1201
|
Abstract
Type 1 diabetes mellitus (T1D) is a chronic, multifactorial autoimmune disease that involves the progressive destruction of pancreatic β-cells, ultimately resulting in the loss of insulin production and secretion. The goal of clinical intervention is to prevent or arrest the onset and progression of autoimmunity, reverse β-cell destruction, and restore glycometabolic and immune homeostasis. Despite promising outcomes observed with islet transplantation and advancements in immunomodulatory therapies, the need for an effective cell replacement strategy for curing T1D still persists. Stem cell therapy offers a solution to the cited challenges of islet transplantation. While the regenerative potential of stem cells can be harnessed to make available a self-replenishing supply of glucose-responsive insulin-producing cells, their immunomodulatory properties may potentially be used to prevent, arrest, or reverse autoimmunity, ameliorate innate/alloimmune graft rejection, and prevent recurrence of the disease. Herein, we discuss the therapeutic potential of stem cells derived from a variety of sources for the cure of T1D, for example, embryonic stem cells, induced pluripotent stem cells, bone marrow-derived hematopoietic stem cells, and multipotent mesenchymal stromal cells derived from bone marrow, umbilical cord blood, and adipose tissue. The benefits of combinatorial approaches designed to ensure the successful clinical translation of stem cell therapeutic strategies, such as approaches combining effective stem cell strategies with islet transplantation, immunomodulatory drug regimens, and/or novel bioengineering techniques, are also discussed. To conclude, the application of stem cell therapy in the cure for T1D appears extremely promising.
Collapse
Affiliation(s)
- Preeti Chhabra
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kenneth L. Brayman
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
1202
|
Paracrine activity of stem cells in therapy for acute lung injury and adult respiratory distress syndrome. J Trauma Acute Care Surg 2013. [DOI: 10.1097/01586154-201305000-00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
1203
|
Pluchino S, Cossetti C. How stem cells speak with host immune cells in inflammatory brain diseases. Glia 2013; 61:1379-401. [PMID: 23633288 DOI: 10.1002/glia.22500] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/01/2013] [Indexed: 12/14/2022]
Abstract
Advances in stem cell biology have raised great expectations that diseases and injuries of the central nervous system (CNS) may be ameliorated by the development of non-hematopoietic stem cell medicines. Yet, the application of adult stem cells as CNS therapeutics is challenging and the interpretation of some of the outcomes ambiguous. In fact, the initial idea that stem cell transplants work only via structural cell replacement has been challenged by the observation of consistent cellular signaling between the graft and the host. Cellular signaling is the foundation of coordinated actions and flexible responses, and arises via networks of exchanging and interacting molecules that transmit patterns of information between cells. Sustained stem cell graft-to-host communication leads to remarkable trophic effects on endogenous brain cells and beneficial modulatory actions on innate and adaptive immune responses in vivo, ultimately promoting the healing of the injured CNS. Among a number of adult stem cell types, mesenchymal stem cells (MSCs) and neural stem/precursor cells (NPCs) are being extensively investigated for their ability to signal to the immune system upon transplantation in experimental CNS diseases. Here, we focus on the main cellular signaling pathways that grafted MSCs and NPCs use to establish a therapeutically relevant cross talk with host immune cells, while examining the role of inflammation in regulating some of the bidirectionality of these communications. We propose that the identification of the players involved in stem cell signaling might contribute to the development of innovative, high clinical impact therapeutics for inflammatory CNS diseases.
Collapse
Affiliation(s)
- Stefano Pluchino
- Department of Clinical Neurosciences, John van Geest Cambridge Centre for Brain Repair and Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, United Kingdom.
| | | |
Collapse
|
1204
|
Wang YC, Wang SH, Wei YN, Du DW, Xu H, Gao CC, Zheng MH, Xie J, Li JC, Dong GY, Li L, Xiao Y, Han H. Notch-RBP-J signaling is required by bone marrow stromal cells for the treatment of acute graft versus host disease. Stem Cell Res 2013; 11:721-35. [PMID: 23735298 DOI: 10.1016/j.scr.2013.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 04/20/2013] [Accepted: 04/22/2013] [Indexed: 12/29/2022] Open
Abstract
Recent evidence has shown that bone marrow stromal cells (BMSCs) may exhibit immuno-suppression activities through soluble mediators and direct cell-cell contact, but how these processes are modulated has been poorly understood. In this study, we show that the Notch signaling pathway participates in the modulation of BMSCs to elicit their immuno-suppressive roles. In a murine lethal acute graft versus host disease (aGvHD) model, BMSCs deficient for RBP-J, the critical transcription factor mediating signaling from all four mammalian Notch receptors, failed to delay the development of the disease. RBP-J deficient BMSCs were not able to inhibit the proliferation and activation of allogenic T-cells. Moreover, RBP-J deficient BMSCs could not down-regulate the expression of MHC II and co-stimulation molecules CD80 and CD86 on dendritic cells (DCs). The antigen presentation capacity of DCs co-cultured with RBP-J deficient BMSCs was not impaired in contrast to wild type BMSCs. Furthermore, we showed that the productions of IL-6 and PGE2, two critical molecules mediating the immuno-suppressive activities of BMSCs, were reduced significantly in RBP-J deficient BMSCs. Both of the two molecules were importantly involved in the regulation of BMSCs by Notch signaling. In conclusion, our data suggests that the immuno-suppressive effects of BMSCs in aGvHD are dependent on Notch-RBP-J signaling, which regulates the productions of IL-6 and PGE2.
Collapse
Affiliation(s)
- Yao-Chun Wang
- Center for Cell Therapy and Research, General Hospital of Guangzhou Military Command of PLA, Guangzhou 510010, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1205
|
Kéramidas M, de Fraipont F, Karageorgis A, Moisan A, Persoons V, Richard MJ, Coll JL, Rome C. The dual effect of mesenchymal stem cells on tumour growth and tumour angiogenesis. Stem Cell Res Ther 2013; 4:41. [PMID: 23628074 PMCID: PMC3706993 DOI: 10.1186/scrt195] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 02/11/2013] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Understanding the multiple biological functions played by human mesenchymal stem cells (hMSCs) as well as their development as therapeutics in regenerative medicine or in cancer treatment are major fields of research. Indeed, it has been established that hMSCs play a central role in the pathogenesis and progression of tumours, but their impact on tumour growth remains controversial. METHODS In this study, we investigated the influence of hMSCs on the growth of pre-established tumours. We engrafted nude mice with luciferase-positive mouse adenocarcinoma cells (TSA-Luc+) to obtain subcutaneous or lung tumours. When tumour presence was confirmed by non-invasive bioluminescence imaging, hMSCs were injected into the periphery of the SC tumours or delivered by systemic intravenous injection in mice bearing either SC tumours or lung metastasis. RESULTS Regardless of the tumour model and mode of hMSC injection, hMSC administration was always associated with decreased tumour growth due to an inhibition of tumour cell proliferation, likely resulting from deep modifications of the tumour angiogenesis. Indeed, we established that although hMSCs can induce the formation of new blood vessels in a non-tumoural cellulose sponge model in mice, they do not modify the overall amount of haemoglobin delivered into the SC tumours or lung metastasis. We observed that these tumour vessels were reduced in number but were longer. CONCLUSIONS Our results suggest that hMSCs injection decreased solid tumour growth in mice and modified tumour vasculature, which confirms hMSCs could be interesting to use for the treatment of pre-established tumours.
Collapse
|
1206
|
Kim KS, Kim HS, Park JM, Kim HW, Park MK, Lee HS, Lim DS, Lee TH, Chopp M, Moon J. Long-term immunomodulatory effect of amniotic stem cells in an Alzheimer's disease model. Neurobiol Aging 2013; 34:2408-20. [PMID: 23623603 DOI: 10.1016/j.neurobiolaging.2013.03.029] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 02/18/2013] [Accepted: 03/24/2013] [Indexed: 12/13/2022]
Abstract
Amyloid beta (Aβ) plays a major role in Alzheimer's disease (AD), and neuroinflammatory processes mediated by Aβ plaque-induced microglial cells and astrocytes contribute to AD pathogenesis. The present study examined human placenta amniotic membrane-derived mesenchymal stem cells (AMSCs), which have potent immunomodulatory and paracrine effects in a Tg2576 (APPswe) transgenic mouse model of AD. AMSCs secreted high levels of transforming growth factor-β under in vitro inflammatory environment conditions. Six weeks after the intravenous injection of AMSCs, APPswe mice showed evidence of improved spatial learning, which significantly correlated with the observation of fewer Aβ plaques in brain. The number of ED1-positive phagocytic microglial cells associated with Aβ plaques was higher in AMSC-injected mice than in phosphate-buffered saline-injected mice, and the level of Aβ-degrading enzymes (matrix metallopeptidase-9 and insulin-degrading enzyme) was also significantly higher. Furthermore, the level of proinflammatory cytokines, interleukin-1 and tumor necrosis factor-α, was lower and that of anti-inflammatory cytokines, interleukin-10 and transforming growth factor-β, was higher in AMSC-injected mice than phosphate-buffered saline-injected mice. These effects lasted until 12 weeks after AMSC injection. Taken together, these results collectively suggest that injection of AMSCs might show significant long-lasting improvement in AD pathology and memory function via immunomodulatory and paracrine mechanisms.
