51
|
Cooper AE, Cho JH, Menges S, Masood S, Xie J, Yang J, Klassen H. Immunosuppressive Treatment Can Alter Visual Performance in the Royal College of Surgeons Rat. J Ocul Pharmacol Ther 2016; 32:296-303. [PMID: 27008099 PMCID: PMC6453503 DOI: 10.1089/jop.2015.0134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/21/2016] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Immunosuppression is frequently employed to enhance survival of xenografted human cells as part of translational proof-of-concept studies. However, the potential effects of this treatment are easily overlooked. METHODS As part of baseline testing in the dark-eyed variant of the dystrophic Royal College of Surgeons (RCS) rat, we documented the time course of retinal degenerative changes versus Long Evans controls using bright field retinal imaging, fluorescein angiography, and histology and examined the impact of immunosuppression on visual function. Rats received either no treatment or systemic immunosuppression with oral cyclosporine A and injectable dexamethasone and subsequently underwent functional evaluation by optomotor response testing and electroretinography (ERG) at multiple intervals from P45 to P180. RESULTS Immunosuppressed RCS animals demonstrated poorer performance on functional tests than age-matched untreated rats during the earlier stages of degeneration, including significantly lower spatial acuities on optomotor threshold testing and significantly lower b-wave amplitudes on scotopic and photopic ERGs. Retinal imaging documented the progression of degenerative changes in the RCS fundus and histologic evaluation of the RCS retina confirmed progressive thinning of the outer nuclear layer. CONCLUSIONS A standard regimen of cyclosporine A plus dexamethasone, administered to RCS rats, results in demonstrable systemic side effects and depressed scores on behavioral and electrophysiological testing at time points before P90. The source of the functional impairment was not identified. This finding has implications for the interpretation of data generated using this commonly used translational model.
Collapse
Affiliation(s)
- Ann E. Cooper
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, Davis, California
| | - Jang-Hyeon Cho
- Gavin Herbert Eye Institute, Stem Cell Research Center, University of California, Irvine, California
| | - Steven Menges
- Gavin Herbert Eye Institute, Stem Cell Research Center, University of California, Irvine, California
| | - Sahar Masood
- Gavin Herbert Eye Institute, Stem Cell Research Center, University of California, Irvine, California
| | - Jun Xie
- Gavin Herbert Eye Institute, Stem Cell Research Center, University of California, Irvine, California
| | - Jing Yang
- Gavin Herbert Eye Institute, Stem Cell Research Center, University of California, Irvine, California
| | - Henry Klassen
- Gavin Herbert Eye Institute, Stem Cell Research Center, University of California, Irvine, California
| |
Collapse
|
52
|
Gene expression changes in the retina following subretinal injection of human neural progenitor cells into a rodent model for retinal degeneration. Mol Vis 2016; 22:472-90. [PMID: 27217715 PMCID: PMC4872275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/13/2016] [Indexed: 12/03/2022] Open
Abstract
PURPOSE Retinal degenerative diseases (RDDs) affect millions of people and are the leading cause of vision loss. Although treatment options for RDDs are limited, stem and progenitor cell-based therapies have great potential to halt or slow the progression of vision loss. Our previous studies have shown that a single subretinal injection of human forebrain derived neural progenitor cells (hNPCs) into the Royal College of Surgeons (RCS) retinal degenerate rat offers long-term preservation of photoreceptors and visual function. Furthermore, neural progenitor cells are currently in clinical trials for treating age-related macular degeneration; however, the molecular mechanisms of stem cell-based therapies are largely unknown. This is the first study to analyze gene expression changes in the retina of RCS rats following subretinal injection of hNPCs using high-throughput sequencing. METHODS RNA-seq data of retinas from RCS rats injected with hNPCs (RCS(hNPCs)) were compared to sham surgery in RCS (RCS(sham)) and wild-type Long Evans (LE(sham)) rats. Differential gene expression patterns were determined with in silico analysis and confirmed with qRT-PCR. Function, biologic, cellular component, and pathway analyses were performed on differentially expressed genes and investigated with immunofluorescent staining experiments. RESULTS Analysis of the gene expression data sets identified 1,215 genes that were differentially expressed between RCS(sham) and LE(sham) samples. Additionally, 283 genes were differentially expressed between the RCS(hNPCs) and RCS(sham) samples. Comparison of these two gene sets identified 68 genes with inverse expression (termed rescue genes), including Pdc, Rp1, and Cdc42ep5. Functional, biologic, and cellular component analyses indicate that the immune response is enhanced in RCS(sham). Pathway analysis of the differential expression gene sets identified three affected pathways in RCS(hNPCs), which all play roles in phagocytosis signaling. Immunofluorescent staining detected the increased presence of macrophages and microglia in RCS(sham) retinas, which decreased in RCS(hNPCs) retinas similar to the patterns detected in LE(sham). CONCLUSIONS The results from this study provide evidence of the gene expression changes that occur following treatment with hNPCs in the degenerating retina. This information can be used in future studies to potentially enhance or predict responses to hNPC and other stem cell therapies for retinal degenerative diseases.
Collapse
|
53
|
Stem Cell Therapy for Treatment of Ocular Disorders. Stem Cells Int 2016; 2016:8304879. [PMID: 27293447 PMCID: PMC4884591 DOI: 10.1155/2016/8304879] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/10/2016] [Indexed: 12/30/2022] Open
Abstract
Sustenance of visual function is the ultimate focus of ophthalmologists. Failure of complete recovery of visual function and complications that follow conventional treatments have shifted search to a new form of therapy using stem cells. Stem cell progenitors play a major role in replenishing degenerated cells despite being present in low quantity and quiescence in our body. Unlike other tissues and cells, regeneration of new optic cells responsible for visual function is rarely observed. Understanding the transcription factors and genes responsible for optic cells development will assist scientists in formulating a strategy to activate and direct stem cells renewal and differentiation. We review the processes of human eye development and address the strategies that have been exploited in an effort to regain visual function in the preclinical and clinical state. The update of clinical findings of patients receiving stem cell treatment is also presented.
Collapse
|
54
|
Hong HS, Kim S, Nam S, Um J, Kim YH, Son Y. Effect of substance P on recovery from laser-induced retinal degeneration. Wound Repair Regen 2016; 23:268-77. [PMID: 25682893 DOI: 10.1111/wrr.12264] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/13/2015] [Accepted: 01/26/2015] [Indexed: 12/16/2022]
Abstract
Retinal degeneration is caused by neovascularization and persistent inflammation in the retinal pigment epithelium (RPE) and choroid, and causes serious eye disease including age-related macular degeneration (AMD). Thus, inhibiting inflammation and neovascularization may be a primary approach to protect the retina from degeneration. The purpose of this study was to determine whether substance P (SP), which can suppress inflammation and mobilize stem cells, can protect the RPE from degeneration. The effect of SP was evaluated by analyzing systemic inflammation, cell survival, and neovascularization within the argon laser-injured retina of mice. At 1 week postinjury, the SP-treated group had lower tumor necrosis factor-alpha and higher interleukin-10 serum concentrations, and a more intact retinal structure compared to the vehicle-treated group. In mice administered SP repeatedly for 4 weeks, the retinal structure appeared normal and showed sparse neovascularization, whereas the vehicle-treated group showed severe retinal destruction and dense neovascularization. Moreover, the efficacy of SP was identical to that of mesenchymal stem cells that were transplanted into the vitreous after retinal injury. This study highlights the potential for the endogenous neuropeptide SP as a treatment for retinal damage to prevent conditions such as AMD.
Collapse
Affiliation(s)
- Hyun Sook Hong
- College of Medicine/ East-West Medical Research Institute, Kyung Hee University
| | - Suna Kim
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University
| | - Seungwoo Nam
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University
| | - Jihyun Um
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University
| | - Yeong Hoon Kim
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, St. Paul's Hospital, Seoul, Korea
| | - Youngsook Son
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University
| |
Collapse
|
55
|
Abstract
Diabetes is one of the main economic burdens in health care, which threatens to worsen dramatically if prevalence forecasts are correct. What makes diabetes harmful is the multi-organ distribution of its microvascular and macrovascular complications. Regenerative medicine with cellular therapy could be the dam against life-threatening or life-altering complications. Bone marrow-derived stem cells are putative candidates to achieve this goal. Unfortunately, the bone marrow itself is affected by diabetes, as it can develop a microangiopathy and neuropathy similar to other body tissues. Neuropathy leads to impaired stem cell mobilization from marrow, the so-called mobilopathy. Here, we review the role of bone marrow-derived stem cells in diabetes: how they are affected by compromised bone marrow integrity, how they contribute to other diabetic complications, and how they can be used as a treatment for these. Eventually, we suggest new tactics to optimize stem cell therapy.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS28HW UK
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS28HW UK
| |
Collapse
|
56
|
Bakondi B, Lv W, Lu B, Jones MK, Tsai Y, Kim KJ, Levy R, Akhtar AA, Breunig JJ, Svendsen CN, Wang S. In Vivo CRISPR/Cas9 Gene Editing Corrects Retinal Dystrophy in the S334ter-3 Rat Model of Autosomal Dominant Retinitis Pigmentosa. Mol Ther 2016; 24:556-63. [PMID: 26666451 PMCID: PMC4786918 DOI: 10.1038/mt.2015.220] [Citation(s) in RCA: 225] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/05/2015] [Indexed: 11/09/2022] Open
Abstract
Reliable genome editing via Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/Cas9 may provide a means to correct inherited diseases in patients. As proof of principle, we show that CRISPR/Cas9 can be used in vivo to selectively ablate the rhodopsin gene carrying the dominant S334ter mutation (Rho(S334)) in rats that model severe autosomal dominant retinitis pigmentosa. A single subretinal injection of guide RNA/Cas9 plasmid in combination with electroporation generated allele-specific disruption of Rho(S334), which prevented retinal degeneration and improved visual function.
Collapse
Affiliation(s)
- Benjamin Bakondi
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Wenjian Lv
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Current address: Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Bin Lu
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Melissa K Jones
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yuchun Tsai
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kevin J Kim
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Rachelle Levy
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shaomei Wang
- Board of Governors Regenerative Medicine Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
57
|
Coulson-Thomas VJ, Coulson-Thomas YM, Gesteira TF, Kao WWY. Extrinsic and Intrinsic Mechanisms by Which Mesenchymal Stem Cells Suppress the Immune System. Ocul Surf 2016; 14:121-34. [PMID: 26804815 DOI: 10.1016/j.jtos.2015.11.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 11/12/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are a group of fibroblast-like multipotent mesenchymal stromal cells that have the ability to differentiate into osteoblasts, adipocytes, and chondrocytes. Recent studies have demonstrated that MSCs possess a unique ability to exert suppressive and regulatory effects on both adaptive and innate immunity in an autologous and allogeneic manner. A vital step in stem cell transplantation is overcoming the potential graft-versus-host disease, which is a limiting factor to transplantation success. Given that MSCs attain powerful differentiation capabilities and also present immunosuppressive properties, which enable them to survive host immune rejection, MSCs are of great interest. Due to their ability to differentiate into different cell types and to suppress and modulate the immune system, MSCs are being developed for treating a plethora of diseases, including immune disorders. Moreover, in recent years, MSCs have been genetically engineered to treat and sometimes even cure some diseases, and the use of MSCs for cell therapy presents new perspectives for overcoming tissue rejection. In this review, we discuss the potential extrinsic and intrinsic mechanisms that underlie MSCs' unique ability to modulate inflammation, and both innate and adaptive immunity.
