stem cell infusion is currently tested in numerous clinical trials. However, therapy-induced thrombotic consequences have been reported in several patients. The aim of this study was to optimize protocols for heterologous human adult liver-derived progenitor cell (HHALPC) infusion, in order to eliminate acute thrombogenesis in liver-based metabolic or acute decompensated cirrhotic (ADC) patients. In rats, thrombotic effects were absent when HHALPCs were infused at low cell dose (5 × 106 cells/kg), or at high cell dose (5 × 107 cells/kg) when combined with anticoagulants. When HHALPCs were exposed to human blood in a whole blood perfusion assay, blocking of the tissue factor (TF) coagulation pathway suppressed fibrin generation and platelet activation. In a Chandler tubing loop model, HHALPCs induced less explosive activation of coagulation with blood from ADC patients, when compared to blood from healthy controls, without alterations in coagulation factor levels other than fibrinogen. These studies confirm a link between TF and thrombogenesis, when TF-expressing cells are exposed to human blood. This phenomenon however, could be controlled using either a low, or a high cell dose combined with anticoagulants. In clinical practice, this points to the suitability of a low HHALPC dose infusion to cirrhotic patients, provided that platelet and fibrinogen levels are monitored.
Collapse
|
20
|
Zannoni J, Mauz N, Seyve L, Meunier M, Pernet-Gallay K, Brault J, Jouzier C, Laurin D, Pezet M, Pernollet M, Cahn JY, Cognasse F, Polack B, Park S. Tumor microenvironment and clonal monocytes from chronic myelomonocytic leukemia induce a procoagulant climate. Blood Adv 2019; 3:1868-1880. [PMID: 31221660 PMCID: PMC6595258 DOI: 10.1182/bloodadvances.2018026955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/14/2019] [Indexed: 01/22/2023] Open
Abstract
Chronic myelomonocytic leukemia (CMML) is a myeloid hematological malignancy with overlapping features of myelodysplastic syndromes (MDSs) and myeloproliferative neoplasms (MPNs). The knowledge of the role of the tumor microenvironment (TME), particularly mesenchymal stromal cells (MSCs), in MDS pathogenesis is increasing. Generally, cancer is associated with a procoagulant state participating in tumor development. Monocytes release procoagulant, tissue factor (TF)-bearing microparticles. We hypothesized that MSCs and clonal monocytes release procoagulant extracellular vesicles (EVs) within the CMML TME, inducing a procoagulant state that could modify hematopoietic stem cell (HSC) homeostasis. We isolated and cultured MSCs and monocytes from CMML patients and MSCs from healthy donors (HDs). Their medium EVs and small EVs (sEVs) were collected after iterative ultracentrifugations and characterized by nanoparticle tracking analysis. Their impact on hemostasis was studied with a thrombin generation assay and fibrinography. CMML or HD HSCs were exposed to sEVs from either CMML or HD MSCs. CMML MSC sEVs increased HD HSC procoagulant activity, suggesting a transfer of TF from the CMML TME to HD HSCs. The presence of TF on sEVs was shown by electron microscopy and western blot. Moreover, CMML monocyte EVs conferred a procoagulant activity to HD MSCs, which was reversed by an anti-TF antibody, suggesting the presence of TF on the EVs. Our findings revealed a procoagulant "climate" within the CMML environment related to TF-bearing sEVs secreted by CMML MSCs and monocytes.
Collapse
Affiliation(s)
- Johanna Zannoni
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
| | - Natacha Mauz
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Landry Seyve
- Techniques de l'Ingénierie Médicale et de la Complexité Informatique, Mathématiques et Applications-Thérapeutique Recombinante Expérimentale, UMR 5525 Centre National de la Recherche Scientifique, Grenoble Alpes University, Grenoble, France
- Laboratory of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Mathieu Meunier
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Karin Pernet-Gallay
- Grenoble Institute for Neurosciences, INSERM U1216, Plateforme de Microscopie Electronique, Grenoble, France
| | - Julie Brault
- Techniques de l'Ingénierie Médicale et de la Complexité Informatique, Mathématiques et Applications-Thérapeutique Recombinante Expérimentale, UMR 5525 Centre National de la Recherche Scientifique, Grenoble Alpes University, Grenoble, France
- Centre de Diagnostic de la Granulomatose Septique Diagnosis and Research Center, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Claire Jouzier
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - David Laurin
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Etablissement Français du Sang Rhône-Alpes-Auvergne, Grenoble, France
| | - Mylène Pezet
- Plateforme de Microscopie Photonique, Cytométrie en Flux, Institute for Advanced Biosciences, Grenoble, France
| | - Martine Pernollet
- Institut de Biologie et de Pathologie, Laboratoire d'Immunologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Jean-Yves Cahn
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Fabrice Cognasse
- Etablissement Français du Sang Rhône-Alpes-Auvergne, Saint-Etienne, France; and
- GIMAP-EA3064, Lyon University, Saint-Etienne, France
| | - Benoît Polack
- Techniques de l'Ingénierie Médicale et de la Complexité Informatique, Mathématiques et Applications-Thérapeutique Recombinante Expérimentale, UMR 5525 Centre National de la Recherche Scientifique, Grenoble Alpes University, Grenoble, France
- Laboratory of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Sophie Park
- Institute for Advanced Biosciences, INSERM U1209 and Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5309, Grenoble Alpes University, Grenoble, France
- Department of Hematology, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| |
Collapse
|
21
|
Silachev DN, Goryunov KV, Shpilyuk MA, Beznoschenko OS, Morozova NY, Kraevaya EE, Popkov VA, Pevzner IB, Zorova LD, Evtushenko EA, Starodubtseva NL, Kononikhin AS, Bugrova AE, Evtushenko EG, Plotnikov EY, Zorov DB, Sukhikh GT. Effect of MSCs and MSC-Derived Extracellular Vesicles on Human Blood Coagulation. Cells 2019; 8:cells8030258. [PMID: 30893822 PMCID: PMC6468445 DOI: 10.3390/cells8030258] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/09/2019] [Accepted: 03/15/2019] [Indexed: 01/10/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a potent therapeutic tool for the treatment of a number of pathologies, including immune pathologies. However, unwelcome effects of MSCs on blood coagulation have been reported, motivating us to explore the thrombotic properties of human MSCs from the umbilical cord. We revealed strong procoagulant effects of MSCs on human blood and platelet-free plasma using rotational thromboelastometry and thrombodynamic tests. A similar potentiation of clotting was demonstrated for MSC-derived extracellular vesicles (EVs). To offer approaches to avoid unwanted effects, we studied the impact of a heparin supplement on MSC procoagulative properties. However, MSCs still retained procoagulant activity toward blood from children receiving a therapeutic dose of unfractionated heparin. An analysis of the mechanisms responsible for the procoagulant effect of MSCs/EVs revealed the presence of tissue factor and other proteins involved in coagulation-associated pathways. Also, we found that some MSCs and EVs were positive for annexin V, which implies the presence of phosphatidylserine on their surfaces, which can potentiate clot formation. Thus, we revealed procoagulant activity of MSCs/EVs associated with the presence of phosphatidylserine and tissue factor, which requires further analysis to avoid adverse effects of MSC therapy in patients with a risk of thrombosis.
Collapse
Affiliation(s)
- Denis N. Silachev
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Kirill V. Goryunov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Margarita A. Shpilyuk
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Olga S. Beznoschenko
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Natalya Y. Morozova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Elizaveta E. Kraevaya
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
| | - Vasily A. Popkov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Irina B. Pevzner
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Ljubava D. Zorova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | | | - Natalia L. Starodubtseva
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Moscow Institute of Physics and Technology, Moscow 141701, Russia
| | - Alexey S. Kononikhin
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Moscow Institute of Physics and Technology, Moscow 141701, Russia
| | - Anna E. Bugrova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | | | - Egor Y. Plotnikov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P. & D.B.Z.)
| | - Dmitry B. Zorov
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P. & D.B.Z.)
| | - Gennady T. Sukhikh
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (D.N.S.); (K.V.G.); (M.A.S.); (N.Y.M.); (E.E.K.); (V.A.P.); (I.B.P.); (L.D.Z.); (N.L.S.); (A.S.K.); (A.E.B.); (G.T.S.)