Collapse
Affiliation(s)
- Kyung-Sul Kim
- Department of Bioengineering, College of Life Science, CHA University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1207
|
Mesenchymal stem cells attenuate ischemic acute kidney injury by inducing regulatory T cells through splenocyte interactions. Kidney Int 2013; 84:521-31. [PMID: 23615497 PMCID: PMC3778762 DOI: 10.1038/ki.2013.114] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 01/22/2013] [Accepted: 01/24/2013] [Indexed: 02/06/2023]
Abstract
The mechanism of mesenchymal stem cell therapy in acute kidney injury remains uncertain. Previous studies indicated that mesenchymal stem cells could attenuate inflammation-related organ injury by induction of regulatory T cells. Whether regulatory T-cell induction is a potential mechanism of mesenchymal stem cell therapy in ischemic acute kidney injury and how these induced regulatory T cells orchestrate local inflammation are unknown. Here we found that mesenchymal stem cells decrease serum creatinine and urea nitrogen levels, improve tubular injury, and downregulate IFN-γ production of T cells in the ischemic kidney. In addition to the lung, mesenchymal stem cells persisted mostly in the spleen. Mesenchymal stem cells increased the percentage of regulatory T cells in the spleen and the ischemic kidney. Antibody-dependent depletion of regulatory T cells blunted the therapeutic effect of mesenchymal stem cells, while coculture of splenocytes with mesenchymal stem cells caused an increase in the percentage of regulatory T cells. Splenectomy abrogated attenuation of ischemic injury, and downregulated IFN-γ production and the induction of regulatory T cells by mesenchymal stem cells. Thus, mesenchymal stem cells ameliorate ischemic acute kidney injury by inducing regulatory T cells through interactions with splenocytes. Accumulated regulatory T cells in ischemic kidney might be involved in the downregulation of IFN-γ production.
Collapse
|
1208
|
Cell transplantation as an initiator of endogenous stem cell-based tissue repair. Curr Opin Organ Transplant 2013; 17:670-4. [PMID: 23111645 DOI: 10.1097/mot.0b013e328359a617] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to summarize recent findings of endogenous cell-based tissue repair triggered by a multitude of approaches, especially stem cell therapy. RECENT FINDINGS The emerging evidence has demonstrated that an important role of transplanted cells is to act as an initiator to trigger endogenous stem cell-mediated tissue repair after injury. Multiple approaches may orchestrate endogenous regeneration. The low efficacy of endogenous stem cells may be due to deficiency of cytokines to activate and induce endogenous stem cell homing, relatively insufficient endogenous stem cell pool, diseases or aging-related dysfunction of endogenous stem cells, and hostile microenvironments that limit the capacity of endogenous stem cells to repair damaged tissue. In certain situations, external stimulation and/or exogenous stem cells may be required to catalyze the repair. SUMMARY Endogenous stem cells are playing an important role in tissue repair. An important role of transplanted cells is to act as an initiator to trigger endogenous stem cell-based tissue repair.
Collapse
|
1209
|
Abstract
PURPOSE OF REVIEW Stem cell-based therapies for preventing and treating chronic end-organ dysfunction have captured the imagination of the lay public and spurred scientific and clinical development in multiple disciplines and disease states. The goal of this review is to build a framework around the different approaches being deployed to heal or treat end-organ dysfunction and discuss how within this framework future developments may occur. RECENT FINDINGS In this review, we divide the development of regenerative therapies into two broad categories. The first 'Stem Cells as the Student' focuses on the fact that we need to coax/teach the stem cells to differentiate in an efficient manner into the cells of interest, then using tissue engineering, we need to integrate them in an appropriate delivery system/matrix, and then generate a blood supply, sufficient to allow for their survival following engraftment. In the second category 'Stem Cells as the Teacher,' we learn from studies on stem cell biology, critical pathways that are dysregulated in tissue repair. By identifying these critical pathways, we can develop drug and biologics that can enhance tissue repair and end-organ function. SUMMARY Regenerative therapies have exciting potential to improve patient outcomes in a variety of acute and chronic disease states. There is significant excitement in general public, and the scientific and clinical communities. Early studies have been variably successful. As we move forward and understand the biology and engineering principles involved, significant advances with greater chances of success and efficacy will come.
Collapse
|
1210
|
Zhang H, Fang J, Wu Y, Mai Y, Lai W, Su H. Mesenchymal stem cells protect against neonatal rat hyperoxic lung injury. Expert Opin Biol Ther 2013; 13:817-29. [PMID: 23534609 DOI: 10.1517/14712598.2013.778969] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Bronchopulmonary dysplasia (BPD) is a significant global health problem and currently lacks effective therapy. We established a neonatal rat model of BPD to investigate therapeutic potential of bone marrow-derived mesenchymal stem cells (BMSCs) in neonatal hyperoxic lung injury. METHODS BMSCs were isolated, identified, and transfected by lentiviral vector carrying green fluorescent protein gene in vitro. Neonatal Sprague-Dawley rats were injected intravenously with either BMSCs or phosphate-buffered saline following 95% oxygen exposure, and assessed for the survival rate and alveolar injury during recovery. RESULTS Treatment with BMSCs after oxygen exposure for 7 days improved survival of neonatal rat during recovery. BMSCs protected against neonatal rat hyperoxic lung injury during recovery as demonstrated by enhanced expression of AQP5 and SP-C, likely due to the suppression of alveolar cell apoptosis and lung inflammation responses to oxygen with up-regulation of the expression of BCL-2 gene and down-regulation of the expression of BAX gene and stimulation of vascular endothelial growth factor and so on. CONCLUSIONS BMSCs protect against O2-mediated injury partially through stimulation of potent mediators that participate in tissue repair.
Collapse
Affiliation(s)
- Hongshan Zhang
- Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Department of Pediatric , Yanjiang Road 107, Guangzhou, Guangdong 510120 , China.
| | | | | | | | | | | |
Collapse
|
1211
|
Therapeutic potential of umbilical cord mesenchymal stromal cells transplantation for cerebral palsy: a case report. Case Rep Transplant 2013; 2013:146347. [PMID: 23533920 PMCID: PMC3603664 DOI: 10.1155/2013/146347] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 02/13/2013] [Indexed: 12/27/2022] Open
Abstract
Cerebral palsy is the most common motor disability in childhood. In current paper, we first report our clinical data regarding administration of umbilical cord mesenchymal stem cells (MSCs) transplantation in treatment of cerebral palsy. A 5-year-old girl with cerebral palsy was treated with multiple times of intravenous and intrathecal administration of MSCs derived from her young sister and was followed up for 28 months. The gross motor dysfunction was improved. Other benefits included enhanced immunity, increased physical strength, and adjusted speech and comprehension. Temporary low-grade fever was the only side effect during the treatment. MSCs may be a safe and effective therapy to improve symptoms in children with cerebral palsy.
Collapse
|
1212
|
Kim J, Ma T. Endogenous extracellular matrices enhance human mesenchymal stem cell aggregate formation and survival. Biotechnol Prog 2013; 29:441-51. [PMID: 23296993 DOI: 10.1002/btpr.1686] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 12/07/2012] [Indexed: 02/06/2023]
Abstract
Human mesenchymal stem or stromal cell (hMSC) therapies have promise across a wide range of diseases. However, inefficient cell delivery and low cell survival at injury sites reduce efficacy and are the major barriers in hMSC-based therapy. Formation of three-dimensional (3D) hMSC aggregates has been found to activate hMSC functions from enhancing secretion of therapeutic factors for improving cell migration and survival. As the stromal cells in bone marrow, hMSCs are significant sources of extracellular matrix (ECM) proteins and growth factors, which form an interactive microenvironment to influence hMSC fate via paracrine and autocrine actions. To date, however, the impact of the extracellular microenvironment on hMSC properties in the aggregates remains unknown. In the present study, we investigated the role of endogenous ECM matrices on hMSC aggregate formation and survival under ischemic stress. The results demonstrated that the preservation of endogenous ECM in the aggregates formed by thermal lifting (termed TLAs) as opposed to the aggregates formed by enzymatically detached hMSCs (termed EDAs) enhanced cell proliferation, multilineage potential, and survival under ischemic stress. The improved cell proliferation and viability in the TLAs is attributed to the incorporation of endogenous ECM proteins in the TLAs and their promitotic and antioxidant properties. The results demonstrate a novel method for the formation of hMSC aggregates via thermal responsive surface and highlight the significant contribution of the ECM in preserving hMSC properties in the 3D aggregates.