Collapse
Affiliation(s)
- Vivien J Coulson-Thomas
- Department of Ophthalmology, University of Cincinnati, Ohio, USA; John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK.
| | | | | | - Winston W-Y Kao
- Department of Ophthalmology, University of Cincinnati, Ohio, USA.
| |
Collapse
|
58
|
Stem Cell Imaging: Tools to Improve Cell Delivery and Viability. Stem Cells Int 2016; 2016:9240652. [PMID: 26880997 PMCID: PMC4736428 DOI: 10.1155/2016/9240652] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/06/2015] [Accepted: 10/15/2015] [Indexed: 01/01/2023] Open
Abstract
Stem cell therapy (SCT) has shown very promising preclinical results in a variety of regenerative medicine applications. Nevertheless, the complete utility of this technology remains unrealized. Imaging is a potent tool used in multiple stages of SCT and this review describes the role that imaging plays in cell harvest, cell purification, and cell implantation, as well as a discussion of how imaging can be used to assess outcome in SCT. We close with some perspective on potential growth in the field.
Collapse
|
59
|
Kramerov AA, Ljubimov AV. Stem cell therapies in the treatment of diabetic retinopathy and keratopathy. Exp Biol Med (Maywood) 2015; 241:559-68. [PMID: 26454200 DOI: 10.1177/1535370215609692] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Nonproliferative diabetic retinopathy (DR) is characterized by multiple degenerative changes that could be potentially corrected by stem cell therapies. Most studies so far have attempted to alleviate typical abnormalities of early retinopathy, including vascular hyperpermeability, capillary closure and pericyte dropout. Success was reported with adult stem cells (vascular progenitors or adipose stem cells), as well as induced pluripotent stem cells from cord blood. The cells were able to associate with damaged vessels in both pericyte and endothelial lining positions in models of DR and ischemia-reperfusion. In some diabetic models, functional amelioration of vasculature and electroretinograms was noted. Another approach for endogenous progenitor cell therapy is to normalize dysfunctional diabetic bone marrow and residing endothelial progenitors using NO donors, PPAR-δ and -γ agonists, or inhibition of TGF-β. A potentially important strategy would be to reduce neuropathy by stem cell inoculations, either naïve (e.g., paracrine-acting adipose stem cells) or secreting specific neuroprotectants, such as ciliary neurotrophic factor or brain-derived neurotrophic factor that showed benefit in amyotrophic lateral sclerosis and Parkinson's disease. Recent advances in stem cell therapies for diabetic retinal microangiopathy may form the basis of first clinical trials in the near future. Additionally, stem cell therapies may prove beneficial for diabetic corneal disease (diabetic keratopathy) with pronounced epithelial stem cell dysfunction.
Collapse
Affiliation(s)
- Andrei A Kramerov
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Alexander V Ljubimov
- Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
60
|
Stem cell based therapies for age-related macular degeneration: The promises and the challenges. Prog Retin Eye Res 2015; 48:1-39. [PMID: 26113213 DOI: 10.1016/j.preteyeres.2015.06.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 12/21/2022]
|
61
|
Nafissi N, Foldvari M. Neuroprotective therapies in glaucoma: I. Neurotrophic factor delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:240-54. [PMID: 26306832 DOI: 10.1002/wnan.1361] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 06/15/2015] [Accepted: 07/04/2015] [Indexed: 12/11/2022]
Abstract
Glaucoma is a neurodegenerative eye disease that causes permanent blindness at the progressive stage and the number of people affected worldwide is expected to reach over 79 million by 2020. Currently, glaucoma management relies on pharmacological and invasive surgical treatments mainly by reducing the intraocular pressure (IOP), which is the most important risk factor for the progression of the visual field loss. Recent research suggests that neuroprotective or neuroregenerative approaches are necessary to prevent retinal ganglion cells (RGCs) loss and visual impairment over time. Neuroprotection is a new therapeutic strategy that attempts to keep RGCs alive and functional. New gene and cell therapeutics encoding neurotrophic factors (NTFs) are emerging for both neuroprotection and regenerative treatments for retinal diseases. This article briefly reviews the role of NTFs in glaucoma and the potential delivery systems.
Collapse
Affiliation(s)
- Nafiseh Nafissi
- School of Pharmacy and Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada
| | - Marianna Foldvari
- School of Pharmacy and Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
62
|
Roubeix C, Denoyer A, Brignole-Baudouin F, Baudouin C. [Mesenchymal stem cell therapy, a new hope for eye disease]. J Fr Ophtalmol 2015. [PMID: 26215486 DOI: 10.1016/j.jfo.2015.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Mesenchymal stem cells (MSC) are adult stem cells, first identified in skeletal tissues and then found in the entire body. MSC are able to not only differentiate into specialized cells within skeletal tissue - chondrocytes, osteocytes, adipocytes and fibroblasts - but also secrete a large range of soluble mediators defining their secretome and allowing their interaction with a number of cell protagonists. Thus, in a general sense, MSC are involved in tissue homeostasis through their secretome and are specifically responsible for cell turn-over in skeletal tissues. For a decade and a half, safety and efficiency of MSC has led to the development of many clinical trials in various fields. However, results were often disappointing, probably because of difficulties in methods and evaluation. At a time when the first clinical trials using MSC are emerging in ophthalmology, the goal of this literature review is to gather and put into perspective preclinical and clinical results in order to better predict the future of this innovative therapeutic pathway.
Collapse
Affiliation(s)
- C Roubeix
- Inserm, U968, 75012 Paris, France; UMR_S 968, institut de la vision, UPMC université Paris 06, 17, rue Moreau, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France.
| | - A Denoyer
- Inserm, U968, 75012 Paris, France; UMR_S 968, institut de la vision, UPMC université Paris 06, 17, rue Moreau, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France; Inserm-DHOS CIC 503, centre hospitalier national d'ophtalmologie des Quinze-Vingts, 75012 Paris, France
| | - F Brignole-Baudouin
- Inserm, U968, 75012 Paris, France; UMR_S 968, institut de la vision, UPMC université Paris 06, 17, rue Moreau, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France; Inserm-DHOS CIC 503, centre hospitalier national d'ophtalmologie des Quinze-Vingts, 75012 Paris, France; Faculté de pharmacie de Paris, université Paris Descartes, Sorbonne Paris-Cité, 75006 Paris, France
| | - C Baudouin
- Inserm, U968, 75012 Paris, France; UMR_S 968, institut de la vision, UPMC université Paris 06, 17, rue Moreau, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France; Inserm-DHOS CIC 503, centre hospitalier national d'ophtalmologie des Quinze-Vingts, 75012 Paris, France; Service d'ophtalmologie, hôpital Ambroise-Paré, AP-HP, 92100 Boulogne, France; Université Versailles-Saint-Quentin-en-Yvelines, 78180 Montigny-le-Bretonneux, France
| |
Collapse
|
63
|
Buzhor E, Leshansky L, Blumenthal J, Barash H, Warshawsky D, Mazor Y, Shtrichman R. Cell-based therapy approaches: the hope for incurable diseases. Regen Med 2015; 9:649-72. [PMID: 25372080 DOI: 10.2217/rme.14.35] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cell therapies aim to repair the mechanisms underlying disease initiation and progression, achieved through trophic effect or by cell replacement. Multiple cell types can be utilized in such therapies, including stem, progenitor or primary cells. This review covers the current state of cell therapies designed for the prominent disorders, including cardiovascular, neurological (Parkinson's disease, amyotrophic lateral sclerosis, stroke, spinal cord injury), autoimmune (Type 1 diabetes, multiple sclerosis, Crohn's disease), ophthalmologic, renal, liver and skeletal (osteoarthritis) diseases. Various cell therapies have reached advanced clinical trial phases with potential marketing approvals in the near future, many of which are based on mesenchymal stem cells. Advances in pluripotent stem cell research hold great promise for regenerative medicine. The information presented in this review is based on the analysis of the cell therapy collection detailed in LifeMap Discovery(®) (LifeMap Sciences Inc., USA) the database of embryonic development, stem cell research and regenerative medicine.
Collapse
|
64
|
Psychophysical testing in rodent models of glaucomatous optic neuropathy. Exp Eye Res 2015; 141:154-63. [PMID: 26144667 DOI: 10.1016/j.exer.2015.06.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 06/08/2015] [Accepted: 06/29/2015] [Indexed: 12/14/2022]
Abstract
Processing of visual information begins in the retina, with photoreceptors converting light stimuli into neural signals. Ultimately, signals are transmitted to the brain through signaling networks formed by interneurons, namely bipolar, horizontal and amacrine cells providing input to retinal ganglion cells (RGCs), which form the optic nerve with their axons. As part of the chronic nature of glaucomatous optic neuropathy, the increasing and irreversible damage and ultimately loss of neurons, RGCs in particular, occurs following progressive damage to the optic nerve head (ONH), eventually resulting in visual impairment and visual field loss. There are two behavioral assays that are typically used to assess visual deficits in glaucoma rodent models, the visual water task and the optokinetic drum. The visual water task can assess an animal's ability to distinguish grating patterns that are associated with an escape from water. The optokinetic drum relies on the optomotor response, a reflex turning of the head and neck in the direction of the visual stimuli, which usually consists of rotating black and white gratings. This reflex is a physiological response critical for keeping the image stable on the retina. Driven initially by the neuronal input from direction-selective RGCs, this reflex is comprised of a number of critical sensory and motor elements. In the presence of repeatable and defined stimuli, this reflex is extremely well suited to analyze subtle changes in the circuitry and performance of retinal neurons. Increasing the cycles of these alternating gratings per degree, or gradually reducing the contrast of the visual stimuli, threshold levels can be determined at which the animal is no longer tracking the stimuli, and thereby visual function of the animal can be determined non-invasively. Integrating these assays into an array of outcome measures that determine multiple aspects of visual function is a central goal in vision research and can be realized, for example, by the combination of measuring optomotor reflex function with electroretinograms (ERGs) and optical coherence tomography (OCT) of the retina. These structure-function correlations in vivo are urgently needed to identify disease mechanisms as potential new targets for drug development. Such a combination of the experimental assessment of the optokinetic reflex (OKR) or optomotor response (OMR) with other measures of retinal structure and function is especially valuable for research on GON. The chronic progression of the disease is characterized by a gradual decrease in function accompanied by a concomitant increase in structural damage to the retina, therefore the assessment of subtle changes is key to determining the success of novel intervention strategies.