- Department of Obstetrics, Gynecology, Perinatology and Reproductology, Institute of Professional Education, First Moscow State Medical University Named after I.M. Sechenov, Moscow 119992, Russia
| |
Collapse
|
22
|
Moll G, Ankrum JA, Kamhieh-Milz J, Bieback K, Ringdén O, Volk HD, Geissler S, Reinke P. Intravascular Mesenchymal Stromal/Stem Cell Therapy Product Diversification: Time for New Clinical Guidelines. Trends Mol Med 2019; 25:149-163. [PMID: 30711482 DOI: 10.1016/j.molmed.2018.12.006] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/04/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022]
Abstract
Intravascular infusion is the most popular route for therapeutic multipotent mesenchymal stromal/stem cell (MSC) delivery in hundreds of clinical trials. Meta-analysis has demonstrated that bone marrow MSC infusion is safe. It is not clear if this also applies to diverse new cell products derived from other sources, such as adipose and perinatal tissues. Different MSC products display varying levels of highly procoagulant tissue factor (TF) and may adversely trigger the instant blood-mediated inflammatory reaction (IBMIR). Suitable strategies for assessing and controlling hemocompatibility and optimized cell delivery are crucial for the development of safer and more effective MSC therapies.
Collapse
Affiliation(s)
- Guido Moll
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany.
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Olle Ringdén
- Translational Cell Therapy Research (TCR), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Institute of Medical Immunology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Berlin Center for Advanced Therapies (BECAT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Equal contribution senior authorship
| | - Sven Geissler
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Julius Wolff Institute (JWI), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Equal contribution senior authorship
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Berlin Center for Advanced Therapies (BECAT), Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin (FUB), Humboldt-Universität zu Berlin (HUB), and Berlin Institute of Health (BIH), Berlin, Germany; Equal contribution senior authorship
| |
Collapse
|
23
|
Perlee D, van Vught LA, Scicluna BP, Maag A, Lutter R, Kemper EM, van ‘t Veer C, Punchard MA, González J, Richard MP, Dalemans W, Lombardo E, de Vos AF, van der Poll T. Intravenous Infusion of Human Adipose Mesenchymal Stem Cells Modifies the Host Response to Lipopolysaccharide in Humans: A Randomized, Single-Blind, Parallel Group, Placebo Controlled Trial. Stem Cells 2018; 36:1778-1788. [DOI: 10.1002/stem.2891] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/06/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Desiree Perlee
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Lonneke A. van Vught
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Brendon P. Scicluna
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Department of Clinical Epidemiology and Biostatistics, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Anja Maag
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - René Lutter
- Department of Experimental Immunology & Respiratory Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Elles M. Kemper
- Department of Pharmacy, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Cornelis van ‘t Veer
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | | | | | | | | | | | - Alex F. de Vos
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Tom van der Poll
- Center of Experimental & Molecular Medicine, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Division of Infectious Diseases, Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|
24
|
George MJ, Prabhakara K, Toledano-Furman NE, Wang YW, Gill BS, Wade CE, Olson SD, Cox CS. Clinical Cellular Therapeutics Accelerate Clot Formation. Stem Cells Transl Med 2018; 7:731-739. [PMID: 30070065 PMCID: PMC6186273 DOI: 10.1002/sctm.18-0015] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/06/2018] [Indexed: 02/04/2023] Open
Abstract
Clinical cellular therapeutics (CCTs) have shown preliminary efficacy in reducing inflammation after trauma, preserving cardiac function after myocardial infarction, and improving functional recovery after stroke. However, most clinically available cell lines express tissue factor (TF) which stimulates coagulation. We sought to define the degree of procoagulant activity of CCTs as related to TF expression. CCT samples from bone marrow, adipose, amniotic fluid, umbilical cord, multi-potent adult progenitor cell donors, and bone marrow mononuclear cells were tested. TF expression and phenotype were quantified using flow cytometry. Procoagulant activity of the CCTs was measured in vitro with thromboelastography and calibrated thrombogram. Fluorescence-activated cell sorting (FACS) separated samples into high- and low-TF expressing populations to isolate the contribution of TF to coagulation. A TF neutralizing antibody was incubated with samples to demonstrate loss of procoagulant function. All CCTs tested expressed procoagulant activity that correlated with expression of tissue factor. Time to clot and thrombin formation decreased with increasing TF expression. High-TF expressing cells decreased clotting time more than low-TF expressing cells when isolated from a single donor using FACS. A TF neutralizing antibody restored clotting time to control values in some, but not all, CCT samples. CCTs demonstrate wide variability in procoagulant activity related to TF expression. Time to clot and thrombin formation decreases as TF load increases and this procoagulant effect is neutralized by a TF blocking antibody. Clinical trials using CCTs are in progress and TF expression may emerge as a safety release criterion. Stem Cells Translational Medicine 2018;7:731-739.
Collapse
Affiliation(s)
- Mitchell J George
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA.,Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA
| | - Karthik Prabhakara
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA
| | - Naama E Toledano-Furman
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA
| | - Yao-Wei Wang
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA
| | - Brijesh S Gill
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA
| | - Charles E Wade
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
25
|
Netsch P, Elvers-Hornung S, Uhlig S, Klüter H, Huck V, Kirschhöfer F, Brenner-Weiß G, Janetzko K, Solz H, Wuchter P, Bugert P, Bieback K. Human mesenchymal stromal cells inhibit platelet activation and aggregation involving CD73-converted adenosine. Stem Cell Res Ther 2018; 9:184. [PMID: 29973267 PMCID: PMC6033237 DOI: 10.1186/s13287-018-0936-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/08/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023] Open
Abstract
Background Mesenchymal stromal cells (MSCs) are promising cell therapy candidates. Clinical application is considered safe. However, minor side effects have included thromboembolism and instant blood-mediated inflammatory reactions suggesting an effect of MSC infusion on hemostasis. Previous studies focusing on plasmatic coagulation as a secondary hemostasis step detected both procoagulatory and anticoagulatory activities of MSCs. We now focus on primary hemostasis and analyzed whether MSCs can promote or inhibit platelet activation. Methods Effects of MSCs and MSC supernatant on platelet activation and function were studied using flow cytometry and further platelet function analyses. MSCs from bone marrow (BM), lipoaspirate (LA) and cord blood (CB) were compared to human umbilical vein endothelial cells or HeLa tumor cells as inhibitory or activating cells, respectively. Results BM-MSCs and LA-MSCs inhibited activation and aggregation of stimulated platelets independent of the agonist used. This inhibitory effect was confirmed in diagnostic point-of-care platelet function analyses in platelet-rich plasma and whole blood. Using inhibitors of the CD39–CD73–adenosine axis, we showed that adenosine produced by CD73 ectonucleotidase activity was largely responsible for the LA-MSC and BM-MSC platelet inhibitory action. With CB-MSCs, batch-dependent responses were obvious, with some batches exerting inhibition and others lacking this effect. Conclusions Studies focusing on plasmatic coagulation suggested both procoagulatory and anticoagulatory activities of MSCs. We now show that MSCs can, dependent on their tissue origin, inhibit platelet activation involving adenosine converted from adenosine monophosphate by CD73 ectonucleotidase activity. These data may have strong implications for safety and risk/benefit assessment regarding MSCs from different tissue sources and may help to explain the tissue protective mode of action of MSCs. The adenosinergic pathway emerges as a key mechanism by which MSCs exert hemostatic and immunomodulatory functions. Electronic supplementary material The online version of this article (10.1186/s13287-018-0936-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- P Netsch
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Friedrich-Ebert Straße 107, 68167, Mannheim, Germany
| | - S Elvers-Hornung
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Friedrich-Ebert Straße 107, 68167, Mannheim, Germany
| | - S Uhlig
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Friedrich-Ebert Straße 107, 68167, Mannheim, Germany.,Flow Core Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - H Klüter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Friedrich-Ebert Straße 107, 68167, Mannheim, Germany
| | - V Huck
- Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - F Kirschhöfer
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - G Brenner-Weiß
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - K Janetzko
- Institute for Clinical Chemistry, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - H Solz
- Mannheim Clinic for Plastic Surgery, Mannheim, Germany
| | - P Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Friedrich-Ebert Straße 107, 68167, Mannheim, Germany
| | - P Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Friedrich-Ebert Straße 107, 68167, Mannheim, Germany
| | - K Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Friedrich-Ebert Straße 107, 68167, Mannheim, Germany.
