1
|
Chen J, Shi X, Deng Y, Dang J, Liu Y, Zhao J, Liang R, Zeng D, Wu W, Xiong Y, Yuan J, Chen Y, Wang J, Lin W, Chen X, Huang W, Olsen N, Pan Y, Fu Q, Zheng SG. miRNA-148a-containing GMSC-derived EVs modulate Treg/Th17 balance via IKKB/NF-κB pathway and treat a rheumatoid arthritis model. JCI Insight 2024; 9:e177841. [PMID: 38652539 PMCID: PMC11141912 DOI: 10.1172/jci.insight.177841] [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: 11/22/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have demonstrated potent immunomodulatory properties that have shown promise in the treatment of autoimmune diseases, including rheumatoid arthritis (RA). However, the inherent heterogeneity of MSCs triggered conflicting therapeutic outcomes, raising safety concerns and limiting their clinical application. This study aimed to investigate the potential of extracellular vesicles derived from human gingival mesenchymal stem cells (GMSC-EVs) as a therapeutic strategy for RA. Through in vivo experiments using an experimental RA model, our results demonstrate that GMSC-EVs selectively homed to inflamed joints and recovered Treg and Th17 cell balance, resulting in the reduction of arthritis progression. Our investigations also uncovered miR-148a-3p as a critical contributor to the Treg/Th17 balance modulation via IKKB/NF-κB signaling orchestrated by GMSC-EVs, which was subsequently validated in a model of human xenograft versus host disease (xGvHD). Furthermore, we successfully developed a humanized animal model by utilizing synovial fibroblasts obtained from patients with RA (RASFs). We found that GMSC-EVs impeded the invasiveness of RASFs and minimized cartilage destruction, indicating their potential therapeutic efficacy in the context of patients with RA. Overall, the unique characteristics - including reduced immunogenicity, simplified administration, and inherent ability to target inflamed tissues - position GMSC-EVs as a viable alternative for RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Jingrong Chen
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Shi
- Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanan Deng
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junlong Dang
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Liu
- Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Zhao
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongzhen Liang
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | - Yiding Xiong
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Yuan
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ye Chen
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Julie Wang
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weidong Lin
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangfang Chen
- Department of Endocrinology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Nancy Olsen
- Division of Rheumatology, Department of Medicine, The Penn State University Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Yunfeng Pan
- Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qingling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Wang Y, Lv Y, Li Y, Bao H, Yu C, Li X, Xu J, Huang J, Zhang Z. Ferromagnetic Vortex Iron Oxide Nanorings Modified with Integrin β1 Antibody for Targeted MRI Tracking of Human Mesenchymal Stem Cells. J Biomed Nanotechnol 2022; 18:1044-1051. [PMID: 35854460 DOI: 10.1166/jbn.2022.3319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Mesenchymal stem cells (MSCs) have demonstrated great potential for tissue engineering and regenerative medicine applications. Noninvasive and real-term tracking of transplanted MSCs in vivo is crucial for studying the distribution and migration of MSCs, and their role in tissue injury repair. This study reports on the use of ferrimagnetic vortex iron oxide (FVIO) nanorings modified with anti-human integrin β1 for specific recognition and magnetic resonance imaging (MRI) tracking of human MSCs (hMSCs). Integrin β1 is highly expressed at all stem cell proliferation and differentiation stages. Therefore, the anti-integrin β1 antibody (Ab) introduced in FVIO targets integrin β1, thus enabling FVIO to target stem cells at any stage. This is unlike the traditional MRI-based monitoring of transplanted stem cells, which usually requires pre-labeling the stem cells with tracers before injection. Because of the ability to recognize hMSCs, the Ab-modified FVIO nanotracers (FVIO-Ab) have the advantage of not requiring pre-labeling before stem cell transplantation. Furthermore, the FVIO-Ab nanotracers have high T*₂ contrast resulting from the unique magnetic properties of FVIO which can improve the MRI tracking efficiency of stem cells. This work may provide a new way for stem cell labeling and in vivo MRI tracking, thus reducing the risks associated with stem cell transplantation and promoting clinical translation.