Collapse
Affiliation(s)
- Junho Kim
- Dept. of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
| | | |
Collapse
|
1213
|
Dreher L, Elvers-Hornung S, Brinkmann I, Huck V, Henschler R, Gloe T, Klüter H, Bieback K. Cultivation in Human Serum Reduces Adipose Tissue-Derived Mesenchymal Stromal Cell Adhesion to Laminin and Endothelium and Reduces Capillary Entrapment. Stem Cells Dev 2013; 22:791-803. [DOI: 10.1089/scd.2012.0051] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Lena Dreher
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Susanne Elvers-Hornung
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Irena Brinkmann
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Volker Huck
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Reinhard Henschler
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Institute of Transfusion Medicine and Immune Haematology, Frankfurt, Germany
| | - Torsten Gloe
- Cardiovascular Physiology, Medical Faculty Mannheim, Centre for Biomedicine and Medical Technology, Heidelberg University, Mannheim, Germany
| | - Harald Klüter
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Karen Bieback
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| |
Collapse
|
1214
|
Menard C, Pacelli L, Bassi G, Dulong J, Bifari F, Bezier I, Zanoncello J, Ricciardi M, Latour M, Bourin P, Schrezenmeier H, Sensebé L, Tarte K, Krampera M. Clinical-grade mesenchymal stromal cells produced under various good manufacturing practice processes differ in their immunomodulatory properties: standardization of immune quality controls. Stem Cells Dev 2013; 22:1789-801. [PMID: 23339531 DOI: 10.1089/scd.2012.0594] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Clinical-grade mesenchymal stromal cells (MSCs) are usually expanded from bone marrow (BMMSCs) or adipose tissue (ADSCs) using processes mainly differing in the use of fetal calf serum (FCS) or human platelet lysate (PL). We aimed to compare immune modulatory properties of clinical-grade MSCs using a combination of fully standardized in vitro assays. BMMSCs expanded with FCS (BMMSC-FCS) or PL (BMMSC-PL), and ADSC-PL were analyzed in quantitative phenotypic and functional experiments, including their capacity to inhibit the proliferation of T, B, and NK cells. The molecular mechanisms supporting T-cell inhibition were investigated. These parameters were also evaluated after pre-stimulation of MSCs with inflammatory cytokines. BMMSC-FCS, BMMSC-PL, and ADSC-PL displayed significant differences in expression of immunosuppressive and adhesion molecules. Standardized functional assays revealed that resting MSCs inhibited proliferation of T and NK cells, but not B cells. ADSC-PL were the most potent in inhibiting T-cell growth, a property ascribed to interferon-γ-dependent indoleamine 2,3-dioxygenase activity. MSCs did not stimulate allogeneic T cell proliferation but were efficiently lysed by activated NK cells. The systematic use of quantitative and reproducible validation techniques highlights differences in immunological properties of MSCs produced using various clinical-grade processes. ADSC-PL emerge as a promising candidate for future clinical trials.
Collapse
Affiliation(s)
- Cedric Menard
- SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1215
|
Cárdenes N, Cáceres E, Romagnoli M, Rojas M. Mesenchymal stem cells: a promising therapy for the acute respiratory distress syndrome. Respiration 2013; 85:267-78. [PMID: 23428562 DOI: 10.1159/000347072] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a pulmonary syndrome with growing prevalence and high mortality and morbidity that increase with age. There is no current therapy able to restore pulmonary function in ARDS patients. Preclinical models of ARDS have demonstrated that intratracheal or systemic administration of mesenchymal stem cells (MSCs) protects the lung against injury. The mechanisms responsible for the protective effects are multiple, including the secretion of multiple paracrine factors capable of modulating the immune response and restoring epithelial and endothelial integrity. Recent studies have demonstrated that MSCs can also control oxidative stress, transfer functional mitochondria to the damaged cells, and control bacterial infection by secretion of antibacterial peptides. These characteristics make MSCs promising candidates for ARDS therapy.
Collapse
Affiliation(s)
- Nayra Cárdenes
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| | | | | | | |
Collapse
|
1216
|
Mesenchymal stromal cells: misconceptions and evolving concepts. Cytotherapy 2013; 15:140-5. [DOI: 10.1016/j.jcyt.2012.11.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 11/01/2012] [Accepted: 11/06/2012] [Indexed: 12/17/2022]
|
1217
|
Molecular characteristics of bone marrow mesenchymal stem cells, source of regenerative medicine. Int J Cardiol 2013; 163:125-31. [DOI: 10.1016/j.ijcard.2011.11.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2011] [Revised: 11/03/2011] [Accepted: 11/04/2011] [Indexed: 12/22/2022]
|
1218
|
Ylöstalo JH, Bartosh TJ, Coble K, Prockop DJ. Human mesenchymal stem/stromal cells cultured as spheroids are self-activated to produce prostaglandin E2 that directs stimulated macrophages into an anti-inflammatory phenotype. Stem Cells 2013; 30:2283-96. [PMID: 22865689 DOI: 10.1002/stem.1191] [Citation(s) in RCA: 329] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Culturing cells in three dimension (3D) provides an insight into their characteristics in vivo. We previously reported that human mesenchymal stem/stromal cells (hMSCs) cultured as 3D spheroids acquire enhanced anti-inflammatory properties. Here, we explored the effects of hMSC spheroids on macrophages that are critical cells in the regulation of inflammation. Conditioned medium (CM) from hMSC spheroids inhibited lipopolysaccharide-stimulated macrophages from secreting proinflammatory cytokines TNFα, CXCL2, IL6, IL12p40, and IL23. CM also increased the secretion of anti-inflammatory cytokines IL10 and IL1ra by the stimulated macrophages, and augmented expression of CD206, a marker of alternatively activated M2 macrophages. The principal anti-inflammatory activity in CM had a small molecular weight, and microarray data suggested that it was prostaglandin E2 (PGE2). This was confirmed by the observations that PGE2 levels were markedly elevated in hMSC spheroid-CM, and that the anti-inflammatory activity was abolished by an inhibitor of cyclooxygenase-2 (COX-2), a silencing RNA for COX-2, and an antibody to PGE2. The anti-inflammatory effects of the PGE2 on stimulated macrophages were mediated by the EP4 receptor. Spheroids formed by human adult dermal fibroblasts produced low levels of PGE2 and displayed negligible anti-inflammatory effects on stimulated macrophages, suggesting the features as unique to hMSCs. Moreover, production of PGE2 by hMSC spheroids was dependent on the activity of caspases and NFκB activation in the hMSCs. The results indicated that hMSCs in 3D-spheroid cultures are self-activated, in part by intracellular stress responses, to produce PGE2 that can change stimulated macrophages from a primarily proinflammatory M1 phenotype to a more anti-inflammatory M2 phenotype.
Collapse
Affiliation(s)
- Joni H Ylöstalo
- Texas A & M Health Science Center College of Medicine, Institute for Regenerative Medicine at Scott & White, Temple, Texas 76502, USA
| | | | | | | |
Collapse
|
1219
|
Zhang S, Danchuk SD, Imhof KMP, Semon JA, Scruggs BA, Bonvillain RW, Strong AL, Gimble JM, Betancourt AM, Sullivan DE, Bunnell BA. Comparison of the therapeutic effects of human and mouse adipose-derived stem cells in a murine model of lipopolysaccharide-induced acute lung injury. Stem Cell Res Ther 2013; 4:13. [PMID: 23360775 PMCID: PMC3706907 DOI: 10.1186/scrt161] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/02/2013] [Accepted: 01/23/2013] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Adipose-derived stem cells (ASCs) have emerged as important regulators of inflammatory/immune responses in vitro and in vivo and represent attractive candidates for cell-based therapies for diseases that involve excessive inflammation. Acute lung injury (ALI) is an inflammatory condition for which treatment is mainly supportive due to lack of effective therapies. In this study, the therapeutic effects of ASC-based therapy were assessed in vivo by comparison of the anti-inflammatory properties of both human and murine ASCs in a mouse model of lipopolysaccharide (LPS)-induced ALI. METHODS Human ASCs (hASCs) or mouse ASCs (mASCs) were delivered to C57Bl/6 mice (7.5 × 105 total cells/mouse) by oropharyngeal aspiration (OA) four hours after the animals were challenged with lipopolysaccharide (15 mg/kg). Mice were sacrificed 24 and 72 hours after LPS exposure, and lung histology examined for evaluation of inflammation and injury. Bronchoalveolar lavage fluid (BALF) was analyzed to determine total and differential cell counts, total protein and albumin concentrations, and myeloperoxidase (MPO) activity. Cytokine expression in the injured lungs was measured at the steady-state mRNA levels and protein levels for assessment of the degree of lung inflammation. RESULTS Both human and mouse ASC treatments provided protective anti-inflammatory responses. There were decreased levels of leukocyte (for example neutrophil) migration into the alveoli, total protein and albumin concentrations in BALF, and MPO activity after the induction of ALI following both therapies. Additionally, cell therapy with both cell types effectively suppressed the expression of proinflammatory cytokines and increased the anti-inflammatory cytokine interleukin 10 (IL-10). Overall, the syngeneic mASC therapy had a more potent therapeutic effect than the xenogeneic hASC therapy in this model. CONCLUSIONS Treatment with hASCs or mASCs significantly attenuated LPS-induced acute lung injury in mice. These results suggest a potential benefit for using an ASC-based therapy to treat clinical ALI and may possibly prevent the development of acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Shijia Zhang
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
- Department of Pharmacology, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-83, New Orleans, LA 70112, USA
| | - Svitlana D Danchuk
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Kathleen MP Imhof
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, 6400 Freret Street, New Orleans, LA 70118, USA
| | - Julie A Semon
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Brittni A Scruggs
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
- Department of Pharmacology, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-83, New Orleans, LA 70112, USA
| | - Ryan W Bonvillain
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Amy L Strong
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Jeffrey M Gimble
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Aline M Betancourt
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
| | - Deborah E Sullivan
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-99, New Orleans, LA 70112, USA
- Department of Pharmacology, School of Medicine, Tulane University, 1430 Tulane Avenue, SL-83, New Orleans, LA 70112, USA
- Division of Regenerative Medicine, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA 70433, USA
| |
Collapse
|
1220
|
Giunti D, Parodi B, Usai C, Vergani L, Casazza S, Bruzzone S, Mancardi G, Uccelli A. Mesenchymal stem cells shape microglia effector functions through the release of CX3CL1. Stem Cells 2013; 30:2044-53. [PMID: 22821677 DOI: 10.1002/stem.1174] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSC) display a remarkable ability to modulate the immune response and protect the central nervous system mainly through the release of soluble factors in a paracrine fashion, affecting the functional behavior of cells in the tissues. Here we investigated the effect of the interaction between MSC and microglia in vitro, and we dissected the molecular and cellular mechanisms of this crosstalk. We demonstrated that MSC impair microglia activation by inflammatory cues through the inhibition of the expression and release of inflammatory molecules and stress-associated proteins. We showed that MSC significantly increase microglial expression and release of molecules associated with a neuroprotective phenotype such as CX3CR1, nuclear receptor 4 family, CD200 receptor, and insulin growth factor 1. Interestingly, MSC can enhance functional changes on microglia as depicted by the increase of intracellular calcium concentration and phagocytic activity. This last event is associated with an increased expression of triggering receptor expressed on myeloid cells-2, an innate immune receptor involved in phagocytosis in the absence of inflammation. The observed effects on CX3CR1-expressing microglia are due to the release of CX3CL1 by MSC, driven by inflammatory signals, as demonstrated by the reversal of the observed results when CX3CL1 expression was silenced in MSC or its release was blocked. Finally, we showed that exogenous CX3CL1 induce phenotypic and functional changes of microglia similar to those induced by MSC. These findings demonstrate that MSC instruct, through the release of CX3CL1, microglia responsiveness to proinflammatory signals by modulating constitutive "calming" receptors, typically expressed by "steady-state microglia" thus switching microglia from a detrimental phenotype to a neuroprotective one.