Collapse
|
65
|
Safety and Efficacy of Human Wharton's Jelly-Derived Mesenchymal Stem Cells Therapy for Retinal Degeneration. PLoS One 2015; 10:e0128973. [PMID: 26107378 PMCID: PMC4479609 DOI: 10.1371/journal.pone.0128973] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/04/2015] [Indexed: 01/01/2023] Open
Abstract
Purpose To investigate the safety and efficacy of subretinal injection of human Wharton’s Jelly-derived mesenchymal stem cells (hWJ-MSCs) on retinal structure and function in Royal College of Surgeons (RCS) rats. Methods RCS rats were divided into 2 groups: hWJ-MSCs treated group (n = 8) and placebo control group (n = 8). In the treatment group, hWJ-MSCs from healthy donors were injected into the subretinal space in one eye of each rat at day 21. Control group received saline injection of the same volume. Additional 3 animals were injected with nanogold-labelled stem cells for in vivo tracking of cells localisation using a micro-computed tomography (microCT). Retinal function was assessed by electroretinography (ERG) 3 days before the injection and repeated at days 15, 30 and 70 after the injection. Eyes were collected at day 70 for histology, cellular and molecular studies. Results No retinal tumor formation was detected by histology during the study period. MicroCT scans showed that hWJ-MSCs stayed localised in the eye with no systemic migration. Transmission electron microscopy showed that nanogold-labelled cells were located within the subretinal space. Histology showed preservation of the outer nuclear layer (ONL) in the treated group but not in the control group. However, there were no significant differences in the ERG responses between the groups. Confocal microscopy showed evidence of hWJ-MSCs expressing markers for photoreceptor, Müller cells and bipolar cells. Conclusions Subretinal injection of hWJ-MSCs delay the loss of the ONL in RCS rats. hWJ-MSCs appears to be safe and has potential to differentiate into retinal-like cells. The potential of this cell-based therapy for the treatment of retinal dystrophies warrants further studies.
Collapse
|
66
|
Tsai Y, Lu B, Bakondi B, Girman S, Sahabian A, Sareen D, Svendsen CN, Wang S. Human iPSC-Derived Neural Progenitors Preserve Vision in an AMD-Like Model. Stem Cells 2015; 33:2537-49. [PMID: 25869002 DOI: 10.1002/stem.2032] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/28/2015] [Indexed: 12/14/2022]
Abstract
Pluripotent stem cell-derived retinal pigment epithelial (RPE) cells are currently being tested for cell replacement in late-stage age-related macular degeneration (AMD). However, preserving vision at early-stages may also be possible. Here, we demonstrate that transplantation of neural progenitor cells (NPCs) derived from induced pluripotent stem cells (iNPCs) limits disease progression in the Royal College of Surgeons rat, a preclinical model of AMD. Grafted-iNPCs survived, remained undifferentiated, and distributed extensively in a laminar fashion in the subretinal space. Retinal pathology resulting from the accumulation of undigested photoreceptor outer segments (POS) was significantly reduced in iNPC-injected rats compared with controls. Phagosomes within grafted-iNPCs contained POS, suggesting that iNPCs had compensated for defective POS phagocytosis by host-RPE. The iNPC-treated eyes contained six to eight rows of photoreceptor nuclei that spanned up to 5 mm in length in transverse retinal sections, compared with only one row of photoreceptors in controls. iNPC treatment fully preserved visual acuity measured by optokinetic response. Electrophysiological recordings revealed that retina with the best iNPC-protected areas were 140-fold more sensitive to light stimulation than equivalent areas of contralateral eyes. The results described here support the therapeutic utility of iNPCs as autologous grafts for early-stage of AMD.
Collapse
Affiliation(s)
- Yuchun Tsai
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Bin Lu
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Benjamin Bakondi
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sergey Girman
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Anais Sahabian
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dhruv Sareen
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Clive N Svendsen
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shaomei Wang
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
67
|
Retinitis Pigmentosa Treatment with Western Medicine and Traditional Chinese Medicine Therapies. J Ophthalmol 2015; 2015:421269. [PMID: 26124961 PMCID: PMC4466478 DOI: 10.1155/2015/421269] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/15/2015] [Accepted: 02/27/2015] [Indexed: 02/05/2023] Open
Abstract
Current management of retinitis pigmentosa (RP) includes an attempt at slowing down the degenerative process through therapies that use either Western or traditional Chinese medicine (TCM). Novel therapies in Western medicine (WM) include use of tailor-made gene therapy, transplantation of stem cells, or neuroprotection treatment. TCM treatment includes two major approaches. These are orally applied herbal decoctions and acupuncture. In fact, all TCM treatments are based on the differentiation of a symptom-complex, which is the characteristic essence of TCM. Thus, diagnosed RP may be treated via the liver, the kidney, and the spleen. The principle behind these treatments is to invigorate the blood and brighten the eyes by toning up the liver and the kidney. Also treatments to cope with deficiencies in the two concepts that are unique and fundamental to TCM are considered: Qi or “vital energy” and Yin and Yang or the harmony of all the opposite elements and forces that make up existence. In particular, the Qi deficiency that results from blood stasis is addressed in these treatments. This paper also puts forward the existing problems and the prospect of the future development on integrating TCM with WM.
Collapse
|
68
|
Jankowiak W, Kruszewski K, Flachsbarth K, Skevas C, Richard G, Rüther K, Braulke T, Bartsch U. Sustained Neural Stem Cell-Based Intraocular Delivery of CNTF Attenuates Photoreceptor Loss in the nclf Mouse Model of Neuronal Ceroid Lipofuscinosis. PLoS One 2015; 10:e0127204. [PMID: 25992714 PMCID: PMC4439090 DOI: 10.1371/journal.pone.0127204] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/13/2015] [Indexed: 01/10/2023] Open
Abstract
A sustained intraocular administration of neurotrophic factors is among the strategies aimed at establishing treatments for currently untreatable degenerative retinal disorders. In the present study we have analyzed the neuroprotective effects of a continuous neural stem (NS) cell-based intraocular delivery of ciliary neurotrophic factor (CNTF) on photoreceptor cells in the nclf mouse, an animal model of the neurodegenerative lysosomal storage disorder variant late infantile neuronal ceroid lipofuscinosis (vLINCL). To this aim, we genetically modified adherently cultivated NS cells with a polycistronic lentiviral vector encoding a secretable variant of CNTF together with a Venus reporter gene (CNTF-NS cells). NS cells for control experiments (control-NS cells) were modified with a vector encoding the reporter gene tdTomato. Clonal CNTF-NS and control-NS cell lines were established using fluorescent activated cell sorting and intravitreally grafted into 14 days old nclf mice at the onset of retinal degeneration. The grafted cells preferentially differentiated into astrocytes that were attached to the posterior side of the lenses and the vitreal side of the retinas and stably expressed the transgenes for at least six weeks, the latest post-transplantation time point analyzed. Integration of donor cells into host retinas, ongoing proliferation of grafted cells or adverse effects of the donor cells on the morphology of the host eyes were not observed. Quantitative analyses of host retinas two, four and six weeks after cell transplantation revealed the presence of significantly more photoreceptor cells in eyes with grafted CNTF-NS cells than in eyes with grafted control-NS cells. This is the first demonstration that a continuous intraocular administration of a neurotrophic factor attenuates retinal degeneration in an animal model of neuronal ceroid lipofuscinosis.
Collapse
Affiliation(s)
- Wanda Jankowiak
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Kruszewski
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Flachsbarth
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christos Skevas
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gisbert Richard
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Rüther
- Department of Ophthalmology, Sankt Gertrauden-Krankenhaus, Berlin, Germany
| | - Thomas Braulke
- Department of Biochemistry, Children’s Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Bartsch
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
69
|
|
70
|
Olmos LC, Nazari H, Rodger DC, Humayun MS. Stem Cell Therapy for the Treatment of Dry Age-Related Macular Degeneration. CURRENT OPHTHALMOLOGY REPORTS 2015. [DOI: 10.1007/s40135-014-0058-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
71
|
Dang Y, Zhang C, Zhu Y. Stem cell therapies for age-related macular degeneration: the past, present, and future. Clin Interv Aging 2015; 10:255-64. [PMID: 25609937 PMCID: PMC4298283 DOI: 10.2147/cia.s73705] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In the developed world, age-related macular degeneration (AMD) is one of the major causes of irreversible blindness in the elderly. Although management of neovascular AMD (wet AMD) has dramatically progressed, there is still no effective treatment for nonneovascular AMD (dry AMD), which is characterized by retinal pigment epithelial (RPE) cell death (or dysfunction) and microenvironmental disruption in the retina. Therefore, RPE replacement and microenvironmental regulation represent viable treatments for dry AMD. Recent advances in cell biology have demonstrated that RPE cells can be easily generated from several cell types (pluripotent stem cells, multipotent stem cells, or even somatic cells) by spontaneous differentiation, coculturing, defined factors or cell reprogramming, respectively. Additionally, in vivo studies also showed that the restoration of visual function could be obtained by transplanting functional RPE cells into the subretinal space of recipient. More importantly, clinical trials approved by the US government have shown promising prospects in RPE transplantation. However, key issues such as implantation techniques, immune rejection, and xeno-free techniques are still needed to be further investigated. This review will summarize recent advances in cell transplantation for dry AMD. The obstacles and prospects in this field will also be discussed.
Collapse
Affiliation(s)
- Yalong Dang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, People's Republic of China ; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, People's Republic of China ; Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Chun Zhang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, People's Republic of China ; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, People's Republic of China
| | - Yu Zhu
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
72
|
Mu Y, Zhao M, Su G. Stem cell-based therapies for age-related macular degeneration: current status and prospects. Int J Clin Exp Med 2014; 7:3843-3852. [PMID: 25550892 PMCID: PMC4276150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 10/23/2014] [Indexed: 06/04/2023]
Abstract
Age-related macular degeneration (AMD) is one of the major causes of irreversible blindness both in developed and developing countries. During the past decades, the managements of neovascular AMD (wet AMD) have dramatically progressed. However, still no effective treatment for non-neovascular AMD (dry AMD) which was characterized by geographic macular atrophy. Recent advances in stem cell sciences have demonstrated that retinal pigment epithelium (RPE) cells can be generated from several types of stem cells (including embryonic stem cells, induced pluripotent stem cells, mesenchymal stem cells, et al) by cell co-culturing or defined factors. Additionally, studies also showed that visual function could be recovered by transplantation of these cells into subretinal space in vivo. Moreover, the United States Food and Drug Administration already approved several clinical trials to evaluate the efficiencies of stem cell based cell transplantation for dry AMD patients. Till now, a few patients enrolled in these studies achieved promising outcomes. This review will summarize recent advances in stem cell based RPE differentiation, transplantation, and the preliminary results of clinical trials. The obstacles and prospects in this field will also be discussed.