Collapse
Affiliation(s)
- Yujie Wang
- New Energy and Sensing Technology Lab, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| | - Yinjuan Lv
- New Energy and Sensing Technology Lab, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| | - Yuxuan Li
- Chinese Academy of Sciences Key Laboratory of Nano-Bio Interface, Division of Nano Biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Hongying Bao
- Chinese Academy of Sciences Key Laboratory of Nano-Bio Interface, Division of Nano Biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Chenggong Yu
- Chinese Academy of Sciences Key Laboratory of Nano-Bio Interface, Division of Nano Biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xiaodi Li
- Chinese Academy of Sciences Key Laboratory of Nano-Bio Interface, Division of Nano Biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jiaqiang Xu
- New Energy and Sensing Technology Lab, Department of Chemistry, College of Science, Shanghai University, Shanghai, 200444, China
| | - Jie Huang
- Chinese Academy of Sciences Key Laboratory of Nano-Bio Interface, Division of Nano Biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Zhijun Zhang
- Chinese Academy of Sciences Key Laboratory of Nano-Bio Interface, Division of Nano Biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| |
Collapse
|
3
|
Wang Y, Yu C, Li Y, Bao H, Li X, Fan H, Huang J, Zhang Z. In vivo MRI tracking and therapeutic efficacy of transplanted mesenchymal stem cells labeled with ferrimagnetic vortex iron oxide nanorings for liver fibrosis repair. NANOSCALE 2022; 14:5227-5238. [PMID: 35315848 DOI: 10.1039/d1nr08544a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mesenchymal stem cells (MSCs) have showed promising effects in the treatment of liver fibrosis. Long-term and noninvasive in vivo tracking of transplanted MSCs is essential for understanding the therapeutic mechanism of MSCs during the therapy of liver fibrosis. In this study, we report the development of a ferrimagnetic vortex iron oxide nanoring (FVIO)-based nanotracer for the long-term visualization of transplanted human MSCs (hMSCs) by magnetic resonance imaging (MRI). The FVIOs were prepared by a hydrothermal reaction followed by hydrogen reduction. To endow the FVIOs with biocompatibility, polyethylene glycol amine (mPEG-NH2) was covalently coupled on the surface of FVIOs, forming FVIO@PEG nanotracers with high contrast enhancement and intracellular uptake. The hMSCs labeled with FVIO@PEG nanotracers exhibited enhanced MRI contrast than those labeled with a commercial contrast agent, and could be continuously monitored by MRI in liver fibrosis mice for 28 days after transplantation, clearly clarifying the migration behavior of hMSCs in vivo. Moreover, we explored the therapeutic mechanism of the FVIO@PEG labeled hMSCs in the amelioration of liver fibrosis, including the reduction in inflammation and oxidative stress, the inhibition of hepatic fibrosis-caused histopathological damage, as well as the down-regulation of the expression of relevant cytokines. The results obtained in this work may deepen our understanding of the behavior and role of hMSCs in the treatment of liver fibrosis, which is key to the clinical application of stem cells in the therapy of liver diseases.
Collapse
Affiliation(s)
- Yujie Wang
- College of Science, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Chenggong Yu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Yuxuan Li
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Hongying Bao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Xiaodi Li
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Haiming Fan
- College of Chemistry and Materials Science, Northwest University, 1 Xue Fu Avenue, Xi'an, 710127, China.
| | - Jie Huang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| | - Zhijun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| |
Collapse
|
4
|
James S, Neuhaus K, Murphy M, Leahy M. Contrast agents for photoacoustic imaging: a review of stem cell tracking. Stem Cell Res Ther 2021; 12:511. [PMID: 34563237 PMCID: PMC8467005 DOI: 10.1186/s13287-021-02576-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022] Open
Abstract
With the advent of stem cell therapy for spinal cord injuries, stroke, burns, macular degeneration, heart diseases, diabetes, rheumatoid arthritis and osteoarthritis; the need to track the survival, migration pathways, spatial destination and differentiation of transplanted stem cells in a clinical setting has gained increased relevance. Indeed, getting regulatory approval to use these therapies in the clinic depends on biodistribution studies. Although optoacoustic imaging (OAI) or photoacoustic imaging can detect functional information of cell activities in real-time, the selection and application of suitable contrast agents is essential to achieve optimal sensitivity and contrast for sensing at clinically relevant depths and can even provide information about molecular activity. This review explores OAI methodologies in conjunction with the specific application of exogenous contrast agents in comparison to other imaging modalities and describes the properties of exogenous contrast agents for quantitative and qualitative monitoring of stem cells. Specific characteristics such as biocompatibility, the absorption coefficient, and surface functionalization are compared and how the labelling efficiency translates to both short and long-term visualization of mesenchymal stem cells is explored. An overview of novel properties of recently developed optoacoustic contrast agents and their capability to detect disease and recovery progression in clinical settings is provided which includes newly developed exogenous contrast agents to monitor stem cells in real-time for multimodal sensing.
