1
|
Le N, Awad S, Palazzo I, Hoang T, Blackshaw S. Viral-mediated Pou5f1 (Oct4) overexpression and inhibition of Notch signaling synergistically induce neurogenic competence in mammalian Müller glia. eLife 2025; 14:RP106450. [PMID: 40388211 PMCID: PMC12088672 DOI: 10.7554/elife.106450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025] Open
Abstract
Retinal Müller glia in cold-blooded vertebrates can reprogram into neurogenic progenitors to replace neurons lost to injury, but mammals lack this ability. While recent studies have shown that transgenic overexpression of neurogenic bHLH factors and glial-specific disruption of NFI family transcription factors and Notch signaling induce neurogenic competence in mammalian Müller glia, induction of neurogenesis in wildtype glia has thus far proven elusive. Here, we report that viral-mediated overexpression of the pluripotency factor Pou5f1 (Oct4) induces transdifferentiation of mouse Müller glia into bipolar neurons, and synergistically stimulates glial-derived neurogenesis in parallel with Notch loss of function. Single-cell multiomic analysis shows that Pou5f1 overexpression leads to widespread changes in gene expression and chromatin accessibility, inducing activity of both the neurogenic transcription factor Rfx4 and the Yamanaka factors Sox2 and Klf4. This study demonstrates that viral-mediated overexpression of Pou5f1 induces neurogenic competence in adult mouse Müller glia, identifying mechanisms that could be used in cell-based therapies for treating retinal dystrophies.
Collapse
Affiliation(s)
- Nguyet Le
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Sherine Awad
- Department of Ophthalmology and Visual Sciences, University of Michigan School of MedicineAnn ArborUnited States
- Department of Cell and Developmental Biology, University of Michigan School of MedicineAnn ArborUnited States
- Michigan Neuroscience Institute, University of Michigan School of MedicineAnn ArborUnited States
| | - Isabella Palazzo
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Thanh Hoang
- Department of Ophthalmology and Visual Sciences, University of Michigan School of MedicineAnn ArborUnited States
- Department of Cell and Developmental Biology, University of Michigan School of MedicineAnn ArborUnited States
- Michigan Neuroscience Institute, University of Michigan School of MedicineAnn ArborUnited States
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Ophthalmology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
2
|
Rayatpour A, Foolad F, Javan M. Deferiprone promoted remyelination and functional recovery through enhancement of oligodendrogenesis in experimental demyelination animal model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:715-727. [PMID: 39046528 DOI: 10.1007/s00210-024-03314-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Remyelination refers to myelin regeneration, which reestablishes metabolic supports to axons. However, remyelination often fails in multiple sclerosis (MS), leading to chronic demyelination and axonal degeneration. Therefore, pharmacological approaches toward enhanced remyelination are highly demanded. Recently, deferiprone (DFP) was reported to exert neuroprotective effects, besides its iron-chelating ability. Since DFP exerts protective effects through various mechanisms, which share several factors with myelin formation process, we aimed to investigate the effects of DFP treatment on remyelination. Focal demyelination was induced by injection of lysolecithin, into the optic nerve of male C57BL/6J mice. The animals were treated with DFP/vehicle, starting from day 7 and continued during the myelin repair period. Histopathological, electrophysiological, and behavioral studies were used to evaluate the outcomes. Results showed that DFP treatment enhanced remyelination, decreased g-ratio and increased myelin thickness. At the mechanistic level, DFP enhanced oligodendrogenesis and ameliorated gliosis during the remyelination period. Furthermore, our results indicated that enhanced remyelination led to functional recovery as evaluated by the electrophysiological and behavioral tests. Even though the exact molecular mechanisms by which DFP-enhanced myelin repair remain to be elucidated, these results raise the possibility of using deferiprone as a therapeutic agent for remyelination therapy in MS.
Collapse
Affiliation(s)
- Atefeh Rayatpour
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Forough Foolad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Brain and Cognition, 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
|
3
|
Yang J. Partial Cell Fate Transitions to Promote Cardiac Regeneration. Cells 2024; 13:2002. [PMID: 39682750 PMCID: PMC11640292 DOI: 10.3390/cells13232002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Heart disease, including myocardial infarction (MI), remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective regenerative therapies. Direct reprogramming of cardiomyocyte-like cells from resident fibroblasts offers a promising avenue for myocardial regeneration, but its efficiency and consistency in generating functional cardiomyocytes remain limited. Alternatively, reprogramming induced cardiac progenitor cells (iCPCs) could generate essential cardiac lineages, but existing methods often involve complex procedures. These limitations underscore the need for advanced mechanistic insights and refined reprogramming strategies to improve reparative outcomes in the heart. Partial cellular fate transitions, while still a relatively less well-defined area and primarily explored in longevity and neurobiology, hold remarkable promise for cardiac repair. It enables the reprogramming or rejuvenation of resident cardiac cells into a stem or progenitor-like state with enhanced cardiogenic potential, generating the reparative lineages necessary for comprehensive myocardial recovery while reducing safety risks. As an emerging strategy, partial cellular fate transitions play a pivotal role in reversing myocardial infarction damage and offer substantial potential for therapeutic innovation. This review will summarize current advances in these areas, including recent findings involving two transcription factors that critically regulate stemness and cardiogenesis. It will also explore considerations for further refining these approaches to enhance their therapeutic potential and safety.
