1
|
Shiri H, Javan M. Sox2-mediated transdifferentiation of hAT-MSCs into induced neural progenitor-like cells for remyelination therapies. Tissue Cell 2024; 91:102553. [PMID: 39255744 DOI: 10.1016/j.tice.2024.102553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/03/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
Mesenchymal stem cells (MSCs) are converted to neural cells using growth factors and chemicals. Although these neural cells are effective at modulating disease symptoms, they are less effective at replacing lost neural cells. Direct transdifferentiation seems to be a promising method for generating the required cells for regenerative medicine applications. Sox2 is a key transcription factor in neural progenitor (NP) fate determination and has been frequently used for transdifferentiating different cell types to NPs. Here, we demonstrated that the overexpression of a single transcription factor, Sox2, in human adipose tissue-derived mesenchymal stem cells (hAT-MSCs) led to the generation of induced NPs-like cells that were clonogenic, proliferative and passageable, and showed the potential to differentiate into three neural lineages. NPs are known as progenitors with the potential to differentiate into oligodendrocytes. In vivo, following transplantation into demyelinated adult mouse brains, they survived, differentiated and integrated into the adult brain while participating in the remyelination process and behavioral improvement. This report introduces a beneficial, low-cost and effective approach for generating NPs from an accessible adult source for autologous applications in treating neurodegenerative diseases, including remyelination therapies for multiple sclerosis and other demyelinating diseases.
Collapse
Affiliation(s)
- Hamed Shiri
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran; Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
2
|
Lapshin EV, Gershovich YG, Karabelsky AV. The Potential and Application of iPSCs in Gene and Cell Therapy for Retinopathies and Optic Neuropathies. Acta Naturae 2023; 15:56-64. [PMID: 38234607 PMCID: PMC10790360 DOI: 10.32607/actanaturae.25454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/05/2023] [Indexed: 01/19/2024] Open
Abstract
This review focuses on in vitro modeling of diseases and the development of therapeutic strategies using iPSCs for the two most common types of optical pathologies: hereditary neuropathies and retinopathies. Degeneration of retinal ganglion cells and the subsequent optic nerve atrophy leads to various types of neuropathies. Damage to photoreceptor cells or retinal pigment epithelium cells causes various retinopathies. Human iPSCs can be used as a model for studying the pathological foundations of diseases and for developing therapies to restore visual function. In recent years, significant progress has also been made in creating ganglionic and retinal organoids from iPSCs. Different research groups have published data pertaining to the potential of using iPSCs for the modeling of optic neuropathies such as glaucoma, Leber hereditary optic neuropathy, etc., including in the development of therapeutic approaches using gene editing tools.
Collapse
Affiliation(s)
- E. V. Lapshin
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, Krasnodar Region, Sirius, 354340 Russian Federation
| | - Y. G. Gershovich
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, Krasnodar Region, Sirius, 354340 Russian Federation
| | - A. V. Karabelsky
- Gene Therapy Department, Science Center for Translational Medicine, Sirius University of Science and Technology, Krasnodar Region, Sirius, 354340 Russian Federation
| |
Collapse
|
3
|
Ferroptosis inhibition by deferiprone, attenuates myelin damage and promotes neuroprotection in demyelinated optic nerve. Sci Rep 2022; 12:19630. [PMID: 36385152 PMCID: PMC9668997 DOI: 10.1038/s41598-022-24152-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease, which leads to focal demyelination in the brain and spinal cord. Studies showed that iron released during the course of myelin breakdown exacerbates tissue damage, which is in agreement with the features of iron-dependent cell death, ferroptosis. Here, we aimed to investigate the possible contribution of ferroptosis in the demyelinated optic nerve, and to explore the effectiveness of ferroptosis inhibitor, deferiprone (DFP), on the extent of demyelination, inflammation and axonal damage. For this purpose, focal demyelination was induced by injection of lysolecithin (LPC), into the optic nerve of male C57BL/6J mice. Afterward, optic nerves were harvested at different time points from as early as 6 h up to 7 days post-LPC injection. Next, to evaluate the effectiveness of DFP two groups of animals received daily intraperitoneal injection of DFP for 3 or 7 continuous days. Vehicle groups received saline. Iron deposition was observed at different time points post-LPC injection from 6 h to 7 days post injection. Examining ferroptosis markers showed a significant reduction in glutathione content along with increased level of malondialdehyde and upregulated ferroptosis marker genes at early time points after injection. Besides, DFP treatment during the inflammatory phase of the model resulted in decreased microgliosis and inflammation. Reduced demyelination, microgliosis and astrogliosis was shown in mice that received DFP for 7 days. Moreover, DFP protected against axonal damage and retinal ganglion cells loss. Our results suggest the possible contribution of ferroptosis pathway in the process of demyelination. The therapeutic strategies targeting iron deposition, e.g. DFP treatment might thus represent a promising therapeutic target for patients with MS.