Collapse
Affiliation(s)
- Soorya James
- Tissue Optics and Microcirculation Imaging facility,School of Physics, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Kai Neuhaus
- Tissue Optics and Microcirculation Imaging facility,School of Physics, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Mary Murphy
- The Regenerative Medicine Institute, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Martin Leahy
- Tissue Optics and Microcirculation Imaging facility,School of Physics, National University of Ireland, Galway, University Road, Galway, Ireland
- ICFO-Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Spain
| |
Collapse
|
5
|
Cseresnyes Z, Hassan MIA, Dahse HM, Voigt K, Figge MT. Quantitative Impact of Cell Membrane Fluorescence Labeling on Phagocytosis Measurements in Confrontation Assays. Front Microbiol 2020; 11:1193. [PMID: 32582113 PMCID: PMC7289966 DOI: 10.3389/fmicb.2020.01193] [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: 03/17/2020] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Phagocytosis is series of steps where the pathogens and the immune cells interact during an invasion. This starts with the adhesion process between the host and pathogen cells, and is followed by the engulfment of the pathogens. Many analytical methods that are applied to characterize phagocytosis based on imaging the host-pathogen confrontation assays rely on the fluorescence labeling of cells. However, the potential effect of the membrane labeling on the quantitative results of the confrontation assays has not been studied in detail. In this study, we determine whether the fluorescence labeling processes themselves influence the results of the phagocytosis measurements. Here, alveolar macrophages, which form one of the most important compartments of the innate immune system, were used as an example of host cells, whereas Aspergillus fumigatus and Lichtheimia corymbifera that cause aspergillosis and mucormycosis, respectively, were studied as examples for pathogens. At first, our study investigated the importance of the sequence of steps of the fixation process when preparing the confrontation assay sample for microscopy studies. Here we showed that applying the fixation agent before the counter-staining causes miscalculations during the determination of the phagocytic measures. Furthermore, we also found that staining the macrophages with various concentrations of DID, as a typical membrane label, in most cases altered the capability of macrophages to phagocytose FITC-stained A. fumigatus and L. corymbifera spores in comparison with unlabeled macrophages. This effect of the DID staining showed a differential character dependent upon the labeling status and the specific type of pathogen. Moreover, labeling the spores of A. fumigatus and L. corymbifera with FITC increased the phagocytic measures during confrontation with unlabeled macrophages when compared to label-free spores. Overall, our study confirms that the staining process itself may significantly manipulate the quantitative outcome of the confrontation assay. As a result of our study, we also developed a user-friendly image analysis tool that analyses confrontation assays both with and without fluorescence labeling of the host cells and of the pathogens. Our image analysis algorithm saves experimental work effort and time, provides more precise results when calculating the phagocytic measures, and delivers a convenient analysis tool for the biologists to monitor host-pathogen interactions as they happen without the artifacts that fluorescence labeling imposes on biological interactions.