Collapse
Affiliation(s)
- Jianchang Yang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
4
|
Sharifi-Kelishadi M, Zare L, Fathollahi Y, Javan M. Conversion of Astrocyte Cell Lines to Oligodendrocyte Progenitor Cells Using Small Molecules and Transplantation to Animal Model of Multiple Sclerosis. J Mol Neurosci 2024; 74:40. [PMID: 38594388 DOI: 10.1007/s12031-024-02206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
Astrocytes, the most prevalent cells in the central nervous system (CNS), can be transformed into neurons and oligodendrocyte progenitor cells (OPCs) using specific transcription factors and some chemicals. In this study, we present a cocktail of small molecules that target different signaling pathways to promote astrocyte conversion to OPCs. Astrocytes were transferred to an OPC medium and exposed for five days to a small molecule cocktail containing CHIR99021, Forskolin, Repsox, LDN, VPA and Thiazovivin before being preserved in the OPC medium for an additional 10 days. Once reaching the OPC morphology, induced cells underwent immunocytofluorescence evaluation for OPC markers while checked for lacking the astrocyte markers. To test the in vivo differentiation capabilities, induced OPCs were transplanted into demyelinated mice brains treated with cuprizone over 12 weeks. Two distinct lines of astrocytes demonstrated the potential of conversion to OPCs using this small molecule cocktail as verified by morphological changes and the expression of PDGFR and O4 markers as well as the terminal differentiation to oligodendrocytes expressing MBP. Following transplantation into demyelinated mice brains, induced OPCs effectively differentiated into mature oligodendrocytes. The generation of OPCs from astrocytes via a small molecule cocktail may provide a new avenue for producing required progenitors necessary for myelin repair in diseases characterized by the loss of myelin such as multiple sclerosis.
Collapse
Affiliation(s)
| | - Leila Zare
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Brain and Cognition, 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
|
5
|
Cho HE, Lee S, Seo JH, Kang SW, Choi WA, Cho SR. In Vivo Reprogramming Using Yamanaka Factors in the CNS: A Scoping Review. Cells 2024; 13:343. [PMID: 38391956 PMCID: PMC10886652 DOI: 10.3390/cells13040343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
Central nervous system diseases, particularly neurodegenerative disorders, pose significant challenges in medicine. These conditions, characterized by progressive neuronal loss, have remained largely incurable, exacting a heavy toll on individuals and society. In recent years, in vivo reprogramming using Yamanaka factors has emerged as a promising approach for central nervous system regeneration. This technique involves introducing transcription factors, such as Oct4, Sox2, Klf4, and c-Myc, into adult cells to induce their conversion into neurons. This review summarizes the current state of in vivo reprogramming research in the central nervous system, focusing on the use of Yamanaka factors. In vivo reprogramming using Yamanaka factors has shown promising results in several animal models of central nervous system diseases. Studies have demonstrated that this approach can promote the generation of new neurons, improve functional outcomes, and reduce scar formation. However, there are still several challenges that need to be addressed before this approach can be translated into clinical practice. These challenges include optimizing the efficiency of reprogramming, understanding the cell of origin for each transcription factor, and developing methods for reprogramming in non-subventricular zone areas. Further research is needed to overcome the remaining challenges, but this approach has the potential to revolutionize the way we treat central nervous system disorders.
Collapse
Affiliation(s)
- Han Eol Cho
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (H.E.C.); (S.-W.K.)
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Seoul 06229, Republic of Korea
| | - Siwoo Lee
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Department of Rehabilitation Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Jung Hwa Seo
- Department of Rehabilitation Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seong-Woong Kang
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (H.E.C.); (S.-W.K.)
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Seoul 06229, Republic of Korea
| | - Won Ah Choi
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (H.E.C.); (S.-W.K.)
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Seoul 06229, Republic of Korea
| | - Sung-Rae Cho
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (H.E.C.); (S.-W.K.)
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Department of Rehabilitation Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
6
|
Gurdita A, Kwiecien JM, Choh V. Development of a new surgical technique to infuse kynurenic acid to optic nerves in chickens for studying loss of myelination. Heliyon 2023; 9:e14361. [PMID: 36938412 PMCID: PMC10020079 DOI: 10.1016/j.heliyon.2023.e14361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/14/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Abstract
Prolonged infusion of a high dose of kynurenic acid (KYNA) reduces the myelin content in the rat spinal cord with preservation of the axonal integrity and without inducing an inflammatory response. We hypothesized that subdural infusion of a high concentration of KYNA can induce myelin loss in the optic nerves (ONs) of chickens. However, existing methods to deliver agents to the ON are inefficient, unlocalized and provide only acute exposure. Thus, we developed a surgical approach for sustained delivery of KYNA to the chicken ON. In brief, the novel surgical technique, which does not include excision of the extraocular muscles, involves incision of the skin and underlying fascial sheath to access the optic nerve within the muscle cone, implantation of a catheter in the dura of the optic nerve, the other end of which exits the orbit under the skin. The catheter runs under the skin near the lateral canthus, over the ears to the back of the neck, where a second incision is made to both implant the osmotic pump and to attach the catheter to the osmotic pump. India ink was used to confirm prolonged sustained administration to the optic nerves and across the chiasm. This surgical model was used to investigate KYNA's effect(s) on myelin loss in the ON. ONs of 7-day old chickens were infused with 50 mM KYNA or phosphate buffered saline (PBS) for seven days. Analysis of KYNA-infused contralateral ON g-ratios and protein levels indicated a reduction in myelin. These findings demonstrate the utility of our surgical approach for sustained delivery of KYNA into the ON and suggest a role for KYNA in modulating CNS myelination.