Collapse
|
4
|
Failed remyelination of the nonhuman primate optic nerve leads to axon degeneration, retinal damages, and visual dysfunction. Proc Natl Acad Sci U S A 2022; 119:e2115973119. [PMID: 35235463 PMCID: PMC8916013 DOI: 10.1073/pnas.2115973119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Promotion of remyelination has become a new therapeutic avenue to prevent neuronal degeneration and promote recovery in white matter diseases, such as multiple sclerosis (MS). To date most of these strategies have been developed in short-lived rodent models of demyelination, which spontaneously repair. Well-defined nonhuman primate models closer to man would allow us to efficiently advance therapeutic approaches. Here we present a nonhuman primate model of optic nerve demyelination that recapitulates several features of MS lesions. The model leads to failed remyelination, associated with progressive axonal degeneration and visual dysfunction, thus providing the missing link to translate emerging preclinical therapies to the clinic for myelin disorders such as MS. White matter disorders of the central nervous system (CNS), such as multiple sclerosis (MS), lead to failure of nerve conduction and long-lasting neurological disabilities affecting a variety of sensory and motor systems, including vision. While most disease-modifying therapies target the immune and inflammatory response, the promotion of remyelination has become a new therapeutic avenue to prevent neuronal degeneration and promote recovery. Most of these strategies have been developed in short-lived rodent models of demyelination, which spontaneously repair and do not reflect the size, organization, and biology of the human CNS. Thus, well-defined nonhuman primate models are required to efficiently advance therapeutic approaches for patients. Here, we followed the consequence of long-term toxin-induced demyelination of the macaque optic nerve on remyelination and axon preservation, as well as its impact on visual functions. Findings from oculomotor behavior, ophthalmic examination, electrophysiology, and retinal imaging indicate visual impairment involving the optic nerve and retina. These visual dysfunctions fully correlated at the anatomical level, with sustained optic nerve demyelination, axonal degeneration, and alterations of the inner retinal layers. This nonhuman primate model of chronic optic nerve demyelination associated with axonal degeneration and visual dysfunction, recapitulates several key features of MS lesions and should be instrumental in providing the missing link to translate emerging repair promyelinating/neuroprotective therapies to the clinic for myelin disorders, such as MS.
Collapse
|
5
|
Nemati S, Seiedrazizadeh Z, Simorgh S, Hesaraki M, Kiani S, Javan M, Pakdel F, Satarian L. Mouse Degenerating Optic Axons Survived by Human Embryonic Stem Cell-Derived Neural Progenitor Cells. CELL JOURNAL 2022; 24:120-126. [PMID: 35451581 PMCID: PMC9035230 DOI: 10.22074/cellj.2022.7873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/04/2020] [Indexed: 11/06/2022]
Abstract
Objective Any damage to the optic nerve can potentially lead to degeneration of non-regenerating axons and ultimately death of retinal ganglion cells (RGCs) that in most cases, are not curable by surgery or medication. Neuroprotective functions of different types of stem cells in the nervous system have been evaluated in many studies investigating the effectiveness of these cells in various retinal disease models. Neural progenitor cells (NPCs) secrete an assortment of trophic factors that are vital to the protection of the visual system. We aimed to assess the therapeutic potentials of NPCs in an ONC mouse model. Materials and Methods In this experimental study, NPCs were produced using noggin and retinoic acid from human embryonic stem cells (hESCs). Fifty mice were divided into the following three groups: i. Intact , ii. Vehicle [optic nerve crush+Hank's balanced salt solution (HBSS)], and iii. Treatment (optic nerve crush+NPCs). The visual behavior of the mice was examined using the Visual Cliff test, and in terms of RGC numbers, they were assessed by Brn3a immunostaining and retrograde tracing using DiI injection. Results Intravenous injection of 50,000 NPCs through visual cliff did not produce any visual improvement. However, our data suggest that the RGCs protection was more than two-times in NPCs compared to the vehicle group as examined by Brn3a staining and retrograde tracing. Conclusion Our study indicated that intravenous injection of NPCs could protect RGCs probably mediated by trophic factors. Due to this ability and good manufacturing practices (GMP) grade production feasibility, NPCs may be used for optic nerve protection.
Collapse
Affiliation(s)
- Shiva Nemati
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Zahra Seiedrazizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Susan Simorgh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Sahar Kiani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Mohammad Javan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farzad Pakdel
- Ophthalmic Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Satarian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,P.O.Box: 16635-148Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan
Institute for Stem Cell Biology and TechnologyACECRTehranIran
| |
Collapse
|
6
|
Harvey JP, Sladen PE, Yu-Wai-Man P, Cheetham ME. Induced Pluripotent Stem Cells for Inherited Optic Neuropathies-Disease Modeling and Therapeutic Development. J Neuroophthalmol 2022; 42:35-44. [PMID: 34629400 DOI: 10.1097/wno.0000000000001375] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Inherited optic neuropathies (IONs) cause progressive irreversible visual loss in children and young adults. There are limited disease-modifying treatments, and most patients progress to become severely visually impaired, fulfilling the legal criteria for blind registration. The seminal discovery of the technique for reprogramming somatic nondividing cells into induced pluripotent stem cells (iPSCs) has opened several exciting opportunities in the field of ION research and treatment. EVIDENCE ACQUISITION A systematic review of the literature was conducted with PubMed using the following search terms: autosomal dominant optic atrophy, ADOA, dominant optic atrophy, DOA, Leber hereditary optic neuropathy, LHON, optic atrophy, induced pluripotent stem cell, iPSC, iPSC derived, iPS, stem cell, retinal ganglion cell, and RGC. Clinical trials were identified on the ClinicalTrials.gov website. RESULTS This review article is focused on disease modeling and the therapeutic strategies being explored with iPSC technologies for the 2 most common IONs, namely, dominant optic atrophy and Leber hereditary optic neuropathy. The rationale and translational advances for cell-based and gene-based therapies are explored, as well as opportunities for neuroprotection and drug screening. CONCLUSIONS iPSCs offer an elegant, patient-focused solution to the investigation of the genetic defects and disease mechanisms underpinning IONs. Furthermore, this group of disorders is uniquely amenable to both the disease modeling capability and the therapeutic potential that iPSCs offer. This fast-moving area will remain at the forefront of both basic and translational ION research in the coming years, with the potential to accelerate the development of effective therapies for patients affected with these blinding diseases.