Collapse
Affiliation(s)
- Zoltan Cseresnyes
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Mohamed I. Abdelwahab Hassan
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
- Department of Pests and Plant Protection, National Research Centre, Giza, Egypt
| | - Hans-Martin Dahse
- Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
6
|
Zhao J, Chen J, Huang F, Wang J, Su W, Zhou J, Qi Q, Cao F, Sun B, Liu Z, Bellanti JA, Zheng S. Human gingiva tissue-derived MSC ameliorates immune-mediated bone marrow failure of aplastic anemia via suppression of Th1 and Th17 cells and enhancement of CD4+Foxp3+ regulatory T cells differentiation. Am J Transl Res 2019; 11:7627-7643. [PMID: 31934306 PMCID: PMC6943455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023]
Abstract
Accumulating evidence has revealed that human gingiva-derived mesenchymal stem cells (GMSCs) are emerging as a new line of mesenchymal stem cells and may have the potential to control or even treat autoimmune diseases through maintaining the balance between Th and Treg cells. Given that GMSCs have a robust immune regulatory function and regenerative ability, we investigated the effect of GMSCs on preventing T cell-mediated bone marrow failure (BMF) in a mouse model. We observed that GMSCs markedly improved mice survival and attenuated histological bone marrow (BM) damage. Moreover, we found GMSCs significantly reduced cell infiltration of CD8+ cells, Th1 and Th17 cells, whereas increased CD4+Foxp3+ regulatory T cells (Tregs) differentiation in lymph nodes. GMSCs also suppressed the levels of TNF-α, IFN-γ, IL-17A and IL-6, but IL-10 was increased in serum. The live in vivo imaging identified that GMSCs can home into inflammatory location on BM. Our results demonstrate that GMSCs attenuate T cell-mediated BMF through regulating the balance of Th1, Th17 and Tregs, implicating that application of GMSCs may provide a promising approach in prevention and treatment of patients with aplastic anemia.
Collapse
Affiliation(s)
- Jianzhi Zhao
- Division of Hematology, Shaoxing Central HospitalShaoxing, China
- Division of Rheumatology, Penn State University College of MedicineHershey, USA
| | - Jingrong Chen
- Department of Clinical Immunology in Third Affiliated Hospital of The Sun Yat-sen UniversityGuangzhou, China
| | - Feng Huang
- Department of Clinical Immunology in Third Affiliated Hospital of The Sun Yat-sen UniversityGuangzhou, China
| | - Julie Wang
- Division of Rheumatology and Immunology, Department of Internal Medicine at The Ohio State University College of MedicineColumbus, OH, USA
| | - Wenru Su
- Department of Clinical Immunology in Third Affiliated Hospital of The Sun Yat-sen UniversityGuangzhou, China
| | - Jianyao Zhou
- Division of Hematology, Shaoxing Central HospitalShaoxing, China
| | - Quanyin Qi
- State Key Lab at Guiling Medical CollegeGuiling, China
| | - Fenglin Cao
- Department of Internal Medicine in The First Affiliated Hospital at The Harbin Medical UniversityHarbin, China
| | - Baoqing Sun
- Department of Allergy and Clinical Immunology, The First Affiliated Hospital at The Guangzhou Medical UniversityGuangzhou, China
| | - Zhongmin Liu
- Center of Stem Cell, Shanghai East Hospital at The Tongji UniversityShanghai, China
| | - Joseph A Bellanti
- Department of Pediatrics and Microbiology-Immunology, Georgetown University Medical CenterWashington, DC, USA
| | - Songguo Zheng
- Division of Rheumatology and Immunology, Department of Internal Medicine at The Ohio State University College of MedicineColumbus, OH, USA
| |
Collapse
|
7
|
Shi Y, Wang Y, Li Q, Liu K, Hou J, Shao C, Wang Y. Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases. Nat Rev Nephrol 2019; 14:493-507. [PMID: 29895977 DOI: 10.1038/s41581-018-0023-5] [Citation(s) in RCA: 783] [Impact Index Per Article: 130.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs; also referred to as mesenchymal stromal cells) have attracted much attention for their ability to regulate inflammatory processes. Their therapeutic potential is currently being investigated in various degenerative and inflammatory disorders such as Crohn's disease, graft-versus-host disease, diabetic nephropathy and organ fibrosis. The mechanisms by which MSCs exert their therapeutic effects are multifaceted, but in general, these cells are thought to enable damaged tissues to form a balanced inflammatory and regenerative microenvironment in the presence of vigorous inflammation. Studies over the past few years have demonstrated that when exposed to an inflammatory environment, MSCs can orchestrate local and systemic innate and adaptive immune responses through the release of various mediators, including immunosuppressive molecules, growth factors, exosomes, chemokines, complement components and various metabolites. Interestingly, even nonviable MSCs can exert beneficial effects, with apoptotic MSCs showing immunosuppressive functions in vivo. Because the immunomodulatory capabilities of MSCs are not constitutive but rather are licensed by inflammatory cytokines, the net outcomes of MSC activation might vary depending on the levels and the types of inflammation within the residing tissues. Here, we review current understanding of the immunomodulatory mechanisms of MSCs and the issues related to their therapeutic applications.