| |
Collapse
|
26
|
Torres Crigna A, Daniele C, Gamez C, Medina Balbuena S, Pastene DO, Nardozi D, Brenna C, Yard B, Gretz N, Bieback K. Stem/Stromal Cells for Treatment of Kidney Injuries With Focus on Preclinical Models. Front Med (Lausanne) 2018; 5:179. [PMID: 29963554 PMCID: PMC6013716 DOI: 10.3389/fmed.2018.00179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022] Open
Abstract
Within the last years, the use of stem cells (embryonic, induced pluripotent stem cells, or hematopoietic stem cells), Progenitor cells (e.g., endothelial progenitor cells), and most intensely mesenchymal stromal cells (MSC) has emerged as a promising cell-based therapy for several diseases including nephropathy. For patients with end-stage renal disease (ESRD), dialysis or finally organ transplantation are the only therapeutic modalities available. Since ESRD is associated with a high healthcare expenditure, MSC therapy represents an innovative approach. In a variety of preclinical and clinical studies, MSC have shown to exert renoprotective properties, mediated mainly by paracrine effects, immunomodulation, regulation of inflammation, secretion of several trophic factors, and possibly differentiation to renal precursors. However, studies are highly diverse; thus, knowledge is still limited regarding the exact mode of action, source of MSC in comparison to other stem cell types, administration route and dose, tracking of cells and documentation of therapeutic efficacy by new imaging techniques and tissue visualization. The aim of this review is to provide a summary of published studies of stem cell therapy in acute and chronic kidney injury, diabetic nephropathy, polycystic kidney disease, and kidney transplantation. Preclinical studies with allogeneic or xenogeneic cell therapy were first addressed, followed by a summary of clinical trials carried out with autologous or allogeneic hMSC. Studies were analyzed with respect to source of cell type, mechanism of action etc.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Cristina Daniele
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Medical Faculty Mannheim, Orthopaedic and Trauma Surgery Centre (OUZ), Heidelberg University, Mannheim, Germany
| | - Sara Medina Balbuena
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Diego O. Pastene
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniela Nardozi
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Cinzia Brenna
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Benito Yard
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Karen Bieback
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
27
|
Abstract
The field of mesenchymal stromal cell (MSC) biology and clinical cellular therapy has grown exponentially over the last few decades. With discovery of multiple tissue specific sources of stromal cells, invariably being termed MSCs, and their increasing clinical application, there is a need to further delineate the true definition of a mesenchymal stromal cell and to recognise the inherit differences between cell sources; both their potential and limitations. In this review, we discuss the importance of considering every stromal cell source as an independent entity and the need to critically evaluate and appreciate the true phenotype of these cells and their safety when considering their use in novel cell therapies.
Collapse
Affiliation(s)
- Katarina Le Blanc
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Division of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Lindsay C Davies
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
Oeller M, Laner-Plamberger S, Hochmann S, Ketterl N, Feichtner M, Brachtl G, Hochreiter A, Scharler C, Bieler L, Romanelli P, Couillard-Despres S, Russe E, Schallmoser K, Strunk D. Selection of Tissue Factor-Deficient Cell Transplants as a Novel Strategy for Improving Hemocompatibility of Human Bone Marrow Stromal Cells. Am J Cancer Res 2018; 8:1421-1434. [PMID: 29507631 PMCID: PMC5835947 DOI: 10.7150/thno.21906] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 12/09/2017] [Indexed: 12/14/2022] Open
Abstract
Intravascular transplantation of tissue factor (TF)-bearing cells elicits an instant blood-mediated inflammatory reaction (IBMIR) resulting in thrombotic complications and reduced engraftment. Here we studied the hemocompatibility of commonly used human white adipose tissue (WAT), umbilical cord (UC) and bone marrow stromal cells (BMSC) and devised a possible strategy for safe and efficient stromal cell transplantation. Methods: Stromal cell identity, purity, and TF expression was tested by RTQ-PCR, flow cytometry and immunohistochemistry. Pro-coagulant activity and fibrin clot formation/stabilization was measured In Vitro by viscoelastic rotational plasma-thromboelastometry and in vivo by injecting sorted human stromal cells intravenously into rats. The impact of TF was verified in factor VII-deficient plasma and by sort-depleting TF/CD142+ BMSC. Results: We found significantly less TF expression by a subpopulation of BMSC corresponding to reduced pro-coagulant activity. UC and WAT stroma showed broad TF expression and durable clotting. Higher cell numbers significantly increased clot formation partially dependent on coagulation factor VII. Depleting the TF/CD142+ subpopulation significantly ameliorated BMSC's hemocompatibility without affecting immunomodulation. TF-deficient BMSC did not produce thromboembolism in vivo, comparing favorably to massive intravascular thrombosis induction by TF-expressing stromal cells. Conclusion: We demonstrate that plasma-based thromboelastometry provides a reliable tool to detect pro-coagulant activity of therapeutic cells. Selecting TF-deficient BMSC is a novel strategy for improving cell therapy applicability by reducing cell dose-dependent IBMIR risk. The particularly strong pro-coagulant activity of UC and WAT preparations sounds an additional note of caution regarding uncritical systemic application of stromal cells, particularly from non-hematopoietic extravascular sources.
Collapse
|
29
|
Sokal EM, Lombard CA, Roelants V, Najimi M, Varma S, Sargiacomo C, Ravau J, Mazza G, Jamar F, Versavau J, Jacobs V, Jacquemin M, Eeckhoudt S, Lambert C, Stéphenne X, Smets F, Hermans C. Biodistribution of Liver-Derived Mesenchymal Stem Cells After Peripheral Injection in a Hemophilia A Patient. Transplantation 2017; 101:1845-1851. [PMID: 28738402 DOI: 10.1097/tp.0000000000001773] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND With the exception of liver transplantation, there is no cure for hemophilia, which is currently managed by preemptive replacement therapy. Liver-derived stem cells are in clinical development for inborn and acquired liver diseases and could represent a curative treatment for hemophilia A. The liver is a major factor VIII (FVIII) synthesis site, and mesenchymal stem cells have been shown to control joint bleeding in animal models of hemophilia. Adult-derived human liver stem cells (ADHLSCs) have mesenchymal characteristics and have been shown able to engraft in and repopulate both animal and human livers. Thus, the objectives were to evaluate the potency of ADHLSCs to control bleeding in a hemophilia A patient and assess the biodistribution of the cells after intravenous injection. METHODS A patient suffering from hemophilia A was injected with repeated doses of ADHLSCs via a peripheral vein (35 million In-oxine-labeled cells, followed by 125 million cells the next day, and 3 infusions of 250 million cells every 2 weeks thereafter; total infusion period, 50 days). RESULTS After cell therapy, we found a temporary (15 weeks) decrease in the patient's FVIII requirements and severe bleeding complications, despite a lack of increase in circulating FVIII. The cells were safely administered to the patient via a peripheral vein. Biodistribution analysis revealed an initial temporary entrapment of the cells in the lungs, followed by homing to the liver and to a joint afflicted with hemarthrosis. CONCLUSION These results suggest the potential use of ADHLSCs in the treatment of hemophilia A.
