1
|
Hintermayer MA, Juźwik CA, Morquette B, Hua E, Zhang J, Drake S, Shi SS, Rambaldi I, Vangoor V, Pasterkamp J, Moore C, Fournier AE. A miR-383-5p Signaling Hub Coordinates the Axon Regeneration Response to Inflammation. J Neurosci 2024; 44:e1822232024. [PMID: 39266301 PMCID: PMC11529811 DOI: 10.1523/jneurosci.1822-23.2024] [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: 09/25/2023] [Revised: 08/07/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024] Open
Abstract
Neuroinflammation can positively influence axon regeneration following injury in the central nervous system. Inflammation promotes the release of neurotrophic molecules and stimulates intrinsic proregenerative molecular machinery in neurons, but the detailed mechanisms driving this effect are not fully understood. We evaluated how microRNAs are regulated in retinal neurons in response to intraocular inflammation to identify their potential role in axon regeneration. We found that miR-383-5p is downregulated in retinal ganglion cells in response to zymosan-induced intraocular inflammation. MiR-383-5p downregulation in neurons is sufficient to promote axon growth in vitro, and the intravitreal injection of a miR-383-5p inhibitor into the eye promotes axon regeneration following optic nerve crush. MiR-383-5p directly targets ciliary neurotrophic factor (CNTF) receptor components, and miR-383-5p inhibition sensitizes adult retinal neurons to the outgrowth-promoting effects of CNTF. Interestingly, we also demonstrate that CNTF treatment is sufficient to reduce miR-383-5p levels in neurons, constituting a positive-feedback module, whereby initial CNTF treatment reduces miR-383-5p levels, which then disinhibits CNTF receptor components to sensitize neurons to the ligand. Additionally, miR-383-5p inhibition derepresses the mitochondrial antioxidant protein peroxiredoxin-3 (PRDX3) which was required for the proregenerative effects associated with miR-383-5p loss-of-function in vitro. We have thus identified a positive-feedback mechanism that facilitates neuronal CNTF sensitivity in neurons and a new molecular signaling module that promotes inflammation-induced axon regeneration.
Collapse
Affiliation(s)
- Matthew A Hintermayer
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Camille A Juźwik
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Barbara Morquette
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Elizabeth Hua
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Julia Zhang
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Sienna Drake
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Shan Shan Shi
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Isabel Rambaldi
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Vamshi Vangoor
- Department of Translation Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht 3584 CG, Netherlands
| | - Jeroen Pasterkamp
- Department of Translation Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht 3584 CG, Netherlands
| | - Craig Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Alyson E Fournier
- Montréal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| |
Collapse
|
2
|
Nouri Z, Barfar A, Perseh S, Motasadizadeh H, Maghsoudian S, Fatahi Y, Nouri K, Yektakasmaei MP, Dinarvand R, Atyabi F. Exosomes as therapeutic and drug delivery vehicle for neurodegenerative diseases. J Nanobiotechnology 2024; 22:463. [PMID: 39095888 PMCID: PMC11297769 DOI: 10.1186/s12951-024-02681-4] [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: 05/11/2024] [Accepted: 06/30/2024] [Indexed: 08/04/2024] Open
Abstract
Neurodegenerative disorders are complex, progressive, and life-threatening. They cause mortality and disability for millions of people worldwide. Appropriate treatment for neurodegenerative diseases (NDs) is still clinically lacking due to the presence of the blood-brain barrier (BBB). Developing an effective transport system that can cross the BBB and enhance the therapeutic effect of neuroprotective agents has been a major challenge for NDs. Exosomes are endogenous nano-sized vesicles that naturally carry biomolecular cargoes. Many studies have indicated that exosome content, particularly microRNAs (miRNAs), possess biological activities by targeting several signaling pathways involved in apoptosis, inflammation, autophagy, and oxidative stress. Exosome content can influence cellular function in healthy or pathological ways. Furthermore, since exosomes reflect the features of the parental cells, their cargoes offer opportunities for early diagnosis and therapeutic intervention of diseases. Exosomes have unique characteristics that make them ideal for delivering drugs directly to the brain. These characteristics include the ability to pass through the BBB, biocompatibility, stability, and innate targeting properties. This review emphasizes the role of exosomes in alleviating NDs and discusses the associated signaling pathways and molecular mechanisms. Furthermore, the unique biological features of exosomes, making them a promising natural transporter for delivering various medications to the brain to combat several NDs, are also discussed.
Collapse
Affiliation(s)
- Zeinab Nouri
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashkan Barfar
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahra Perseh
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Motasadizadeh
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samane Maghsoudian
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Keyvan Nouri
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Rassoul Dinarvand
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Wang D, Wu Y, Liu Y, Ji Q, Luo Y, Yan J. Dysregulated MiR-223-5p Modulates Inflammation and Oxidative Stress in Traumatic Spinal Cord Injury. Immunol Invest 2024; 53:947-961. [PMID: 38814140 DOI: 10.1080/08820139.2024.2359531] [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] [Indexed: 05/31/2024]
Abstract
AIM This study aimed to evaluate the miR-223-5p expression in patients with spinal cord injury (SCI) and to determine its role in the pathogenesis of SCI. METHODS The serum miR-223-5p levels were analyzed using quantitative real-time polymerase chain reaction. The diagnostic accuracy of miR-223-5p was evaluated using the receiving operating characteristic curves. LPS-induced PC12 cells were established as an in vitro inflammatory cell model. Cell apoptosis, inflammation and oxidative stress were examined. The SCI rat model was constructed to evaluate the effects of miR-223-5p on inflammatory response and motor function in rats. RESULTS MiR-223-5p expression was upregulated in SCI patients. MiR-223-5p expression in the complete SCI group was significantly higher than that in incomplete SCI group. ROC analysis showed that miR-223-5p can distinguish SCI patients from healthy volunteers. In vitro experiments demonstrated that LPS upregulated apoptosis and inflammation in PC12 cells. Treatment with miR-223-5p inhibitor alleviated the changes in LPS-induced PC12 cells . Inhibition of miR-223-5p can alleviate the activation of inflammatory response and the effects of SCI on the motor function in rats. CONCLUSIONS MiR-223-5p is a potential diagnostic marker for SCI, and it can promote the SCI progression by regulating nerve cell survival, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Dawei Wang
- Department of Orthopedics, Zhangjiakou First Hospital, Zhangjiakou, Hebei, China
| | - Yingshuang Wu
- Department of Paediatrics, Zhangjiakou First Hospital, Zhangjiakou, Hebei, China
| | - Yongxiang Liu
- Department of Orthopedics, Yantaishan Hospital, Yantai, Shandong, China
| | - Qinghui Ji
- Department of Orthopedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, China
| | - Yi Luo
- Department of Orthopedics, Zhangjiakou First Hospital, Zhangjiakou, Hebei, China
| | - Jinglong Yan
- Department of Orthopedics, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
4
|
Jiang Y, Cai Y, Yang N, Gao S, Li Q, Pang Y, Su P. Molecular mechanisms of spinal cord injury repair across vertebrates: A comparative review. Eur J Neurosci 2024; 60:4552-4568. [PMID: 38978308 DOI: 10.1111/ejn.16462] [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: 11/16/2023] [Revised: 05/09/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024]
Abstract
In humans and other adult mammals, axon regeneration is difficult in axotomized neurons. Therefore, spinal cord injury (SCI) is a devastating event that can lead to permanent loss of locomotor and sensory functions. Moreover, the molecular mechanisms of axon regeneration in vertebrates are not very well understood, and currently, no effective treatment is available for SCI. In striking contrast to adult mammals, many nonmammalian vertebrates such as reptiles, amphibians, bony fishes and lampreys can spontaneously resume locomotion even after complete SCI. In recent years, rapid progress in the development of next-generation sequencing technologies has offered valuable information on SCI. In this review, we aimed to provide a comparison of axon regeneration process across classical model organisms, focusing on crucial genes and signalling pathways that play significant roles in the regeneration of individually identifiable descending neurons after SCI. Considering the special evolutionary location and powerful regenerative ability of lamprey and zebrafish, they will be the key model organisms for ongoing studies on spinal cord regeneration. Detailed study of SCI in these model organisms will help in the elucidation of molecular mechanisms of neuron regeneration across species.