Collapse
Affiliation(s)
- Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China. .,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Yu Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Keli Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianquan Hou
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
8
|
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
|
9
|
Sarmah D, Kaur H, Saraf J, Pravalika K, Goswami A, Kalia K, Borah A, Wang X, Dave KR, Yavagal DR, Bhattacharya P. Getting Closer to an Effective Intervention of Ischemic Stroke: The Big Promise of Stem Cell. Transl Stroke Res 2017; 9:356-374. [PMID: 29075984 DOI: 10.1007/s12975-017-0580-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/12/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022]
Abstract
Stem cell therapy for ischemic stroke has widely been explored. Results from both preclinical and clinical studies have immensely supported the judicious use of stem cells as therapy. These provide an attractive means for preserving and replacing the damaged brain tissues following an ischemic attack. Since the past few years, researchers have used various types of stem cells to replenish insulted neuronal and glial cells in neurological disorders. In the present review, we discuss different types of stem cells employed for the treatment of ischemic stroke and mechanisms and challenges these cells face once introduced into the living system. Further, we also present different ways to maneuver and overcome challenges to translate the advances made at the preclinical level to clinics.
Collapse
Affiliation(s)
- Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Jackson Saraf
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Kanta Pravalika
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Avirag Goswami
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dileep R Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India.
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Effect of DiD Carbocyanine Dye Labeling on Immunoregulatory Function and Differentiation of Mice Mesenchymal Stem Cells. Stem Cells Int 2014; 2014:457614. [PMID: 25580134 PMCID: PMC4279147 DOI: 10.1155/2014/457614] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 11/18/2014] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been used to treat a variety of degenerative disorders. Labeling of MSCs with an appropriate tracer is vital to demonstrate the in vivo engraftment and differentiation of transplanted MSCs. DiD is a lipophilic fluorescent dye with near infrared emission spectra that makes it suitable for in vivo tracing. Therefore, in the present study the consequences of DiD labeling on induction of oxidative stress and apoptosis as well as inhibition of biological functions of mesenchymal stem cells (MSCs) were investigated. DiD labeling did not provoke the production of ROS, induction of apoptosis, or inhibition of production of immunosuppressive factors (PGE2 and IL-10) by MSCs. In addition, there were no statistical differences between DiD-labeled and unlabeled MSCs in suppression of proliferation and cytokine production (IFN-γ and IL-17) by in vitro stimulated splenocytes or improvement of clinical score in EAE after in vivo administration. In addition, DiD labeling did not alter the differentiation capacity of MSCs. Taken together, DiD can be considered as a safe dye for in vivo tracking of MSCs.
Collapse
|
11
|
Williams LB, Tessier L, Koenig JB, Koch TG. Post-thaw non-cultured and post-thaw cultured equine cord blood mesenchymal stromal cells equally suppress lymphocyte proliferation in vitro. PLoS One 2014; 9:e113615. [PMID: 25438145 PMCID: PMC4249887 DOI: 10.1371/journal.pone.0113615] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 10/29/2014] [Indexed: 01/27/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSC) are receiving increased attention for their non-progenitor immunomodulatory potential. Cryopreservation is commonly used for long-term storage of MSC. Post-thaw MSC proliferation is associated with a lag-phase in vitro. How this lag-phase affect MSC immunomodulatory properties is unknown. We hypothesized that in vitro there is no difference in lymphocyte suppression potential between quick-thawed cryopreserved equine cord blood (CB) MSC immediately included in mixed lymphocyte reaction (MLR) and same MSC allowed post-thaw culture time prior to inclusion in MLR. Cryopreserved CB-MSC from five unrelated foals were compared using two-way MLR. For each of the five unrelated MSC cultures, paired MLR assays of MSC allowed five days of post-thaw culture and MSC included in MLR assay immediately post-thawing were evaluated. We report no difference in the suppression of lymphocyte proliferation by CB-MSC that had undergone post-thaw culture and MSC not cultured post-thaw (p<0.0001). Also, there was no inter-donor variability between the lymphocyte suppressive properties of MSC harvested from the five different donors (p = 0.13). These findings suggest that cryopreserved CB-MSC may have clinical utility immediately upon thawing. One implication hereof is the possibility of using cryopreserved CB-MSC at third party locations without the need for cell culture equipment or competencies.