Collapse
Affiliation(s)
- Akshay Gurdita
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Vivian Choh
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
- Corresponding author. University of Waterloo, 200 Columbia St W, Waterloo, ON N2L 3G1
| |
Collapse
|
7
|
Cis-p-tau plays crucial role in lysolecithin-induced demyelination and subsequent axonopathy in mouse optic chiasm. Exp Neurol 2023; 359:114262. [PMID: 36343678 DOI: 10.1016/j.expneurol.2022.114262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease that leads to axon degeneration as the major cause of everlasting neurological disability. The cis-phosphorylated tau (cis-p-tau) is an isoform of tau phosphorylated on threonine 231 and causes tau fails to bind micro-tubules and promotes assembly. It gains toxic function and forms tangles in the cell which finally leads to cell death. An antibody raised against cis- p-tau (cis mAb) detects this isoform and induces its clearance. Here, we investigated the formation of cis-p-tau in a lysophosphatidylcholine (LPC)-induced prolonged demyelination model as well as the beneficial effects of its clearance using cis mAb. Cis -p-tau was increased in the lesion site, especially in axons and microglia. Behavioral and functional studies were performed using visual cliff test, visual placing test, and visual evoked potential recording. Cis-p-tau clearance resulted in decreased gliosis, protected myelin and reduced axon degeneration. Analysis of behavioral and electrophysiological data showed that clearance of cis-p-tau by cis mAb treatment improved the visual acuity along with the integrity of the optic pathway. Our results highlight the opportunity of using cis mAb as a new therapy for protecting myelin and axons in patients suffering from MS.
Collapse
|
8
|
Yun W, Kim YJ, Lee G. Direct Conversion to Achieve Glial Cell Fates: Oligodendrocytes and Schwann Cells. Int J Stem Cells 2022; 15:14-25. [PMID: 35220289 PMCID: PMC8889328 DOI: 10.15283/ijsc22008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
Glia have been known for its pivotal roles in physiological and pathological conditions in the nervous system. To study glial biology, multiple approaches have been applied to utilize glial cells for research, including stem cell-based technologies. Human glial cells differentiated from pluripotent stem cells are now available, allowing us to study the structural and functional roles of glia in the nervous system, although the efficiency is still low. Direct conversion is an advanced strategy governing fate conversion of diverse cell types directly into the desired lineage. This novel strategy stands as a promising approach for preliminary research and regenerative medicine. Direct conversion employs genetic and environmental cues to change cell fate to that with the required functional cell properties while retaining maturity-related molecular features. As an alternative method, it is now possible to obtain a variety of mature cell populations that could not be obtained using conventional differentiation methods. This review summarizes current achievements in obtaining glia, particularly oligodendrocytes and Schwann cells.
Collapse
Affiliation(s)
- Wonjin Yun
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yong Jun Kim
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
OCT4-induced oligodendrocyte progenitor cells promote remyelination and ameliorate disease. NPJ Regen Med 2022; 7:4. [PMID: 35027563 PMCID: PMC8758684 DOI: 10.1038/s41536-021-00199-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 11/30/2021] [Indexed: 12/23/2022] Open
Abstract
The generation of human oligodendrocyte progenitor cells (OPCs) may be therapeutically valuable for human demyelinating diseases such as multiple sclerosis. Here, we report the direct reprogramming of human somatic cells into expandable induced OPCs (iOPCs) using a combination of OCT4 and a small molecule cocktail. This method enables generation of A2B5+ (an early marker for OPCs) iOPCs within 2 weeks retaining the ability to differentiate into MBP-positive mature oligodendrocytes. RNA-seq analysis revealed that the transcriptome of O4+ iOPCs was similar to that of O4+ OPCs and ChIP-seq analysis revealed that putative OCT4-binding regions were detected in the regulatory elements of CNS development-related genes. Notably, engrafted iOPCs remyelinated the brains of adult shiverer mice and experimental autoimmune encephalomyelitis mice with MOG-induced 14 weeks after transplantation. In conclusion, our study may contribute to the development of therapeutic approaches for neurological disorders, as well as facilitate the understanding of the molecular mechanisms underlying glial development.