Collapse
Affiliation(s)
- Debora Giunti
- Department of Neurosciences, Ophthalmology and Genetics, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
1221
|
Yang X, Hou J, Han Z, Wang Y, Hao C, Wei L, Shi Y. One cell, multiple roles: contribution of mesenchymal stem cells to tumor development in tumor microenvironment. Cell Biosci 2013; 3:5. [PMID: 23336752 PMCID: PMC3693909 DOI: 10.1186/2045-3701-3-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/20/2012] [Indexed: 12/13/2022] Open
Abstract
The discovery of tissue reparative and immunosuppressive abilities of mesenchymal stem cells (MSCs) has drawn more attention to tumor microenvironment and its role in providing the soil for the tumor cell growth. MSCs are recruited to tumor which is referred as the never healing wound and altered by the inflammation environment, thereby helping to construct the tumor microenvironment. The environment orchestrated by MSCs and other factors can be associated with angiogenesis, immunosuppression, inhibition of apoptosis, epithelial-mesenchymal transition (EMT), survival of cancer stem cells, which all contribute to tumor growth and progression. In this review, we will discuss how MSCs are recruited to the tumor microenvironment and what effects they have on tumor progression.
Collapse
Affiliation(s)
- Xue Yang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, the Second Military Medicial University, 225 Changhai Road, Shanghai 200438, China.
| | | | | | | | | | | | | |
Collapse
|
1222
|
Jiang D, Qi Y, Walker NG, Sindrilaru A, Hainzl A, Wlaschek M, MacNeil S, Scharffetter-Kochanek K. The effect of adipose tissue derived MSCs delivered by a chemically defined carrier on full-thickness cutaneous wound healing. Biomaterials 2013; 34:2501-15. [PMID: 23317813 DOI: 10.1016/j.biomaterials.2012.12.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 12/14/2012] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem cells (MSCs) have properties which make them promising for the treatment of chronic non-healing wounds. A major so far unmet challenge is the efficient, safe and painless delivery of MSCs to skin wounds. Recently, a surface carrier of medical-grade silicone coated by plasma polymerisation with a thin layer of acrylic acid (ppAAc) was developed, and shown to successfully deliver MSCs to deepithelialised human dermis in vitro. Here we studied the potential of the ppAAc carrier to deliver human adipose tissue derived MSCs (AT-MSCs) to murine full-thickness excisional skin wounds in vivo. Further we investigate the mechanism of action of MSCs in accelerating wound healing in these wounds. AT-MSCs cultured on ppAAc carriers for 4 days or longer fully retained their cell surface marker expression profile, colony-forming-, differentiation- and immunosuppressive potential. Importantly, AT-MSCs delivered to murine wounds by ppAAc carriers significantly accelerated wound healing, similar to AT-MSCs delivered by intradermal injection. More than 80% of AT-MSCs were transferred from carriers to wounds in 3 days. AT-MSCs were detectable in wounds for at least 5 days after wounding. Carrier delivered AT-MSCs were demonstrated to have the capacity to down-modulate TNF-α-dependent inflammation, increase anti-inflammatory M2 macrophage numbers, and induce TGF-β(1)-dependent angiogenesis, myofibroblast differentiation and granulation tissue formation, thereby enhancing overall tissue repair.
Collapse
Affiliation(s)
- Dongsheng Jiang
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm 89081, Germany
| | | | | | | | | | | | | | | |
Collapse
|
1223
|
The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine. Nat Med 2013; 19:35-42. [PMID: 23296015 DOI: 10.1038/nm.3028] [Citation(s) in RCA: 889] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 10/29/2012] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are the focus of intensive efforts worldwide directed not only at elucidating their nature and unique properties but also developing cell-based therapies for a diverse range of diseases. More than three decades have passed since the original formulation of the concept, revolutionary at the time, that multiple connective tissues could emanate from a common progenitor or stem cell retained in the postnatal bone marrow. Despite the many important advances made since that time, substantial ambiguities still plague the field regarding the nature, identity, function, mode of isolation and experimental handling of MSCs. These uncertainties have a major impact on their envisioned therapeutic use.
Collapse
|
1224
|
Kholodenko IV, Konieva AA, Kholodenko RV, Yarygin KN. Molecular mechanisms of migration and homing of intravenously transplanted mesenchymal stem cells. ACTA ACUST UNITED AC 2013. [DOI: 10.7243/2050-1218-2-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
1225
|
He S, Khan J, Gleason J, Eliav E, Fik-Rymarkiewicz E, Herzberg U, Albert V, Hariri R. Placenta-derived adherent cells attenuate hyperalgesia and neuroinflammatory response associated with perineural inflammation in rats. Brain Behav Immun 2013; 27:185-92. [PMID: 23103445 DOI: 10.1016/j.bbi.2012.10.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/05/2012] [Accepted: 10/17/2012] [Indexed: 10/27/2022] Open
Abstract
Neuropathic pain is a debilitating condition of the somatosensory system caused by pathology of the nervous system. Current drugs treat symptoms but largely fail to target the underlying mechanisms responsible for the pathological changes seen in the central or peripheral nervous system. We investigated the therapeutic effects of PDA-001, a culture expanded placenta-derived adherent cell, in the rat neuritis model. Pain is induced in the model by applying carrageenan to the sciatic nerve trunk, causing perineural inflammation of the sciatic nerve. PDA-001, at doses ranging from 0.4×10(6) to 4×10(6) cells/animal, or vehicle control was intravenously administrated to assess the biological activity of the cells. A dose-dependent effect of PDA-001 on pain relief was demonstrated. PDA-001 at doses of 1×10(6) and 4×10(6), but not 0.4×10(6), reduced mechanical hyperalgesia within 24h following treatment and through day 8 after induction of neuritis. The mechanism underlying PDA-001-mediated reduction of neuroinflammatory pain was also explored. Ex vivo tissue analyses demonstrated that PDA-001 suppressed homing, maturation and differentiation of dendritic cells, thus inhibiting T-cell priming and activation in draining lymph nodes. PDA-001 also reduced interferon gamma and IL-17 in draining lymph nodes and in the ispilateral sciatic nerve, and increased the levels of IL-10 in draining lymph nodes and plasma, pointing to T-cell modulation as a possible mechanism mediating the observed anti-hyperalgesic effects. Furthermore, in the ipsilateral sciatic nerve, significantly less leukocyte infiltration was observed in PDA-001-treated animals. The results suggest that PDA-001may provide a novel therapeutic approach in the management of inflammatory neuropathic pain and similar conditions.
Collapse
Affiliation(s)
- Shuyang He
- Celgene Cellular Therapeutics, Warren, NJ, USA.
| | | | | | | | | | | | | | | |
Collapse
|
1226
|
Popp N, Chu XK, Shen D, Tuo J, Chan CC. Evaluating Potential Therapies in a Mouse Model of Focal Retinal Degeneration with Age-related Macular Degeneration (AMD)-Like Lesions. ACTA ACUST UNITED AC 2013; 4:1000296. [PMID: 24432192 PMCID: PMC3890246 DOI: 10.4172/2155-9570.1000296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although the mouse has no macula leutea, its neuroretina and retinal pigment epithelium (RPE) can develop lesions mimicking certain features of age-related macular degeneration (AMD). Differences between the Ccl2 and Cx3cr1 double deficient mouse on Crb1rd8(rd8) background (DKOrd8) and the Crb1rd8 mouse in photoreceptor and RPE pathology, as well as ocularA2E contents and immune responses, show that DKOrd8 recapitulates some human AMD-like features in addition to rd8 retinal dystrophy/degeneration. Different therapeutic interventions have been demonstrated to be effective on the AMD-like features of DKOrd8 mice. The use of the DKOrd8 model and C57BL/6N (wild type, WT) mice as group controls (4 groups) to test treatments such as high omega-3 polyunsaturated fatty acid (n-3) diet has, for example, shown the beneficial effect of n-3 on AMD-like lesions by anti-inflammatory action of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA). The use of self-control in the DKOrd8 mouse by treating one eye and using the contralateral eye as the control for the same mouse allows for appropriate interventional experiments and evaluates various novel therapeutic agents. Three examples will be briefly presented and discussed: (1) tumor necrosis factor-inducible gene 6 recombinant protein (TSG-6) arrests the AMD-like lesions via modulation of ocular immunological gene expression, e.g., Il-17a; (2) adeno-associated virus encoding sIL-17R (AAV2.sIL17R) stabilizes the AMD-like lesions; and (3) pigment epithelium-derived factor (PEDF) ameliorates the AMD-lesions by its anti-inflammatory, anti-apoptotic and neuroprotective roles. Therefore, the DKOrd8 mouse model can be useful and appropriate for therapeutic compound screening in the management of human AMD.