Collapse
Affiliation(s)
- Joshua Paul Harvey
- UCL Institute of Ophthalmology (JPH, PES, PY-W-M, MC), London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust (JPH, PY-W-M), London, United Kingdom; Department of Clinical Neurosciences (PY-W-M), Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom; and Department of Clinical Neurosciences (PY-W-M), John van Geest Centre for Brain Repair and MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
7
|
Esmaeilnejad S, Semnanian S, Javan M. Metformin Protects Myelin from Degeneration in A Mouse Model of Iysophosphatidylcholine-Induced Demyelination in The Optic Chiasm. CELL JOURNAL 2021; 23:119-128. [PMID: 33650828 PMCID: PMC7944130 DOI: 10.22074/cellj.2021.7174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022]
Abstract
Objective Multiple sclerosis (MS) is a demyelinating disease of the central nervous system. The autoimmune
pathology and long-term inflammation lead to substantial demyelination. These events lead to a substantial loss
of oligodendrocytes (OLs), which in a longer period, results in axonal loss and long-term disabilities. Neural cells
protection approaches decelerate or inhibit the disease progress to avoid further disability. Previous studies showed
that metformin has beneficial effects against neurodegenerative conditions. In this experimental study, we examined
possible protective effects of metformin on toxin-induced myelin destruction in adult mice brains.
Materials and Methods Lysophosphatidylcholine (LPC) was used to induce demyelination in mice optic chiasm. We
examined the extent of demyelination at different time points post LPC injection using myelin staining and evaluated the
severity of inflammation. Functional state of optic pathway was evaluated by visual evoked potential (VEP) recording.
Results Metformin attenuated LPC-induced demyelination (P<0.05) and inflammation (P<0.05) and protected against
significant decrease (P<0.05) in functional conductivity of optic tract. These data indicated that metformin administration
attenuates the myelin degeneration following LPC injection which led to functional enhancement.
Conclusion Our findings suggest metformin for combination therapy for patients suffering from the myelin degenerative
diseases, especially multiple sclerosis; however, additional mechanistic studies are required.
Collapse
Affiliation(s)
- Saman Esmaeilnejad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran. .,Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
8
|
Dehghan S, Aref E, Raoufy MR, Javan M. An optimized animal model of lysolecithin induced demyelination in optic nerve; more feasible, more reproducible, promising for studying the progressive forms of multiple sclerosis. J Neurosci Methods 2021; 352:109088. [PMID: 33508411 DOI: 10.1016/j.jneumeth.2021.109088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/01/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Multiple Sclerosis (MS) is a demyelinating disease leading to long-term neurological deficit due to unsuccessful remyelination and axonal loss. Currently, there are no satisfactory treatments for progressive MS somewhat due to the lack of an adequate animal model for studying the mechanisms of disease progression and screening new drugs. NEW METHOD Lysolecithin (LPC) or agarose-gel loaded LPC (AL-LPC) were applied to mouse optic nerve behind the globe via a minor surgery. Agarose loading was used to achieve longer time of LPC exposure and subsequently long-lasting demyelination. RESULTS The lesion sites characterized by luxol fast blue (LFB), FluoroMyelin, Bielschowsky's staining, and immunostaining showed extensive demyelination and axonal damage. The loss of Retinal ganglion cells (RGCs) in the corresponding retinal layer was shown by immunostaining and H&E staining. Visual evoked potential (VEP) recordings showed a significant increase in the latency of the P1 wave and a decrease in the amplitude of the P1N1 wave. COMPARISON WITH EXISTING METHODS The new approach with a very minor surgery seems to be more feasible and reproducible compared to stereotaxic LPC injection to optic chiasm. Our data revealed prolonged demyelination, axonal degeneration and RGCs loss in both AL-LPC and LPC groups; however, these pathologies were more extensive in the AL-LPC group. CONCLUSION The optimized model provides a longer demyelination time frame and axonal damage followed by RGC degeneration; which is of exceptional interest in investigating axonal degeneration mechanisms and screening the new drugs for progressive MS.
Collapse
Affiliation(s)
- Samaneh Dehghan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran
| | - Ehsan Aref
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, P.O. Box:14115-331, Tehran, Iran; Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box:14115-331, Tehran, Iran.
| |
Collapse
|
9
|
Hereditary Optic Neuropathies: Induced Pluripotent Stem Cell-Based 2D/3D Approaches. Genes (Basel) 2021; 12:genes12010112. [PMID: 33477675 PMCID: PMC7831942 DOI: 10.3390/genes12010112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/10/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Inherited optic neuropathies share visual impairment due to the degeneration of retinal ganglion cells (RGCs) as the hallmark of the disease. This group of genetic disorders are caused by mutations in nuclear genes or in the mitochondrial DNA (mtDNA). An impaired mitochondrial function is the underlying mechanism of these diseases. Currently, optic neuropathies lack an effective treatment, and the implementation of induced pluripotent stem cell (iPSC) technology would entail a huge step forward. The generation of iPSC-derived RGCs would allow faithfully modeling these disorders, and these RGCs would represent an appealing platform for drug screening as well, paving the way for a proper therapy. Here, we review the ongoing two-dimensional (2D) and three-dimensional (3D) approaches based on iPSCs and their applications, taking into account the more innovative technologies, which include tissue engineering or microfluidics.