Collapse
|
10
|
In Vivo Expression of Reprogramming Factor OCT4 Ameliorates Myelination Deficits and Induces Striatal Neuroprotection in Huntington's Disease. Genes (Basel) 2021; 12:genes12050712. [PMID: 34068799 PMCID: PMC8150572 DOI: 10.3390/genes12050712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/27/2021] [Accepted: 05/01/2021] [Indexed: 12/26/2022] Open
Abstract
White matter atrophy has been shown to precede the massive loss of striatal GABAergic neurons in Huntington’s disease (HD). This study investigated the effects of in vivo expression of reprogramming factor octamer-binding transcription factor 4 (OCT4) on neural stem cell (NSC) niche activation in the subventricular zone (SVZ) and induction of cell fate specific to the microenvironment of HD. R6/2 mice randomly received adeno-associated virus 9 (AAV9)-OCT4, AAV9-Null, or phosphate-buffered saline into both lateral ventricles at 4 weeks of age. The AAV9-OCT4 group displayed significantly improved behavioral performance compared to the control groups. Following AAV9-OCT4 treatment, the number of newly generated NSCs and oligodendrocyte progenitor cells (OPCs) significantly increased in the SVZ, and the expression of OPC-related genes and glial cell-derived neurotrophic factor (GDNF) significantly increased. Further, amelioration of myelination deficits in the corpus callosum was observed through electron microscopy and magnetic resonance imaging, and striatal DARPP32+ GABAergic neurons significantly increased in the AAV9-OCT4 group. These results suggest that in situ expression of the reprogramming factor OCT4 in the SVZ induces OPC proliferation, thereby attenuating myelination deficits. Particularly, GDNF released by OPCs seems to induce striatal neuroprotection in HD, which explains the behavioral improvement in R6/2 mice overexpressing OCT4.
Collapse
|
11
|
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
|
12
|
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
|
13
|
PPAR-γ Is Critical for HDAC3-Mediated Control of Oligodendrocyte Progenitor Cell Proliferation and Differentiation after Focal Demyelination. Mol Neurobiol 2020; 57:4810-4824. [PMID: 32803489 DOI: 10.1007/s12035-020-02060-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022]
Abstract
Disruption of remyelination contributes to neurodegeneration and cognitive impairment in chronically disabled patients. Valproic acid (VPA) inhibits histone deacetylase (HDAC) function and probably promotes oligodendrocyte progenitor cell (OPC) proliferation and differentiation; however, the relevant molecular mechanisms remain unknown. Here, focal demyelinating lesions (FDLs) were generated in mice by two-point stereotactic injection of lysophosphatidylcholine (LPC) into the corpus callosum. Cognitive functions, sensorimotor abilities and histopathological changes were assessed for up to 28 days post-injury with or without VPA treatment. Primary OPCs were harvested and used to study the effect of VPA on OPC differentiation under inflammatory conditions. VPA dose-dependently attenuated learning and memory deficits and robustly protected white matter after FDL induction, as demonstrated by reductions in SMI-32 and increases in myelin basic protein staining. VPA also promoted OPC proliferation and differentiation and increased subsequent remyelination efficiency by day 28 post-FDL induction. VPA treatment did not affect HDAC1, HDAC2 or HDAC8 expression but reduced HDAC3 protein levels. In vitro, VPA improved the survival of mouse OPCs and promoted their differentiation into oligodendrocytes following lipopolysaccharide (LPS) stimulation. LPS caused OPCs to overexpress HDAC3, which translocated from the cytoplasm into the nucleus, where it directly interacted with the nuclear transcription factor PPAR-γ and negatively regulated PPAR-γ expression. VPA decreased the expression of HDAC3 and promoted remyelination and functional neurological recovery after FDL. These findings may support the use of strategies modulating HDAC3-mediated regulation of protein acetylation for the treatment of demyelination-related cognitive dysfunction.
Collapse
|
14
|
Fingolimod (FTY720) improves the functional recovery and myelin preservation of the optic pathway in focal demyelination model of rat optic chiasm. Brain Res Bull 2019; 153:109-121. [DOI: 10.1016/j.brainresbull.2019.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/09/2019] [Accepted: 08/18/2019] [Indexed: 12/21/2022]
|
15
|
Yavarpour‐Bali H, Nakhaei‐Nejad M, Yazdi A, Ghasemi‐Kasman M. Direct conversion of somatic cells towards oligodendroglial lineage cells: A novel strategy for enhancement of myelin repair. J Cell Physiol 2019; 235:2023-2036. [DOI: 10.1002/jcp.29195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | | | - Azadeh Yazdi
- Department of Physiology, Faculty of Medical Sciences 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
| |
Collapse
|
16
|
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
|
17
|
Mesentier-Louro LA, Dodd R, Domizi P, Nobuta H, Wernig M, Wernig G, Liao YJ. Direct targeting of the mouse optic nerve for therapeutic delivery. J Neurosci Methods 2019; 313:1-5. [PMID: 30389488 PMCID: PMC10870831 DOI: 10.1016/j.jneumeth.2018.10.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Animal models of optic nerve injury are often used to study central nervous system (CNS) degeneration and regeneration, and targeting the optic nerve is a powerful approach for axon-protective or remyelination therapy. However, the experimental delivery of drugs or cells to the optic nerve is rarely performed because injections into this structure are difficult in small animals, especially in mice. NEW METHOD We investigated and developed methods to deliver drugs or cells to the mouse optic nerve through 3 different routes: a) intraorbital, b) through the optic foramen and c) transcranial. RESULTS The methods targeted different parts of the mouse optic nerve: intraorbital proximal (intraorbital), intracranial middle (optic-foramen) or intracranial distal (transcranial) portion. COMPARISON WITH EXISTING METHODS Most existing methods target the optic nerve indirectly. For instance, intravitreally delivered cells often cannot cross the inner limiting membrane to reach retinal neurons and optic nerve axons. Systemic delivery, eye drops and intraventricular injections do not always successfully target the optic nerve. Intraorbital and transcranial injections into the optic nerve or chiasm have been performed but these methods have not been well described. We approached the optic nerve with more selective and precise targeting than existing methods. CONCLUSIONS We successfully targeted the murine optic nerve intraorbitally, through the optic foramen, and transcranially. Of all methods, the injection through the optic foramen is likely the most innovative and fastest. These methods offer additional approaches for therapeutic intervention to be used by those studying white matter damage and axonal regeneration in the CNS.