Collapse
Affiliation(s)
- Nicholas Popp
- Immunopathology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, 10 Center Dr., 10/10N103, NIH/NEI, Bethesda, MD, 20892-1857, USA
| | - Xi K Chu
- Immunopathology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, 10 Center Dr., 10/10N103, NIH/NEI, Bethesda, MD, 20892-1857, USA
| | - Defen Shen
- Immunopathology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, 10 Center Dr., 10/10N103, NIH/NEI, Bethesda, MD, 20892-1857, USA
| | - Jingsheng Tuo
- Immunopathology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, 10 Center Dr., 10/10N103, NIH/NEI, Bethesda, MD, 20892-1857, USA
| | - Chi-Chao Chan
- Immunopathology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, 10 Center Dr., 10/10N103, NIH/NEI, Bethesda, MD, 20892-1857, USA
| |
Collapse
|
1227
|
Shin L, Peterson DA. Human mesenchymal stem cell grafts enhance normal and impaired wound healing by recruiting existing endogenous tissue stem/progenitor cells. Stem Cells Transl Med 2012; 2:33-42. [PMID: 23283490 DOI: 10.5966/sctm.2012-0041] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been investigated as a clinical therapy to promote tissue repair. However, the disappearance of grafted cells soon after engraftment suggests a possible role as initiators of repair rather than effectors. We evaluated the relative contribution of grafted human MSCs and host stem/progenitor cells in promoting wound healing by using a novel asymmetric wound model in normal and impaired healing diabetic (db/db) mice to discriminate between the effect of direct engraftment and the subsequent systemic response. Experimental animals received paired wounds, with one wound receiving human mesenchymal stem cells (hMSCs) and the other wound receiving vehicle to assess local and systemic effects, respectively. Control animals received vehicle in both wounds. Grafted hMSCs significantly improved healing in both normal and impaired healing animals; produced significant elevation of signals such as Wnt3a, vascular endothelial growth factor, and platelet-derived growth factor receptor-α; and increased the number of pre-existing host MSCs recruited to the wound bed. Improvement was also seen in both the grafted and nongrafted sides, suggesting a systemic response to hMSC engraftment. Healing was enhanced despite the rapid loss of hMSCs, suggesting that mobilizing the host response is the major outcome of grafting MSCs to tissue repair. We validate that hMSCs evoke a host response that is clinically relevant, and we suggest that therapeutic efforts should focus on maximizing the mobilization of host MSCs.
Collapse
Affiliation(s)
- Laura Shin
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | | |
Collapse
|
1228
|
Wagner B, Henschler R. Fate of intravenously injected mesenchymal stem cells and significance for clinical application. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 130:19-37. [PMID: 23334265 DOI: 10.1007/10_2012_155] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) have initially been characterized as a fibroblastlike cell population that can be expanded readily in vitro, and is able to support hematopoiesis in vitro and in vivo. By serendipity it was discovered that MSCs can also be administered into the bloodstream. This mode of application formed a major breakthrough in the clinical use of MSCs, because MSC transplantation was found to cure severe immune hyperactivation states such as graft-versus-host disease after allogeneic bone marrow transplantation, or bacterial sepsis. However, MSCs were found difficult to trace and consensus to date is lacking in the scientific community as to where transplanted MSCs end up in the body and which major principles are responsible for the therapeutic effects of MSCs. This chapter gives an overview of the current knowledge on interactions of freshly transplanted MSCs with the cells in the blood stream and the vessel wall, with major organs such as lung, liver, gut, and spleen, and discusses the limitations of the methodologies used to trace transplanted MSCs. The findings will be put into perspective on how therapeutically applied, culture-expanded MSCs may exert beneficial effects.
Collapse
Affiliation(s)
- Beate Wagner
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, Ludwig Maximilians University München, Marchioninistrasse 15, 81433, Munich, Germany
| | | |
Collapse
|
1229
|
Secreted factors from bone marrow stromal cells upregulate IL-10 and reverse acute kidney injury. Stem Cells Int 2012; 2012:392050. [PMID: 23319959 PMCID: PMC3539665 DOI: 10.1155/2012/392050] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 11/02/2012] [Indexed: 12/26/2022] Open
Abstract
Acute kidney injury is a devastating syndrome that afflicts over 2,000,000 people in the US per year, with an associated mortality of greater than 70% in severe cases. Unfortunately, standard-of-care treatments are not sufficient for modifying the course of disease. Many groups have explored the use of bone marrow stromal cells (BMSCs) for the treatment of AKI because BMSCs have been shown to possess unique anti-inflammatory, cytoprotective, and regenerative properties in vitro and in vivo. It is yet unresolved whether the primary mechanisms controlling BMSC therapy in AKI depend on direct cell infusion, or whether BMSC-secreted factors alone are sufficient for mitigating the injury. Here we show that BMSC-secreted factors are capable of providing a survival benefit to rats subjected to cisplatin-induced AKI. We observed that when BMSC-conditioned medium (BMSC-CM) is administered intravenously, it prevents tubular apoptosis and necrosis and ameliorates AKI. In addition, we observed that BMSC-CM causes IL-10 upregulation in treated animals, which is important to animal survival and protection of the kidney. In all, these results demonstrate that BMSC-secreted factors are capable of providing support without cell transplantation, and the IL-10 increase seen in BMSC-CM-treated animals correlates with attenuation of severe AKI.
Collapse
|
1230
|
Abstract
The past decade has seen tremendous growth in the clinical application of cell-based therapies, and the number of planned human clinical trials to evaluate these therapies continues to increase in number and scope at a rapid pace. A considerable effort on this front has been devoted to evaluating the therapeutic potential of mesenchymal stem cells (MSCs), which were initially characterized as connective tissue progenitors resident in bone marrow. MSCs are now known to possess potent tissue reparative properties that have been linked to secretion of paracrine-acting angiogenic, trophic, anti-inflammatory, and immunomodulatory factors. Accordingly, MSC-based therapies are being evaluated for the treatment of a broad array of ischemic, inflammatory, and immunological disorders. Nevertheless, knowledge regarding how the wide-ranging activities of MSCs vary between and are specified within populations remains largely unexplored. Lack of such knowledge makes it difficult to predict and/or control how sampling bias and ex vivo expansion of populations alters their biological activity and therapeutic potency. Herein, we discuss how heterogeneity of MSC populations may explain, in part, disparate outcomes in both experimental animal and human clinical trial data, and discuss several strategies to achieve more reproducible and efficacious outcomes for MSC-based therapies.
Collapse
|
1231
|
Wang N, Shao Y, Mei Y, Zhang L, Li Q, Li D, Shi S, Hong Q, Lin H, Chen X. Novel mechanism for mesenchymal stem cells in attenuating peritoneal adhesion: accumulating in the lung and secreting tumor necrosis factor α-stimulating gene-6. Stem Cell Res Ther 2012; 3:51. [PMID: 23217986 PMCID: PMC3580481 DOI: 10.1186/scrt142] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 12/04/2012] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION We previously found that mesenchymal stem cells (MSCs) injected intravenously could attenuate peritoneal adhesion by secreting tumor necrosis alpha-stimulating gene (TSG)-6, while MSCs injected intraperitoneally could not. However, the underlying mechanism remains unclear. This study was designed to investigate the means by which MSCs exert their effects. METHODS Rat bone marrow-derived MSCs/red fluorescent protein (RFP) were injected either intraperitoneally or intravenously into Sprague-Dawley (SD) rats at different time points after peritoneal scraping. Peritoneal adhesions were evaluated macroscopically at day 14 after scraping. The distribution of MSCs injected intraperitoneally or intravenously was traced by two-photon fluorescence confocal imaging and immunofluorescence microscopy. The co-localization of MSCs and macrophages in the lung and the spleen, and the expression of TSG-6 in MSCs trapped in the lung or the spleen were evaluated by immunofluorescence microscopy. The concentration of TSG-6 in serum was evaluated by ELISA. After intravenous injection of TSG-6- small interfering (si) RNA-MSCs, the expression of TSG-6 in MSCs and the concentration of TSG-6 in serum were reevaluated, and peritoneal adhesions were evaluated macroscopically and histologically. RESULTS MSCs injected intraperitoneally failed to reduce peritoneal adhesion, and MSCs injected intravenously markedly improved peritoneal adhesion. Two-photon fluorescence confocal imaging showed that MSCs injected intravenously accumulated mainly in the lung, where they remained for seven days, and immunofluorescence microscopy showed few MSCs phagocytosed by macrophages. In contrast, large numbers of MSCs accumulated in the spleen with obvious phagocytosis by macrophages even at 4 hours after intraperitoneal injection. Immunofluorescence microscopy showed that MSCs that accumulated in the lung after intravenous injection could express TSG-6 within 12 hours, but TSG-6-siRNA-MSCs or MSCs accumulated in the spleen after intraperitoneal injection did not. ELISA showed that the concentration of TSG-6 in serum was increased at 4 hours after intravenous injection of MSCs, while there was no increase after injection of TSG-6-siRNA-MSCs or after intraperitoneal injection of MSCs. Moreover, intravenous injection of TSG-6-siRNA-MSCs failed to attenuate peritoneal adhesion. CONCLUSIONS Our findings suggest that intravenously injected MSCs accumulated in the lung and attenuated peritoneal adhesion by secreting TSG-6, but intraperitoneally injected MSCs were phagocytosed by macrophages in the spleen and failed to attenuate peritoneal adhesion.