Collapse
Affiliation(s)
- Yalin Mu
- Department of Ophthalmology, Yellow River Hospital, Henan University of Science and Technology Sanmenxia City, Henan Province, People's Republic of China
| | - Manli Zhao
- Department of Ophthalmology, Yellow River Hospital, Henan University of Science and Technology Sanmenxia City, Henan Province, People's Republic of China
| | - Guangming Su
- Department of Ophthalmology, Yellow River Hospital, Henan University of Science and Technology Sanmenxia City, Henan Province, People's Republic of China
| |
Collapse
|
73
|
Yu H, Cheng L, Cho KS. The potential of stem cell-based therapy for retinal repair. Neural Regen Res 2014; 9:1100-3. [PMID: 25206766 PMCID: PMC4146102 DOI: 10.4103/1673-5374.135311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2014] [Indexed: 12/20/2022] Open
Affiliation(s)
- Honghua Yu
- Department of Ophthalmology, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong Province, China ; Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| | - Lin Cheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China ; Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, China ; Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| | - Kin-Sang Cho
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| |
Collapse
|
74
|
Recent advances of stem cell therapy for retinitis pigmentosa. Int J Mol Sci 2014; 15:14456-74. [PMID: 25141102 PMCID: PMC4159862 DOI: 10.3390/ijms150814456] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/24/2014] [Accepted: 08/11/2014] [Indexed: 12/22/2022] Open
Abstract
Retinitis pigmentosa (RP) is a group of inherited retinal disorders characterized by progressive loss of photoreceptors and eventually leads to retina degeneration and atrophy. Until now, the exact pathogenesis and etiology of this disease has not been clear, and many approaches for RP therapies have been carried out in animals and in clinical trials. In recent years, stem cell transplantation-based attempts made some progress, especially the transplantation of bone marrow-derived mesenchymal stem cells (BMSCs). This review will provide an overview of stem cell-based treatment of RP and its main problems, to provide evidence for the safety and feasibility for further clinical treatment.
Collapse
|
75
|
Abstract
Diabetic retinopathy is the leading cause of blindness in the working population. We now understand that the pathogenesis of the disease contains both a neurodegenerative and vasodegenerative component. Yet despite this, current treatment is still limited to combating the proliferative end stage component of the disease rather than addressing its underlying causes. In recent years, much basic research has focused on demonstrating the potential that several classes of stem cells have in conferring both neuro- and vasoprotection on the diabetic retina. Further, progress has been made in using stem cells to stimulate both neuro and vascular regeneration. This review will discuss the current understanding as to what mechanisms underlie diabetic retinopathy while highlighting the types of stem cells which offer hope as potential novel therapies for diabetic retinopathy, including those that are now in clinical trial.
Collapse
Affiliation(s)
- Roly Megaw
- Scottish Center for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK,
| | | |
Collapse
|
76
|
Ng TK, Fortino VR, Pelaez D, Cheung HS. Progress of mesenchymal stem cell therapy for neural and retinal diseases. World J Stem Cells 2014; 6:111-119. [PMID: 24772238 PMCID: PMC3999769 DOI: 10.4252/wjsc.v6.i2.111] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/14/2014] [Accepted: 03/04/2014] [Indexed: 02/07/2023] Open
Abstract
Complex circuitry and limited regenerative power make central nervous system (CNS) disorders the most challenging and difficult for functional repair. With elusive disease mechanisms, traditional surgical and medical interventions merely slow down the progression of the neurodegenerative diseases. However, the number of neurons still diminishes in many patients. Recently, stem cell therapy has been proposed as a viable option. Mesenchymal stem cells (MSCs), a widely-studied human adult stem cell population, have been discovered for more than 20 years. MSCs have been found all over the body and can be conveniently obtained from different accessible tissues: bone marrow, blood, and adipose and dental tissue. MSCs have high proliferative and differentiation abilities, providing an inexhaustible source of neurons and glia for cell replacement therapy. Moreover, MSCs also show neuroprotective effects without any genetic modification or reprogramming. In addition, the extraordinary immunomodulatory properties of MSCs enable autologous and heterologous transplantation. These qualities heighten the clinical applicability of MSCs when dealing with the pathologies of CNS disorders. Here, we summarize the latest progress of MSC experimental research as well as human clinical trials for neural and retinal diseases. This review article will focus on multiple sclerosis, spinal cord injury, autism, glaucoma, retinitis pigmentosa and age-related macular degeneration.
Collapse
|
77
|
Ezquer F, Ezquer M, Arango-Rodriguez M, Conget P. Could donor multipotent mesenchymal stromal cells prevent or delay the onset of diabetic retinopathy? Acta Ophthalmol 2014; 92:e86-95. [PMID: 23773776 DOI: 10.1111/aos.12113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a complex metabolic disease that has become a global epidemic with more than 285 million cases worldwide. Major medical advances over the past decades have substantially improved its management, extending patients' survival. The latter is accompanied by an increased risk of developing chronic macro- and microvascular complications. Amongst them, diabetic retinopathy (DR) is the most common and frightening. Furthermore, during the past two decades, it has become the leading cause of visual loss. Irrespective of the type of diabetes, DR follows a well-known clinical and temporal course characterized by pericytes and neuronal cell loss, formation of acellular-occluded capillaries, occasional microaneurysms, increased leucostasis and thickening of the vascular basement membrane. These alterations progressively affect the integrity of retinal microvessels, leading to the breakdown of the blood-retinal barrier, widespread haemorrhage and neovascularization. Finally, tractional retinal detachment occurs leading to blindness. Nowadays, there is growing evidence that local inflammation and oxidative stress play pivotal roles in the pathogenesis of DR. Both processes have been associated with pericytes and neuronal degeneration observed early during DR progression. They may also be linked to sustained retinal vasculature damage that results in abnormal neovascularization. Currently, DR therapeutic options depend on highly invasive surgical procedures performed only at advanced stages of the disease, and which have proved to be ineffective to restore visual acuity. Therefore, the availability of less invasive and more effective strategies aimed to prevent or delay the onset of DR is highly desirable. Multipotent mesenchymal stromal cells, also referred to as mesenchymal stem cells (MSCs), are promising healing agents as they contribute to tissue regeneration by pleiotropic mechanisms, with no evidence of significant adverse events. Here, we revise the pathophysiology of DR to identify therapeutic targets for donor MSCs. Also, we discuss whether an MSC-based therapy could prevent or delay the onset of DR.
Collapse
Affiliation(s)
- Fernando Ezquer
- Institute of Science, Faculty of Medicine Clinica Alemana Universidad del Desarrollo, Lo Barnechea, Santiago, Chile
| | | | | | | |
Collapse
|
78
|
Bharti K, Rao M, Hull SC, Stroncek D, Brooks BP, Feigal E, van Meurs JC, Huang CA, Miller SS. Developing cellular therapies for retinal degenerative diseases. Invest Ophthalmol Vis Sci 2014; 55:1191-202. [PMID: 24573369 DOI: 10.1167/iovs.13-13481] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Biomedical advances in vision research have been greatly facilitated by the clinical accessibility of the visual system, its ease of experimental manipulation, and its ability to be functionally monitored in real time with noninvasive imaging techniques at the level of single cells and with quantitative end-point measures. A recent example is the development of stem cell-based therapies for degenerative eye diseases including AMD. Two phase I clinical trials using embryonic stem cell-derived RPE are already underway and several others using both pluripotent and multipotent adult stem cells are in earlier stages of development. These clinical trials will use a variety of cell types, including embryonic or induced pluripotent stem cell-derived RPE, bone marrow- or umbilical cord-derived mesenchymal stem cells, fetal neural or retinal progenitor cells, and adult RPE stem cells-derived RPE. Although quite distinct, these approaches, share common principles, concerns and issues across the clinical development pipeline. These considerations were a central part of the discussions at a recent National Eye Institute meeting on the development of cellular therapies for retinal degenerative disease. At this meeting, emphasis was placed on the general value of identifying and sharing information in the so-called "precompetitive space." The utility of this behavior was described in terms of how it could allow us to remove road blocks in the clinical development pipeline, and more efficiently and economically move stem cell-based therapies for retinal degenerative diseases toward the clinic. Many of the ocular stem cell approaches we discuss are also being used more broadly, for nonocular conditions and therefore the model we develop here, using the precompetitive space, should benefit the entire scientific community.
Collapse
Affiliation(s)
- Kapil Bharti
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Tsai Y, Lu B, Ljubimov AV, Girman S, Ross-Cisneros FN, Sadun AA, Svendsen CN, Cohen RM, Wang S. Ocular changes in TgF344-AD rat model of Alzheimer's disease. Invest Ophthalmol Vis Sci 2014; 55:523-34. [PMID: 24398104 DOI: 10.1167/iovs.13-12888] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by progressive decline in learning, memory, and executive functions. In addition to cognitive and behavioral deficits, vision disturbances have been reported in early stage of AD, well before the diagnosis is clearly established. To further investigate ocular abnormalities, a novel AD transgenic rat model was analyzed. METHODS Transgenic (Tg) rats (TgF344-AD) heterozygous for human mutant APPswe/PS1ΔE9 and age-matched wild type (WT) rats, as well as 20 human postmortem retinal samples from both AD and healthy donors were used. Visual function in the rodent was analyzed using the optokinetic response and luminance threshold recording from the superior colliculus. Immunohistochemistry on retinal and brain sections was used to detect various markers including amyloid-β (Aβ) plaques. RESULTS As expected, Aβ plaques were detected in the hippocampus, cortex, and retina of Tg rats. Plaque-like structures were also found in two AD human whole-mount retinas. The choroidal thickness was significantly reduced in both Tg rat and in AD human eyes when compared with age-matched controls. Tg rat eyes also showed hypertrophic retinal pigment epithelial cells, inflammatory cells, and upregulation of complement factor C3. Although visual acuity was lower in Tg than in WT rats, there was no significant difference in the retinal ganglion cell number and retinal vasculature. CONCLUSIONS In this study, we observed pathological changes in the choroid and in RPE cells in the TgF344-AD rat model; choroidal thinning was observed further in human AD retina. Along with Ab deposition, the inflammatory response was manifested by microglial recruitment and complement activation. Further studies are needed to elucidate the significance and mechanisms of these pathological changes [corrected].