Collapse
Affiliation(s)
- Lynn B. Williams
- Department of Clinical Studies, University of Guelph, Guelph, Ontario, Canada
| | - Laurence Tessier
- Department of Biomedical Science, University of Guelph, Guelph, Ontario, Canada
| | - Judith B. Koenig
- Department of Clinical Studies, University of Guelph, Guelph, Ontario, Canada
| | - Thomas G. Koch
- Department of Biomedical Science, University of Guelph, Guelph, Ontario, Canada
- Department of Clinical Studies, Orthopaedic Research Lab, Aarhus University, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
12
|
Roche ET, Hastings CL, Lewin SA, Shvartsman D, Brudno Y, Vasilyev NV, O'Brien FJ, Walsh CJ, Duffy GP, Mooney DJ. Comparison of biomaterial delivery vehicles for improving acute retention of stem cells in the infarcted heart. Biomaterials 2014; 35:6850-6858. [PMID: 24862441 DOI: 10.1016/j.biomaterials.2014.04.114] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/27/2014] [Indexed: 12/22/2022]
Abstract
Cell delivery to the infarcted heart has emerged as a promising therapy, but is limited by very low acute retention and engraftment of cells. The objective of this study was to compare a panel of biomaterials to evaluate if acute retention can be improved with a biomaterial carrier. Cells were quantified post-implantation in a rat myocardial infarct model in five groups (n = 7-8); saline injection (current clinical standard), two injectable hydrogels (alginate, chitosan/β-glycerophosphate (chitosan/ß-GP)) and two epicardial patches (alginate, collagen). Human mesenchymal stem cells (hMSCs) were delivered to the infarct border zone with each biomaterial. At 24 h, retained cells were quantified by fluorescence. All biomaterials produced superior fluorescence to saline control, with approximately 8- and 14-fold increases with alginate and chitosan/β-GP injectables, and 47 and 59-fold increases achieved with collagen and alginate patches, respectively. Immunohistochemical analysis qualitatively confirmed these findings. All four biomaterials retained 50-60% of cells that were present immediately following transplantation, compared to 10% for the saline control. In conclusion, all four biomaterials were demonstrated to more efficiently deliver and retain cells when compared to a saline control. Biomaterial-based delivery approaches show promise for future development of efficient in vivo delivery techniques.