Collapse
Affiliation(s)
| | - Robert Dodd
- Department of Neurosurgery Stanford University School of Medicine, Palo Alto, CA, USA
| | - Pablo Domizi
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Hiroko Nobuta
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Marius Wernig
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Gerlinde Wernig
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Yaping Joyce Liao
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA; Department of Neurology, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
18
|
Farhangi S, Dehghan S, Totonchi M, Javan M. In vivo conversion of astrocytes to oligodendrocyte lineage cells in adult mice demyelinated brains by Sox2. Mult Scler Relat Disord 2019; 28:263-272. [PMID: 30639828 DOI: 10.1016/j.msard.2018.12.041] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/11/2018] [Accepted: 12/31/2018] [Indexed: 12/17/2022]
Abstract
Sox2 transcription factor has been frequently used for reprograming starting cells to neural progenitor/stem cells. In vivo administration of Sox2 in the adult mouse brain reprogrammed the transduced astrocytes to neurons. In searching for adequate cell source for repairing the myelin insults, here, we studied the possible conversion of astrocytes to oligodendrocyte lineage cells by Sox2, while an extensive demyelination exists in animal brain. Lentiviral particles expressing Sox2-GFP were injected into the corpora callosa of animals fed with cuprizone diet for 12 weeks. Transduced cells were mainly astrocytes that changed their fate to oligodendrocyte lineage cells by time. For further conformation astrocytes received the vector in culture and then transplanted to the animal brains. Tracing the fate of transplanted cells showed their conversion to oligodendrocyte lineage cells. In vitro transduced cell were also maintained in the oligodendrocyte progenitor cell (OPC) induction medium. Produced OPC-like cells were positive for specific markers. This study provides a new strategy for endogenous production of myelinating cells. After optimizing the experimental conditions for safety and feasibility, this approach may contribute into future cell based therapies in patients with white matter insults as like as those with multiple sclerosis.
Collapse
Affiliation(s)
- Sahar Farhangi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O.Box:14115-331, Tehran, Iran
| | - Samaneh Dehghan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O.Box:14115-331, Tehran, Iran
| | - Mehdi Totonchi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, 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, Tehran, Iran.
| |
Collapse
|
19
|
Crocker CE, Tibbo PG. Confused Connections? Targeting White Matter to Address Treatment Resistant Schizophrenia. Front Pharmacol 2018; 9:1172. [PMID: 30405407 PMCID: PMC6201564 DOI: 10.3389/fphar.2018.01172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Abstract
Despite development of comprehensive approaches to treat schizophrenia and other psychotic disorders and improve outcomes, there remains a proportion (approximately one-third) of patients who are treatment resistant and will not have remission of psychotic symptoms despite adequate trials of pharmacotherapy. This level of treatment response is stable across all stages of the spectrum of psychotic disorders, including early phase psychosis and chronic schizophrenia. Our current pharmacotherapies are beneficial in decreasing positive symptomology in most cases, however, with little to no impact on negative or cognitive symptoms. Not all individuals with treatment resistant psychosis unfortunately, even benefit from the potential pharmacological reductions in positive symptoms. The existing pharmacotherapy for psychosis is targeted at neurotransmitter receptors. The current first and second generation antipsychotic medications all act on dopamine type 2 receptors with the second generation drugs also interacting significantly with serotonin type 1 and 2 receptors, and with varying pharmacodynamic profiles overall. This focus on developing dopaminergic/serotonergic antipsychotics, while beneficial, has not reduced the proportion of patients experiencing treatment resistance to date. Another pharmacological approach is imperative to address treatment resistance both for response overall and for negative symptoms in particular. There is research suggesting that changes in white matter integrity occur in schizophrenia and these may be more associated with cognition and even negative symptomology. Here we review the evidence that white matter abnormalities in the brain may be contributing to the symptomology of psychotic disorders. Additionally, we propose that white matter may be a viable pharmacological target for pharmacoresistant schizophrenia and discuss current treatments in development for schizophrenia that target white matter.