Collapse
|
10
|
Li J, Bai X, Guan X, Yuan H, Xu X. Treatment of Optic Canal Decompression Combined with Umbilical Cord Mesenchymal Stem (Stromal) Cells for Indirect Traumatic Optic Neuropathy: A Phase 1 Clinical Trial. Ophthalmic Res 2020; 64:398-404. [PMID: 33091914 DOI: 10.1159/000512469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/21/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE This study was aimed to investigate the safety and feasibility of umbilical cord-derived mesenchymal stem cell (MSC) transplantation in patients with traumatic optic neuropathy (TON). METHODS This is a single-center, prospective, open-labeled phase 1 study that enrolled 20 patients with TON. Patients consecutively underwent either optic canal decompression combined with MSC local implantation treatment (group 1) or only optic canal decompression (group 2). Patients were evaluated on the first day, seventh day, first month, third month, and sixth month postoperatively. Adverse events, such as fever, urticarial lesions, nasal infection, and death, were recorded at each visit. The primary outcome was changes in best-corrected visual acuity. The secondary outcomes were changes in color vision, relative afferent pupillary defect, and flash visual evoked potential. RESULTS All 20 patients completed the 6-month follow-up. None of them had any systemic or ocular complications. The change in best-corrected visual acuity at follow-up was not significantly different between group 1 and group 2 (p > 0.05); however, group 1 showed better visual outcome than group 2. Both groups showed significant improvements in vision compared with the baseline (p < 0.05); however, there were no statistically significant differences between the groups (p > 0.05). In addition, no adverse events related to local transplantation were observed in the patients. CONCLUSIONS A single, local MSC transplantation in the optic nerve is safe for patients with TON.
Collapse
Affiliation(s)
- Jia Li
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xu Bai
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoyue Guan
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, China
| | - Hongfeng Yuan
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiang Xu
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China,
| |
Collapse
|
11
|
Mao Y, Ma J, Xia Y, Xie X. The Overexpression of Epidermal Growth Factor (EGF) in HaCaT Cells Promotes the Proliferation, Migration, Invasion and Transdifferentiation to Epidermal Stem Cell Immunophenotyping of Adipose-Derived Stem Cells (ADSCs). Int J Stem Cells 2020; 13:93-103. [PMID: 32114740 PMCID: PMC7119215 DOI: 10.15283/ijsc18146] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives The application of adipose derived stem cells (ADSCs) in skin repair has attracted much attention nowadays. Epidermal growth factor (EGF) participates in the progress of skin proliferation, differentiation and so forth. We aimed to explore the role of EGF in the proliferation, invasion, migration and transdifferentiation into epidermal cell phenotypes of ADSCs. Methods and Results ADSCs were extracted from adipose tissues from patient. Immunophenotyping was determined by flow cytometry. Overexpressed EGF or siEGF was transfected by lentiviruses. EGF was determined by enzyme linked immunosorbent assay (ELISA) or western blot. ADSCs and HaCaT cells were co-cultured by Transwell chambers. Conditioned medium (CM) was obtained from cultured HaCaT cells and used for the culturing of ADSCs. Cell viability was tested by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Invasion rate was measured by Transwell invasion assay and migration rate by wound healing test. mRNA and protein levels were measured by qPCR and western blot respectively. The extracted cells from adipose tissues were identified as ADSCs by morphology and immunophenotyping. The expression of EGF was up or down regulated constantly in HaCaT cell line after transfection. EGF overexpression upregulated the proliferation, migration and invasion rates of ADSCs, and EGF expression regulated the expression of cytokeratin-19 (CK19) and integrin-β as well. Conclusions EGF could be served as a stimulus to promote the proliferation, migration, and invasion as well as the transdifferentiation into epidermal stem cell immunophenotyping of ADSCs. The results showed that EGF had a promising effect on the repair of skin wound.