Collapse
Affiliation(s)
- Ellen T Roche
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford street, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, 60 Oxford street, Cambridge, MA 02138, USA
| | - Conn L Hastings
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin 2, Ireland
| | - Sarah A Lewin
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford street, Cambridge, MA 02138, USA
| | - Dmitry Shvartsman
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford street, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, 60 Oxford street, Cambridge, MA 02138, USA
| | - Yevgeny Brudno
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford street, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, 60 Oxford street, Cambridge, MA 02138, USA
| | | | - Fergal J O'Brien
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin 2, Ireland
| | - Conor J Walsh
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford street, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, 60 Oxford street, Cambridge, MA 02138, USA
| | - Garry P Duffy
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin 2, Ireland
| | - David J Mooney
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford street, Cambridge, MA 02138, USA.,Wyss Institute for Biologically Inspired Engineering, 60 Oxford street, Cambridge, MA 02138, USA
| |
Collapse
|
13
|
Grange C, Tapparo M, Bruno S, Chatterjee D, Quesenberry PJ, Tetta C, Camussi G. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int J Mol Med 2014; 33:1055-63. [PMID: 24573178 PMCID: PMC4020482 DOI: 10.3892/ijmm.2014.1663] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/17/2014] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) contribute to the recovery of tissue injury, providing a paracrine support. Cell-derived extracellular vesicles (EVs), carrying membrane and cytoplasmatic constituents of the cell of origin, have been described as a fundamental mechanism of intercellular communication. We previously demonstrated that EVs derived from human MSCs accelerated recovery following acute kidney injury (AKI) in vivo. The aim of the present study was to investigate the biodistribution and the renal localization of EVs in AKI. For this purpose, two methods for EV labeling suitable for in vivo tracking with optical imaging (OI), were employed using near infrared (NIR) dye (DiD): i) labeled EVs were generated by MSCs pre-incubated with NIR dye and collected from cell supernatants; ii) purified EVs were directly labeled with NIR dye. EVs obtained with these two procedures were injected intravenously (i.v.) into mice with glycerol-induced AKI and into healthy mice to compare the efficacy of the two labeling methods for in vivo detection of EVs at the site of damage. We found that the labeled EVs accumulated specifically in the kidneys of the mice with AKI compared with the healthy controls. After 5 h, the EVs were detectable in whole body images and in dissected kidneys by OI with both types of labeling procedures. The directly labeled EVs showed a higher and brighter fluorescence compared with the labeled EVs produced by cells. The signal generated by the directly labeled EVs was maintained in time, but provided a higher background than that of the labeled EVs produced by cells. The comparison of the two methods indicated that the latter displayed a greater specificity for the injured kidney.
Collapse
Affiliation(s)
- Cristina Grange
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Marta Tapparo
- Translational Center for Regenerative Medicine, University of Torino, Torino, Italy
| | - Stefania Bruno
- Department of Molecular Biotechnology and Health Science, University of Torino, Torino, Italy
| | - Devasis Chatterjee
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Peter J Quesenberry
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ciro Tetta
- EMEA LA Medical Board, Fresenius Medical Care, Bad Homburg, Germany
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
14
|
Ma S, Xie N, Li W, Yuan B, Shi Y, Wang Y. Immunobiology of mesenchymal stem cells. Cell Death Differ 2013; 21:216-25. [PMID: 24185619 PMCID: PMC3890955 DOI: 10.1038/cdd.2013.158] [Citation(s) in RCA: 569] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 10/03/2013] [Accepted: 10/04/2013] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be isolated from almost all tissues and effectively expanded in vitro. Although their true in situ properties and biological functions remain to be elucidated, these in vitro expanded cells have been shown to possess potential to differentiate into specific cell lineages. It is speculated that MSCs in situ have important roles in tissue cellular homeostasis by replacing dead or dysfunctional cells. Recent studies have demonstrated that in vitro expanded MSCs of various origins have great capacity to modulate immune responses and change the progression of different inflammatory diseases. As tissue injuries are often accompanied by inflammation, inflammatory factors may provide cues to mobilize MSCs to tissue sites with damage. Before carrying out tissue repair functions, MSCs first prepare the microenvironment by modulating inflammatory processes and releasing various growth factors in response to the inflammation status. In this review, we focus on the crosstalk between MSCs and immune responses and their potential clinical applications, especially in inflammatory diseases.