Collapse
Affiliation(s)
- Etienne M Sokal
- 1 Université Catholique de Louvain, Cliniques Universitaires St Luc, Service de Gastroentérologie & Hépatologie Pédiatrique, Brussels, Belgium.2 Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Brussels, Belgium.3 Centre de Thérapie Cellulaire et Tissulaire, Cliniques Universitaires St Luc, Brussels, Belgium.4 Service de Médecine Nucléaire, Cliniques Universitaires St Luc, Brussels, Belgium.5 Division of Medicine, Institute for Liver and Digestive Health, Royal Free Hospital, University College of London, London, United Kingdom.6 Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Belgium.7 Service d'Hématologie, Cliniques Universitaires St Luc, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cryopreserved or Fresh Mesenchymal Stromal Cells: Only a Matter of Taste or Key to Unleash the Full Clinical Potential of MSC Therapy? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 951:77-98. [PMID: 27837556 DOI: 10.1007/978-3-319-45457-3_7] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) harbor great therapeutic potential for numerous diseases. From early clinical trials, success and failure analysis, bench-to-bedside and back-to-bench approaches, there has been a great gain in knowledge, still leaving a number of questions to be answered regarding optimal manufacturing and quality of MSCs for clinical application. For treatment of many acute indications, cryobanking may remain a prerequisite, but great uncertainty exists considering the therapeutic value of freshly thawed (thawed) and continuously cultured (fresh) MSCs. The field has seen an explosion of new literature lately, outlining the relevance of the topic. MSCs appear to have compromised immunomodulatory activity directly after thawing for clinical application. This may provide a possible explanation for failure of early clinical trials. It is not clear if and how quickly MSCs recover their full therapeutic activity, and if the "cryo stun effect" is relevant for clinical success. Here, we will share our latest insights into the relevance of these observations for clinical practice that will be discussed in the context of the published literature. We argue that the differences of fresh and thawed MSCs are limited but significant. A key issue in evaluating potency differences is the time point of analysis after thawing. To date, prospective double-blinded randomized clinical studies to evaluate potency of both products are lacking, although recent progress was made with preclinical assessment. We suggest refocusing therapeutic MSC development on potency and safety assays with close resemblance of the clinical reality.
Collapse
|
31
|
Wu Z, Zhang S, Zhou L, Cai J, Tan J, Gao X, Zeng Z, Li D. Thromboembolism Induced by Umbilical Cord Mesenchymal Stem Cell Infusion: A Report of Two Cases and Literature Review. Transplant Proc 2017; 49:1656-1658. [PMID: 28838459 DOI: 10.1016/j.transproceed.2017.03.078] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/16/2016] [Accepted: 03/15/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate the thromboembolism induced by blood-mediated inflammatory reactions against infused cells during the clinical application of stem cells. METHODS Two patients with renal transplantation and chronic kidney disease, respectively, experienced thromboembolism after umbilical cord mesenchymal stem cell (UCMSC) infusion. The clinical manifestations and the laboratory test results were collected and analyzed. RESULTS The patients received stem cell infusion through the peripheral veins and presented with a swollen and painful forearm postinfusion. Doppler ultrasound showed venous clots at the proximal end of the puncture site. Urokinase and warfarin were used for thrombolytic therapy. The swelling and pain were relieved and cured. CONCLUSION Safety concerns are still a primary hurdle for stem cell therapy, and thromboembolism as a critical complication should be prevented appropriately.
Collapse
Affiliation(s)
- Z Wu
- Department of Hepatobiliary Disease, Fuzhou General Hospital (Donfang Hospital), Xiamen University, Fuzhou, China
| | - S Zhang
- Department of Hepatobiliary Disease, Fuzhou General Hospital (Donfang Hospital), Xiamen University, Fuzhou, China
| | - L Zhou
- Department of Hepatobiliary Disease, Fuzhou General Hospital (Donfang Hospital), Xiamen University, Fuzhou, China
| | - J Cai
- Organ Transplant Institute, Fuzhou General Hospital (Dongfang Hospital), Xiamen University, Fuzhou, China
| | - J Tan
- Organ Transplant Institute, Fuzhou General Hospital (Dongfang Hospital), Xiamen University, Fuzhou, China
| | - X Gao
- Organ Transplant Institute, Fuzhou General Hospital (Dongfang Hospital), Xiamen University, Fuzhou, China
| | - Z Zeng
- Department of Hepatobiliary Disease, Fuzhou General Hospital (Donfang Hospital), Xiamen University, Fuzhou, China
| | - D Li
- Department of Hepatobiliary Disease, Fuzhou General Hospital (Donfang Hospital), Xiamen University, Fuzhou, China.
| |
Collapse
|
32
|
Abstract
BACKGROUND Allogeneic mesenchymal stem cells (MSCs) show great potential for the treatment of military and civilian trauma based on their reduced immunogenicity and ability to modulate inflammation and immune function in the recipient. Although generally considered to be safe, MSCs express tissue factor (TF), a potent activator of coagulation. In the current study, we evaluated multiple MSC populations for tissue factor expression and procoagulant activity to characterize safety considerations for systemic use of MSCs in trauma patients who may have altered coagulation homeostasis. METHODS Multiple MSC populations derived from either human adipose tissue or bone marrow were expanded in the recommended stem cell media. Stem cell identity was confirmed using a well-characterized panel of positive and negative markers. Tissue factor expression on the cell surface was evaluated by flow cytometry with anti-CD142 antibody. Effects on blood coagulation were determined by thromboelastography and calibrated automated thrombogram assays using platelet-poor plasma or whole blood. RESULTS Mesenchymal stem cells express tissue factor on their surfaces and are procoagulant in the presence of blood or plasma. The adipose-derived MSCs (Ad-MSC) evaluated were more procoagulant and expressed more tissue factor than bone marrow MSCs (BM-MSCs), which showed a greater variability in TF expression. Bone marrow MSCs were identified that exhibited low procoagulant activity, whereas all Ad-MSCs examined exhibited high procoagulant activity. The percentage of cells in a given population expressing surface tissue factor correlates roughly with functional procoagulant activity. Mesenchymal stem cell tissue factor expression and procoagulant activity change over time in culture. CONCLUSIONS All MSC populations are not equivalent; care should be taken to select cells for clinical use that minimize potential safety problems and maximize chance of patient benefit. Adipose-derived MSCs seem more consistently procoagulant than BM-MSCs, presenting a potential safety concern for systemic administration in coagulopathic patients. Donor variation exists between different cell populations, and culture handling conditions may also determine coagulation activity. Cells must be routinely monitored during preparation to ensure that they retain the desired characteristics before patient administration.
Collapse
|
33
|
Baygan A, Aronsson-Kurttila W, Moretti G, Tibert B, Dahllöf G, Klingspor L, Gustafsson B, Khoein B, Moll G, Hausmann C, Svahn BM, Westgren M, Remberger M, Sadeghi B, Ringden O. Safety and Side Effects of Using Placenta-Derived Decidual Stromal Cells for Graft-versus-Host Disease and Hemorrhagic Cystitis. Front Immunol 2017; 8:795. [PMID: 28744284 PMCID: PMC5504152 DOI: 10.3389/fimmu.2017.00795] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/22/2017] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are increasingly used in regenerate medicine. Placenta-derived decidual stromal cells (DSCs) are a novel therapy for acute graft-versus-host-disease (GVHD) and hemorrhagic cystitis (HC) after allogeneic hematopoietic stem cell transplantation (HSCT). DSCs are more immunosuppressive than MSCs. We assessed adverse events and safety using DSCs among 44 treated patients and 40 controls. The median dose of infused cells was 1.5 (range 0.9–2.9) × 106 DSCs/kg. The patients were given 2 (1–5) doses, with a total of 82 infusions. Monitoring ended 3 months after the last DSC infusion. Three patients had transient reactions during DSC infusion. Laboratory values, hemorrhages, and transfusions were similar in the two groups. The frequency of leukemic relapse (2/2, DSC/controls) and invasive fungal infections (6/6) were the same in the two groups. Causes of death were those seen in HSCT patients: infections (5/3), respiratory failure (1/1), circulatory failure (3/1), thromboembolism (1/0), multiorgan failure (0/1), and GVHD and others (2/7). One-year survival for the DSC patients with GVHD was 67%, which was significantly better than achieved previously at our center. One-year survival was 90% in the DSC-treated HC group. DSC infusions appear safe. Randomized studies are required to prove efficacy.