Collapse
Affiliation(s)
- Yueping Mao
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianchi Ma
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yue Xia
- Department of Dermatology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyuan Xie
- Department of Dermatology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
12
|
Niknam P, Raoufy MR, Fathollahi Y, Javan M. Modulating proteoglycan receptor PTPσ using intracellular sigma peptide improves remyelination and functional recovery in mice with demyelinated optic chiasm. Mol Cell Neurosci 2019; 99:103391. [DOI: 10.1016/j.mcn.2019.103391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 11/29/2022] Open
|
13
|
Yazdi A, Ghasemi‐Kasman M, Javan M. Possible regenerative effects of fingolimod (FTY720) in multiple sclerosis disease: An overview on remyelination process. J Neurosci Res 2019; 98:524-536. [DOI: 10.1002/jnr.24509] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Azadeh Yazdi
- Department of Physiology, School of Medicine Isfahan University of Medical Sciences Isfahan Iran
| | - Maryam Ghasemi‐Kasman
- Cellular and Molecular Biology Research Center Health Research Institute, Babol University of Medical Sciences Babol Iran
- Neuroscience Research Center Health Research Institute, Babol University of Medical Sciences Babol Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences Tarbiat Modares University Tehran Iran
- Department of Brain and Cognitive Sciences, Cell Science Research Center Royan Institute for Stem Cell Biology and Technology, ACECR Tehran Iran
| |
Collapse
|
14
|
Bucan V, Fliess M, Schnabel R, Peck CT, Vaslaitis D, Fülbier A, Reimers K, Strauss S, Vogt PM, Radtke C. In vitro enhancement and functional characterization of neurite outgrowth by undifferentiated adipose-derived stem cells. Int J Mol Med 2018; 43:593-602. [PMID: 30431135 DOI: 10.3892/ijmm.2018.3979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/22/2018] [Indexed: 11/05/2022] Open
Abstract
Adipose‑derived stem cells (ASCs) can easily be obtained and expanded in vitro for use in autologous cell therapy. Via their production of cytokines and neurotrophic factors, transplanted ASCs provide neuroprotection, neovascularization and induction of axonal sprouting. However, the influencing mechanism of undifferentiated ASCs on nerve regeneration is currently only partially understood. In the present study, undifferentiated ASCs and cutaneous primary afferent dorsal root ganglion (DRG) neurons were co‑cultured in order to investigate their interaction. ASCs were isolated from adult rat fat tissue. The presence of characteristic stem cell markers was determined by flow cytometry in three subsequent passages. Adipogenic, osteogenic, chondrogenic and glial differentiation was performed in order to evaluate their differentiation capacity. A direct co‑culture system with DRG cells was established to determine the effect of undifferentiated pluripotent ASCs on neurite elongation. Neurite outgrowth, length and number was examined in the co‑culture and compared with single‑culture cells and cells stimulated with nerve growth factor (NGF). In ASC cultures, NGF expression was assessed by ELISA. The present results demonstrated that the specific mesenchymal stem cell surface markers CD44, CD73 and CD90 were detected in all three subsequent passages of the isolated ASCs. In accordance, ASC differentiation into adipogenic, osteogenic, chondrogenic and Schwann cell phenotype was conducted successfully. Neurite outgrowth of DRG neurons was enhanced following co‑culture with ASCs, resulting in increased neurite length after 24 h of cultivation. Furthermore, neurite outgrowth of DRG neurons was directed towards the undifferentiated ASC and direct cell‑to‑cell contact was observed. In summary, the results of the present study revealed an interaction between the two cell types with guidance of neurite growth towards the undifferentiated ASC. These findings suggest that the use of undifferentiated ASC optimizing tissue‑engineered constructs may be promising for peripheral nerve repair.
Collapse
Affiliation(s)
- Vesna Bucan
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Malte Fliess
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Reinhild Schnabel
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Claas-Tido Peck
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Desiree Vaslaitis
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Angela Fülbier
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Kerstin Reimers
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Sarah Strauss
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Peter M Vogt
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Christine Radtke
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| |
Collapse
|
15
|
Rabesandratana O, Goureau O, Orieux G. Pluripotent Stem Cell-Based Approaches to Explore and Treat Optic Neuropathies. Front Neurosci 2018; 12:651. [PMID: 30294255 PMCID: PMC6158340 DOI: 10.3389/fnins.2018.00651] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Sight is a major sense for human and visual impairment profoundly affects quality of life, especially retinal degenerative diseases which are the leading cause of irreversible blindness worldwide. As for other neurodegenerative disorders, almost all retinal dystrophies are characterized by the specific loss of one or two cell types, such as retinal ganglion cells, photoreceptor cells, or retinal pigmented epithelial cells. This feature is a critical point when dealing with cell replacement strategies considering that the preservation of other cell types and retinal circuitry is a prerequisite. Retinal ganglion cells are particularly vulnerable to degenerative process and glaucoma, the most common optic neuropathy, is a frequent retinal dystrophy. Cell replacement has been proposed as a potential approach to take on the challenge of visual restoration, but its application to optic neuropathies is particularly challenging. Many obstacles need to be overcome before any clinical application. Beyond their survival and differentiation, engrafted cells have to reconnect with both upstream synaptic retinal cell partners and specific targets in the brain. To date, reconnection of retinal ganglion cells with distal central targets appears unrealistic since central nervous system is refractory to regenerative processes. Significant progress on the understanding of molecular mechanisms that prevent central nervous system regeneration offer hope to overcome this obstacle in the future. At the same time, emergence of reprogramming of human somatic cells into pluripotent stem cells has facilitated both the generation of new source of cells with therapeutic potential and the development of innovative methods for the generation of transplantable cells. In this review, we discuss the feasibility of stem cell-based strategies applied to retinal ganglion cells and optic nerve impairment. We present the different strategies for the generation, characterization and the delivery of transplantable retinal ganglion cells derived from pluripotent stem cells. The relevance of pluripotent stem cell-derived retinal organoid and retinal ganglion cells for disease modeling or drug screening will be also introduced in the context of optic neuropathies.