Collapse
Affiliation(s)
- Ying Jiang
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Yang Cai
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Ning Yang
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Si Gao
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Qingwei Li
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Yue Pang
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Peng Su
- College of Life Science, Liaoning Normal University, Dalian, China
- Lamprey Research Center, Liaoning Normal University, Dalian, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
5
|
Shen W, Cai J, Li J, Li W, Shi P, Zhao X, Feng S. Regulation of MicroRNAs After Spinal Cord Injury in Adult Zebrafish. J Mol Neurosci 2024; 74:66. [PMID: 38990400 DOI: 10.1007/s12031-024-02242-2] [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: 06/09/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
Spinal cord injury (SCI) is a central nerve injury that often leads to loss of motor and sensory functions at or below the level of the injury. Zebrafish have a strong ability to repair after SCI, but the role of microRNAs (miRNAs) after SCI remains unclear. Locomotor behavior analysis showed that adult zebrafish recovered about 30% of their motor ability at 2 weeks and 55% at 3 weeks after SCI, reflecting their strong ability to repair SCI. Through miRNA sequencing, mRNA sequencing, RT-qPCR experiment verification, and bioinformatics predictive analysis, the key miRNAs and related genes in the repair of SCI were screened. A total of 38 miRNAs were significantly different, the top ten miRNAs were verified by RT-qPCR. The prediction target genes were verified by the mRNAs sequencing results at the same time point. Finally, 182 target genes were identified as likely to be networked regulated by the 38 different miRNAs. GO and KEGG enrichment analysis found that miRNAs targeted gene regulation of many key pathways, such as membrane tissue transport, ribosome function, lipid binding, and peroxidase activity. The PPI network analysis showed that miRNAs were involved in SCI repair through complex network regulation, among which dre-miR-21 may enhance cell reversibility through nop56, and that dre-miR-125c regulates axon growth through kpnb1 to repair SCI.
Collapse
Affiliation(s)
- Wenyuan Shen
- Department of Orthopedics, the Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong, 250033, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jun Cai
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, 300020, China
| | - Jinze Li
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Wenchang Li
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, 300020, China
| | - Pengcheng Shi
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, 300020, China
| | - Xiumei Zhao
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, 300020, China.
| | - Shiqing Feng
- Department of Orthopedics, the Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong, 250033, China.
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
6
|
Sintakova K, Romanyuk N. The role of small extracellular vesicles and microRNA as their cargo in the spinal cord injury pathophysiology and therapy. Front Neurosci 2024; 18:1400413. [PMID: 38774785 PMCID: PMC11106386 DOI: 10.3389/fnins.2024.1400413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating condition with a complex pathology that affects a significant portion of the population and causes long-term consequences. After primary injury, an inflammatory cascade of secondary injury occurs, followed by neuronal cell death and glial scar formation. Together with the limited regenerative capacity of the central nervous system, these are the main reasons for the poor prognosis after SCI. Despite recent advances, there is still no effective treatment. Promising therapeutic approaches include stem cells transplantation, which has demonstrated neuroprotective and immunomodulatory effects in SCI. This positive effect is thought to be mediated by small extracellular vesicles (sEVs); membrane-bound nanovesicles involved in intercellular communication through transport of functional proteins and RNA molecules. In this review, we summarize the current knowledge about sEVs and microRNA as their cargo as one of the most promising therapeutic approaches for the treatment of SCI. We provide a comprehensive overview of their role in SCI pathophysiology, neuroprotective potential and therapeutic effect.
Collapse
Affiliation(s)
- Kristyna Sintakova
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czechia
| | - Nataliya Romanyuk
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
7
|
Alzahrani FA, Riza YM, Eid TM, Almotairi R, Scherschinski L, Contreras J, Nadeem M, Perez SE, Raikwar SP, Jha RM, Preul MC, Ducruet AF, Lawton MT, Bhatia K, Akhter N, Ahmad S. Exosomes in Vascular/Neurological Disorders and the Road Ahead. Cells 2024; 13:670. [PMID: 38667285 PMCID: PMC11049650 DOI: 10.3390/cells13080670] [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: 03/22/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), stroke, and aneurysms, are characterized by the abnormal accumulation and aggregation of disease-causing proteins in the brain and spinal cord. Recent research suggests that proteins linked to these conditions can be secreted and transferred among cells using exosomes. The transmission of abnormal protein buildup and the gradual degeneration in the brains of impacted individuals might be supported by these exosomes. Furthermore, it has been reported that neuroprotective functions can also be attributed to exosomes in neurodegenerative diseases. The potential neuroprotective functions may play a role in preventing the formation of aggregates and abnormal accumulation of proteins associated with the disease. The present review summarizes the roles of exosomes in neurodegenerative diseases as well as elucidating their therapeutic potential in AD, PD, ALS, HD, stroke, and aneurysms. By elucidating these two aspects of exosomes, valuable insights into potential therapeutic targets for treating neurodegenerative diseases may be provided.
Collapse
Affiliation(s)
- Faisal A. Alzahrani
- Department of Biochemistry, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yasir M. Riza
- Department of Biochemistry, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Thamir M. Eid
- Department of Biochemistry, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Reema Almotairi
- Department of Medical Laboratory Technology, Prince Fahad bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Lea Scherschinski
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Jessica Contreras
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Muhammed Nadeem
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Sylvia E. Perez
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Sudhanshu P. Raikwar
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Ruchira M. Jha
- Department of Neurology, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Mark C. Preul
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Andrew F. Ducruet
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Michael T. Lawton
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Kanchan Bhatia
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA
| | - Naseem Akhter
- Department of Biology, Arizona State University, Lake Havasu City, AZ 86403, USA
| | - Saif Ahmad
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
- Phoenix Veterans Affairs (VA) Health Care System, Phoenix, AZ 85012, USA
| |
Collapse
|
8
|
Yu JYH, Chen TC, Danilov CA. MicroRNA-133b Dysregulation in a Mouse Model of Cervical Contusion Injury. Int J Mol Sci 2024; 25:3058. [PMID: 38474302 DOI: 10.3390/ijms25053058] [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: 02/01/2024] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Our previous research studies have demonstrated the role of microRNA133b (miR133b) in healing the contused spinal cord when administered either intranasally or intravenously 24 h following an injury. While our data showed beneficial effects of exogenous miR133b delivered within hours of a spinal cord injury (SCI), the kinetics of endogenous miR133b levels in the contused spinal cord and rostral/caudal segments of the injury were not fully investigated. In this study, we examined the miR133b dysregulation in a mouse model of moderate unilateral contusion injury at the fifth cervical (C5) level. Between 30 min and 7 days post-injury, mice were euthanized and tissues were collected from different areas of the spinal cord, ipsilateral and contralateral prefrontal motor cortices, and off-targets such as lung and spleen. The endogenous level of miR133b was determined by RT-qPCR. We found that after SCI, (a) most changes in miR133b level were restricted to the injured area with very limited alterations in the rostral and caudal parts relative to the injury site, (b) acute changes in the endogenous levels were predominantly specific to the lesion site with delayed miR133b changes in the motor cortex, and (c) ipsilateral and contralateral hemispheres responded differently to unilateral SCI. Our results suggest that the therapeutic window for exogenous miR133b therapy begins earlier than 24 h post-injury and potentially lasts longer than 7 days.
Collapse
Affiliation(s)
- James Young Ho Yu
- Department of Neurological Surgery, University of Southern California, 1200 N State St., Suite 3300, Los Angeles, CA 90033, USA
| | - Thomas C Chen
- Department of Neurological Surgery, University of Southern California, 1200 N State St., Suite 3300, Los Angeles, CA 90033, USA
| | - Camelia A Danilov
- Department of Neurological Surgery, University of Southern California, 2011 Zonal Ave., Los Angeles, CA 90089, USA
| |
Collapse
|
9
|
Wu Z, Feng K, Huang J, Ye X, Yang R, Huang Q, Jiang Q. Brain region changes following a spinal cord injury. Neurochem Int 2024; 174:105696. [PMID: 38354751 DOI: 10.1016/j.neuint.2024.105696] [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: 08/11/2023] [Revised: 01/16/2024] [Accepted: 02/10/2024] [Indexed: 02/16/2024]
Abstract
Brain-related complications are common in clinical practice after spinal cord injury (SCI); however, the molecular mechanisms of these complications are still unclear. Here, we reviewed the changes in the brain regions caused by SCI from three perspectives: imaging, molecular analysis, and electrophysiology. Imaging studies revealed abnormal functional connectivity, gray matter volume atrophy, and metabolic abnormalities in brain regions after SCI, leading to changes in the structure and function of brain regions. At the molecular level, chemokines, inflammatory factors, and damage-associated molecular patterns produced in the injured area were retrogradely transmitted through the corticospinal tract, cerebrospinal fluid, or blood circulation to the specific brain area to cause pathologic changes. Electrophysiologic recordings also suggested abnormal changes in brain electrical activity after SCI. Transcranial magnetic stimulation, transcranial direct current stimulation, and deep brain stimulation alleviated pain and improved motor function in patients with SCI; therefore, transcranial therapy may be a new strategy for the treatment of patients with SCI.