Collapse
Affiliation(s)
- Arjang Baygan
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Wictor Aronsson-Kurttila
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Gianluca Moretti
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Babylonia Tibert
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Göran Dahllöf
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Klingspor
- Department of Microbiology, Uppsala University Hospital, Uppsala, Sweden
| | - Britt Gustafsson
- Department of Pediatrics, Uppsala University Hospital, Uppsala, Sweden
| | - Bita Khoein
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Guido Moll
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden.,Charité Universitätsmedizin, Berlin, Germany
| | - Charlotta Hausmann
- Center for Allogeneic Stem Cell Transplantation, Department of Pathology/Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Britt-Marie Svahn
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Westgren
- Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Department of Pathology/Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Behnam Sadeghi
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| | - Olle Ringden
- Translational Cell Therapy Research Group (TCR), Division of Therapeutic Immunology, Department of LabMed, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Grudzenski S, Baier S, Ebert A, Pullens P, Lemke A, Bieback K, Dijkhuizen RM, Schad LR, Alonso A, Hennerici MG, Fatar M. The effect of adipose tissue-derived stem cells in a middle cerebral artery occlusion stroke model depends on their engraftment rate. Stem Cell Res Ther 2017; 8:96. [PMID: 28446216 PMCID: PMC5407025 DOI: 10.1186/s13287-017-0545-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/25/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In the field of experimental stem cell therapy, intra-arterial (IA) delivery yields the best results concerning, for example, migrated cell number at the targeted site. However, IA application also appears to be associated with increased mortality rates and infarction. Since many rodent studies systemically apply 1 × 106 cells, this could also be a consequence of engrafted cell number. The aim of this study was therefore to investigate the effect of different doses of adipose tissue-derived stem cells (ASCs) on engraftment rates and stroke outcome measured in vivo using 9.4-T high-field magnetic resonance imaging (MRI). METHODS Male Wistar rats (n = 43) underwent a middle cerebral artery occlusion (MCAo) for 45 or 90 min, followed by IA delivery of either saline or 1 × 106, 3 × 105, or 5 × 104 ASCs pre-labelled with very small superparamagnetic iron oxide particles (VSOPs). MRI (9.4-T) analysis was performed 48 h and 9 days post-MCAo. Lesion volumes were assessed by analysis of T2-weighted images and cell signal tracking showing cell engraftment and active cell migration by an improved T2*-analysis. RESULTS The ASC-derived signal intensity increased in the affected hemisphere 48 h post MCAo with injected cell number (p < 0.05). The analysis of stroke volumes revealed an increased infarction after injection of 1 × 106 ASCs compared to controls or application of 5 × 104 ASCs (p < 0.05). At 9 days post-MCAo, injection of 3 × 105 ASCs resulted in reduced infarct volumes (p < 0.05). Correspondingly, MRI analysis revealed no changes in cell numbers between both MRI examinations but showed active ASC migration to the site of infarction. CONCLUSION Our results confirm that IA injection is an efficient way of targeting damaged brain tissue but its usefulness strongly depends on the right dose of delivered stem cells since this factor has a strong influence on migration rate and infarct volume, with better results for doses below 1 × 106 cells. Future challenges will include the determination of therapeutic doses for best cellular engraftment and stroke outcome.
Collapse
Affiliation(s)
- Saskia Grudzenski
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167, Mannheim, Germany.
| | - Sebastian Baier
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Anne Ebert
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167, Mannheim, Germany
| | - Pim Pullens
- Department of Radiology, UZ-Brussel, Vrije Universiteit (VUB), 1090, Brussels, Belgium
| | - Andreas Lemke
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Lothar R Schad
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Angelika Alonso
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167, Mannheim, Germany
| | - Michael G Hennerici
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167, Mannheim, Germany
| | - Marc Fatar
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167, Mannheim, Germany
| |
Collapse
|
35
|
Liao L, Shi B, Chang H, Su X, Zhang L, Bi C, Shuai Y, Du X, Deng Z, Jin Y. Heparin improves BMSC cell therapy: Anticoagulant treatment by heparin improves the safety and therapeutic effect of bone marrow-derived mesenchymal stem cell cytotherapy. Theranostics 2017; 7:106-116. [PMID: 28042320 PMCID: PMC5196889 DOI: 10.7150/thno.16911] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Systemic infusion of bone marrow-derived mesenchymal stem cells (BMSCs) has become a promising strategy for disease treatment and tissue regeneration. Strategies to enhance the efficiency of BMSC cell therapy are crucial to promote its clinical application. Here, we aimed to improve BMSC cell therapy by inhibiting the BMSC-induced coagulation reaction. Intravenous injection of gradient BMSCs into mice showed that BMSCs were not fully compatible with blood. Large doses of BMSCs induced a series of symptoms of respiratory failure and heart failure. Histological and homeostasis analysis confirmed that large doses of BMSCs induced disseminated intravascular thrombosis, exhaustion of platelets and coagulation factors, and prolonged prothrombin time (PT) and activated partial thromboplastin time (APTT). Similar to mouse BMSCs, goat and human BMSCs also induced coagulation reactions in vitro and in vivo. The coagulation was induced mostly by tissue factor, the overexpression of which enhanced the procoagulant activity of BMSCs during in vitro culture. Notably, clinical doses of BMSCs in cell therapy also induced mild and reversible coagulation, which increased BMSC lung embolism and clearance. Anticoagulation treatment by heparin (400 U/kg) prevented BMSC-induced coagulation and the acute adverse effects of large-dose BMSCs infusion efficiently. Importantly, heparin treatment led to decreased BMSC lung embolism and enhanced migration and maintenance of BMSCs to target organs in cell therapy. Based on an experimental colitis model, we confirmed that heparin treatment enhanced the effect of BMSC therapy efficiently to reduce mortality, prevent weight loss, suppress inflammation reaction and alleviate tissue injury. In conclusion, BMSCs possess procoagulant activity that could induce disseminated coagulation and thrombosis in recipients. Anticoagulation treatment by heparin is a practical strategy to improve both the safety and therapeutic effect of BMSC therapy.
Collapse
|
36
|
Comprehensive Screening of Cell Surface Markers Expressed by Adult-Derived Human Liver Stem/Progenitor Cells Harvested at Passage 5: Potential Implications for Engraftment. Stem Cells Int 2016; 2016:9302537. [PMID: 27956903 PMCID: PMC5124472 DOI: 10.1155/2016/9302537] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/13/2016] [Accepted: 10/17/2016] [Indexed: 01/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are known to have potential therapeutic benefits for a number of diseases. However, many studies report low engraftment levels, regardless of the target organ. One possible explanation could be that MSCs do not express the necessary receptors for engraftment. Indeed, MSCs appear to use a similar mechanism to leukocytes to engraft into injured organs, relying on various receptors for rolling, firm adhesion, and transmigration. In this study, we conducted an extensive surface molecule screening of adult-derived human liver stem/progenitor cells (ADHLSC) in an attempt to shed some light on this subject. We observed that ADHLSCs lack expression of most of the costimulatory molecules tested. Furthermore, study of the adhesion molecule profile of ADHLSCs revealed that they do not express selectin ligands or LFA-1 which are, respectively, involved in the rolling process and the firm adhesion. In addition, ADHLSCs slightly express VLA-4 and lose expression of CXCR4 altogether on their surface during culture expansion. However, ADHLSCs express all the integrin couples and matrix metalloproteinases needed to bind and integrate the extracellular matrix once the endothelial barrier is crossed. Collectively, these results suggest that binding to the endothelium may be the critical weak point in the engraftment process.