Collapse
Affiliation(s)
| | - Olivier Goureau
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Gaël Orieux
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
16
|
Yazdi A, Mokhtarzadeh Khanghahi A, Baharvand H, Javan M. Fingolimod Enhances Oligodendrocyte Differentiation of Transplanted Human Induced Pluripotent Stem Cell-Derived Neural Progenitors. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:1444-1457. [PMID: 30568702 PMCID: PMC6269577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease which affects myelin in the central nervous system (CNS) and leads to serious disability. Currently available treatments for MS mainly suppress the immune system. Regenerative medicine-based approaches attempt to increase myelin repair by targeting endogenous progenitors or transplanting stem cells or their derivatives. Fingolimod exerts anti-inflammatory effects and directly affects neural cells. In this study we assessed the effect of fingolimod on transplanted human induced pluripotent stem cell derived neural progenitors (hiPSC-NPs). hiPSC-NPs were labeled by green fluorescence protein (GFP) and transplanted into the corpus callosum of mice which were chronically demyelinated after cuprizone (CPZ) feedings for 10 weeks. The animals received fingolimod from 1 day prior to NPs transplantation via gavage as well as daily intraperitoneal cyclosporine A from 2 days before cell transplantation until the time of sampling. At either 7 or 21 days after NPs transplantation, the animals were sacrificed and their brains were histologically evaluated for the number of transplanted cells and their fate. In the animals treated with fingolimod, we observed higher numbers of NPs within the injection site compared to the animals who did not receive fingolimod showing that hiPSC- NPs were more efficiently differentiated to the oligodendrocyte lineage. These data have suggested that repetitive treatment with fingolimod, beside its anti-inflammatory effect, may enhance the survival and differentiation of transplanted NPs to oligodendrocyte lineage cells to participate in myelin repair.
Collapse
Affiliation(s)
- Azadeh Yazdi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Akram Mokhtarzadeh Khanghahi
- Department of Brain Sciences and Cognition, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Hossein Baharvand
- Department of Brain Sciences and Cognition, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. ,Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran. ,Department of Brain Sciences and Cognition, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. ,Corresponding author: E-mail:
| |
Collapse
|
17
|
The use of induced pluripotent stem cells for studying and treating optic neuropathies. Curr Opin Organ Transplant 2017; 21:484-9. [PMID: 27517502 DOI: 10.1097/mot.0000000000000348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW The present review aims to provide an update of applications of induced pluripotent stem cells (iPSCs) for disease modeling, cell/gene therapy, and drug screening for optic neuropathies. RECENT FINDINGS Degeneration of retinal ganglion cells (RGCs) is a characteristic of optic neuropathies. Human iPSCs can serve as a model to investigate disease pathology and potential repair mechanisms. In recent years, significant progress has been made in generating RGCs from iPSCs. Various groups have reported the potential of iPSCs for modeling optic neuropathies, such as glaucoma. The literature also highlights the potential to use iPSC-derived cells for high-throughput drug and toxicity screening. SUMMARY The present review summarizes current work in the field of iPSCs in optic neuropathies. Future studies to characterize iPSC-derived RGCs in a more in-depth manner will help expand the use of iPSCs to model and treat optic neuropathic diseases. Furthermore, iPSC modeling can be used in drug development by offering a new avenue to test novel therapeutic drugs for optic neuropathies.
Collapse
|
18
|
Wegscheid ML, Anastasaki C, Gutmann DH. Human stem cell modeling in neurofibromatosis type 1 (NF1). Exp Neurol 2017; 299:270-280. [PMID: 28392281 DOI: 10.1016/j.expneurol.2017.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/15/2017] [Accepted: 04/05/2017] [Indexed: 01/03/2023]
Abstract
The future of precision medicine is heavily reliant on the use of human tissues to identify the key determinants that account for differences between individuals with the same disorder. This need is exemplified by the neurofibromatosis type 1 (NF1) neurogenetic condition. As such, individuals with NF1 are born with a germline mutation in the NF1 gene, but may develop numerous distinct neurological problems, ranging from autism and attention deficit to brain and peripheral nerve sheath tumors. Coupled with accurate preclinical mouse models, the availability of NF1 patient-derived induced pluripotent stem cells (iPSCs) provides new opportunities to define the critical factors that underlie NF1-associated nervous system disease pathogenesis and progression. In this review, we discuss the generation and potential applications of iPSC technology to the study of NF1.
Collapse
Affiliation(s)
- Michelle L Wegscheid
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States.
| |
Collapse
|
19
|
Selvaraj K, Gowthamarajan K, Karri VVSR, Barauah UK, Ravisankar V, Jojo GM. Current treatment strategies and nanocarrier based approaches for the treatment and management of diabetic retinopathy. J Drug Target 2017; 25:386-405. [DOI: 10.1080/1061186x.2017.1280809] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Kousalya Selvaraj
- Department of Pharmaceutics, JSS College of Pharmacy, Ootacamund, JSS University, Mysuru, India
| | - Kuppusamy Gowthamarajan
- Department of Pharmaceutics, JSS College of Pharmacy, Ootacamund, JSS University, Mysuru, India
| | | | - Uday K. Barauah
- Department of Pharmaceutics, JSS College of Pharmacy, Ootacamund, JSS University, Mysuru, India
| | - Vanka Ravisankar
- Department of Pharmaceutics, JSS College of Pharmacy, Ootacamund, JSS University, Mysuru, India
| | - Gifty M. Jojo
- Department of Pharmaceutics, JSS College of Pharmacy, Ootacamund, JSS University, Mysuru, India
| |
Collapse
|
20
|
Induced Pluripotent Stem Cells: Development in the Ophthalmologic Field. Stem Cells Int 2016; 2016:2361763. [PMID: 27594887 PMCID: PMC4995319 DOI: 10.1155/2016/2361763] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/30/2016] [Indexed: 12/22/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) are a type of stem cells that can be derived from human somatic cells by introducing certain transcription factors. Induced pluripotent stem cells can divide indefinitely and are able to differentiate into every cell type, which make them viable for transplantation and individual disease modeling. Recently, various ocular cells, including corneal epithelial-like cells, retinal pigment epithelium (RPE) cells displaying functions similar to native RPE, photoreceptors, and retinal ganglion cells, have all been successfully derived from iPSCs. Transplantation of these cells in animal models showed great promise for reversing blindness, and the first clinical trial on humans started in 2013. Despite these promising results, more research is in demand for preventing inadvertent tumor growth, developing precise functionality of the cells, and promoting integration into the host tissue.