Collapse
Affiliation(s)
- Zhiwu Wu
- Department of Neurosurgery, Ganzhou People's Hospital (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16th Mei-guan Avenue, Ganzhou, 341000, China
| | - Kaiming Feng
- Department of Neurosurgery, Ganzhou People's Hospital (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16th Mei-guan Avenue, Ganzhou, 341000, China
| | - Jinqing Huang
- Department of Neurosurgery, Ganzhou People's Hospital (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16th Mei-guan Avenue, Ganzhou, 341000, China
| | - Xinyun Ye
- Department of Neurosurgery, Ganzhou People's Hospital (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16th Mei-guan Avenue, Ganzhou, 341000, China
| | - Ruijin Yang
- Department of Neurosurgery, Ganzhou People's Hospital (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16th Mei-guan Avenue, Ganzhou, 341000, China
| | - Qianliang Huang
- Department of Neurosurgery, Ganzhou People's Hospital (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16th Mei-guan Avenue, Ganzhou, 341000, China.
| | - Qiuhua Jiang
- Department of Neurosurgery, Ganzhou People's Hospital (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16th Mei-guan Avenue, Ganzhou, 341000, China.
| |
Collapse
|
10
|
Gupta S, Dutta S, Hui SP. Regenerative Potential of Injured Spinal Cord in the Light of Epigenetic Regulation and Modulation. Cells 2023; 12:1694. [PMID: 37443728 PMCID: PMC10341208 DOI: 10.3390/cells12131694] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 07/15/2023] Open
Abstract
A spinal cord injury is a form of physical harm imposed on the spinal cord that causes disability and, in many cases, leads to permanent mammalian paralysis, which causes a disastrous global issue. Because of its non-regenerative aspect, restoring the spinal cord's role remains one of the most daunting tasks. By comparison, the remarkable regenerative ability of some regeneration-competent species, such as some Urodeles (Axolotl), Xenopus, and some teleost fishes, enables maximum functional recovery, even after complete spinal cord transection. During the last two decades of intensive research, significant progress has been made in understanding both regenerative cells' origins and the molecular signaling mechanisms underlying the regeneration and reconstruction of damaged spinal cords in regenerating organisms and mammals, respectively. Epigenetic control has gradually moved into the center stage of this research field, which has been helped by comprehensive work demonstrating that DNA methylation, histone modifications, and microRNAs are important for the regeneration of the spinal cord. In this review, we concentrate primarily on providing a comparison of the epigenetic mechanisms in spinal cord injuries between non-regenerating and regenerating species. In addition, we further discuss the epigenetic mediators that underlie the development of a regeneration-permissive environment following injury in regeneration-competent animals and how such mediators may be implicated in optimizing treatment outcomes for spinal cord injurie in higher-order mammals. Finally, we briefly discuss the role of extracellular vesicles (EVs) in the context of spinal cord injury and their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Samudra Gupta
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India;
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
| | - Subhra Prakash Hui
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India;
| |
Collapse
|
11
|
Lim WQ, Michelle Luk KH, Lee KY, Nurul N, Loh SJ, Yeow ZX, Wong QX, Daniel Looi QH, Chong PP, How CW, Hamzah S, Foo JB. Small Extracellular Vesicles' miRNAs: Biomarkers and Therapeutics for Neurodegenerative Diseases. Pharmaceutics 2023; 15:pharmaceutics15041216. [PMID: 37111701 PMCID: PMC10143523 DOI: 10.3390/pharmaceutics15041216] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 04/29/2023] Open
Abstract
Neurodegenerative diseases are critical in the healthcare system as patients suffer from progressive diseases despite currently available drug management. Indeed, the growing ageing population will burden the country's healthcare system and the caretakers. Thus, there is a need for new management that could stop or reverse the progression of neurodegenerative diseases. Stem cells possess a remarkable regenerative potential that has long been investigated to resolve these issues. Some breakthroughs have been achieved thus far to replace the damaged brain cells; however, the procedure's invasiveness has prompted scientists to investigate using stem-cell small extracellular vesicles (sEVs) as a non-invasive cell-free therapy to address the limitations of cell therapy. With the advancement of technology to understand the molecular changes of neurodegenerative diseases, efforts have been made to enrich stem cells' sEVs with miRNAs to increase the therapeutic efficacy of the sEVs. In this article, the pathophysiology of various neurodegenerative diseases is highlighted. The role of miRNAs from sEVs as biomarkers and treatments is also discussed. Lastly, the applications and delivery of stem cells and their miRNA-enriched sEVs for treating neurodegenerative diseases are emphasised and reviewed.
Collapse
Affiliation(s)
- Wei Qing Lim
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Kie Hoon Michelle Luk
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Kah Yee Lee
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Nasuha Nurul
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Sin Jade Loh
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Zhen Xiong Yeow
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Qi Xuan Wong
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Qi Hao Daniel Looi
- My CytoHealth Sdn. Bhd., Lab 6, DMC Level 2, Hive 5, Taman Teknologi MRANTI, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Pan Pan Chong
- National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Sharina Hamzah
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
- Medical Advancement for Better Quality of Life Impact Lab, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
- Medical Advancement for Better Quality of Life Impact Lab, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| |
Collapse
|
12
|
Schepici G, Silvestro S, Mazzon E. Regenerative Effects of Exosomes-Derived MSCs: An Overview on Spinal Cord Injury Experimental Studies. Biomedicines 2023; 11:biomedicines11010201. [PMID: 36672709 PMCID: PMC9855467 DOI: 10.3390/biomedicines11010201] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition usually induced by the initial mechanical insult that can lead to permanent motor and sensory deficits. At present, researchers are investigating potential therapeutic strategies to ameliorate the neuro-inflammatory cascade that occurs post-injury. Although the use of mesenchymal stromal/stem (MSCs) as a potential therapy in application to regenerative medicine promoted anti-inflammatory and neuroprotective effects, several disadvantages limit their use. Therefore, recent studies have reported the effects of exosomes-derived MSCs (MSC-EXOs) as an innovative therapeutic option for SCI patients. It is noteworthy that MSC-EXOs can maintain the integrity of the blood-spinal cord barrier (BSCB), promoting angiogenic, proliferative, and anti-oxidant effects, as well as immunomodulatory, anti-inflammatory, and antiapoptotic properties. Therefore, in this study, we summarized the preclinical studies reported in the literature that have shown the effects of MSC-EXOs as a new molecular target to counteract the devastating effects of SCI.
Collapse
|
13
|
Sadrifar S, Abbasi-Dokht T, Forouzandeh S, Malek F, Yousefi B, Salek Farrokhi A, Karami J, Baharlou R. Immunomodulatory effects of probiotic supplementation in patients with asthma: a randomized, double-blind, placebo-controlled trial. Allergy Asthma Clin Immunol 2023; 19:1. [PMID: 36593510 PMCID: PMC9806812 DOI: 10.1186/s13223-022-00753-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/12/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Asthma is considered to be a chronic inflammatory disorder of the airways. Probiotics are living microorganisms that are found in the human gut and have protective effects against a wide range of diseases such as allergies. The aim of this study was to investigate the improvement of clinical asthma symptoms and changes in the expression pattern of selective microRNAs in patients with asthma and the changes in IL-4 and IFN-γ plasma levels after receiving probiotics. MATERIALS AND METHODS The present study was a randomized, double-blind, placebo-controlled trial that enrolled 40 asthmatic patients. They were treated with probiotics or placebo: 1 capsule/day for 8 weeks. Pulmonary function tests, IL-4 and IFN-γ levels, and expression of microRNAs were assessed at baseline and after treatment. RESULTS The results showed that the expression of miR-16, miR146-a and IL-4 levels in patients with asthma after receiving probiotic supplementation was significantly reduced and miR-133b expression was increased. In addition, pulmonary function tests showed a significant improvement in Forced Expiratory Volume in 1 s and Forced Vital Capacity after receiving probiotics. CONCLUSION In our study, 8-week treatment with probiotic supplementation led to reduced Th2 cells-associated IL-4 and improved Forced Expiratory Volume and Forced Vital Capacity. It appears probiotics can be used in addition to common asthma treatments.