Collapse
|
37
|
Herrero A, Prigent J, Lombard C, Rosseels V, Daujat-Chavanieu M, Breckpot K, Najimi M, Deblandre G, Sokal EM. Adult-Derived Human Liver Stem/Progenitor Cells Infused 3 Days Postsurgery Improve Liver Regeneration in a Mouse Model of Extended Hepatectomy. Cell Transplant 2016; 26:351-364. [PMID: 27657746 DOI: 10.3727/096368916x692960] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There is growing evidence that cell therapy constitutes a promising strategy for liver regenerative medicine. In the setting of hepatic cancer treatments, cell therapy could prove a useful therapeutic approach for managing the acute liver failure that occurs following extended hepatectomy. In this study, we examined the influence of delivering adult-derived human liver stem/progenitor cells (ADHLSCs) at two different early time points in an immunodeficient mouse model (Rag2-/-IL2Rγ-/-) that had undergone a 70% hepatectomy procedure. The hepatic mesenchymal cells were intrasplenically infused either immediately after surgery (n = 26) or following a critical 3-day period (n = 26). We evaluated the cells' capacity to engraft at day 1 and day 7 following transplantation by means of human Alu qPCR quantification, along with histological assessment of human albumin and α-smooth muscle actin. In addition, cell proliferation (anti-mouse and human Ki-67 staining) and murine liver weight were measured in order to evaluate liver regeneration. At day 1 posttransplantation, the ratio of human to mouse cells was similar in both groups, whereas 1 week posttransplantation this ratio was significantly improved (p < 0.016) in mice receiving ADHLSC injection at day 3 posthepatectomy (1.7%), compared to those injected at the time of surgery (1%). On the basis of liver weight, mouse liver regeneration was more extensive 1 week posttransplantation in mice transplanted with ADHLSCs (+65.3%) compared to that of mice from the sham vehicle group (+42.7%). In conclusion, infusing ADHLSCs 3 days after extensive hepatectomy improves the cell engraftment and murine hepatic tissue regeneration, thereby confirming that ADHLSCs could be a promising cell source for liver cell therapy and hepatic tissue repair.
Collapse
|
38
|
Herzig MC, Cap AP. Challenges in translating mesenchymal stem cell therapies for trauma and critical care. Transfusion 2016; 56:20S-5S. [PMID: 27079318 DOI: 10.1111/trf.13566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Maryanne C Herzig
- Coagulation and Blood Research Program, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, Texas
| | - Andrew P Cap
- Coagulation and Blood Research Program, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, Texas
| |
Collapse
|
39
|
Najimi M, Defresne F, Sokal EM. Concise Review: Updated Advances and Current Challenges in Cell Therapy for Inborn Liver Metabolic Defects. Stem Cells Transl Med 2016; 5:1117-25. [PMID: 27245366 DOI: 10.5966/sctm.2015-0260] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/14/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED : The development of liver cell transplantation (LCT), considered a major biotechnological breakthrough, was intended to provide more accessible treatments for liver disease patients. By preserving the native recipient liver and decreasing hospitalization time, this innovative approach has progressively gained interest among clinicians. LCT initially targets inborn errors of liver metabolism, enabling the compensation of deficient metabolic functions for up to 18 months post-transplantation, supporting its use at least as a bridge to transplantation. The rigorous clinical development and widespread use of LCT depends strongly on controlled and consistent clinical trial data, which may help improve several critical factors, including the standardization of raw biological material and immunosuppression regimens. Substantial effort has also been made in defining and optimizing the most efficient cell population to be transplanted in the liver setting. Although isolated hepatocytes remain the best cell type, showing positive clinical results, their widespread use is hampered by their poor resistance to both cryopreservation and in vitro culture, as well as ever-more-significant donor shortages. Hence, there is considerable interest in developing more standardized and widely accessible cell medicinal products to improve engraftment permanency and post-cell transplantation metabolic effects. SIGNIFICANCE In this therapeutic approach to liver disease, new solutions are being designed and evaluated to bypass the documented limitations and move forward toward wide clinical use. Future developments also require a deep knowledge of regulatory framework to launch specific clinical trials that will allow clear assessment of cell therapy and help patients with significant unmet medical needs.
Collapse
Affiliation(s)
- Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain and Cliniques Universitaires St Luc, Brussels, Belgium
| | - Florence Defresne
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain and Cliniques Universitaires St Luc, Brussels, Belgium
| | - Etienne M Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain and Cliniques Universitaires St Luc, Brussels, Belgium
| |
Collapse
|
40
|
Lee CA, Dhawan A, Smith RA, Mitry RR, Fitzpatrick E. Instant Blood-Mediated Inflammatory Reaction in Hepatocyte Transplantation: Current Status and Future Perspectives. Cell Transplant 2016; 25:1227-36. [PMID: 26996786 DOI: 10.3727/096368916x691286] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hepatocyte transplantation (HT) is emerging as a promising alternative to orthotopic liver transplantation (OLT) in patients with certain liver-based metabolic disease and acute liver failure. Hepatocytes are generally infused into the portal venous system, from which they migrate into the liver cell plates of the native organ. One of the major hurdles to the sustained success of this therapy is early cell loss, with up to 70% of hepatocytes lost immediately following infusion. This is largely thought to be due to the instant blood-mediated inflammatory reaction (IBMIR), resulting in the activation of complement and coagulation pathways. Transplanted hepatocytes produce and release tissue factor (TF), which activates the coagulation pathway, leading to the formation of thrombin and fibrin clots. Thrombin can further activate a number of complement proteins, leading to the activation of the membrane attack complex (MAC) and subsequent hepatocyte cell death. Inflammatory cells including granulocytes, monocytes, Kupffer cells, and natural killer (NK) cells have been shown to cluster around transplanted hepatocytes, leading to their rapid clearance shortly after transplantation. Current research aims to improve cell engraftment and prevent early cell loss. This has been proven successful in vitro using pharmacological interventions such as melagatran, low-molecular-weight dextran sulphate, and N-acetylcysteine (NAC). Effective inhibition of IBMIR would significantly improve hepatocyte engraftment, proliferation, and function, providing successful treatment for patients with liver-based metabolic diseases.
Collapse
Affiliation(s)
- Charlotte A Lee
- Institute of Liver Studies, King's College London, School of Life Sciences and Medicine, King's College Hospital, London, UK
| | | | | | | | | |
Collapse
|
41
|
Moll G, Alm JJ, Davies LC, von Bahr L, Heldring N, Stenbeck-Funke L, Hamad OA, Hinsch R, Ignatowicz L, Locke M, Lönnies H, Lambris JD, Teramura Y, Nilsson-Ekdahl K, Nilsson B, Le Blanc K. Do cryopreserved mesenchymal stromal cells display impaired immunomodulatory and therapeutic properties? Stem Cells 2015; 32:2430-42. [PMID: 24805247 DOI: 10.1002/stem.1729] [Citation(s) in RCA: 276] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 04/01/2014] [Accepted: 04/09/2014] [Indexed: 12/17/2022]
Abstract
We have recently reported that therapeutic mesenchymal stromal cells (MSCs) have low engraftment and trigger the instant blood mediated inflammatory reaction (IBMIR) after systemic delivery to patients, resulting in compromised cell function. In order to optimize the product, we compared the immunomodulatory, blood regulatory, and therapeutic properties of freeze-thawed and freshly harvested cells. We found that freeze-thawed MSCs, as opposed to cells harvested from continuous cultures, have impaired immunomodulatory and blood regulatory properties. Freeze-thawed MSCs demonstrated reduced responsiveness to proinflammatory stimuli, an impaired production of anti-inflammatory mediators, increased triggering of the IBMIR, and a strong activation of the complement cascade compared to fresh cells. This resulted in twice the efficiency in lysis of thawed MSCs after 1 hour of serum exposure. We found a 50% and 80% reduction in viable cells with freshly detached as opposed to thawed in vitro cells, indicating a small benefit for fresh cells. In evaluation of clinical response, we report a trend that fresh cells, and cells of low passage, demonstrate improved clinical outcome. Patients treated with freshly harvested cells in low passage had a 100% response rate, twice the response rate of 50% observed in a comparable group of patients treated with freeze-thawed cells at higher passage. We conclude that cryobanked MSCs have reduced immunomodulatory and blood regulatory properties directly after thawing, resulting in faster complement-mediated elimination after blood exposure. These changes seem to be paired by differences in therapeutic efficacy in treatment of immune ailments after hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Guido Moll
- Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Hematology and Regenerative Medicine Centre at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bayon Y, Vertès AA, Ronfard V, Culme-Seymour E, Mason C, Stroemer P, Najimi M, Sokal E, Wilson C, Barone J, Aras R, Chiesi A. Turning Regenerative Medicine Breakthrough Ideas and Innovations into Commercial Products. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:560-71. [PMID: 26179129 DOI: 10.1089/ten.teb.2015.0068] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The TERMIS-Europe (EU) Industry committee intended to address the two main critical issues in the clinical/commercial translation of Advanced Therapeutic Medicine Products (ATMP): (1) entrepreneurial exploitation of breakthrough ideas and innovations, and (2) regulatory market approval. Since January 2012, more than 12,000 publications related to regenerative medicine and tissue engineering have been accepted for publications, reflecting the intense academic research activity in this field. The TERMIS-EU 2014 Industry Symposium provided a reflection on the management of innovation and technological breakthroughs in biotechnology first proposed to contextualize the key development milestones and constraints of allocation of financial resources, in the development life-cycle of radical innovation projects. This was illustrated with the biofuels story, sharing similarities with regenerative medicine. The transition was then ensured by an overview of the key identified challenges facing the commercialization of cell therapy products as ATMP examples. Real cases and testimonies were then provided by a palette of medical technologies and regenerative medicine companies from their commercial development of cell and gene therapy products. Although the commercial development of ATMP is still at the proof-of-concept stage due to technology risks, changing policies, changing markets, and management changes, the sector is highly dynamic with a number of explored therapeutic approaches, developed by using a large diversity of business models, both proposed by the experience, pitfalls, and successes of regenerative medicine pioneers, and adapted to the constraint resource allocation and environment in radical innovation projects.