Collapse
|
1232
|
Preactivation of Human MSCs with TNF-α Enhances Tumor-Suppressive Activity. Cell Stem Cell 2012; 11:825-35. [DOI: 10.1016/j.stem.2012.10.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 07/23/2012] [Accepted: 10/04/2012] [Indexed: 12/24/2022]
|
1233
|
Ziadloo A, Burks SR, Gold EM, Lewis BK, Chaudhry A, Merino MJ, Frenkel V, Frank JA. Enhanced homing permeability and retention of bone marrow stromal cells by noninvasive pulsed focused ultrasound. Stem Cells 2012; 30:1216-27. [PMID: 22593018 DOI: 10.1002/stem.1099] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bone marrow stromal cells (BMSCs) have shown significant promise in the treatment of disease, but their therapeutic efficacy is often limited by inefficient homing of systemically administered cells, which results in low number of cells accumulating at sites of pathology. BMSC home to areas of inflammation where local expression of integrins and chemokine gradients is present. We demonstrated that nondestructive pulsed focused ultrasound (pFUS) exposures that emphasize the mechanical effects of ultrasound-tissue interactions induced local and transient elevations of chemoattractants (i.e., cytokines, integrins, and growth factors) in the murine kidney. pFUS-induced upregulation of cytokines occurred through approximately 1 day post-treatment and returned to contralateral kidney levels by day 3. This window of significant increases in cytokine expression was accompanied by local increases of other trophic factors and integrins that have been shown to promote BMSC homing. When BMSCs were intravenously administered following pFUS treatment to a single kidney, enhanced homing, permeability, and retention of BMSC was observed in the treated kidney versus the contralateral kidney. Histological analysis revealed up to eight times more BMSC in the peritubular regions of the treated kidneys on days 1 and 3 post-treatment. Furthermore, cytokine levels in pFUS-treated kidneys following BMSC administration were found to be similar to controls, suggesting modulation of cytokine levels by BMSC. pFUS could potentially improve cell-based therapies as a noninvasive modality to target homing by establishing local chemoattractant gradients and increasing expression of integrins to enhance tropism of cells toward treated tissues.
Collapse
Affiliation(s)
- Ali Ziadloo
- Laboratory of Diagnostic Radiology Research, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
1234
|
Millard SM, Fisk NM. Mesenchymal stem cells for systemic therapy: Shotgun approach or magic bullets? Bioessays 2012. [DOI: 10.1002/bies.201200087] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
1235
|
Kim JY, Kim DH, Kim JH, Yang YS, Oh W, Lee EH, Chang JW. Umbilical cord blood mesenchymal stem cells protect amyloid-β42 neurotoxicity via paracrine. World J Stem Cells 2012; 4:110-116. [PMID: 23293711 PMCID: PMC3536832 DOI: 10.4252/wjsc.v4.i11.110] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 12/04/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To understand the neuroprotective mechanism of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) against amyloid-β42 (Aβ42) exposed rat primary neurons.
METHODS: To evaluate the neuroprotective effect of hUCB-MSCs, the cells were co-cultured with Aβ42-exposed rat primary neuronal cells in a Transwell apparatus. To assess the involvement of soluble factors released from hUCB-MSCs in neuroprotection, an antibody-based array using co-cultured media was conducted. The neuroprotective roles of the identified hUCB-MSC proteins was assessed by treating recombinant proteins or specific small interfering RNAs (siRNAs) for each candidate protein in a co-culture system.
RESULTS: The hUCB-MSCs secreted elevated levels of decorin and progranulin when co-cultured with rat primary neuronal cells exposed to Aβ42. Treatment with recombinant decorin and progranulin protected from Aβ42-neurotoxicity in vitro. In addition, siRNA-mediated knock-down of decorin and progranulin production in hUCB-MSCs reduced the anti-apoptotic effects of hUCB-MSC in the co-culture system.
CONCLUSION: Decorin and progranulin may be involved in anti-apoptotic activity of hUCB-MSCs exposed to Aβ42.
Collapse
|
1236
|
Ren G, Zhao X, Wang Y, Zhang X, Chen X, Xu C, Yuan ZR, Roberts AI, Zhang L, Zheng B, Wen T, Han Y, Rabson AB, Tischfield JA, Shao C, Shi Y. CCR2-dependent recruitment of macrophages by tumor-educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFα. Cell Stem Cell 2012; 11:812-24. [PMID: 23168163 DOI: 10.1016/j.stem.2012.08.013] [Citation(s) in RCA: 272] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 03/12/2012] [Accepted: 08/30/2012] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) tend to infiltrate into tumors and form a major component of the tumor microenvironment. These tumor-resident MSCs are known to affect tumor growth, but the mechanisms are largely unknown. We found that MSCs isolated from spontaneous lymphomas in mouse (L-MSCs) strikingly enhanced tumor growth in comparison to bone marrow MSCs (BM-MSCs). L-MSCs contributed to greater recruitment of CD11b(+)Ly6C(+) monocytes, F4/80(+) macrophages, and CD11b(+)Ly6G(+) neutrophils to the tumor. Depletion of monocytes/macrophages, but not neutrophils, completely abolished tumor promotion of L-MSCs. Furthermore, L-MSCs expressed high levels of CCR2 ligands, and monocyte/macrophage accumulation and L-MSC-mediated tumor promotion were largely abolished in CCR2(-/-) mice. Intriguingly, TNFα-pretreated BM-MSCs mimicked L-MSCs in their chemokine production profile and ability to promote tumorigenesis of lymphoma, melanoma, and breast carcinoma. Therefore, our findings demonstrate that, in an inflammatory environment, tumor-resident MSCs promote tumor growth by recruiting monocytes/macrophages.
Collapse
Affiliation(s)
- Guangwen Ren
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1237
|
Gene-modified mesenchymal stem cells protect against radiation-induced lung injury. Mol Ther 2012; 21:456-65. [PMID: 23299797 DOI: 10.1038/mt.2012.183] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Radiation-induced lung injury (RILI) presents a common and major obstacle in the radiotherapy of thoracic cancers. The aim of this study was to examine whether RILI could be alleviated by mesenchymal stem cells (MSCs) expressing soluble transforming growth factor-β (TGF-β) type II receptor via an adenovirus (Ad-sTβR). Here, we systemically administered male MSCs into female mice challenged with thoracic irradiation. The data showed that either MSCs or Ad-sTβR transduced MSCs (Ad-sTβR-MSCs) specifically migrated into radiation-injured lung. Ad-sTβR-MSCs obviously alleviated lung injury, as reflected by survival and histopathology data, as well as the assays of malondialdehyde (MDA), hydroxyproline, plasma cytokines, and the expression of connective tissue growth factor (CTGF) and α-smooth muscle actin (α-SMA). Furthermore, MSCs and Ad-sTβR-MSCs could adopt the characteristics of alveolar type II (ATII) cells. However, the MSCs levels in the lungs were relatively low to account for the noted therapeutic effects, suggesting the presence of other mechanisms. In vivo, MSCs-conditioned medium (MSCs CM) significantly attenuated RILI. In vitro, MSCs CM protected ATII cells against radiation-induced apoptosis and DNA damage, and modulated the inflammatory response, indicating the beneficial effects of MSCs are largely due to its paracrine activity. Our results provide a novel insight for RILI therapy that currently lack efficient treatments.
Collapse
|
1238
|
A new vision of mesenchymal stromal cells. Mol Ther 2012; 20:2017-8. [PMID: 23131854 DOI: 10.1038/mt.2012.221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
1239
|
Bianchi G, Morandi F, Cilli M, Daga A, Bocelli-Tyndall C, Gambini C, Pistoia V, Raffaghello L. Close interactions between mesenchymal stem cells and neuroblastoma cell lines lead to tumor growth inhibition. PLoS One 2012; 7:e48654. [PMID: 23119082 PMCID: PMC3485378 DOI: 10.1371/journal.pone.0048654] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 09/28/2012] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted much interest in oncology since they exhibit marked tropism for the tumor microenvironment and support or suppress malignant cell growth depending on the tumor model tested. The aim of this study was to investigate the role of MSCs in the control of the growth of neuroblastoma (NB), which is the second most common solid tumor in children. In vivo experiments showed that systemically administered MSCs, under our experimental conditions, did not home to tumor sites and did not affect tumor growth or survival. However, MSCs injected intratumorally in an established subcutaneous NB model reduced tumor growth through inhibition of proliferation and induction of apoptosis of NB cells and prolonged the survival of hMSC-treated mice. The need for contact between MSCs and NB cells was further supported by in vitro experiments. In particular, MSCs were found to be attracted by NB cells, and to affect NB cell proliferation with different results depending on the cell line tested. Moreover, NB cells, after pre-incubation with hMSCs, acquired a more invasive behavior towards CXCL12 and the bone marrow, i.e., the primary site of NB metastases. In conclusion, this study demonstrates that functional cross-talk between MSCs and NB cell lines used in our experiments can occur only within short range interaction. Thus, this report does not support the clinical use of MSCs as vehicles for selective delivery of antitumor drugs at the NB site unless chemotherapy and/or radiotherapy create suitable local conditions for MSCs recruitment.