Collapse
Affiliation(s)
- Yuchun Tsai
- Cedars-Sinai Regenerative Medicine Institute, Los Angeles, California
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Jiang XS, Ni YQ, Liu TJ, Zhang M, Jiang R, Xu GZ. Generation and characterization of immortalized rat retinal microglial cell lines. J Neurosci Res 2014; 92:424-31. [PMID: 24452530 DOI: 10.1002/jnr.23337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/29/2013] [Accepted: 11/04/2013] [Indexed: 02/05/2023]
Abstract
Retinal microglia play an important role as resident immunocompetent and phagocytic cells in the event of injury and disease. Retinal microglia and microglia precursor transplantation show a rescue effect in ischemic retina and retinal degeneration. However, studies of retinal microglia have been hampered by the difficulty of obtaining sufficient numbers of microglia. One way to circumvent this difficulty is to establish permanent retinal microglia cell lines. In the present study, we report the generation of immortalized retinal microglia, T-MG cells, from postnatal day 3 rat retinal tissue using a lentiviral vector encoding SV40 large T antigen. The T-MG cells exhibited cell-type-specific antigens for monocyte/macrophage lineage cells, including CD11b (OX42), ED1 (OX6), and Iba1, and actively phagocytosed latex beads. In addition to primary retinal microglia, T-MG cells also have the ability to recruit into chemokines. Treatment of T-MG cells with lipopolysaccharide (LPS) led to increased levels of tumor necrosis factor-α, interleukin-1β, and inducible nitric oxide synthase. Genome-wide microarray analysis showed a less than 1% difference in the genes between the T-MG cells and the control primary retinal microglia. The T-MG cells exhibited properties similar to those of the primary retinal microglia and should have considerable utility as an in vitro model for the study of retinal microglia in health and as a curative therapy and an in vivo model for the study of retinal microglia in disease.
Collapse
Affiliation(s)
- Xiao-shuang Jiang
- Department of Ophthalmology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China; Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
81
|
Luo J, Baranov P, Patel S, Ouyang H, Quach J, Wu F, Qiu A, Luo H, Hicks C, Zeng J, Zhu J, Lu J, Sfeir N, Wen C, Zhang M, Reade V, Patel S, Sinden J, Sun X, Shaw P, Young M, Zhang K. Human retinal progenitor cell transplantation preserves vision. J Biol Chem 2014; 289:6362-6371. [PMID: 24407289 DOI: 10.1074/jbc.m113.513713] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cell transplantation is a potential therapeutic strategy for retinal degenerative diseases involving the loss of photoreceptors. However, it faces challenges to clinical translation due to safety concerns and a limited supply of cells. Human retinal progenitor cells (hRPCs) from fetal neural retina are expandable in vitro and maintain an undifferentiated state. This study aimed to investigate the therapeutic potential of hRPCs transplanted into a Royal College of Surgeons (RCS) rat model of retinal degeneration. At 12 weeks, optokinetic response showed that hRPC-grafted eyes had significantly superior visual acuity compared with vehicle-treated eyes. Histological evaluation of outer nuclear layer (ONL) characteristics such as ONL thickness, spread distance, and cell count demonstrated a significantly greater preservation of the ONL in hRPC-treated eyes compared with both vehicle-treated and control eyes. The transplanted hRPCs arrested visual decline over time in the RCS rat and rescued retinal morphology, demonstrating their potential as a therapy for retinal diseases. We suggest that the preservation of visual acuity was likely achieved through host photoreceptor rescue. We found that hRPC transplantation into the subretinal space of RCS rats was well tolerated, with no adverse effects such as tumor formation noted at 12 weeks after treatment.
Collapse
Affiliation(s)
- Jing Luo
- Department of Ophthalmology, Second Xiangya Hospital and International Academy of Translational Medicine, Central South University, Changsha, Hunan 410011, China; Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - Petr Baranov
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114
| | - Sherrina Patel
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - Hong Ouyang
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - John Quach
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - Frances Wu
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - Austin Qiu
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093; Molecular Medicine Research Center, West China Hospital, Chengdu, Sichuan 610041, China
| | - Hongrong Luo
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093; Molecular Medicine Research Center, West China Hospital, Chengdu, Sichuan 610041, China
| | - Caroline Hicks
- ReNeuron Ltd., Guildford, Surrey GU2 7AF, United Kingdom
| | - Jing Zeng
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093; Department of Ophthalmology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jing Zhu
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093; Molecular Medicine Research Center, West China Hospital, Chengdu, Sichuan 610041, China
| | - Jessica Lu
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - Nicole Sfeir
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - Cindy Wen
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - Meixia Zhang
- Molecular Medicine Research Center, West China Hospital, Chengdu, Sichuan 610041, China
| | | | - Sara Patel
- Molecular Medicine Research Center, West China Hospital, Chengdu, Sichuan 610041, China
| | - John Sinden
- Molecular Medicine Research Center, West China Hospital, Chengdu, Sichuan 610041, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200080, China; Eye Research Institute, Shanghai JiaoTong University, Shanghai 200080, China.
| | - Peter Shaw
- Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093
| | - Michael Young
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114
| | - Kang Zhang
- Department of Ophthalmology, Second Xiangya Hospital and International Academy of Translational Medicine, Central South University, Changsha, Hunan 410011, China; Institute for Genomic Medicine and Shiley Eye Center, University of California at San Diego, La Jolla, California 92093; Molecular Medicine Research Center, West China Hospital, Chengdu, Sichuan 610041, China; Veterans Affairs Healthcare System, San Diego, California 92161.
| |
Collapse
|
82
|
Davey GC, Patil SB, O'Loughlin A, O'Brien T. Mesenchymal stem cell-based treatment for microvascular and secondary complications of diabetes mellitus. Front Endocrinol (Lausanne) 2014; 5:86. [PMID: 24936198 PMCID: PMC4047679 DOI: 10.3389/fendo.2014.00086] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/23/2014] [Indexed: 12/22/2022] Open
Abstract
The worldwide increase in the prevalence of Diabetes mellitus (DM) has highlighted the need for increased research efforts into treatment options for both the disease itself and its associated complications. In recent years, mesenchymal stromal cells (MSCs) have been highlighted as a new emerging regenerative therapy due to their multipotency but also due to their paracrine secretion of angiogenic factors, cytokines, and immunomodulatory substances. This review focuses on the potential use of MSCs as a regenerative medicine in microvascular and secondary complications of DM and will discuss the challenges and future prospects of MSCs as a regenerative therapy in this field. MSCs are believed to have an important role in tissue repair. Evidence in recent years has demonstrated that MSCs have potent immunomodulatory functions resulting in active suppression of various components of the host immune response. MSCs may also have glucose lowering properties providing another attractive and unique feature of this therapeutic approach. Through a combination of the above characteristics, MSCs have been shown to exert beneficial effects in pre-clinical models of diabetic complications prompting initial clinical studies in diabetic wound healing and nephropathy. Challenges that remain in the clinical translation of MSC therapy include issues of MSC heterogeneity, optimal mode of cell delivery, homing of these cells to tissues of interest with high efficiency, clinically meaningful engraftment, and challenges with cell manufacture. An issue of added importance is whether an autologous or allogeneic approach will be used. In summary, MSC administration has significant potential in the treatment of diabetic microvascular and secondary complications but challenges remain in terms of engraftment, persistence, tissue targeting, and cell manufacture.
Collapse
Affiliation(s)
- Grace C Davey
- Regenerative Medicine Institute (REMEDI) and Biosciences Building, National University of Ireland , Galway , Ireland
| | - Swapnil B Patil
- Regenerative Medicine Institute (REMEDI) and Biosciences Building, National University of Ireland , Galway , Ireland
| | - Aonghus O'Loughlin
- Department of Medicine, Galway University Hospital (GUH) , Galway , Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI) and Biosciences Building, National University of Ireland , Galway , Ireland ; Department of Medicine, Galway University Hospital (GUH) , Galway , Ireland
| |
Collapse
|
83
|
Tzameret A, Sher I, Belkin M, Treves AJ, Meir A, Nagler A, Levkovitch-Verbin H, Barshack I, Rosner M, Rotenstreich Y. Transplantation of human bone marrow mesenchymal stem cells as a thin subretinal layer ameliorates retinal degeneration in a rat model of retinal dystrophy. Exp Eye Res 2013; 118:135-44. [PMID: 24239509 DOI: 10.1016/j.exer.2013.10.023] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/24/2013] [Accepted: 10/31/2013] [Indexed: 02/06/2023]
Abstract
Vision incapacitation and blindness associated with retinal degeneration affect millions of people worldwide. Cell based therapy and specifically transplantation of human adult bone marrow-derived stem cells (hBM-MSCs) present possible treatment strategy. Subretinal transplantation of human or rat BM-MSCs was shown previously to improve retinal function in Royal College Surgeons (RCS) rats. In those studies cells were transplanted via a transscleral-transchoroidal approach, creating a localized subretinal bleb. Limited number of cells could be injected and photoreceptor rescue was restricted to areas in proximity to the injection site. Here we describe a new surgical method for subretinal transplantation that facilitates uniform distribution of transplanted cells as a thin layer along most of the subretinal space. We assessed the therapeutic effect of hBM-MSCs on RCS rats when transplanted either subretinally or intravitreally. We also examined whether a second transplantation can prolong the therapeutic effect. A cell suspension of 2.5 × 10(6) cells in 5 μl was injected subretinally or intravitreally in RCS rats at 28 days postnatal. In the subretinal group, hBM-MSCs were transplanted posterior to the limbus in the superotemporal part of the eye through a longitudinal triangular scleral tunnel reaching the choroid. In the intravitreal group, the cells were injected into the superotemporal part of the vitreous cavity. In cross sections of subretinally transplanted eyes, removed 2 h following transplantation, hBM-MSCs were distributed as a near-homogenous thin layer along most of the subretinal space. In some animals the cells were also detected in the choroid. In the intravitreal injection group, hBM-MSCs were clustered in the vitreous cavity. Transplanted cells could be detected up to 2 weeks after transplantation but not at later time points. Retinal function and structure were assessed by electroretinogram (ERG) and histology analysis, respectively. Six weeks post transplantation, the mean maximal scotopic ERG b-wave amplitude response recorded in RCS control eyes was 1.2 μV. By contrast, in transplanted eyes mean responses of 56.4 μV and 66.2 μV were recorded in the intravitreally and subretinally transplanted eyes, respectively. In the subretinal group, retinal function was significantly higher in transplanted compared with control eyes up to 20 weeks following transplantation. By contrast, in the intravitreal group, rescue of retinal function persisted only up to 12 weeks following transplantation. Histological analysis revealed that 8 weeks following subretinal transplantation, the retinas of control eyes were dystrophic, with outer nuclear layer (ONL) containing a single cell layer. An extensive photoreceptor rescue was demonstrated in transplanted eyes at this time point, with 3-4 cell layers in the ONL along the entire retina. A second subretinal transplantation at 70 days postnatal did not enhance or prolong the therapeutic effect of hBM-MSCs. No immunosuppressants were used and long-term safety analysis demonstrated no gross or microscopic adverse effects. Taken together our findings suggest that transplantation of hBM-MSCs as a thin subretinal layer enhances the therapeutic effect and the safety of cell transplantation.