Collapse
Affiliation(s)
- Yves Bayon
- 1 Medtronic-Sofradim Production , Trévoux, France
| | - Alain A Vertès
- 2 London Business School , London, United Kingdom .,3 NxR Biotechnologies GmbH , Basel, Switzerland
| | - Vincent Ronfard
- 4 College of Pharmacy UNT Health Science Center , Fort Worth, Texas
| | | | - Chris Mason
- 6 Advanced Centre for Biochemical Engineering, University College London , London, United Kingdom
| | | | - Mustapha Najimi
- 8 Institute of Experimental and Clinical Research , Université catholique de Louvain and Cliniques St-Luc, Brussels, Belgium .,9 PROMETHERA Biosciences, Mont-St.-Guibert, Belgium
| | - Etienne Sokal
- 8 Institute of Experimental and Clinical Research , Université catholique de Louvain and Cliniques St-Luc, Brussels, Belgium .,9 PROMETHERA Biosciences, Mont-St.-Guibert, Belgium
| | | | - Joe Barone
- 11 Juventas Therapeutics, Inc. , Cleveland, Ohio
| | - Rahul Aras
- 11 Juventas Therapeutics, Inc. , Cleveland, Ohio
| | | |
Collapse
|
43
|
Moll G, Ignatowicz L, Catar R, Luecht C, Sadeghi B, Hamad O, Jungebluth P, Dragun D, Schmidtchen A, Ringdén O. Different Procoagulant Activity of Therapeutic Mesenchymal Stromal Cells Derived from Bone Marrow and Placental Decidua. Stem Cells Dev 2015; 24:2269-79. [PMID: 26192403 DOI: 10.1089/scd.2015.0120] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
While therapeutic mesenchymal stromal/stem cells (MSCs) have usually been obtained from bone marrow, perinatal tissues have emerged as promising new sources of cells for stromal cell therapy. In this study, we present a first safety follow-up on our clinical experience with placenta-derived decidual stromal cells (DSCs), used as supportive immunomodulatory and regenerative therapy for patients with severe complications after allogeneic hematopoietic stem cell transplantation (HSCT). We found that DSCs are smaller, almost half the volume of MSCs, which may favor microvascular passage. DSCs also show different hemocompatibility, with increased triggering of the clotting cascade after exposure to human blood and plasma in vitro. After infusion of DSCs in HSCT patients, we observed a weak activation of the fibrinolytic system, but the other blood activation markers remained stable, excluding major adverse events. Expression profiling identified differential levels of key factors implicated in regulation of hemostasis, such as a lack of prostacyclin synthase and increased tissue factor expression in DSCs, suggesting that these cells have intrinsic blood-activating properties. The stronger triggering of the clotting cascade by DSCs could be antagonized by optimizing the cell graft reconstitution before infusion, for example, by use of low-dose heparin anticoagulant in the cell infusion buffer. We conclude that DSCs are smaller and have stronger hemostatic properties than MSCs, thus triggering stronger activation of the clotting system, which can be antagonized by optimizing the cell graft preparation before infusion. Our results highlight the importance of hemocompatibility safety testing for every novel cell therapy product before clinical use, when applied using systemic delivery.
Collapse
Affiliation(s)
- Guido Moll
- 1 Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden .,2 Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universtätsmedizin Berlin , Berlin, Germany .,3 Department of Nephrology and Intensive Care Medicine, Charité Universtätsmedizin Berlin , Berlin, Germany
| | - Lech Ignatowicz
- 4 Department of Clinical Sciences, Lund University , Lund, Sweden
| | - Rusan Catar
- 3 Department of Nephrology and Intensive Care Medicine, Charité Universtätsmedizin Berlin , Berlin, Germany
| | - Christian Luecht
- 3 Department of Nephrology and Intensive Care Medicine, Charité Universtätsmedizin Berlin , Berlin, Germany
| | - Behnam Sadeghi
- 1 Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden .,5 Center for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge , Stockholm, Sweden
| | - Osama Hamad
- 6 Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University , Stockholm, Sweden
| | - Philipp Jungebluth
- 7 Advanced Center for Translational Regenerative Medicine (ACTREM), Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet , Stockholm, Sweden .,8 Department of Thoracic Surgery, Thoraxklinik, Heidelberg University , Heidelberg, Germany
| | - Duska Dragun
- 2 Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universtätsmedizin Berlin , Berlin, Germany .,3 Department of Nephrology and Intensive Care Medicine, Charité Universtätsmedizin Berlin , Berlin, Germany
| | | | - Olle Ringdén
- 1 Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet , Stockholm, Sweden .,5 Center for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge , Stockholm, Sweden
| |
Collapse
|
44
|
Gleeson BM, Martin K, Ali MT, Kumar AHS, Pillai MGK, Kumar SPG, O'Sullivan JF, Whelan D, Stocca A, Khider W, Barry FP, O'Brien T, Caplice NM. Bone Marrow-Derived Mesenchymal Stem Cells Have Innate Procoagulant Activity and Cause Microvascular Obstruction Following Intracoronary Delivery: Amelioration by Antithrombin Therapy. Stem Cells 2015; 33:2726-37. [PMID: 25969127 DOI: 10.1002/stem.2050] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 04/02/2015] [Accepted: 04/07/2015] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) are currently under investigation as tools to preserve cardiac structure and function following acute myocardial infarction (AMI). However, concerns have emerged regarding safety of acute intracoronary (IC) MSC delivery. This study aimed to characterize innate prothrombotic activity of MSC and identify means of its mitigation toward safe and efficacious therapeutic IC MSC delivery post-AMI. Expression of the initiator of the coagulation cascade tissue factor (TF) on MSC was detected and quantified by immunofluorescence, FACS, and immunoblotting. MSC-derived TF antigen was catalytically active and capable of supporting thrombin generation in vitro. Addition of MSCs to whole citrated blood enhanced platelet thrombus deposition on collagen at arterial shear, an effect abolished by heparin coadministration. In a porcine AMI model, IC infusion of 25 × 10(6) MSC during reperfusion was associated with a decrease in coronary flow reserve but not when coadministered with an antithrombin agent (heparin). Heparin reduced MSC-associated thrombosis incorporating platelets and VWF within the microvasculature. Heparin-assisted therapeutic MSC delivery also reduced apoptosis in the infarct border zone at 24 hours, significantly improved infarct size, left ventricular (LV) ejection fraction, LV volumes, wall motion, and attenuated histologic evidence of scar formation at 6 weeks post-AMI. Heparin alone or heparin-assisted fibroblast control cell delivery had no such effect. Procoagulant TF activity of therapeutic MSCs is associated with reductions in myocardial perfusion when delivered IC may be successfully managed by heparin coadministration. This study highlights an important mechanistic insight into safety concerns associated with therapeutic IC MSC delivery for AMI.