Collapse
Affiliation(s)
| | - Fabio Morandi
- Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Michele Cilli
- Technology Transfer, Animal Research Facility, San Martino-National Institute for Cancer Research, Genoa, Italy
| | - Antonio Daga
- Department of Translational Oncology, National Institute for Cancer Research, Genoa, Italy
| | | | - Claudio Gambini
- Laboratory of Pathology, Istituto Giannina Gaslini, Genoa, Italy
| | - Vito Pistoia
- Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Lizzia Raffaghello
- Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
- * E-mail:
| |
Collapse
|
1240
|
Abstract
Multipotent mesenchymal stromal cells (MSCs) have generated considerable interest in the fields of regenerative medicine, cell therapy and immune modulation. Over the past 5 years, the initial observations that MSCs could enhance regeneration and modulate immune responses have been significantly advanced and we now have a clearer picture of the effects that MSCs have on the immune system particularly in the context of inflammatory-mediated disorders. A number of mechanisms of action have been reported in MSC immunomodulation, which encompass the secretion of soluble factors, induction of anergy, apoptosis, regulatory T cells and tolerogenic dendritic cells. It is clear that MSCs modulate both innate and adaptive responses and evidence is now emerging that the local microenvironment is key in the activation or licensing of MSCs to become immunosuppressive. More recently, studies have suggested that MSCs have the capacity to sense their environment and have a role in pathogen clearance in conjunction with the resolution of insult or injury. This review focuses on the mechanisms of MSC immunomodulation discussing the multistep process of MSC localisation at sites of inflammation, the cross talk between MSCs and the local microenvironment as well as the subsequent mechanisms of action used to resolve inflammation.
Collapse
|
1241
|
Lee JK, Schuchman EH, Jin HK, Bae JS. Soluble CCL5 derived from bone marrow-derived mesenchymal stem cells and activated by amyloid β ameliorates Alzheimer's disease in mice by recruiting bone marrow-induced microglia immune responses. Stem Cells 2012; 30:1544-55. [PMID: 22570192 DOI: 10.1002/stem.1125] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microglia have the ability to eliminate amyloid β (Aβ) by a cell-specific phagocytic mechanism, and bone marrow (BM) stem cells have shown a beneficial effect through endogenous microglia activation in the brains of Alzheimer's disease (AD) mice. However, the mechanisms underlying BM-induced activation of microglia have not been resolved. Here we show that BM-derived mesenchymal stem cells (MSCs) induced the migration of microglia when exposed to Aβ in vitro. Cytokine array analysis of the BM-MSC media obtained after stimulation by Aβ further revealed elevated release of the chemoattractive factor, CCL5. We also observed that CCL5 was increased when BM-MSCs were transplanted into the brains of Aβ-deposited AD mice, but not normal mice. Interestingly, alternative activation of microglia in AD mice was associated with elevated CCL5 expression following intracerebral BM-MSC transplantation. Furthermore, by generating an AD-green fluorescent protein chimeric mouse, we ascertained that endogenous BM cells, recruited into the brain by CCL5, induced microglial activation. Additionally, we observed that neprilysin and interleukin-4 derived from the alternative microglia were associated with a reduction in Aβ deposition and memory impairment in AD mice. These results suggest that the beneficial effects observed in AD mice after intracerebral SC transplantation may be explained by alternative microglia activation. The recruitment of the alternative microglia into the brain is driven by CCL5 secretion from the transplanted BM-MSCs, which itself is induced by Aβ deposition in the AD brain.
Collapse
Affiliation(s)
- Jong Kil Lee
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu, South Korea
| | | | | | | |
Collapse
|
1242
|
Moll G, Rasmusson-Duprez I, von Bahr L, Connolly-Andersen AM, Elgue G, Funke L, Hamad OA, Lönnies H, Magnusson PU, Sanchez J, Teramura Y, Nilsson-Ekdahl K, Ringdén O, Korsgren O, Nilsson B, Le Blanc K. Are therapeutic human mesenchymal stromal cells compatible with human blood? Stem Cells 2012; 30:1565-74. [PMID: 22522999 DOI: 10.1002/stem.1111] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multipotent mesenchymal stromal cells (MSCs) are tested in numerous clinical trials. Questions have been raised concerning fate and function of these therapeutic cells after systemic infusion. We therefore asked whether culture-expanded human MSCs elicit an innate immune attack, termed instant blood-mediated inflammatory reaction (IBMIR), which has previously been shown to compromise the survival and function of systemically infused islet cells and hepatocytes. We found that MSCs expressed hemostatic regulators similar to those produced by endothelial cells but displayed higher amounts of prothrombotic tissue/stromal factors on their surface, which triggered the IBMIR after blood exposure, as characterized by formation of blood activation markers. This process was dependent on the cell dose, the choice of MSC donor, and particularly the cell-passage number. Short-term expanded MSCs triggered only weak blood responses in vitro, whereas extended culture and coculture with activated lymphocytes increased their prothrombotic properties. After systemic infusion to patients, we found increased formation of blood activation markers, but no formation of hyperfibrinolysis marker D-dimer or acute-phase reactants with the currently applied dose of 1.0-3.0 × 10(6) cells per kilogram. Culture-expanded MSCs trigger the IBMIR in vitro and in vivo. Induction of IBMIR is dose-dependent and increases after prolonged ex vivo expansion. Currently applied doses of low-passage clinical-grade MSCs elicit only minor systemic effects, but higher cell doses and particularly higher passage cells should be handled with care. This deleterious reaction can compromise the survival, engraftment, and function of these therapeutic cells.
Collapse
Affiliation(s)
- Guido Moll
- Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1243
|
Trophic actions of bone marrow-derived mesenchymal stromal cells for muscle repair/regeneration. Cells 2012; 1:832-50. [PMID: 24710532 PMCID: PMC3901134 DOI: 10.3390/cells1040832] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 09/28/2012] [Accepted: 10/09/2012] [Indexed: 12/30/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) represent the leading candidate cell in tissue engineering and regenerative medicine. These cells can be easily isolated, expanded in vitro and are capable of providing significant functional benefits after implantation in the damaged muscle tissues. Despite their plasticity, the participation of BM-MSCs to new muscle fiber formation is controversial; in fact, emerging evidence indicates that their therapeutic effects occur without signs of long-term tissue engraftment and involve the paracrine secretion of cytokines and growth factors with multiple effects on the injured tissue, including modulation of inflammation and immune reaction, positive extracellular matrix (ECM) remodeling, angiogenesis and protection from apoptosis. Recently, a new role for BM-MSCs in the stimulation of muscle progenitor cells proliferation has been demonstrated, suggesting the potential ability of these cells to influence the fate of local stem cells and augment the endogenous mechanisms of repair/regeneration in the damaged tissues.
Collapse
|
1244
|
Abstract
Research into mesenchymal stromal/stem cells (MSCs) has been particularly exciting in the past five years. Our understanding of mechanisms of MSC-mediated tissue regeneration has undergone considerable evolution. Recent investigation of the primary in situ counterpart of cultured MSCs has led to fresh insights into MSC physiology and its role in the immune system. At the same time, the clinical application of MSCs continues to increase markedly. Taken together, a reappraisal of the definition of MSCs, a review of current research directions, and a reassessment of the approach to clinical investigation are timely and prudent.
Collapse
Affiliation(s)
- Armand Keating
- Cell Therapy Program, Princess Margaret Hospital, Toronto, ON M5G 2M9, Canada.
| |
Collapse
|
1245
|
Yannarelli G, Dayan V, Pacienza N, Lee CJ, Medin J, Keating A. Human umbilical cord perivascular cells exhibit enhanced cardiomyocyte reprogramming and cardiac function after experimental acute myocardial infarction. Cell Transplant 2012; 22:1651-66. [PMID: 23043977 DOI: 10.3727/096368912x657675] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We were interested in evaluating the ability of the mesenchymal stromal cell (MSC) population, human umbilical cord perivascular cells (HUCPVCs), to undergo cardiomyocyte reprogramming in an established coculture system with rat embryonic cardiomyocytes. Results were compared with human bone marrow-derived (BM) MSCs. The transcription factors GATA4 and Mef 2c were expressed in HUCPVCs but not BM-MSCs at baseline and, at 7 days, increased 7.6- and 3.5-fold, respectively, compared with BM-MSCs. Although cardiac-specific gene expression increased in both cell types in coculture, upregulation was more significant in HUCPVCs, consistent with Mef 2c-GATA4 synergism. Using a lentivector with eGFP transcribed from the α-myosin heavy chain (α-MHC) promoter, we found that cardiac gene expression was greater in HUCPVCs than BM-MSCs after 14 days coculture (52±17% vs. 29±6%, respectively). A higher frequency of HUCPVCs expressed α-MHC protein compared with BM-MSCs (11.6±0.9% vs. 5.3±0.3%); however, both cell types retained MSC-associated determinants. We also assessed the ability of the MSC types to mediate cardiac regeneration in a NOD/SCID γ mouse model of acute myocardial infarction (AMI). Fourteen days after AMI, cardiac function was significantly better in cell-treated mice compared with control animals and HUCPVCs exhibited greater improvement. Although human cells persisted in the infarct area, the frequency of α-MHC expression was low. Our results indicate that HUCPVCs exhibit a greater degree of cardiomyocyte reprogramming but that differentiation for both cell types is partial. We conclude that HUCPVCs may be preferable to BM-MSCs in the cell therapy of AMI.