Collapse
Affiliation(s)
- Adi Tzameret
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Ifat Sher
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Michael Belkin
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Avraham J Treves
- Center for Stem Cells and Regenerative Medicine, Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Amilia Meir
- Center for Stem Cells and Regenerative Medicine, Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Arnon Nagler
- Hematology Division, Sheba Medical Center, Tel-Hashomer, Israel
| | - Hani Levkovitch-Verbin
- Rothberg Ophthalmic Molecular Biology Laboratory, Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer, Israel
| | - Iris Barshack
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer, Israel
| | - Mordechai Rosner
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Ygal Rotenstreich
- Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv University, Sheba Medical Center, Tel-Hashomer 52621, Israel.
| |
Collapse
|
84
|
Xu W, Wang X, Xu G, Guo J. Basic fibroblast growth factor expression is implicated in mesenchymal stem cells response to light-induced retinal injury. Cell Mol Neurobiol 2013; 33:1171-9. [PMID: 24030359 PMCID: PMC11497881 DOI: 10.1007/s10571-013-9983-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 08/31/2013] [Indexed: 12/29/2022]
Abstract
Neurotrophic factors are involved in neuroprotection and its expression in mesenchymal stem cells (MSCs) may change during light-induced retinal injury. In this study, neurotrophic factor expression in MSCs was investigated after stimulation by supernatants of homogenized retina (SHR) from normal and light-injured rats. Conditioned media from control MSCs (CM-MSCs), MSCs stimulated by normal SHR (CM-NSHR), and MSCs stimulated by light-injured SHR (CM-ISHR) were examined regarding their ability to prevent degeneration of retinal explants. Basic fibroblast growth factor (bFGF) in MSCs was knockdown by lentivirus-mediated mRNA interference. Transfected MSCs were stimulated by SHR, and retinal preservation was reevaluated in the resultant conditioned media. We detected significant up-regulation of bFGF in CM-ISHR, accompanied by superior retinal neurotrophic effects in CM-ISHR over CM-NSHR and CM-MSCs. Down-regulation of bFGF in MSCs effectively inhibited this protective effect. Adding neutralizing antibody against bFGF to CM-ISHR also induced a similar effect. It is thus concluded that retinal injury may enhance neurotrophic factor expression in MSCs and promote the repair process. bFGF may play a critical role in MSCs' response to retinal injury.
Collapse
Affiliation(s)
- Wei Xu
- Department of Ophthalmology, First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005 China
| | - Xiaoting Wang
- Department of Ophthalmology, First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005 China
| | - Guoxing Xu
- Department of Ophthalmology, First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005 China
- Fujian Institute of Ophthalmology, Fuzhou, China
| | - Jian Guo
- Department of Ophthalmology, First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005 China
- Fujian Institute of Ophthalmology, Fuzhou, China
| |
Collapse
|
85
|
Xu W, Wang XT, Xu GX, Guo J, Huang LB. Stromal cell-derived factor 1α-stimulated mesenchymal stem cells confer enhanced protection against light-induced retinal degeneration in rats. Curr Eye Res 2013; 39:69-78. [PMID: 24074164 DOI: 10.3109/02713683.2013.824988] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE Mesenchymal stem cells (MSCs) are currently considered to be modulators of repair in various tissues. After MSC transplant, photoreceptor rescue has been demonstrated in models of retinal degeneration. Herein, we evaluate the roles of MSCs in modulating the host reaction and photoreceptor preservation in rats suffering from light-induced retinal degeneration. METHODS Unstimulated and stromal cell-derived factor 1α (SDF-1α)-stimulated MSCs were intravenously transplanted into light-injured rats. Their photoreceptor rescue effect was compared with untreated light-injured rats and light-injured rats received only medium injection. Ciliary neurotrophic factor (CNTF) and glial fibrillary acidic protein (GFAP) expression was identified to assess host reaction post-transplantation. Retinal localization and integration of MSCs were determined by green fluorescence protein labeling. RESULTS MSCs were able to migrate and integrate into the host retina, and significantly inhibited retinal cell death. CNTF and GFAP were induced upregluation after MSC injection. SDF-1α stimulation elicited superior effects in both MSC migration and the inhibition of apoptosis. CNTF and GFAP expression in host retinas that received stimulated MSCs were stronger than in retinas that received unstimulated MSCs. CONCLUSIONS Systemic administration of MSCs exerts a protective effect against light-induced retinal degeneration, and upregulates neurotrophin expression in the host retina. MSCs can be stimulated to enhance the therapeutic effect.
Collapse
Affiliation(s)
- Wei Xu
- Department of Ophthalmology, First Affiliated Hospital of Fujian Medical University Fuzhou , China , and
| | | | | | | | | |
Collapse
|
86
|
Swoboda JG, Elliott J, Deshmukh V, de Lichtervelde L, Shen W, Tremblay MS, Peters EC, Cho CY, Lu B, Girman S, Wang S, Schultz PG. Small molecule mediated proliferation of primary retinal pigment epithelial cells. ACS Chem Biol 2013; 8:1407-11. [PMID: 23621521 DOI: 10.1021/cb4001712] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Retinal pigment epithelial (RPE) cells form a monolayer adjacent to the retina and play a critical role in the visual light cycle. Degeneration of RPE cells results in retinal disorders such as age-related macular degeneration. Cell transplant strategies have potential therapeutic value for such disorders; however, risks associated with an inadequate supply of donor cells limit their therapeutic success. The identification of factors that proliferate RPE cells ex vivo could provide a renewable source of cells for transplantation. Here, we report that a small molecule (WS3) can reversibly proliferate primary RPE cells isolated from fetal and adult human donors. Following withdrawal of WS3, RPE cells differentiate into a functional monolayer, as exhibited by their expression of mature RPE genes and phagocytosis of photoreceptor outer segments. Furthermore, chemically expanded RPE cells preserve vision when transplanted into dystrophic Royal College of Surgeons (RCS) rats, a well-established model of retinal degeneration.
Collapse
Affiliation(s)
- Jonathan G. Swoboda
- Department of Chemistry and
the Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La
Jolla, California 92037, United States
| | - Jimmy Elliott
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay
Hopkins Drive, San Diego, California 92121, United States
| | - Vishal Deshmukh
- Department of Chemistry and
the Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La
Jolla, California 92037, United States
| | - Lorenzo de Lichtervelde
- Department of Chemistry and
the Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La
Jolla, California 92037, United States
| | - Weijun Shen
- California Institute for Biomedical Research, 11119 North Torrey Pines Road,
La Jolla, California 92037, United States
| | - Matthew S. Tremblay
- California Institute for Biomedical Research, 11119 North Torrey Pines Road,
La Jolla, California 92037, United States
| | - Eric C. Peters
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay
Hopkins Drive, San Diego, California 92121, United States
| | - Charles Y. Cho
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay
Hopkins Drive, San Diego, California 92121, United States
| | - Bin Lu
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard,
Los Angeles, California 90048, United States
| | - Sergej Girman
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard,
Los Angeles, California 90048, United States
| | - Shaomei Wang
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard,
Los Angeles, California 90048, United States
| | - Peter G. Schultz
- Department of Chemistry and
the Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La
Jolla, California 92037, United States
| |
Collapse
|
87
|
Siqueira RC. Stem cell therapy in retinal diseases? Rev Bras Hematol Hemoter 2013; 34:222-6. [PMID: 23049424 PMCID: PMC3459631 DOI: 10.5581/1516-8484.20120054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 04/18/2012] [Indexed: 12/11/2022] Open
|
88
|
Huang L, Xu W, Xu G. Transplantation of CX3CL1-expressing mesenchymal stem cells provides neuroprotective and immunomodulatory effects in a rat model of retinal degeneration. Ocul Immunol Inflamm 2013; 21:276-85. [PMID: 23718544 DOI: 10.3109/09273948.2013.791925] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE To investigate the neuroprotective and immunomodulatory effects of mesenchymal stem cells (MSCs) engineered to secrete CX3CL1 on the light-injured retinal structure and function. METHODS Normal MSCs and CX3CL1-expressing MSCs (CX3CL1-MSCs) were transplanted into the subretinal space of light-injured rats. By ERG and TUNEL methods, their rescue effect of the host retina was compared with untreated light-injured and vehicle-injected rats. Activated microglia in the retina were stained by ED-1 antibody, and Western blot was performed to quantify cytokines secreted by the retina post-transplantation. RESULTS ERG analysis showed better function in CX3CL1-MSC-injected group than other groups at 21 days after transplantation (p < 0.05). CX3CL1-MSCs inhibited apoptosis of the retinal cells and microglial activation. Neurotrophic factors expression in host retina that received CX3CL1-MSCs was stronger than in the retina that received normal MSCs. Conversely, the expression of proinflammatory factors was downregulated. CONCLUSIONS CX3CL1-MSCs subretinal transplantation may enhance protective effect against light-induced retinal degeneration.
Collapse
Affiliation(s)
- Libin Huang
- Department of Ophthalmology, First Affiliated Hospital of Fujian Medical University, Fujian Institute of Ophthalmology, Fuzhou, China
| | | | | |
Collapse
|
89
|
Peng C, Yin X, Li M, He T, Li G. Construction of a eukaryotic expression plasmid for human retina-derived neurotrophin-3. Neural Regen Res 2013; 8:1031-40. [PMID: 25206397 PMCID: PMC4145883 DOI: 10.3969/j.issn.1673-5374.2013.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 03/26/2013] [Indexed: 11/18/2022] Open
Abstract
Neurotrophin-3 (NT-3) can promote the repair of central nervous system and retinal damage. In previous reports, NT-3 has been expressed by viral vectors. However, plasmid vectors have a safer profile compared with viral vectors in clinical studies. This study recombined amplified human retinal NT-3 with a eukaryotic expression plasmid containing green fluorescent protein (GFP) to construct an NT-3 expression plasmid, pEGFP-N1-NT-3. The transfection efficiency 48 hours after pEGFP-N1-NT-3 transfection to 293T cells was 50.06 ± 2.78%. Abundant NT-3-GFP was expressed in 293T cells as observed by fluorescence microscopy, suggesting the construct pEGFP-N1-NT-3 effectively expressed and secreted NT-3-GFP. Secretory vesicles containing NT-3-GFP were observed in a constant location in cells by laser scan confocal microscopy, indicating the expression and secretion processes of NT-3 in eukaryotic cells were in accordance with the physical synthesis processes of secreted proteins. Western blot assay showed that pro-NT-3-GFP had a molecular weight of 56 kDa, further confirming NT-3-GFP expression. At 48 hours after transfection, the concentration of NT-3 in culture medium was 22.3 ng/mL, suggesting NT-3 produced by pEGFP-N1-NT-3 was efficiently secreted. This study constructed a human retinal-derived NT-3 eukaryotic expression plasmid that efficiently expressed and secreted NT-3.