Collapse
Affiliation(s)
- Sina Sadrifar
- grid.486769.20000 0004 0384 8779Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran ,grid.486769.20000 0004 0384 8779Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Tannaz Abbasi-Dokht
- grid.486769.20000 0004 0384 8779Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran ,grid.486769.20000 0004 0384 8779Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Sarvenaz Forouzandeh
- grid.486769.20000 0004 0384 8779Department of Internal medicine, Kosar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Farhad Malek
- grid.486769.20000 0004 0384 8779Department of Internal medicine, Kosar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- grid.486769.20000 0004 0384 8779Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran ,grid.486769.20000 0004 0384 8779Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Amir Salek Farrokhi
- grid.420169.80000 0000 9562 2611Department of Immunology, Pasteur Institute, Tehran, Iran
| | - Jafar Karami
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
| | - Rasoul Baharlou
- grid.486769.20000 0004 0384 8779Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran ,grid.486769.20000 0004 0384 8779Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
14
|
Li X, Jin DS, Eadara S, Caterina MJ, Meffert MK. Regulation by noncoding RNAs of local translation, injury responses, and pain in the peripheral nervous system. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 13:100119. [PMID: 36798094 PMCID: PMC9926024 DOI: 10.1016/j.ynpai.2023.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 06/18/2023]
Abstract
Neuropathic pain is a chronic condition arising from damage to somatosensory pathways that results in pathological hypersensitivity. Persistent pain can be viewed as a consequence of maladaptive plasticity which, like most enduring forms of cellular plasticity, requires altered expression of specific gene programs. Control of gene expression at the level of protein synthesis is broadly utilized to directly modulate changes in activity and responsiveness in nociceptive pathways and provides an effective mechanism for compartmentalized regulation of the proteome in peripheral nerves through local translation. Levels of noncoding RNAs (ncRNAs) are commonly impacted by peripheral nerve injury leading to persistent pain. NcRNAs exert spatiotemporal regulation of local proteomes and affect signaling cascades supporting altered sensory responses that contribute to hyperalgesia. This review discusses ncRNAs found in the peripheral nervous system (PNS) that are dysregulated following nerve injury and the current understanding of their roles in pathophysiological pain-related responses including neuroimmune interactions, neuronal survival and axon regeneration, Schwann cell dedifferentiation and proliferation, intercellular communication, and the generation of ectopic action potentials in primary afferents. We review progress in the field beyond cataloging, with a focus on the relevant target transcripts and mechanisms underlying pain modulation by ncRNAs.
Collapse
Affiliation(s)
- Xinbei Li
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
| | - Daniel S. Jin
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
| | - Sreenivas Eadara
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
| | - Michael J. Caterina
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
- Department of Neurosurgery and Neurosurgery Pain Research Institute, Johns Hopkins University School of Medicine, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, United States
| | - Mollie K. Meffert
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, United States
| |
Collapse
|
15
|
Wang H, Wang Q, Xiao X, Luo X, Gao L. Clinical Trials of Non-Coding RNAs as Diagnostic and Therapeutic Biomarkers for Central Nervous System Injuries. Curr Neuropharmacol 2023; 21:2237-2246. [PMID: 36443964 PMCID: PMC10556392 DOI: 10.2174/1570159x21666221128090025] [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: 09/06/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Huiqing Wang
- Medical Simulation Centre, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Qiang Wang
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Xiao Xiao
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University and the Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, P.R. China
| | - Xiaolei Luo
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Linbo Gao
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, NHC Key Laboratory of Chronobiology, Sichuan University, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| |
Collapse
|
16
|
Villalobos-Escobedo JM, Martínez-Hernández JP, Pelagio-Flores R, González-De la Rosa PM, Carreras-Villaseñor N, Abreu-Goodger C, Herrera-Estrella AH. Trichoderma atroviride hyphal regeneration and conidiation depend on cell-signaling processes regulated by a microRNA-like RNA. Microb Genom 2022; 8. [PMID: 36239595 DOI: 10.1099/mgen.0.000869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The ability to respond to injury is essential for the survival of an organism and involves analogous mechanisms in animals and plants. Such mechanisms integrate coordinated genetic and metabolic reprogramming events requiring regulation by small RNAs for adequate healing of the wounded area. We have previously reported that the response to injury of the filamentous fungus Trichoderma atroviride involves molecular mechanisms closely resembling those of plants and animals that lead to the formation of new hyphae (regeneration) and the development of asexual reproduction structures (conidiophores). However, the involvement of microRNAs in this process has not been investigated in fungi. In this work, we explore the participation of microRNA-like RNAs (milRNAs) molecules by sequencing messenger and small RNAs during the injury response of the WT strain and RNAi mutants. We found that Dcr2 appears to play an important role in hyphal regeneration and is required to produce the majority of sRNAs in T. atroviride. We also determined that the three main milRNAs produced via Dcr2 are induced during the damage-triggered developmental process. Importantly, elimination of a single milRNA phenocopied the main defects observed in the dcr2 mutant. Our results demonstrate the essential role of milRNAs in hyphal regeneration and asexual development by post-transcriptionally regulating cellular signalling processes involving phosphorylation events. These observations allow us to conclude that fungi, like plants and animals, in response to damage activate fine-tuning regulatory mechanisms.
Collapse
Affiliation(s)
- José M Villalobos-Escobedo
- Laboratorio Nacional de Genómica para la Biodiversidad-Unidad de Genómica Avanzada, Cinvestav. Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Gto, Mexico
| | - J Pedro Martínez-Hernández
- Laboratorio Nacional de Genómica para la Biodiversidad-Unidad de Genómica Avanzada, Cinvestav. Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Gto, Mexico
| | - Ramón Pelagio-Flores
- Laboratorio Nacional de Genómica para la Biodiversidad-Unidad de Genómica Avanzada, Cinvestav. Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Gto, Mexico.,Present address: Facultad de Químico Farmacobiología, Universidad Michoacana de San Nicolás de Hidalgo, C.P. 58030 Morelia, Michoacán, Mexico
| | - Pablo M González-De la Rosa
- Laboratorio Nacional de Genómica para la Biodiversidad-Unidad de Genómica Avanzada, Cinvestav. Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Gto, Mexico.,Present address: Tree of Life, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Nohemí Carreras-Villaseñor
- Laboratorio Nacional de Genómica para la Biodiversidad-Unidad de Genómica Avanzada, Cinvestav. Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Gto, Mexico.,Present address: Red de Estudios Moleculares Avanzados, Instituto de Ecología A.C. C.P. 91070 Xalapa, Veracruz, Mexico
| | - Cei Abreu-Goodger
- Laboratorio Nacional de Genómica para la Biodiversidad-Unidad de Genómica Avanzada, Cinvestav. Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Gto, Mexico.,Present address: Institute of Ecology and Evolution, University of Edinburgh, Edinburgh, UK
| | - Alfredo H Herrera-Estrella
- Laboratorio Nacional de Genómica para la Biodiversidad-Unidad de Genómica Avanzada, Cinvestav. Km 9.6 Libramiento Norte Carretera Irapuato-León, 36824, Irapuato, Gto, Mexico
| |
Collapse
|
17
|
RhoA Signaling in Neurodegenerative Diseases. Cells 2022; 11:cells11091520. [PMID: 35563826 PMCID: PMC9103838 DOI: 10.3390/cells11091520] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023] Open
Abstract
Ras homolog gene family member A (RhoA) is a small GTPase of the Rho family involved in regulating multiple signal transduction pathways that influence a diverse range of cellular functions. RhoA and many of its downstream effector proteins are highly expressed in the nervous system, implying an important role for RhoA signaling in neurons and glial cells. Indeed, emerging evidence points toward a role of aberrant RhoA signaling in neurodegenerative diseases such as Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. In this review, we summarize the current knowledge of RhoA regulation and downstream cellular functions with an emphasis on the role of RhoA signaling in neurodegenerative diseases and the therapeutic potential of RhoA inhibition in neurodegeneration.