Collapse
Affiliation(s)
- Birgitta M Gleeson
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Kenneth Martin
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Mohammed T Ali
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Arun H S Kumar
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - M Gopala-Krishnan Pillai
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Sujith P G Kumar
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - John F O'Sullivan
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Derek Whelan
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Alessia Stocca
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Wisam Khider
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Frank P Barry
- Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Noel M Caplice
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
45
|
Moll G, Le Blanc K. Engineering more efficient multipotent mesenchymal stromal (stem) cells for systemic delivery as cellular therapy. ACTA ACUST UNITED AC 2015. [DOI: 10.1111/voxs.12133] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- G. Moll
- Division of Clinical Immunology and Transfusion Medicine; Department of Laboratory Medicine; Karolinska Institutet; Stockholm Sweden
- Hematology and Regenerative Medicine Centre at Karolinska University Hospital Huddinge; Stockholm Sweden
| | - K. Le Blanc
- Division of Clinical Immunology and Transfusion Medicine; Department of Laboratory Medicine; Karolinska Institutet; Stockholm Sweden
- Hematology and Regenerative Medicine Centre at Karolinska University Hospital Huddinge; Stockholm Sweden
| |
Collapse
|
46
|
Kopec AK, Luyendyk JP. Coagulation in liver toxicity and disease: role of hepatocyte tissue factor. Thromb Res 2014; 133 Suppl 1:S57-9. [PMID: 24759146 DOI: 10.1016/j.thromres.2014.03.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The liver is the primary source of a number of circulating coagulation factors, and acute liver injury and chronic liver disease are each associated with alterations in blood coagulation. Current views of the connection between liver injury and coagulation extend beyond the impact of liver disease on synthesis of coagulation factors to include a role for coagulation factor activity in the initiation and progression of liver disease. Mechanisms of coagulation initiation in liver disease are not completely understood. Compared to other tissues, liver expresses very low levels of tissue factor (TF). Recent studies indicate that expression of TF by hepatocytes comprises the majority of liver procoagulant activity, and that hepatocyte TF activates coagulation induced by liver injury. This review will briefly cover the expression and regulation of TF by hepatocytes, the role of TF in coagulation triggered by liver toxicity, and the contribution of coagulation activity to the progression of liver disease.
Collapse
Affiliation(s)
- Anna K Kopec
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824
| | - James P Luyendyk
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824.
| |
Collapse
|
47
|
Rackham CL, Dhadda PK, Le Lay AM, King AJF, Jones PM. Preculturing Islets With Adipose-Derived Mesenchymal Stromal Cells Is an Effective Strategy for Improving Transplantation Efficiency at the Clinically Preferred Intraportal Site. CELL MEDICINE 2014; 7:37-47. [PMID: 26858891 DOI: 10.3727/215517914x680047] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have recently shown that preculturing islets with kidney-derived mesenchymal stromal cells (MSCs) improves transplantation outcome in streptozotocin-diabetic mice implanted with a minimal mass of islets beneath the kidney capsule. In the present study, we have extended our previous observations to investigate whether preculturing islets with MSCs can also be used to enhance islet function at the clinically used intraportal site. We have used MSCs derived from adipose tissue, which are more readily accessible than alternative sources in human subjects and can be expanded to clinically efficacious numbers, to preculture islets throughout this study. The in vivo efficacy of grafts consisting of islets precultured alone or with MSCs was tested using a syngeneic streptozotocin-diabetic minimal islet mass model at the clinically relevant intraportal site. Blood glucose concentrations were monitored for 1 month. The vascularization of islets precultured alone or with MSCs was investigated both in vitro and in vivo, using immunohistochemistry. Islet insulin content was measured by radioimmunoassay. The effect of preculturing islets with MSCs on islet function in vitro was investigated using static incubation assays. There was no beneficial angiogenic influence of MSC preculture, as demonstrated by the comparable vascularization of islets precultured alone or with MSCs, both in vitro after 3 days and in vivo 1 month after islet transplantation. However, the in vitro insulin secretory capacity of MSC precultured islets was superior to that of islets precultured alone. In vivo, this was associated with improved glycemia at 7, 14, 21, and 28 days posttransplantation, in recipients of MSC precultured islets compared to islets precultured alone. The area of individual islets within the graft-bearing liver was significantly higher in recipients of MSC precultured islets compared to islets precultured alone. Our experimental studies suggest that preculturing islets with MSCs represents a favorable strategy for improving the efficiency of clinical islet transplantation.
Collapse
Affiliation(s)
- Chloe L Rackham
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Paramjeet K Dhadda
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Aurélie M Le Lay
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Aileen J F King
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Peter M Jones
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London , London , UK
| |
Collapse
|
48
|
Liver engraftment and repopulation by in vitro expanded adult derived human liver stem cells in a child with ornithine carbamoyltransferase deficiency. JIMD Rep 2013; 13:65-72. [PMID: 24142276 DOI: 10.1007/8904_2013_257] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 07/28/2013] [Accepted: 08/01/2013] [Indexed: 12/13/2022] Open
Abstract
A 3-year-old girl suffering from ornithine carbamoyltransferase (OTC) deficiency was poorly equilibrated under conventional diet and scavenger treatment. Following unsuccessful cryopreserved hepatocyte transplantation, she received two infusions of Adult Derived Human Liver Stem/Progenitor Cells (ADHLSCs) expanded in vitro under GMP settings, the quantity being equivalent to 0.75% of her calculated liver mass. Using FISH immunostaining for the Y chromosome, the initial biopsy did not detect any male nuclei in the recipient liver. Two liver biopsies taken 100 days after ADHLSC transplantation showed 3% and 5% of male donor cells in the recipient liver, thus suggesting repopulation by donor cells. Although limited follow-up did not allow us to draw conclusions on long-term improvement, these results provide a promising proof of concept that this therapy is feasible in an OTC patient.
Collapse
|
49
|
The effect of nonuniform magnetic targeting of intracoronary-delivering mesenchymal stem cells on coronary embolisation. Biomaterials 2013; 34:9905-16. [PMID: 24055521 DOI: 10.1016/j.biomaterials.2013.08.092] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 08/31/2013] [Indexed: 01/10/2023]
Abstract
Magnetic targeting has been recently introduced to enhance cell retention in animals with acute myocardial infarction. However, it is unclear whether the magnetic accumulation of intravascular cells increases the risk of coronary embolism. Upon finite element analysis, we found that the permanent magnetic field was nonuniform, manifestated as attenuation along the vertical axis and polarisation along the horizontal axis. In the in vitro experiments, iron-labelled mesenchymal stem cells (MSCs) were accumulated in layers predominantly at the edge of the magnet. In an ischaemic rat model subjected to intracavitary MSCs injection, magnetic targeting induced unfavourable vascular embolisation and an inhomogeneous distribution of the donor cells, which prevented the enhanced cell retention from translating into additional functional benefit. These potential complications of magnetic targeting should be thoroughly investigated and overcome before clinical application.
Collapse
|
50
|
Abstract
Because of their high proliferative capacity, resistance to cryopreservation, and ability to differentiate into hepatocyte-like cells, stem and progenitor cells have recently emerged as attractive cell sources for liver cell therapy, a technique used as an alternative to orthotopic liver transplantation in the treatment of various hepatic ailments ranging from metabolic disorders to end-stage liver disease. Although stem and progenitor cells have been isolated from various tissues, obtaining them from the liver could be an advantage for the treatment of hepatic disorders. However, the techniques available to isolate these stem/progenitor cells are numerous and give rise to cell populations with different morphological and functional characteristics. In addition, there is currently no established consensus on the tests that need to be performed to ensure the quality and safety of these cells when used clinically. The purpose of this review is to describe the different types of liver stem/progenitor cells currently reported in the literature, discuss their suitability and limitations in terms of clinical applications, and examine how the culture and transplantation techniques can potentially be improved to achieve a better clinical outcome.
Collapse
Affiliation(s)
- Catherine A. Lombard
- Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Institut de Recherche Expérimentale et Clinique, Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Julie Prigent
- Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Institut de Recherche Expérimentale et Clinique, Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| | - Etienne M. Sokal
- Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Institut de Recherche Expérimentale et Clinique, Pediatric Hepatology and Cell Therapy, Brussels, Belgium
| |
Collapse
|