Collapse
Affiliation(s)
- Candice E Crocker
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada.,Department of Diagnostic Imaging, Nova Scotia Health Authority, Halifax, NS, Canada
| | - Philip G Tibbo
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
20
|
Mokhtarzadeh Khanghahi A, Satarian L, Deng W, Baharvand H, Javan M. In vivo conversion of astrocytes into oligodendrocyte lineage cells with transcription factor Sox10; Promise for myelin repair in multiple sclerosis. PLoS One 2018; 13:e0203785. [PMID: 30212518 PMCID: PMC6136770 DOI: 10.1371/journal.pone.0203785] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/27/2018] [Indexed: 11/18/2022] Open
Abstract
Recent studies demonstrate that astroglial cells can be directly converted into functional neurons or oligodendrocytes. Here, we report that a single transcription factor Sox10 could reprogram astrocytes into oligodendrocyte-like cells, in vivo. For transdifferentiation, Sox10-GFP expressing viral particles were injected into cuprizone-induced demyelinated mice brains after which we assessed for the presence of specific oligodendrocyte lineage cell markers by immunohistofluorescence (IHF). As control, another group of demyelinated mice received GFP expressing viral particles. After 3 weeks, the majority of transduced (GFP+) cells in animals which received control vector were astrocytes, while in animals which received Sox10-GFP vector, the main population of GFP+ cells were positive for oligodendrocyte lineage markers. We also extracted primary astrocytes from mouse pups and purified them. Primary astrocytes were transduced in vitro and then transplanted into demyelinated brains for later fate mapping. After three weeks, in vitro transduced and then transplanted astrocytes showed oligodendrocyte progenitor and mature oligodendrocyte markers. Further confirmation was done by transduction of astrocytes with lentiviral particles that expressed Sox10 and GFP and their culture in the oligodendrocyte progenitor medium. The induced cells expressed oligodendrocyte progenitor cells (iOPCs) markers. Our findings showed the feasibility of reprogramming of astrocytes into oligodendrocyte-like cells in vivo, by using a single transcription factor, Sox10. This finding suggested a master regulatory role for Sox10 which enabled astrocytes to change their fate to OPC-like cells and establish an oligodendroglial phenotype. We hope this approach lead to effective myelin repair in patients suffering from myelination deficit.
Collapse
Affiliation(s)
- Akram Mokhtarzadeh Khanghahi
- Department of Brain Sciences and Cognition, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Leila Satarian
- Department of Brain Sciences and Cognition, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Wenbin Deng
- Institute for Pediatric Regenerative Medicine, University of California, Davis, School of Medicine, Sacramento, California, United States of America
| | - 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 Brain Sciences and Cognition, 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
- * E-mail: ,
| |
Collapse
|
21
|
Luo Q, Ding L, Zhang N, Jiang Z, Gao C, Xue L, Peng B, Wang G. A stable and easily reproducible model of focal white matter demyelination. J Neurosci Methods 2018; 307:230-239. [DOI: 10.1016/j.jneumeth.2018.05.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022]
|
22
|
Altered Brain Cholesterol/Isoprenoid Metabolism in a Rat Model of Autism Spectrum Disorders. Neuroscience 2018; 372:27-37. [PMID: 29309878 DOI: 10.1016/j.neuroscience.2017.12.053] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/28/2017] [Accepted: 12/28/2017] [Indexed: 12/11/2022]
Abstract
Autism spectrum disorders (ASDs) present a wide range of symptoms characterized by altered sociability, compromised communication and stereotypic/repetitive behaviors. These symptoms are caused by developmental changes, but the mechanisms remain largely unknown. Some lines of evidence suggest an impairment of the cholesterol/isoprenoid metabolism in the brain as a possible cause, but systematic analyses in rodent models of ASDs are lacking. Prenatal exposure to the antiepileptic drug valproate (VPA) is a risk factor for ASDs in humans and generates a well-established model for the disease in rodents. Here, we studied cholesterol/isoprenoid metabolism in different brain areas of infant, adolescent and adult rats prenatally exposed to VPA. VPA-treated rats present autistic-like symptoms, they show changes in cholesterol/isoprenoid homeostasis in some brain areas, a decreased number of oligodendrocytes and impaired myelination in the hippocampus. Together, our data suggest a relation between brain cholesterol/isoprenoid homeostasis and ASDs.
Collapse
|
23
|
miR-302/367-induced neurons reduce behavioral impairment in an experimental model of Alzheimer's disease. Mol Cell Neurosci 2018; 86:50-57. [DOI: 10.1016/j.mcn.2017.11.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/03/2017] [Accepted: 11/22/2017] [Indexed: 01/13/2023] Open
|
24
|
Ghasemi-Kasman M, Baharvand H, Javan M. Enhanced neurogenesis in degenerated hippocampi following pretreatment with miR-302/367 expressing lentiviral vector in mice. Biomed Pharmacother 2017; 96:1222-1229. [PMID: 29174574 DOI: 10.1016/j.biopha.2017.11.094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/11/2017] [Accepted: 11/17/2017] [Indexed: 11/15/2022] Open
Abstract
Astrogliosis is the main landmark of neurodegenerative diseases. In vivo reprogramming of reactive astrocytes to functional neurons opened a new horizon in regenerative medicine. However there is little evidence that show possible application of in vivo reprogramming approaches for enhancement of neurogenesis. Cluster miR-302/367 showed high capability in cell reprogramming. Here we show that application of lentiviral particles expressing cluster miR-302/367 along with systemic valproate (VPA) enhanced the capability of mice brains for neurogenesis in CA3 area following kainic acid (KA) induced hippocampal neurodegeneration. Following pretreatment with miR-302/367 expressing viral particles and VPA, transduced cells showed neuroblast and mature neuron markers when neuronal loss was induced by KA. Comparing the neuron counts in CA3 region also showed that neurogenesis was increased in CA3 region in animals which were pretreated with miR-302/367 vector and VPA, only in injected side of the brain. Our data suggest that targeted application of miR-302/367 expressing vector may enhance the capacity of hippocampus and other brain structures for regeneration following neuronal loss.