Collapse
|
18
|
Gelosa P, Castiglioni L, Rzemieniec J, Muluhie M, Camera M, Sironi L. Cerebral derailment after myocardial infarct: mechanisms and effects of the signaling from the ischemic heart to brain. J Mol Med (Berl) 2022; 100:23-41. [PMID: 34674004 PMCID: PMC8724191 DOI: 10.1007/s00109-021-02154-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/07/2021] [Accepted: 10/14/2021] [Indexed: 12/04/2022]
Abstract
Myocardial infarction (MI) is the leading cause of death among ischemic heart diseases and is associated with several long-term cardiovascular complications, such as angina, re-infarction, arrhythmias, and heart failure. However, MI is frequently accompanied by non-cardiovascular multiple comorbidities, including brain disorders such as stroke, anxiety, depression, and cognitive impairment. Accumulating experimental and clinical evidence suggests a causal relationship between MI and stroke, but the precise underlying mechanisms have not yet been elucidated. Indeed, the risk of stroke remains a current challenge in patients with MI, in spite of the improvement of medical treatment among this patient population has reduced the risk of stroke. In this review, the effects of the signaling from the ischemic heart to the brain, such as neuroinflammation, neuronal apoptosis, and neurogenesis, and the possible actors mediating these effects, such as systemic inflammation, immunoresponse, extracellular vesicles, and microRNAs, are discussed.
Collapse
Affiliation(s)
- Paolo Gelosa
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Laura Castiglioni
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Joanna Rzemieniec
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Majeda Muluhie
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - Marina Camera
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
- Centro Cardiologico Monzino, 20138, Milan, Italy
| | - Luigi Sironi
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy.
| |
Collapse
|
19
|
Kim GU, Sung SE, Kang KK, Choi JH, Lee S, Sung M, Yang SY, Kim SK, Kim YI, Lim JH, Seo MS, Lee GW. Therapeutic Potential of Mesenchymal Stem Cells (MSCs) and MSC-Derived Extracellular Vesicles for the Treatment of Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms222413672. [PMID: 34948463 PMCID: PMC8703906 DOI: 10.3390/ijms222413672] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a life-threatening condition that leads to permanent disability with partial or complete loss of motor, sensory, and autonomic functions. SCI is usually caused by initial mechanical insult, followed by a cascade of several neuroinflammation and structural changes. For ameliorating the neuroinflammatory cascades, MSC has been regarded as a therapeutic agent. The animal SCI research has demonstrated that MSC can be a valuable therapeutic agent with several growth factors and cytokines that may induce anti-inflammatory and regenerative effects. However, the therapeutic efficacy of MSCs in animal SCI models is inconsistent, and the optimal method of MSCs remains debatable. Moreover, there are several limitations to developing these therapeutic agents for humans. Therefore, identifying novel agents for regenerative medicine is necessary. Extracellular vesicles are a novel source for regenerative medicine; they possess nucleic acids, functional proteins, and bioactive lipids and perform various functions, including damaged tissue repair, immune response regulation, and reduction of inflammation. MSC-derived exosomes have advantages over MSCs, including small dimensions, low immunogenicity, and no need for additional procedures for culture expansion or delivery. Certain studies have demonstrated that MSC-derived extracellular vesicles (EVs), including exosomes, exhibit outstanding chondroprotective and anti-inflammatory effects. Therefore, we reviewed the principles and patho-mechanisms and summarized the research outcomes of MSCs and MSC-derived EVs for SCI, reported to date.
Collapse
Affiliation(s)
- Gang-Un Kim
- Department of Orthopedic Surgery, Hanil General Hospital, 308 Uicheon-ro, Dobong-gu, Seoul 01450, Korea;
| | - Soo-Eun Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Kyung-Ku Kang
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Joo-Hee Choi
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Sijoon Lee
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Minkyoung Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Seung Yun Yang
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea;
| | - Seul-Ki Kim
- Efficacy Evaluation Team, Food Science R&D Center, KolmarBNH CO., LTD, 61Heolleungro 8-gil, Seocho-gu, Seoul 06800, Korea;
| | | | - Ju-Hyeon Lim
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk 28160, Korea;
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| | - Min-Soo Seo
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
- Correspondence: (M.-S.S.); (G.W.L.); Tel.: +82-53-7905727 (M.S.S.); +82-53-6203642 (G.W.L.)
| | - Gun Woo Lee
- Cellexobio, Co. Ltd., Daegu 42415, Korea;
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
- Correspondence: (M.-S.S.); (G.W.L.); Tel.: +82-53-7905727 (M.S.S.); +82-53-6203642 (G.W.L.)
| |
Collapse
|
20
|
Shen WY, Fu XH, Cai J, Li WC, Fan BY, Pang YL, Zhao CX, Abula M, Kong XH, Yao X, Feng SQ. Identification of key genes involved in recovery from spinal cord injury in adult zebrafish. Neural Regen Res 2021; 17:1334-1342. [PMID: 34782579 PMCID: PMC8643032 DOI: 10.4103/1673-5374.327360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Zebrafish are an effective vertebrate model to study the mechanisms underlying recovery after spinal cord injury. The subacute phase after spinal cord injury is critical to the recovery of neurological function, which involves tissue bridging and axon regeneration. In this study, we found that zebrafish spontaneously recovered 44% of their swimming ability within the subacute phase (2 weeks) after spinal cord injury. During this period, we identified 7762 differentially expressed genes in spinal cord tissue: 2950 were up-regulated and 4812 were down-regulated. These differentially expressed genes were primarily concentrated in the biological processes of the respiratory chain, axon regeneration, and cell-component morphogenesis. The genes were also mostly involved in the regulation of metabolic pathways, the cell cycle, and gene-regulation pathways. We verified the gene expression of two differentially expressed genes, clasp2 up-regulation and h1m down-regulation, in zebrafish spinal cord tissue in vitro. Pathway enrichment analysis revealed that up-regulated clasp2 functions similarly to microtubule-associated protein, which is responsible for axon extension regulated by microtubules. Down-regulated h1m controls endogenous stem cell differentiation after spinal cord injury. This study provides new candidate genes, clasp2 and h1m, as potential therapeutic intervention targets for spinal cord injury repair by neuroregeneration. All experimental procedures and protocols were approved by the Animal Ethics Committee of Tianjin Institute of Medical & Pharmaceutical Sciences (approval No. IMPS-EAEP-Q-2019-02) on September 24, 2019.
Collapse
Affiliation(s)
- Wen-Yuan Shen
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuan-Hao Fu
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Cai
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Wen-Chang Li
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Bao-You Fan
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-Lin Pang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen-Xi Zhao
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Muhtidir Abula
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | | | - Xue Yao
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shi-Qing Feng
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
21
|
Potential of different cells-derived exosomal microRNA cargos for treating spinal cord injury. J Orthop Translat 2021; 31:33-40. [PMID: 34760623 PMCID: PMC8560648 DOI: 10.1016/j.jot.2021.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is a disastrous situation that affects many patients worldwide. A profound understanding of the pathology and etiology of SCI is of great importance in inspiring new therapeutic concepts and treatment. In recent years, exosomes, which are complex lipid membrane structures secreted nearly by all kinds of plants and animal cells, can transport their valuable cargoes (e.g., proteins, lipids, RNAs) to the targeted cells and exert their communication and regulation functions, which open up a new field of treatment of SCI. Notably, the exosome's advantage is transporting the carried material to the target cells across the blood-brain barrier and exerting regulatory functions. Among the cargoes of exosomes, microRNAs, through the modulation of their mRNA targets, emerges with great potentiality in the pathological process, diagnosis and treatment of SCI. In this review, we discuss the role of miRNAs transported by different cell-derived exosomes in SCI that are poised to enhance SCI-specific therapeutic capabilities of exosomes.