Collapse
|
21
|
Wu N, Wang Y, Yang L, Cho KS. Signaling Networks of Retinal Ganglion Cell Formation and the Potential Application of Stem Cell–Based Therapy in Retinal Degenerative Diseases. Hum Gene Ther 2016; 27:609-20. [PMID: 27466076 DOI: 10.1089/hum.2016.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Nan Wu
- 1 Department of Ophthalmology, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Yi Wang
- 1 Department of Ophthalmology, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Lanbo Yang
- 2 Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, Massachusetts
| | - Kin-Sang Cho
- 2 Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
22
|
Venugopalan P, Wang Y, Nguyen T, Huang A, Muller KJ, Goldberg JL. Transplanted neurons integrate into adult retinas and respond to light. Nat Commun 2016; 7:10472. [PMID: 26843334 PMCID: PMC4742891 DOI: 10.1038/ncomms10472] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 12/16/2015] [Indexed: 12/12/2022] Open
Abstract
Retinal ganglion cells (RGCs) degenerate in diseases like glaucoma and are not replaced in adult mammals. Here we investigate whether transplanted RGCs can integrate into the mature retina. We have transplanted GFP-labelled RGCs into uninjured rat retinas in vivo by intravitreal injection. Transplanted RGCs acquire the general morphology of endogenous RGCs, with axons orienting towards the optic nerve head of the host retina and dendrites growing into the inner plexiform layer. Preliminary data show in some cases GFP(+) axons extending within the host optic nerves and optic tract, reaching usual synaptic targets in the brain, including the lateral geniculate nucleus and superior colliculus. Electrophysiological recordings from transplanted RGCs demonstrate the cells' electrical excitability and light responses similar to host ON, ON-OFF and OFF RGCs, although less rapid and with greater adaptation. These data present a promising approach to develop cell replacement strategies in diseased retinas with degenerating RGCs.
Collapse
Affiliation(s)
- Praseeda Venugopalan
- Neuroscience Program, University of Miami, Miami, Florida 33136, USA.,Shiley Eye Center, University of California, San Diego, California 92093, USA
| | - Yan Wang
- Shiley Eye Center, University of California, San Diego, California 92093, USA
| | - Tu Nguyen
- Shiley Eye Center, University of California, San Diego, California 92093, USA
| | - Abigail Huang
- Shiley Eye Center, University of California, San Diego, California 92093, USA
| | - Kenneth J Muller
- Neuroscience Program, University of Miami, Miami, Florida 33136, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Jeffrey L Goldberg
- Neuroscience Program, University of Miami, Miami, Florida 33136, USA.,Shiley Eye Center, University of California, San Diego, California 92093, USA.,Byers Eye Institute, Department of Ophthalmology, Stanford University, Stanford, California 94303, USA
| |
Collapse
|
23
|
Dehghan S, Hesaraki M, Soleimani M, Mirnajafi-Zadeh J, Fathollahi Y, Javan M. Oct4 transcription factor in conjunction with valproic acid accelerates myelin repair in demyelinated optic chiasm in mice. Neuroscience 2016; 318:178-89. [PMID: 26804242 DOI: 10.1016/j.neuroscience.2016.01.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 12/23/2015] [Accepted: 01/13/2016] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis is a demyelinating disease with severe neurological symptoms due to blockage of signal conduction in affected axons. Spontaneous remyelination via endogenous progenitors is limited and eventually fails. Recent reports showed that forced expression of some transcription factors within the brain converted somatic cells to neural progenitors and neuroblasts. Here, we report the effect of valproic acid (VPA) along with forced expression of Oct4 transcription factor on lysolecithin (LPC)-induced experimental demyelination. Mice were gavaged with VPA for one week, and then inducible Oct4 expressing lentiviral particles were injected into the lateral ventricle. After one-week induction of Oct4, LPC was injected into the optic chiasm. Functional remyelination was assessed by visual-evoked potential (VEP) recording. Myelination level was studied using FluoroMyelin staining and immunohistofluorescent (IHF) against proteolipid protein (PLP). IHF was also performed to detect Oct4 and SSEA1 as pluripotency markers and Olig2, Sox10, CNPase and PDGFRα as oligodendrocyte lineage markers. One week after injection of Oct4 expressing vector, pluripotency markers SSEA1 and Oct4 were detected in the rims of the 3rd ventricle. LPC injection caused extensive demyelination and significantly delayed the latency of VEP wave. Animals pre-treated with VPA+Oct4 expressing vector, showed faster recovery in the VEP latency and enhanced myelination. Immunostaining against oligodendrocyte lineage markers showed an increased number of Sox10+ and myelinating cells. Moreover, transdifferentiation of some Oct4-transfected cells (GFP+ cells) to Olig2+ and CNPase+ cells was confirmed by immunostaining. One-week administration of VPA followed by one-week forced expression of Oct4 enhanced myelination by converting transduced cells to myelinating oligodendrocytes. This finding seems promising for enhancing myelin repair within the adult brains.