Collapse
Affiliation(s)
- Gustavo Yannarelli
- Cell Therapy Program, Prince Margaret Hospital, University Health Network, Toronto, ON, Canada M5G2M9.
| | | | | | | | | | | |
Collapse
|
1246
|
Payne NL, Sun G, McDonald C, Layton D, Moussa L, Emerson-Webber A, Veron N, Siatskas C, Herszfeld D, Price J, Bernard CCA. Distinct immunomodulatory and migratory mechanisms underpin the therapeutic potential of human mesenchymal stem cells in autoimmune demyelination. Cell Transplant 2012; 22:1409-25. [PMID: 23057962 DOI: 10.3727/096368912x657620] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are efficacious in a variety of intractable diseases. While bone marrow (BM)-derived MSCs (BM-MSCs) have been widely investigated, MSCs from other tissue sources have also been shown to be effective in several autoimmune and inflammatory disorders. In the present study, we simultaneously assessed the therapeutic efficacy of human BM-MSCs, as well as MSCs isolated from adipose tissue (Ad-MSCs) and umbilical cord Wharton's jelly (UC-MSCs), in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). Prior to in vivo experiments, we characterized the phenotype and function of all three MSC types. We show that BM-MSCs were more efficient at suppressing the in vitro proliferation of mitogen or antigen-stimulated T-cell responses compared to Ad-MSCs and UC-MSCs. Notably BM-MSCs induced the differential expression of cytokines from normal and stimulated T-cells. Paradoxically, intravenous transplantation of BM-MSCs into C57Bl/6 mice with chronic progressive EAE had a negligible effect on the disease course, even when multiple MSC injections were administered over a number of time points. In contrast, Ad-MSCs had the most significant impact on clinical and pathological disease outcomes in chronic progressive and relapsing-remitting EAE models. In vivo tracking studies revealed that Ad-MSCs were able to migrate to the central nervous system (CNS), a property that most likely correlated with their broader expression of homing molecules, while BM-MSCs were not detected in this anatomic region. Collectively, this comparative investigation demonstrates that transplanted Ad-MSCs play a significant role in tissue repair processes by virtue of their ability to suppress inflammation coupled with their enhanced ability to home to the injured CNS. Given the access and relatively ease for harvesting adipose tissue, these data further implicate Ad-MSCs as a cell therapeutic that may be used to treat MS patients.
Collapse
Affiliation(s)
- Natalie L Payne
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1247
|
Horie M, Choi H, Lee RH, Reger RL, Ylostalo J, Muneta T, Sekiya IC, Prockop DJ. Intra-articular injection of human mesenchymal stem cells (MSCs) promote rat meniscal regeneration by being activated to express Indian hedgehog that enhances expression of type II collagen. Osteoarthritis Cartilage 2012; 20:1197-207. [PMID: 22750747 PMCID: PMC3788634 DOI: 10.1016/j.joca.2012.06.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 06/18/2012] [Accepted: 06/20/2012] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Meniscal regeneration was previously shown to be enhanced by injection of mesenchymal stem/stromal cells (MSCs) but the mode of action of the MSCs was not established. The aim of this study was to define how injection of MSCs enhances meniscal regeneration. DESIGN A hemi-meniscectomy model in rats was used. Rat-MSCs (rMSCs) or human-MSCs (hMSCs) were injected into the right knee joint after the surgery, and PBS was injected into the left. The groups were compared macroscopically and histologically at 2, 4, and 8 weeks. The changes in transcription in both human and rat genes were assayed by species-specific microarrays and real-time RT-PCRs. RESULTS Although the number of hMSCs decreased with time, hMSCs enhanced meniscal regeneration in a manner similar to rMSCs. hMSCs injection increased expression of rat type II collagen (rat-Col II), and inhibited osteoarthritis progression. The small fraction of hMSCs was activated to express high levels of a series of genes including Indian hedgehog (Ihh), parathyroid hormone-like hormone (PTHLH), and bone morphogenetic protein 2 (BMP2). The presence of hMSCs triggered the subsequent expression of rat-Col II. An antagonist of hedgehog signaling inhibited the expression of rat-Col II and an agonist increased expression of rat-Col II in the absence of hMSCs. CONCLUSIONS Despite rapid reduction in cell numbers, intra-articular injected hMSCs were activated to express Ihh, PTHLH, and BMP2 and contributed to meniscal regeneration. The hedgehog signaling was essential in enhancing the expression of rat-Col II, but several other factors provided by the hMSCs probably contributed to the repair.
Collapse
Affiliation(s)
- Masafumi Horie
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas 76502
- Section of Orthopedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hosoon Choi
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas 76502
| | - Ryang Hwa Lee
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas 76502
| | - Roxanne L. Reger
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas 76502
| | - Joni Ylostalo
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas 76502
| | - Takeshi Muneta
- Section of Orthopedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - I chiro Sekiya
- Section of Cartilage Regeneration, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Darwin J. Prockop
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas 76502
| |
Collapse
|
1248
|
Reagan MR, Seib FP, McMillin DW, Sage EK, Mitsiades CS, Janes SM, Ghobrial IM, Kaplan DL. Stem Cell Implants for Cancer Therapy: TRAIL-Expressing Mesenchymal Stem Cells Target Cancer Cells In Situ. J Breast Cancer 2012; 15:273-82. [PMID: 23091539 PMCID: PMC3468780 DOI: 10.4048/jbc.2012.15.3.273] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 07/17/2012] [Indexed: 11/30/2022] Open
Abstract
Purpose Tumor-specific delivery of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), an apoptosis-inducing peptide, at effective doses remains challenging. Herein we demonstrate the utility of a scaffold-based delivery system for sustained therapeutic cell release that capitalizes on the tumor-homing properties of mesenchymal stem cells (MSCs) and their ability to express genetically-introduced therapeutic genes. Methods Implants were formed from porous, biocompatible silk scaffolds seeded with full length TRAIL-expressing MSCs (FLT-MSCs). under a doxycycline inducible promoter. In vitro studies with FLT-MSCs demonstrated TRAIL expression and antitumor effects on breast cancer cells. Next, FLT-MSCs were administered to mice using three administration routes (mammary fat pad co-injections, tail vein injections, and subcutaneous implantation on scaffolds). Results In vitro cell-specific bioluminescent imaging measured tumor cell specific growth in the presence of stromal cells and demonstrated FLT-MSC inhibition of breast cancer growth. FLT-MSC implants successfully decreased bone and lung metastasis, whereas liver metastasis decreased only with tail vein and co-injection administration routes. Average tumor burden was decreased when doxycycline was used to induce TRAIL expression for co-injection and scaffold groups, as compared to controls with no induced TRAIL expression. Conclusion This implant-based therapeutic delivery system is an effective and completely novel method of anticancer therapy and holds great potential for clinical applications.
Collapse
Affiliation(s)
- Michaela R Reagan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA. ; Department of Medicine, Harvard Medical School, Boston, USA. ; Department of Biomedical Engineering, Tufts University, Medford, USA
| | | | | | | | | | | | | | | |
Collapse
|
1249
|
Prockop DJ, Oh JY. Medical therapies with adult stem/progenitor cells (MSCs): a backward journey from dramatic results in vivo to the cellular and molecular explanations. J Cell Biochem 2012; 113:1460-9. [PMID: 22213121 DOI: 10.1002/jcb.24046] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
There is currently great interest in the use of mesenchymal stem/stromal cells (MSCs) for the therapy of many diseases of animals and humans. However, we are still left with the serious challenges in explaining the beneficial effects of the cells. Hence, it is essential to work backward from dramatic results obtained in vivo to the cellular and molecular explanations in order to discover the secrets of MSCs. This review will focus on recent data that have changed the paradigms for understanding the therapeutic potentials of MSCs.
Collapse
Affiliation(s)
- Darwin J Prockop
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas 76502, USA.
| | | |
Collapse
|
1250
|
Reekmans K, Praet J, De Vocht N, Daans J, Van der Linden A, Berneman Z, Ponsaerts P. Stem cell therapy for multiple sclerosis: preclinical evidence beyond all doubt? Regen Med 2012; 7:245-59. [PMID: 22397612 DOI: 10.2217/rme.12.5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stem cell transplantation holds great promise for restoration of neural function in various neurodegenerative disorders, including multiple sclerosis (MS). However, many questions remain regarding the true efficacy and precise mode of action of stem cell-based therapeutic approaches. Therefore, in this article, we will first discuss the ideal route and/or timing of stem cell-based therapies for experimental autoimmune encephalomyelitis (EAE), the most used preclinical animal model for MS. Next, we will provide an overview of the proposed mechanisms that contribute to the beneficial effects of stem cell transplantation observed during the treatment of rodent EAE. Reviews of current and past literature clearly demonstrate conceptual changes in the development of stem cell-based approaches for EAE/MS, leading to the identification of several major challenges to be tackled before (stem) cell therapy for rodent EAE can be safely and successfully translated to human therapy for MS.
Collapse
Affiliation(s)
- Kristien Reekmans
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | | | | | | | | | | | | |
Collapse
|