Collapse
|
22
|
Huang W, Lin M, Yang C, Wang F, Zhang M, Gao J, Yu X. Rat Bone Mesenchymal Stem Cell-Derived Exosomes Loaded with miR-494 Promoting Neurofilament Regeneration and Behavioral Function Recovery after Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1634917. [PMID: 34635862 PMCID: PMC8501401 DOI: 10.1155/2021/1634917] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/05/2021] [Accepted: 09/09/2021] [Indexed: 01/08/2023]
Abstract
Exosomes (Exo) exhibit numerous advantages (e.g., good encapsulation, high targeting efficiency, and easy to penetrate the blood-brain barrier to the central nervous system). Exosomes are recognized as prominent carriers of mRNAs, siRNAs, miRNAs, proteins, and other bioactive molecules. As confirmed by existing studies, miR-494 is important to regulate the occurrence, progression, and repair of spinal cord injury (SCI). We constructed miR-494-modified exosomes (Exo-miR-494). As indicated from related research in vitro and vivo, Exo-miR-494 is capable of effectively inhibiting the inflammatory response and neuronal apoptosis in the injured area, as well as upregulating various anti-inflammatory factors and miR-494 to protect neurons. Moreover, it can promote the regeneration of the neurofilament and improve the recovery of behavioral function of SCI rats.
Collapse
Affiliation(s)
- Wei Huang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
- Department of Orthopaedics, Dongguan Tungwah Hospital, Dongguan 523000, China
| | - Miaoman Lin
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Cunheng Yang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Fumin Wang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Meng Zhang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Junxiao Gao
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Xiaobing Yu
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| |
Collapse
|
23
|
Ribeiro AO, de Oliveira AC, Costa JM, Nachtigall PG, Herkenhoff ME, Campos VF, Delella FK, Pinhal D. MicroRNA roles in regeneration: Multiple lessons from zebrafish. Dev Dyn 2021; 251:556-576. [PMID: 34547148 DOI: 10.1002/dvdy.421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/23/2021] [Accepted: 09/07/2021] [Indexed: 12/23/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs with pivotal roles in the control of gene expression. By comparing the miRNA profiles of uninjured vs. regenerating tissues and structures, several studies have found that miRNAs are potentially involved in the regenerative process. By inducing miRNA overexpression or inhibition, elegant experiments have directed regenerative responses validating relevant miRNA-to-target interactions. The zebrafish (Danio rerio) has been the epicenter of regenerative research because of its exceptional capability to self-repair damaged tissues and body structures. In this review, we discuss recent discoveries that have improved our understanding of the impact of gene regulation mediated by miRNAs in the context of the regeneration of fins, heart, retina, and nervous tissue in zebrafish. We compiled what is known about the miRNA control of regeneration in these tissues and investigated the links among up-regulated and down-regulated miRNAs, their putative or validated targets, and the regenerative process. Finally, we briefly discuss the forthcoming prospects, highlighting directions and the potential for further development of this field.
Collapse
Affiliation(s)
- Amanda Oliveira Ribeiro
- Laboratório Genômica e Evolução Molecular (LGEM), Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu, SP, Brazil
| | - Arthur Casulli de Oliveira
- Laboratório Genômica e Evolução Molecular (LGEM), Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu, SP, Brazil
| | - Juliana Mara Costa
- Laboratório Genômica e Evolução Molecular (LGEM), Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu, SP, Brazil
| | - Pedro Gabriel Nachtigall
- Laboratório Genômica e Evolução Molecular (LGEM), Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu, SP, Brazil.,Laboratório Especial de Toxicologia Aplicada (LETA), CeTICS, Instituto Butantan, São Paulo, SP, Brazil
| | - Marcos Edgar Herkenhoff
- Laboratório Genômica e Evolução Molecular (LGEM), Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu, SP, Brazil.,Departamento de Tecnologia Bioquímico-Farmacêutica, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vinicius Farias Campos
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Flávia Karina Delella
- Departamento de Biologia Estrutural e Funcional, Instituto de Biociências, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu, SP, Brazil
| | - Danillo Pinhal
- Laboratório Genômica e Evolução Molecular (LGEM), Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista Júlio de Mesquita Filho (UNESP), Botucatu, SP, Brazil
| |
Collapse
|
24
|
Khyeam S, Lee S, Huang GN. Genetic, Epigenetic, and Post-Transcriptional Basis of Divergent Tissue Regenerative Capacities Among Vertebrates. ADVANCED GENETICS (HOBOKEN, N.J.) 2021; 2:e10042. [PMID: 34423307 PMCID: PMC8372189 DOI: 10.1002/ggn2.10042] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/29/2022]
Abstract
Regeneration is widespread across the animal kingdom but varies vastly across phylogeny and even ontogeny. Adult mammalian regeneration in most organs and appendages is limited, while vertebrates such as zebrafish and salamanders are able to regenerate various organs and body parts. Here, we focus on the regeneration of appendages, spinal cord, and heart - organs and body parts that are highly regenerative among fish and amphibian species but limited in adult mammals. We then describe potential genetic, epigenetic, and post-transcriptional similarities among these different forms of regeneration across vertebrates and discuss several theories for diminished regenerative capacity throughout evolution.
Collapse
Affiliation(s)
- Sheamin Khyeam
- Cardiovascular Research Institute and Department of PhysiologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell ResearchUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Sukjun Lee
- Cardiovascular Research Institute and Department of PhysiologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell ResearchUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Guo N. Huang
- Cardiovascular Research Institute and Department of PhysiologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell ResearchUniversity of CaliforniaSan FranciscoCaliforniaUSA
| |
Collapse
|
25
|
Nozohouri S, Vaidya B, Abbruscato TJ. Exosomes in Ischemic Stroke. Curr Pharm Des 2021; 26:5533-5545. [PMID: 32534564 DOI: 10.2174/1381612826666200614180253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Ischemic stroke, a leading cause of mortality, results in severe neurological outcomes in the patients. Effective stroke therapies may significantly decrease the extent of injury. For this purpose, novel and efficient drug delivery strategies need to be developed. Among a myriad of therapeutic and drug delivery techniques, exosomes have shown promising results in ischemic stroke either by their intrinsic therapeutic characteristics, which can result in angiogenesis and neurogenesis or by acting as competent, biocompatible drug delivery vehicles to transport neurotherapeutic agents into the brain. In this review, we have discussed different methods of exosome isolation and cargo loading techniques, advantages and disadvantages of using exosomes as a drug delivery carrier and the therapeutic applications of exosomes with a focus on ischemic stroke therapy.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| |
Collapse
|
26
|
Ghibaudi M, Boido M, Green D, Signorino E, Berto GE, Pourshayesteh S, Singh A, Di Cunto F, Dalmay T, Vercelli A. miR-7b-3p Exerts a Dual Role After Spinal Cord Injury, by Supporting Plasticity and Neuroprotection at Cortical Level. Front Mol Biosci 2021; 8:618869. [PMID: 33869277 PMCID: PMC8044879 DOI: 10.3389/fmolb.2021.618869] [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] [Received: 10/18/2020] [Accepted: 03/04/2021] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury (SCI) affects 6 million people worldwide with no available treatment. Despite research advances, the inherent poor regeneration potential of the central nervous system remains a major hurdle. Small RNAs (sRNAs) 19-33 nucleotides in length are a set of non-coding RNA molecules that regulate gene expression and have emerged as key players in regulating cellular events occurring after SCI. Here we profiled a class of sRNA known as microRNAs (miRNAs) following SCI in the cortex where the cell bodies of corticospinal motor neurons are located. We identified miR-7b-3p as a candidate target given its significant upregulation after SCI in vivo and we screened by miRWalk PTM the genes predicted to be targets of miR-7b-3p (among which we identified Wipf2, a gene regulating neurite extension). Moreover, 16 genes, involved in neural regeneration and potential miR-7b-3p targets, were found to be downregulated in the cortex following SCI. We also analysed miR-7b-3p function during cortical neuron development in vitro: we observed that the overexpression of miR-7b-3p was important (1) to maintain neurons in a more immature and, likely, plastic neuronal developmental phase and (2) to contrast the apoptotic pathway; however, in normal conditions it did not affect the Wipf2 expression. On the contrary, the overexpression of miR-7b-3p upon in vitro oxidative stress condition (mimicking the SCI environment) significantly reduced the expression level of Wipf2, as observed in vivo, confirming it as a direct miR-7b-3p target. Overall, these data suggest a dual role of miR-7b-3p: (i) the induction of a more plastic neuronal condition/phase, possibly at the expense of the axon growth, (ii) the neuroprotective role exerted through the inhibition of the apoptotic cascade. Increasing the miR-7b-3p levels in case of SCI could reactivate in adult neurons silenced developmental programmes, supporting at the same time the survival of the axotomised neurons.