Collapse
Affiliation(s)
- S Dehghan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - M Hesaraki
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - M Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - J Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Y Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - M Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
24
|
Alonso-Alonso ML, Srivastava GK. Current focus of stem cell application in retinal repair. World J Stem Cells 2015; 7:641-648. [PMID: 25914770 PMCID: PMC4404398 DOI: 10.4252/wjsc.v7.i3.641] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/06/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
The relevance of retinal diseases, both in society’s economy and in the quality of people’s life who suffer with them, has made stem cell therapy an interesting topic for research. Embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adipose derived mesenchymal stem cells (ADMSCs) are the focus in current endeavors as a source of different retinal cells, such as photoreceptors and retinal pigment epithelial cells. The aim is to apply them for cell replacement as an option for treating retinal diseases which so far are untreatable in their advanced stage. ESCs, despite the great potential for differentiation, have the dangerous risk of teratoma formation as well as ethical issues, which must be resolved before starting a clinical trial. iPSCs, like ESCs, are able to differentiate in to several types of retinal cells. However, the process to get them for personalized cell therapy has a high cost in terms of time and money. Researchers are working to resolve this since iPSCs seem to be a realistic option for treating retinal diseases. ADMSCs have the advantage that the procedures to obtain them are easier. Despite advancements in stem cell application, there are still several challenges that need to be overcome before transferring the research results to clinical application. This paper reviews recent research achievements of the applications of these three types of stem cells as well as clinical trials currently based on them.
Collapse
|
25
|
Fairbairn NG, Meppelink AM, Ng-Glazier J, Randolph MA, Winograd JM. Augmenting peripheral nerve regeneration using stem cells: A review of current opinion. World J Stem Cells 2015; 7:11-26. [PMID: 25621102 PMCID: PMC4300921 DOI: 10.4252/wjsc.v7.i1.11] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 09/18/2014] [Accepted: 10/27/2014] [Indexed: 02/06/2023] Open
Abstract
Outcomes following peripheral nerve injury remain frustratingly poor. The reasons for this are multifactorial, although maintaining a growth permissive environment in the distal nerve stump following repair is arguably the most important. The optimal environment for axonal regeneration relies on the synthesis and release of many biochemical mediators that are temporally and spatially regulated with a high level of incompletely understood complexity. The Schwann cell (SC) has emerged as a key player in this process. Prolonged periods of distal nerve stump denervation, characteristic of large gaps and proximal injuries, have been associated with a reduction in SC number and ability to support regenerating axons. Cell based therapy offers a potential therapy for the improvement of outcomes following peripheral nerve reconstruction. Stem cells have the potential to increase the number of SCs and prolong their ability to support regeneration. They may also have the ability to rescue and replenish populations of chromatolytic and apoptotic neurons following axotomy. Finally, they can be used in non-physiologic ways to preserve injured tissues such as denervated muscle while neuronal ingrowth has not yet occurred. Aside from stem cell type, careful consideration must be given to differentiation status, how stem cells are supported following transplantation and how they will be delivered to the site of injury. It is the aim of this article to review current opinions on the strategies of stem cell based therapy for the augmentation of peripheral nerve regeneration.
Collapse
|
26
|
Pourabdolhossein F, Mozafari S, Morvan-Dubois G, Mirnajafi-Zadeh J, Lopez-Juarez A, Pierre-Simons J, Demeneix BA, Javan M. Nogo receptor inhibition enhances functional recovery following lysolecithin-induced demyelination in mouse optic chiasm. PLoS One 2014; 9:e106378. [PMID: 25184636 PMCID: PMC4153612 DOI: 10.1371/journal.pone.0106378] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 07/29/2014] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Inhibitory factors have been implicated in the failure of remyelination in demyelinating diseases. Myelin associated inhibitors act through a common receptor called Nogo receptor (NgR) that plays critical inhibitory roles in CNS plasticity. Here we investigated the effects of abrogating NgR inhibition in a non-immune model of focal demyelination in adult mouse optic chiasm. METHODOLOGY/PRINCIPAL FINDINGS A focal area of demyelination was induced in adult mouse optic chiasm by microinjection of lysolecithin. To knock down NgR levels, siRNAs against NgR were intracerebroventricularly administered via a permanent cannula over 14 days, Functional changes were monitored by electrophysiological recording of latency of visual evoked potentials (VEPs). Histological analysis was carried out 3, 7 and 14 days post demyelination lesion. To assess the effect of NgR inhibition on precursor cell repopulation, BrdU was administered to the animals prior to the demyelination induction. Inhibition of NgR significantly restored VEPs responses following optic chiasm demyelination. These findings were confirmed histologically by myelin specific staining. siNgR application resulted in a smaller lesion size compared to control. NgR inhibition significantly increased the numbers of BrdU+/Olig2+ progenitor cells in the lesioned area and in the neurogenic zone of the third ventricle. These progenitor cells (Olig2+ or GFAP+) migrated away from this area as a function of time. CONCLUSIONS/SIGNIFICANCE Our results show that inhibition of NgR facilitate myelin repair in the demyelinated chiasm, with enhanced recruitment of proliferating cells to the lesion site. Thus, antagonizing NgR function could have therapeutic potential for demyelinating disorders such as Multiple Sclerosis.
Collapse
Affiliation(s)
- Fereshteh Pourabdolhossein
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Sabah Mozafari
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghislaine Morvan-Dubois
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alejandra Lopez-Juarez
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Jacqueline Pierre-Simons
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A. Demeneix
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- * E-mail:
| |
Collapse
|