Collapse
Affiliation(s)
- Maryam Ghasemi-Kasman
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Baharvand
- 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 Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
25
|
Motavaf M, Sadeghizadeh M, Javan M. Attempts to Overcome Remyelination Failure: Toward Opening New Therapeutic Avenues for Multiple Sclerosis. Cell Mol Neurobiol 2017; 37:1335-1348. [PMID: 28224237 PMCID: PMC11482203 DOI: 10.1007/s10571-017-0472-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 02/12/2017] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic immune-mediated disorder of the central nervous system that results in destruction of the myelin sheath wrapped around the axons and eventual axon degeneration. The disease is pathologically heterogeneous; however, perhaps its most frustrating aspect is the lack of efficient regenerative response for remyelination. Current treatment strategies are based on anti-inflammatory or immunomodulatory medications that have the potential to reduce the numbers of newly evolving lesions. However, therapies are still required that can repair already damaged myelin for which current treatments are not effective. A prerequisite for the development of such new treatments is understanding the reasons for insufficient endogenous repair. This review briefly summarizes the currently suggested causes of remyelination failure in MS and possible solutions.
Collapse
Affiliation(s)
- Mahsa Motavaf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Islamic Republic of Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Islamic Republic of Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-331, Tehran, Islamic Republic of Iran.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Islamic Republic of Iran.
| |
Collapse
|
26
|
Taghiyar L, Hesaraki M, Sayahpour FA, Satarian L, Hosseini S, Aghdami N, Baghaban Eslaminejad M. Msh homeobox 1 ( Msx1)- and Msx2-overexpressing bone marrow-derived mesenchymal stem cells resemble blastema cells and enhance regeneration in mice. J Biol Chem 2017; 292:10520-10533. [PMID: 28461333 DOI: 10.1074/jbc.m116.774265] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/29/2017] [Indexed: 01/23/2023] Open
Abstract
Amputation of the proximal region in mammals is not followed by regeneration because blastema cells (BCs) and expression of regenerative genes, such as Msh homeobox (Msx) genes, are absent in this animal group. The lack of BCs and positional information in other cells is therefore the main obstacle to therapeutic approaches for limb regeneration. Hence, this study aimed to create blastema-like cells (BlCs) by overexpressing Msx1 and Msx2 genes in mouse bone marrow-derived mesenchymal stem cells (mBMSCs) to regenerate a proximally amputated digit tip. We transduced mBMSCs with Msx1 and Msx2 genes and compared osteogenic activity and expression levels of several Msx-regulated genes (Bmp4, Fgf8, and keratin 14 (K14)) in BlC groups, including MSX1, MSX2, and MSX1/2 (in a 1:1 ratio) with those in mBMSCs and BCs in vitro and in vivo following injection into the amputation site. We found that Msx gene overexpression increased expression of specific blastemal markers and enhanced the proliferation rate and osteogenesis of BlCs compared with mBMSCs and BCs via activation of Fgf8 and Bmp4 Histological analyses indicated full regrowth of digit tips in the Msx-overexpressing groups, particularly in MSX1/2, through endochondral ossification 6 weeks post-injection. In contrast, mBMSCs and BCs formed abnormal bone and nail. Full digit tip was regenerated only in the MSX1/2 group and was related to boosted Bmp4, Fgf8, and K14 gene expression and to limb-patterning properties resulting from Msx1 and Msx2 overexpression. We propose that Msx-transduced cells that can regenerate epithelial and mesenchymal tissues may potentially be utilized in limb regeneration.