Collapse
Affiliation(s)
- Matilde Ghibaudi
- Department of Neuroscience “Rita Levi Montalcini,” Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
- Polymers and Biomaterials, Italian Institute of Technology, Genova, Italy
| | - Marina Boido
- Department of Neuroscience “Rita Levi Montalcini,” Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Darrell Green
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Elena Signorino
- Department of Neuroscience “Rita Levi Montalcini,” Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Gaia Elena Berto
- Department of Neuroscience “Rita Levi Montalcini,” Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Soraya Pourshayesteh
- Department of Neuroscience “Rita Levi Montalcini,” Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Archana Singh
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Ferdinando Di Cunto
- Department of Neuroscience “Rita Levi Montalcini,” Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Tamas Dalmay
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Alessandro Vercelli
- Department of Neuroscience “Rita Levi Montalcini,” Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| |
Collapse
|
27
|
Liu WZ, Ma ZJ, Li JR, Kang XW. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther 2021; 12:102. [PMID: 33536064 PMCID: PMC7860030 DOI: 10.1186/s13287-021-02153-8] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/07/2021] [Indexed: 12/31/2022] Open
Abstract
Spinal cord injury (SCI) often leads to serious motor and sensory dysfunction of the limbs below the injured segment. SCI not only results in physical and psychological harm to patients but can also cause a huge economic burden on their families and society. As there is no effective treatment method, the prevention, treatment, and rehabilitation of patients with SCI have become urgent problems to be solved. In recent years, mesenchymal stem cells (MSCs) have attracted more attention in the treatment of SCI. Although MSC therapy can reduce injured volume and promote axonal regeneration, its application is limited by tumorigenicity, a low survival rate, and immune rejection. Accumulating literature shows that exosomes have great potential in the treatment of SCI. In this review, we summarize the existing MSC-derived exosome studies on SCI and discuss the advantages and challenges of treating SCI based on exosomes derived from MSCs.
Collapse
Affiliation(s)
- Wen-Zhao Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China
| | - Zhan-Jun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China
| | - Jie-Ru Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Xue-Wen Kang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China.
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China.
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
28
|
Zheng X, Hermann DM, Bähr M, Doeppner TR. The role of small extracellular vesicles in cerebral and myocardial ischemia-Molecular signals, treatment targets, and future clinical translation. Stem Cells 2021; 39:403-413. [PMID: 33432732 DOI: 10.1002/stem.3329] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/09/2020] [Accepted: 12/13/2020] [Indexed: 12/17/2022]
Abstract
The heart and the brain mutually interact with each other, forming a functional axis that is disturbed under conditions of ischemia. Stem cell-derived extracellular vesicles (EVs) show great potential for the treatment of ischemic stroke and myocardial infarction. Due to heart-brain interactions, therapeutic actions of EVs in the brain and the heart cannot be regarded in an isolated way. Effects in each of the two organs reciprocally influence the outcome of the other. Stem cell-derived EVs modulate a large number of signaling pathways in both tissues. Upon ischemia, EVs prevent delayed injury, promote angiogenesis, enhance parenchymal remodeling, and enable functional tissue recovery. The therapeutic effects greatly depend on EV cargos, among which are noncoding RNAs like microRNAs (miRNAs) and proteins, which modulate cell signaling in a differential way that not always corresponds to each other in the two tissues. Interestingly, the same miRNA or protein localized in EVs can modulate different signaling pathways in the ischemic heart and brain, which may have diverse consequences for disease outcomes. Paying careful attention to unveiling these underlying mechanisms may provide new insights into tissue remodeling processes and identify targets for ischemic stroke and myocardial infarction therapies. Some of these mechanisms are discussed in this concise review, and consequences for the clinical translation of EVs are presented.
Collapse
Affiliation(s)
- Xuan Zheng
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
29
|
Li F, Zhang J, Chen A, Liao R, Duan Y, Xu Y, Tao L. Combined transplantation of neural stem cells and bone marrow mesenchymal stem cells promotes neuronal cell survival to alleviate brain damage after cardiac arrest via microRNA-133b incorporated in extracellular vesicles. Aging (Albany NY) 2021; 13:262-278. [PMID: 33436530 PMCID: PMC7835040 DOI: 10.18632/aging.103920] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/25/2020] [Indexed: 02/06/2023]
Abstract
Neural stem cell (NSC) transplantation has prevailed as a promising protective strategy for cardiac arrest (CA)-induced brain damage. Surprisingly, the poor survival of neuronal cells in severe hypoxic condition restricts the utilization of this cell-based therapy. Extracellular vesicles (EVs) transfer microRNAs (miRNAs) between cells are validated as the mode for the release of several therapeutic molecules. The current study reports that the bone marrow mesenchymal stem cells (BMSCs) interact with NSCs via EVs thereby affecting the survival of neuronal cells. Hypoxic injury models of neuronal cells were established using cobalt chloride, followed by co-culture with BMSCs and NSCs alone or in combination. BMSCs combined with NSCs elicited as a superior protocol to stimulate neuronal cell survival. BMSCs-derived EVs could protect neuronal cells against hypoxic injury. Silencing of miR-133b incorporated in BMSCs-derived EVs could decrease the cell viability and the number of NeuN-positive cells and increase the apoptosis in the CA rat model. BMSCs-derived EVs could transfer miR-133b to neuronal cells to activate the AKT-GSK-3β-WNT-3 signaling pathway by targeting JAK1. Our study demonstrates that NSCs promotes the release of miR-133b from BMSCs-derived EVs to promote neuronal cell survival, representing a potential therapeutic strategy for the treatment of CA-induced brain damage.
Collapse
Affiliation(s)
- Fang Li
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunan Province, P.R. China
| | - Jie Zhang
- The 2nd Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunan Province, P.R. China
| | - Anbao Chen
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunan Province, P.R. China
| | - Rui Liao
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunan Province, P.R. China
| | - Yongchun Duan
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunan Province, P.R. China
| | - Yuwei Xu
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunan Province, P.R. China
| | - Lili Tao
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunan Province, P.R. China
| |
Collapse
|
30
|
Rho GTPases Signaling in Zebrafish Development and Disease. Cells 2020; 9:cells9122634. [PMID: 33302361 PMCID: PMC7762611 DOI: 10.3390/cells9122634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 02/08/2023] Open
Abstract
Cells encounter countless external cues and the specificity of their responses is translated through a myriad of tightly regulated intracellular signals. For this, Rho GTPases play a central role and transduce signals that contribute to fundamental cell dynamic and survival events. Here, we review our knowledge on how zebrafish helped us understand the role of some of these proteins in a multitude of in vivo cellular behaviors. Zebrafish studies offer a unique opportunity to explore the role and more specifically the spatial and temporal dynamic of Rho GTPases activities within a complex environment at a level of details unachievable in any other vertebrate organism.
Collapse
|
31
|
Wu WD, Wang LH, Wei NX, Kong DH, Shao G, Zhang SR, Du YS. MicroRNA-15a inhibits inflammatory response and apoptosis after spinal cord injury via targeting STAT3. EUROPEAN REVIEW FOR MEDICAL AND PHARMACOLOGICAL SCIENCES 2020; 23:9189-9198. [PMID: 31773669 DOI: 10.26355/eurrev_201911_19409] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To clarify the function of microRNA-15a in the spinal cord injury (SCI) and its potential mechanism. PATIENTS AND METHODS The plasma levels of microRNA-15a and signal transducer and activator of transcription 3 (STAT3) in SCI patients were determined by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). The correlation between the expressions of microRNA-15a and STAT3 was analyzed. The in vitro SCI model was established in H2O2-induced C8-D1A and C8B4 cells, and in vivo SCI model was established in mice by hitting T10. The mRNA and protein expressions of tumor necrosis factor-α (TNF-α) and interleukin 6 (IL-6) were detected in the SCI model. The apoptosis was examined by flow cytometry or TUNEL staining, respectively. The motor function of mouse hindlimb was evaluated using the Basso Beattie Bresnahan (BBB) standard scale. The target gene of microRNA-15a was predicted by bioinformatics and further verified by dual-luciferase reporter gene assay. The expression changes of target genes in C8-D1A and C8B4 cells with microRNA-15a overexpression or knockdown were examined by qRT-PCR and Western blot. Finally, rescue experiments were performed to evaluate the regulatory effects of microRNA-15a and STAT3 on cell apoptosis. RESULTS MicroRNA-15a was lowly expressed in plasma of SCI patients, while STAT3 was highly expressed with a negative correlation to microRNA-15a. Identically, microRNA-15a was lowly expressed in H2O2-induced C8-D1A and C8B4 cells, and STAT3 was highly expressed. MicroRNA-15a overexpression downregulated mRNA and protein levels of TNF-α and IL-6 in C8-D1A and C8B4 cells. BBB score was markedly low in SCI mice relative to controls. SCI mice injected with microRNA-15a mimics had higher BBB score than those injected with negative control. Besides, SCI mice with microRNA-15a overexpression had downregulated expressions of STAT3, TNF-α, and IL-6 in the impaired spinal cord tissues, as well as lower apoptotic rate. Through bioinformatics, we found binding sites between STAT3 and microRNA-15a. Their binding conditions were further verified by dual-luciferase reporter gene assay. Moreover, STAT3 expression was negatively regulated by microRNA-15a. Finally, rescue experiments showed that STAT3 overexpression could reverse the regulatory effects of microRNA-15a on expressions of TNF-α and IL-6, as well as apoptosis. CONCLUSIONS MicroRNA-15a expression decreases in the SCI model, which participates in the process of SCI by regulating inflammatory response and cell apoptosis via targeting STAT3.