Collapse
Affiliation(s)
- Chunxia Peng
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China
| | - Xiaobei Yin
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China
| | - Mengda Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China
| | - Ting He
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China
| | - Genlin Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology & Visual Sciences, Beijing 100730, China
| |
Collapse
|
90
|
Lu B, Morgans CW, Girman S, Luo J, Zhao J, Du H, Lim S, Ding S, Svendsen C, Zhang K, Wang S. Neural Stem Cells Derived by Small Molecules Preserve Vision. Transl Vis Sci Technol 2013; 2:1. [PMID: 24049711 DOI: 10.1167/tvst.2.1.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 11/28/2012] [Indexed: 12/21/2022] Open
Abstract
PURPOSE The advances in stem cell biology hold a great potential to treat retinal degeneration. Importantly, specific cell types can be generated efficiently with small molecules and maintained stably over numerous passages. Here, we investigated whether neural stem cell (NSC) derived from human embryonic stem cells (hESC) by small molecules can preserve vision following grafting into the Royal College Surgeon (RCS) rats; a model for retinal degeneration. METHODS A cell suspension containing 3 × 104 NSCs or NSCs labeled with green fluorescent protein (GFP) was injected into the subretinal space or the vitreous cavity of RCS rats at postnatal day (P) 22; animals injected with cell-carry medium and those left untreated were used as controls. The efficacy of treatment was evaluated by testing optokinetic response, recording luminance threshold, and examining retinal histology. RESULTS NSCs offered significant preservation of both photoreceptors and visual function. The grafted NSCs survived for long term without evidence of tumor formation. Functionally, NSC treated eyes had significantly better visual acuity and lower luminance threshold than controls. Morphologically, photoreceptors and retinal connections were well preserved. There was an increase in expression of cillary neurotrophic factor (CNTF) in Müller cells in the graft-protected retina. CONCLUSIONS This study reveals that NSCs derived from hESC by small molecules can survive and preserve vision for long term following subretinal transplantation in the RCS rats. These cells migrate extensively in the subretinal space and inner retina; there is no evidence of tumor formation or unwanted changes after grafting into the eyes. TRANSLATIONAL RELEVANCE The NSCs derived from hESC by small molecules can be generated efficiently and provide an unlimited supply of cells for the treatment of some forms of human outer retinal degenerative diseases. The capacity of NSCs migrating into inner retina offers a potential as a vehicle to delivery drugs/factors to treat inner retinal disorders.
Collapse
Affiliation(s)
- Bin Lu
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
|
92
|
Bashar AE, Metcalfe A, Yanai A, Laver C, Häfeli UO, Gregory-Evans CY, Moritz OL, Matsubara JA, Gregory-Evans K. Influence of Iron Oxide Nanoparticles on Innate and Genetically Modified Secretion Profiles of Mesenchymal Stem Cells. IEEE TRANSACTIONS ON MAGNETICS 2013; 49:389-393. [PMID: 24976643 PMCID: PMC4072646 DOI: 10.1109/tmag.2012.2225829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Mesenchymal stem cells (MSCs) have well-established paracrine effects that are proving to be therapeutically useful. This potential is based on the ability of MSCs to secrete a range of neuroprotective and anti-inflammatory molecules. Previous work in our laboratory has demonstrated that intravenous injection of MSCs, treated with superparamagnetic iron oxide nanoparticle fluidMAG-D resulted in enhanced levels of glial-derived neurotrophic factor, ciliary neurotrophic factor, hepatocyte growth factor and interleukin-10 in the dystrophic rat retina. In this present study we investigated whether the concentration of fluidMAG-D in cell culture media affects the secretion of these four molecules in vitro. In addition, we assessed the effect of fluidMAG-D concentration on retinoschisin secretion from genetically modified MSCs. ELISA-assayed secretion of these molecules was measured using escalating concentrations of fluidMAG-D which resulted in MSC iron loads of 0, 7, 120, or 274 pg iron oxide per cell respectively. Our results demonstrated glial-derived neurotrophic factor and hepatocyte growth factor secretion was significantly decreased but only at the 96 hour's time-point whereas no statistically significant effect was seen with ciliary neurotrophic factor secretion. Whereas no effect was observed on culture media concentrations of retinoschisin with increasing iron oxide load, a statistically significant increase in cell lysate retinoschisin concentration (p = 0.01) was observed suggesting that increasing fluidMAG-D concentration did increase retinoschisin production but this did not lead to greater secretion. We hypothesize that higher concentrations of iron-oxide nanoparticle fluidMAG-D have an effect on the innate ability of MSCs to secrete therapeutically useful molecules and also on secretion from genetically modified cells. Further work is required to verify these in vitro finding using in vivo model systems.
Collapse
Affiliation(s)
- Abu Emran Bashar
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Andrew Metcalfe
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Anat Yanai
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Christopher Laver
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Urs O Häfeli
- Department of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl Y Gregory-Evans
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Orson L Moritz
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Kevin Gregory-Evans
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
93
|
Jiang XS, Ni YQ, Liu TJ, Zhang M, Ren H, Jiang R, Huang X, Xu GZ. CCR2 overexpression promotes the efficient recruitment of retinal microglia in vitro. Mol Vis 2012; 18:2982-92. [PMID: 23288990 PMCID: PMC3534147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Accepted: 12/13/2012] [Indexed: 11/01/2022] Open
Abstract
PURPOSE Retinal microglia can be activated and recruited by chemokines and play a protective role in early retinal degeneration. CC-chemokine ligand 2 (CCL2) and its receptor, CC-chemokine receptor 2 (CCR2), have been implicated as key mediators for the trafficking and accumulation of microglial cells in lesioned tissue. The current study investigates whether the overexpression of CCR2 allows microglia to migrate toward CCL2 more efficiently. METHODS Primary microglial cells were transduced with lentivirus carrying green fluorescent protein (GFP)-tagged CCR2 (CCR2-GFP). Overexpression of CCR2 was assessed by western blot analysis and fluorescence-assisted cell sorting. The chemotaxis of primary microglia transduced with lentivirus carrying CCR2-GFP was compared to either those transduced with GFP alone or those not transduced, using a chemotaxis chamber assay. RESULTS Primary microglia showed a high transduction rate following lentivirus application and maintained normal microglial morphology and a significant overexpression of CCR2 protein. We found that CCL2-mediated chemotaxis is concentration and time dependent in microglia. The chemotactic response of microglia cells overexpressing CCR2-GFP was significantly increased compared to that of nontransduced and GFP-expressing microglia. CONCLUSIONS These findings suggest that microglia can be efficiently transduced with CCR2-GFP lentiviral vectors and that the overexpression of CCR2 in retinal microglia promotes their chemotaxis in response to chemokines, suggesting that these cells may be promising targets for cell-based therapeutic manipulation in retinal disease.
Collapse
Affiliation(s)
- Xiao-shuang Jiang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Ying-qin Ni
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Tian-jin Liu
- Institute of Biochemistry, Chinese Academy of Science, Shanghai, People’s Republic of China
| | - Meng Zhang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Hui Ren
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Rui Jiang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Xin Huang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, People’s Republic of China
| | - Ge-zhi Xu
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
94
|
Zhou X, Xia XB, Xiong SQ. Neuro-protection of retinal stem cells transplantation combined with copolymer-1 immunization in a rat model of glaucoma. Mol Cell Neurosci 2012; 54:1-8. [PMID: 23246669 DOI: 10.1016/j.mcn.2012.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 11/06/2012] [Accepted: 12/04/2012] [Indexed: 01/18/2023] Open
Abstract
Glaucoma is a chronic, neurodegenerative disease that often leads to blindness. A common treatment is to reduce intraocular pressure (IOP), but this approach does not halt visual loss caused by the death of retinal ganglion cells (RGCs). Therefore, there is an important need for therapies that protect against RGCs degeneration. The present study in a rat glaucoma model aimed to determine whether retinal stem cells (RSCs) transplantation plus vaccination with a glatiramer acetate copolymer-1 (COP-1) could confer neuroprotection. Rats were immunized with COP-1 on the same day as IOP induction by argon laser photocoagulation of the episcleral veins and limbal plexus. RSCs were cultured and transplanted intravitreally 1week after laser treatment. The expression of brain-derived neurotrophic factor (BDNF) and insulin-like growth factor I (IGF-I) was detected by immunohistochemical staining, RT-PCR, and western blotting. RGCs survival was assessed by TUNEL staining and RGCs counting. We found that the expression of BDNF and IGF-I in the RSCs/COP-1 group was significantly higher than in other groups (P<0.05). In addition, the number of the apoptotic RGCs in the RSCs/COP-1 group was notably lower than in other groups (P<0.05), and the number of RGCs in the RSCs/COP-1 group was higher than in other groups (P<0.05). We conclude, therefore, that the combined effects between RSCs transplantation and COP-1 immunization protect RGCs from apoptosis in our rat model of glaucoma. The increase in levels of secreted BDNF and IGF-I may be one of the mechanisms underlying the neuro-protection of RGCs.
Collapse
Affiliation(s)
- Xia Zhou
- Department of Ophthalmology, Xiangya Hospital of Central South University, Changsha 410008, China
| | | | | |
Collapse
|
95
|
Kotowski M, Safranow K, Kawa MP, Lewandowska J, Kłos P, Dziedziejko V, Paczkowska E, Czajka R, Celewicz Z, Rudnicki J, Machaliński B. Circulating hematopoietic stem cell count is a valuable predictor of prematurity complications in preterm newborns. BMC Pediatr 2012; 12:148. [PMID: 22985188 PMCID: PMC3573966 DOI: 10.1186/1471-2431-12-148] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 08/29/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The frequency of preterm labour has risen over the last few years. Hence, there is growing interest in the identification of markers that may facilitate prediction and prevention of premature birth complications. Here, we studied the association of the number of circulating stem cell populations with the incidence of complications typical of prematurity. METHODS The study groups consisted of 90 preterm (23-36 weeks of gestational age) and 52 full-term (37-41 weeks) infants. Non-hematopoietic stem cells (non-HSCs; CD45-lin-CD184+), enriched in very small embryonic-like stem cells (VSELs), expressing pluripotent (Oct-4, Nanog), early neural (β-III-tubulin), and oligodendrocyte lineage (Olig-1) genes as well as hematopoietic stem cells (HSCs; CD45+lin-CD184+), and circulating stem/progenitor cells (CSPCs; CD133+CD34+; CD133-CD34+) in association with characteristics of prematurity and preterm morbidity were analyzed in cord blood (CB) and peripheral blood (PB) until the sixth week after delivery. Phenotype analysis was performed using flow cytometry methods. Clonogenic assays suitable for detection of human hematopoietic progenitor cells were also applied. The quantitative parameters were compared between groups by the Mann-Whitney test and between time points by the Friedman test. Fisher's exact test was used for qualitative variables. RESULTS We found that the number of CB non-HSCs/VSELs is inversely associated with the birth weight of preterm infants. More notably, a high number of CB HSCs is strongly associated with a lower risk of prematurity complications including intraventricular hemorrhage, respiratory distress syndrome, infections, and anemia. The number of HSCs remains stable for the first six weeks of postnatal life. Besides, the number of CSPCs in CB is significantly higher in preterm infants than in full-term neonates (p < 0.0001) and extensively decreases in preterm babies during next six weeks after birth. Finally, the growth of burst-forming unit of erythrocytes (BFU-E) and colony-forming units of granulocyte-macrophage (CFU-GM) obtained from CB of premature neonates is higher than those obtained from CB of full-term infants and strongly correlates with the number of CB-derived CSPCs. CONCLUSION We conclude that CB HSCs are markedly associated with the development of premature birth complications. Thus, HSCs ought to be considered as the potential target for further research as they may be relevant for predicting and controlling the morbidity of premature infants. Moreover, the observed levels of non-HSCs/VSELs circulating in CB are inversely associated with the birth weight of preterm infants, suggesting non-HSCs/VSELs might be involved in the maturation of fetal organism.