Collapse
Affiliation(s)
- S Ma
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| | - N Xie
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| | - W Li
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| | - B Yuan
- National Institutes for Food and Drug Control, No. 2 Tiantan Xili, Beijing 100050, China
| | - Y Shi
- 1] Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China [2] Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08901, USA
| | - Y Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
15
|
Herrera MB, Fonsato V, Bruno S, Grange C, Gilbo N, Romagnoli R, Tetta C, Camussi G. Human liver stem cells improve liver injury in a model of fulminant liver failure. Hepatology 2013; 57:311-9. [PMID: 22829291 DOI: 10.1002/hep.25986] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 07/16/2012] [Indexed: 12/17/2022]
Abstract
UNLABELLED Liver transplantation is currently the only effective therapy for fulminant liver failure, but its use is limited by the scarcity of organs for transplantation, high costs, and lifelong immunosuppression. Here we investigated whether human liver stem cells (HLSCs) protect from death in a lethal model of fulminant liver failure induced by intraperitoneal injection of D-galactosamine and lipopolysaccharide in SCID mice. We show that injection of HLSCs and of HLSC-conditioned medium (CM) significantly attenuates mouse mortality in this model. Histopathological analysis of liver tissue showed reduction of liver apoptosis and enhancement of liver regeneration. By optical imaging we observed a preferential localization of labeled HLSCs within the liver. HLSCs were detected by immunohistochemistry in large liver vessels (at 24 hours) and in the liver parenchyma (after day 3). Fluorescence in situ hybridization analysis with the human pan-centromeric probe showed that positive cells were cytokeratin-negative at 24 hours. Coexpression of cytokeratin and human chromosome was observed at 7 and, to a lesser extent, at 21 days. HLSC-derived CM mimicked the effect of HLSCs in vivo. Composition analysis of the HLSC-CM revealed the presence of growth factors and cytokines with liver regenerative properties. In vitro experiments showed that HLSC-CM protected human hepatocytes from apoptosis and enhanced their proliferation. CONCLUSION These data suggest that fulminant liver failure may potentially benefit from treatment with HLSCs or HLSC-CM.
Collapse
Affiliation(s)
- Maria Beatriz Herrera
- Department of Internal Medicine and Molecular Biotechnology Center, University of Turin, Italy
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Sutton EJ, Henning TD, Boddington S, Demos S, Krug C, Meier R, Kornak J, Zhao S, Baehner R, Sharifi S, Daldrup-Link H. In vivo magnetic resonance imaging and optical imaging comparison of viable and nonviable mesenchymal stem cells with a bifunctional label. Mol Imaging 2010; 9:278-290. [PMID: 20868628 PMCID: PMC3060764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023] Open
Abstract
The purpose of this study was to compare viable and nonviable bilabeled mesenchymal stem cells (MSCs) in arthritic joints with magnetic resonance imaging (MRI) and optical imaging (OI). MSCs were labeled with ferucarbotran and DiD. MRI and OI of bilabeled cells were compared with controls. Six rats with arthritis received intra-articular injections of bilabeled viable MSCs into the right knee and nonviable MSCs into the left knee. Animals underwent MRI and OI preinjection and at 4, 24, 48, and 72 hours postinjection. The results were analyzed with a mixed random effects model and Fisher probability. Bilabeled MSCs showed increased MRI and OI signals compared to unlabeled controls (p < .0001). After intra-articular injection, bilabeled MSCs caused significant T2 and T2* effect on MRI and fluorescence on OI up to 72 hours postinjection (p < .05). There was no significant difference between viable and nonviable MSC signal in the knee joints; however, some of the viable cells migrated to an adjacent inflamed ankle joint (p < .05). Immunohistochemistry confirmed viable MSCs in right knee and ankle joints and nonviable MSCs in the left knee. Viable and nonviable cells could not be differentiated with MRI or OI signal intensity but were differentiated based on their ability to migrate in vivo.
Collapse
Affiliation(s)
- Elizabeth Jane Sutton
- Department of Radiology, Mount Auburn Hospital, 330 Mount Auburn Street, Cambridge, MA 02138, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sutton EJ, Henning TD, Boddington S, Demos S, Krug C, Meier R, Kornak J, Zhao S, Baehner R, Sharifi S, Daldrup-Link H. In Vivo Magnetic Resonance Imaging and Optical Imaging Comparison of Viable and Nonviable Mesenchymal Stem Cells with a Bifunctional Label. Mol Imaging 2010. [DOI: 10.2310/7290.2010.00029] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Elizabeth Jane Sutton
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Tobias D. Henning
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Sophie Boddington
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Stavros Demos
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Christian Krug
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Reinhardt Meier
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - John Kornak
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Shoujun Zhao
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Rick Baehner
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Sheida Sharifi
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| | - Heike Daldrup-Link
- From the Departments of Radiology and Pathology, Mount Auburn Hospital, Cambridge, MA; Departments of Radiology and Biomedical Imaging, and Pathology, University of California, San Francisco, CA; Department of Radiology, Technical University of Munich, Munich, Germany; Lawrence Livermore National Laboratory, Livermore, CA
| |
Collapse
|