Collapse
Affiliation(s)
- Leila Taghiyar
- From the Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran and.,the Department of Developmental Biology, University of Science and Culture, Tehran 13145-871, Iran
| | - Mahdi Hesaraki
- From the Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran and
| | - Forough Azam Sayahpour
- From the Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran and
| | - Leila Satarian
- From the Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran and
| | - Samaneh Hosseini
- From the Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran and
| | - Naser Aghdami
- From the Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran and
| | - Mohamadreza Baghaban Eslaminejad
- From the Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran and
| |
Collapse
|
27
|
Ghasemi-Kasman M, Zare L, Baharvand H, Javan M. In vivo
conversion of astrocytes to myelinating cells by miR-302/367 and valproate to enhance myelin repair. J Tissue Eng Regen Med 2017; 12:e462-e472. [DOI: 10.1002/term.2276] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Maryam Ghasemi-Kasman
- Department of Physiology, Faculty of Medical Sciences; Tarbiat Modares University; Tehran Iran
| | - Leila Zare
- Department of Physiology, Faculty of Medical Sciences; Tarbiat Modares University; Tehran Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
- Department of Developmental Biology; University of Science and Culture, ACECR; Tehran Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences; Tarbiat Modares University; Tehran Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
| |
Collapse
|
28
|
Eslami-Behroozi M, Pazhoohan S, Aref E, Zare L, Javan M, Hajizadeh S, Raoufy MR. Bronchoconstriction Induces Structural and Functional Airway Alterations in Non-sensitized Rats. Lung 2016; 195:167-171. [PMID: 28025669 DOI: 10.1007/s00408-016-9970-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022]
Abstract
The impact of mechanical forces on pathogenesis of airway remodeling and the functional consequences in asthma remains to be fully established. In the present study, we investigated the effect of repeated bronchoconstriction induced by methacholine (MCh) on airway remodeling and airway hyperresponsiveness (AHR) in rats with or without sensitization to an external allergen. We provide evidence that repeated bronchoconstriction, using MCh, alone induces airway inflammation and remodeling as well as AHR in non-allergen-sensitized rats. Also, we found that the airways are structurally and functionally altered by bronchoconstriction induced by either allergen or MCh in allergen-sensitized animals. This finding provides a new animal model for the development of airway remodeling and AHR in mammals and can be used for studying the complex reciprocal relationship between bronchoconstriction and airway inflammation. Further studies on presented animal models are required to clarify the exact mechanisms underlying airway remodeling due to bronchoconstriction and the functional consequences.
Collapse
Affiliation(s)
- Mehdi Eslami-Behroozi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Nasr Bridge, Jalal Al Ahmad Highway, Tehran, Iran
| | - Saeed Pazhoohan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Nasr Bridge, Jalal Al Ahmad Highway, Tehran, Iran
| | - Ehsan Aref
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Nasr Bridge, Jalal Al Ahmad Highway, Tehran, Iran
| | - Leila Zare
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Nasr Bridge, Jalal Al Ahmad Highway, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Nasr Bridge, Jalal Al Ahmad Highway, Tehran, Iran
| | - Sohrab Hajizadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Nasr Bridge, Jalal Al Ahmad Highway, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Nasr Bridge, Jalal Al Ahmad Highway, Tehran, Iran. .,Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Altinoz MA, Ozcan EM, Ince B, Guloksuz S. Hemoglobins as new players in multiple sclerosis: metabolic and immune aspects. Metab Brain Dis 2016; 31:983-92. [PMID: 27234993 DOI: 10.1007/s11011-016-9845-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
Basic science investigations and clinical observations in recent years indicate that hemoglobins (Hbs) may have important roles in the pathogenesis of multiple sclerosis (MS). These findings can be summarized as follows: 1- Erythrocyte fragility is higher in MS patients, the released free Hb damages blood-brain barrier, myelin basic protein and also triggers iron overload and inflammation. 2- Free Hb may further activate the inflammatory responses through Toll-like receptor 4 (TLR4), present on microglia and other innate immunocytes. 3- Hbs are expressed in neural cells including dopaminergic neurons. Also, several studies have demonstrated that Hbs are expressed in astrocytes and oligodendroglia. 4- Hb overexpression in neural cells upregulate mitochondrial complex I-V subunits. The comparison of the mitochondrial proteome between healthy and patients with MS revealed only four differentially expressed proteins including Hb β-chain. 5- Microarray analysis of 8300 genes in monocytes of twins with and without MS showed a difference in 25 genes that include genes encoding α- and β-globins as well. 6- β- and α-globin gene clusters reside at chromosomal regions 11p15.5 and 16p13.3, respectively. Whole genome screen (WGS) in Sardinian MS families using 327 markers revealed linkage in 3 regions including 11p15.5 loci. Further, 11p15.5 and 16p13.3 were part of the 17 regions identified in the WGS study of 136 sibling-pairs in Nordic countries analyzing 399 microsatellite markers. In the light of these findings, we propose that free Hb released from dying erythrocytes is detrimental. On the contrary, intracellular Hbs in neural cells are protective in MS. The genomic linkage findings can be explained by common haematologically-silent Hb variants that may lower the protective function of intracellular Hbs, and therefore, enhance the risk for MS. In the absence of such variants, aberrations in the translational and post-translational mechanisms controlling synthesis of neural Hbs may also enhance the vulnerability to MS. Alternatively, such genetic variants may perturb the metabolism of anti-inflammatory hemorphins produced via cleavage of Hbs.
Collapse
Affiliation(s)
- Meric A Altinoz
- Immunology Program, Experimental Medicine Research Institute, Istanbul University, Yildirim Apt. No: 5, D:6, Güven Sk, Nurtepe Mh, Kagithane, Istanbul, Turkey.
| | - Emin M Ozcan
- Department of Neurology, Biruni University, Istanbul, Turkey
| | - Bahri Ince
- Department of Psychiatry, Bakirkoy Education and Research Hospital for Psychiatry, Istanbul, Turkey
| | - Sinan Guloksuz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry and Psychology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|