Collapse
Affiliation(s)
- Maciej Kotowski
- Department of General Pathology, Pomeranian Medical University in Szczecin, Powstancow Wlkp, 72, Szczecin 70-111, Poland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Adamus G, Wang S, Kyger M, Worley A, Lu B, Burrows GG. Systemic immunotherapy delays photoreceptor cell loss and prevents vascular pathology in Royal College of Surgeons rats. Mol Vis 2012; 18:2323-37. [PMID: 22977300 PMCID: PMC3441155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 09/04/2012] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Degenerative retinopathies, including retinitis pigmentosa, age-related retinal degeneration, autoimmune retinopathy, and related diseases affect millions of people around the world. Currently, there is no effective treatment for most of those diseases. We investigated systemic recombinant T-cell receptor ligand (RTL) immunotherapy for preventing retinal degeneration and vascular damage in the Royal College of Surgeons (RCS) rat model of retinal degeneration. METHODS RCS rats were treated with RTL220 tethered to interphotoreceptor retinoid binding protein (IRBP) peptide or control RTL101 without peptide by subcutaneous administration starting at the onset of photoreceptor degeneration or after the degenerative process began daily or every other day and performed for a 13-week period. The retinal cross sections and whole mounts were prepared to determine histopathology, leaking vessels, and formation of vascular complexes. Immunofluorescent studies evaluated microglia and monocyte chemoattractant protein-1 chemokine in treated retinas. Optokinetic studies were performed to determine visual acuity. RESULTS Systemic treatment with RTL220 prevented decreases in outer nuclear layer (ONL) thickness and showed a significantly higher number of nuclei than control rats treated with RTL101 or vehicle. RTL220 was also effective in protecting retinal vasculature from leakage and the formation of abnormal vascular complexes even when the treatment was administered after the degenerative process was initiated. Visual acuity measurement showed that rats treated with RTL220 performed significantly better than those with RTL101 and untreated age-matched controls at P60 and P90. Biodistribution studies showed that RTL220 cleared slowly from the administration site. Moreover, RTL220-treated retinas had a significantly reduced number of activated microglia in the subretinal space, decreased monocyte chemoattractant protein-1 production in the retina, inhibited T-cell responses, and reduced anti-interphotoreceptor retinoid binding protein autoantibody titers. Treatment with the control RTL101 (without a specific peptide tethered) or vehicle alone did not inhibit microglia activation or protect photoreceptors or vasculature. CONCLUSIONS RTL therapy augmented photoreceptor cell survival, protected vasculature, and increased visual function in the RTL rat. Targeting chronic autoimmunity with RTLs can be an effective therapeutic alternative in delaying retinal degeneration. Subcutaneous delivery of RTLs alone or combined with other drugs could be an attractive option for long-term therapy for retinal degenerative diseases.
Collapse
Affiliation(s)
- Grazyna Adamus
- Ocular Immunology Laboratory, Casey Eye Institute, Oregon Health & Science University, Portland, OR
| | - Shaomei Wang
- Ocular Immunology Laboratory, Casey Eye Institute, Oregon Health & Science University, Portland, OR
| | - Madison Kyger
- Ocular Immunology Laboratory, Casey Eye Institute, Oregon Health & Science University, Portland, OR
| | - Aneta Worley
- Ocular Immunology Laboratory, Casey Eye Institute, Oregon Health & Science University, Portland, OR
| | - Bin Lu
- Ocular Immunology Laboratory, Casey Eye Institute, Oregon Health & Science University, Portland, OR
| | - Gregory G. Burrows
- Tykeson MS Research Laboratory, Department of Neurology, Oregon Health & Science University, Portland, OR,Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR
| |
Collapse
|
97
|
Ong JM, da Cruz L. A review and update on the current status of stem cell therapy and the retina. Br Med Bull 2012; 102:133-46. [PMID: 22577179 DOI: 10.1093/bmb/lds013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION OR BACKGROUND Many diseases of the retina result in irreversible visual loss. Stem cell (SC) therapy is a rapidly developing field and represents a novel approach to replace non-functioning neuro-retinal cells. SOURCES OF DATA A systematic computerized literature search was conducted on PubMed (http://www.ncbi.nlm.nih.gov/pubmed/). AREAS OF AGREEMENT The use of stem cells (SCs) in animal models of retinal diseases has resulted in improvement in visual function and performance. SC therapy represents an exciting prospect in restoring vision. Areas of controversy The use of human embryonic SCs raises ethical concerns. GROWING POINTS Human trials using SCs in retinal diseases have recently been approved. AREAS TIMELY FOR DEVELOPING RESEARCH The success of SCs in retinal therapy depends not only on implanted cell survival, but also on how well SCs migrate, integrate and form synapses. Further research will be needed to overcome these hurdles.
Collapse
Affiliation(s)
- J M Ong
- National Institute of Health Research, Biomedical Research Centre, Moorfields Eye Hospital, London, UK.
| | | |
Collapse
|
98
|
Yanai A, Häfeli UO, Metcalfe AL, Soema P, Addo L, Gregory-Evans CY, Po K, Shan X, Moritz OL, Gregory-Evans K. Focused magnetic stem cell targeting to the retina using superparamagnetic iron oxide nanoparticles. Cell Transplant 2012; 21:1137-48. [PMID: 22405427 DOI: 10.3727/096368911x627435] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Developing new ways of delivering cells to diseased tissue will be a key factor in translating cell therapeutics research into clinical use. Magnetically targeting cells enables delivery of significant numbers of cells to key areas of specific organs. To demonstrate feasibility in neurological tissue, we targeted cells magnetically to the upper hemisphere of the rodent retina. Rat mesenchymal stem cells (MSCs) were magnetized using superparamagnetic iron oxide nanoparticles (SPIONs). In vitro studies suggested that magnetization with fluidMAG-D was well tolerated, that cells remained viable, and they retained their differentiation capabilities. FluidMAG-D-labeled MSCs were injected intravitreally or via the tail vein of the S334ter-4 transgenic rat model of retinal degeneration with or without placing a gold-plated neodymium disc magnet within the orbit, but outside the eye. Retinal flatmount and cryosection imaging demonstrated that after intravitreal injection cells localized to the inner retina in a tightly confined area corresponding to the position of the orbital magnet. After intravenous injection, similar retinal localization was achieved and remarkably was associated with a tenfold increase in magnetic MSC delivery to the retina. Cryosections demonstrated that cells had migrated into both the inner and outer retina. Magnetic MSC treatment with orbital magnet also resulted in significantly higher retinal concentrations of anti-inflammatory molecules interleukin-10 and hepatocyte growth factor. This suggested that intravenous MSC therapy also resulted in significant therapeutic benefit in the dystrophic retina. With minimal risk of collateral damage, these results suggest that magnetic cell delivery is the best approach for controlled delivery of cells to the outer retina-the focus for disease in age-related macular degeneration and retinitis pigmentosa.
Collapse
Affiliation(s)
- Anat Yanai
- Department of Ophthalmology & Visual Science, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Schmeer CW, Wohl SG, Isenmann S. Cell-replacement therapy and neural repair in the retina. Cell Tissue Res 2012; 349:363-74. [PMID: 22354517 DOI: 10.1007/s00441-012-1335-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 01/18/2012] [Indexed: 01/12/2023]
Abstract
Visual impairment severely affects the quality of life of patients and their families and is also associated with a deep economic impact. The most common pathologies responsible for visual impairment and legally defined blindness in developed countries include age-related macular degeneration, glaucoma and diabetic retinopathy. These conditions share common pathophysiological features: dysfunction and loss of retinal neurons. To date, two main approaches are being taken to develop putative therapeutic strategies: neuroprotection and cell replacement. Cell replacement is a novel therapeutic approach to restore visual capabilities to the degenerated adult neural retina and represents an emerging field of regenerative neurotherapy. The discovery of a population of proliferative cells in the mammalian retina has raised the possibility of harnessing endogenous retinal stem cells to elicit retinal repair. Furthermore, the development of suitable protocols for the reprogramming of differentiated somatic cells to a pluripotent state further increases the therapeutic potential of stem-cell-based technologies for the treatment of major retinal diseases. Stem-cell transplantation in animal models has been most effectively used for the replacement of photoreceptors, although this therapeutic approach is also being used for inner retinal pathologies. In this review, we discuss recent advances in the development of cell-replacement approaches for the treatment of currently incurable degenerative retinal diseases.
Collapse
Affiliation(s)
- Christian W Schmeer
- Hans Berger Clinic of Neurology, University Hospital Jena, Erlanger Allee 101, 07747 Jena, Germany.
| | | | | |
Collapse
|
100
|
Stanniocalcin-1 rescued photoreceptor degeneration in two rat models of inherited retinal degeneration. Mol Ther 2012; 20:788-97. [PMID: 22294148 DOI: 10.1038/mt.2011.308] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Oxidative stress and photoreceptor apoptosis are prominent features of many forms of retinal degeneration (RD) for which there are currently no effective therapies. We previously observed that mesenchymal stem/stromal cells reduce apoptosis by being activated to secrete stanniocalcin-1 (STC-1), a multifunctional protein that reduces oxidative stress by upregulating mitochondrial uncoupling protein-2 (UCP-2). Therefore, we tested the hypothesis that intravitreal injection of STC-1 can rescue photoreceptors. We first tested STC-1 in the rhodopsin transgenic rat characterized by rapid photoreceptor loss. Intravitreal STC-1 decreased the loss of photoreceptor nuclei and transcripts and resulted in measurable retinal function when none is otherwise present in this rapid degeneration. We then tested STC-1 in the Royal College of Surgeons (RCS) rat characterized by a slower photoreceptor degeneration. Intravitreal STC-1 reduced the number of pyknotic nuclei in photoreceptors, delayed the loss of photoreceptor transcripts, and improved function of rod photoreceptors. Additionally, STC-1 upregulated UCP-2 and decreased levels of two protein adducts generated by reactive oxygen species (ROS). Microarrays from the two models demonstrated that STC-1 upregulated expression of a similar profile of genes for retinal development and function. The results suggested that intravitreal STC-1 is a promising therapy for various forms of RD including retinitis pigmentosa and atrophic age-related macular degeneration (AMD).
Collapse
|