Collapse
Affiliation(s)
- W-D Wu
- Anesthesiology Department, Danyang People's Hospital of Jiangsu Province & Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu, China.
| | | | | | | | | | | | | |
Collapse
|
32
|
Downregulation of circular RNA HECTD1 induces neuroprotection against ischemic stroke through the microRNA-133b/TRAF3 pathway. Life Sci 2020; 264:118626. [PMID: 33148417 DOI: 10.1016/j.lfs.2020.118626] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 01/16/2023]
Abstract
AIMS Circular RNAs (circRNAs) have been shown to play crucial roles in various biological processes and human diseases. However, their exact functions in ischemic stroke remain largely unknown. In this study, we explored the functional role of circRNA HECTD1 (circ-HECTD1) and its underlying mechanism in cerebral ischemia/reperfusion injury. METHODS Mouse middle cerebral artery occlusion (MCAO) model and oxygen-glucose deprivation (OGD) model in HT22 cells were used to mimic the cerebral ischemia/reperfusion injury. Brain infarct volume, flow cytometry, caspase 3 activity, NF-κB activity, and TUNEL staining were performed to evaluate the function of circ-HECTD1. Luciferase report assay was used to explore the regulatory mechanism. FINDINGS The results showed that the expression of circ-HECTD1 and tumor necrosis factor receptor-associated factor 3 (TRAF3) was remarkably up-regulated, while miR-133b was down-regulated in oxygen-glucose deprivation (OGD)-induced HT22 cells and mouse middle cerebral artery occlusion (MCAO) model. circ-HECTD1 knockdown relieved OGD-caused neuronal cell death in vitro. Simultaneously, circ-HECTD1 knockdown improved cerebral infarction volume and neuronal apoptosis in MCAO mice. circ-HECTD1 was able to negatively regulate the expression of miR-133b, and TRAF3 is one of the targets of miR-133b. Upregulation of miR-133b inhibited the expression of TRAF3 in OGD-stimulated cells, whereas circ-HECTD1 upregulation reversed this effect. Furthermore, upregulation of miR-133 was able to inhibit OGD-caused cell apoptosis and NF-κB activation, whereas upregulation of circ-HECTD1 attenuated these effects of miR-133b mimics. SIGNIFICANCE Taken together, circ-HECTD1 knockdown inhibited the expression of TRAF3 by targeting miR-133b, thereby attenuating neuronal injury caused by cerebral ischemia.
Collapse
|
33
|
West-Livingston LN, Park J, Lee SJ, Atala A, Yoo JJ. The Role of the Microenvironment in Controlling the Fate of Bioprinted Stem Cells. Chem Rev 2020; 120:11056-11092. [PMID: 32558555 PMCID: PMC7676498 DOI: 10.1021/acs.chemrev.0c00126] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The field of tissue engineering and regenerative medicine has made numerous advances in recent years in the arena of fabricating multifunctional, three-dimensional (3D) tissue constructs. This can be attributed to novel approaches in the bioprinting of stem cells. There are expansive options in bioprinting technology that have become more refined and specialized over the years, and stem cells address many limitations in cell source, expansion, and development of bioengineered tissue constructs. While bioprinted stem cells present an opportunity to replicate physiological microenvironments with precision, the future of this practice relies heavily on the optimization of the cellular microenvironment. To fabricate tissue constructs that are useful in replicating physiological conditions in laboratory settings, or in preparation for transplantation to a living host, the microenvironment must mimic conditions that allow bioprinted stem cells to proliferate, differentiate, and migrate. The advances of bioprinting stem cells and directing cell fate have the potential to provide feasible and translatable approach to creating complex tissues and organs. This review will examine the methods through which bioprinted stem cells are differentiated into desired cell lineages through biochemical, biological, and biomechanical techniques.
Collapse
Affiliation(s)
- Lauren N. West-Livingston
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Jihoon Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
34
|
Beylerli OA, Azizova ST, Konovalov NA, Akhmedov AD, Gareev IF, Belogurov AA. [Non-coding RNAs as therapeutic targets in spinal cord injury]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2020; 84:104-110. [PMID: 32759933 DOI: 10.17116/neiro202084031104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Spinal cord injury (SCI) may be followed by persistent motor dysfunction and somatosensory disturbances that negatively influences the quality of life of patients and creates a significant economic burden. Analysis of secondary biological processes associated with changes in genetic expression is becoming increasingly important every day in understanding the pathophysiology of spinal cord injury. The results of international sequencing of the human genome were analyzed in 2004. These data revealed about 20,000 protein-coding genes covering near 2% of the total genomic sequence. The vast majority of gene transcripts are actually characterized as non-coding RNAs (ncRNAs). These RNA clusters do not encode functional proteins and ensure post-transcriptional regulation of gene expression. The clusters may be small (approximately 20 nucleotides) known as miRNAs or the transcripts can enroll over 200 nucleotides defined as long non-coding RNAs (lncRNAs). Some modern studies describe transient expression of microRNA in case of spinal cord injury. These RNAs are associated with inflammation and apoptosis, functional recovery and regeneration. Large-scale genomic analysis has demonstrated the existence of multiple lncRNAs whose expression is associated with some processes of spinal cord injury. lncRNA can be divided into two categories depending on the position in relation to the coding genes: intergenic and intragenic. Intergenic lncRNAs is currently the most studied class. Intragenic lncRNAs can be subdivided depending on the overlap of the coding genes (antisense, intron, etc.). According to recent studies, long non-coding RNAs are abundantly present in the tissues of central nervous system and may be crucial in the pathogenesis of certain diseases of nervous system. At the cellular level, it has been shown that lncRNAs regulate the expression of protein-coding RNAs. Moreover, these molecules are involved into such processes as neuronal death, demyelination and glia activation. This review is devoted to the role of ncRNAs in the pathogenesis of spinal cord injury and their potential use as targets for the treatment of consequences of spinal cord injury.
Collapse
Affiliation(s)
- O A Beylerli
- Bashkir State Medical University of the Ministry of Health of the Russian Federation, Ufa, Russia
| | - Sh T Azizova
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | - I F Gareev
- Bashkir State Medical University of the Ministry of Health of the Russian Federation, Ufa, Russia
| | - A A Belogurov
- Shemyakin-Ovcinnicov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
35
|
Kitatani Y, Tezuka A, Hasegawa E, Yanagi S, Togashi K, Tsuji M, Kondo S, Parrish JZ, Emoto K. Drosophila miR-87 promotes dendrite regeneration by targeting the transcriptional repressor Tramtrack69. PLoS Genet 2020; 16:e1008942. [PMID: 32764744 PMCID: PMC7439810 DOI: 10.1371/journal.pgen.1008942] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/19/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022] Open
Abstract
To remodel functional neuronal connectivity, neurons often alter dendrite arbors through elimination and subsequent regeneration of dendritic branches. However, the intrinsic mechanisms underlying this developmentally programmed dendrite regeneration and whether it shares common machinery with injury-induced regeneration remain largely unknown. Drosophila class IV dendrite arborization (C4da) sensory neurons regenerate adult-specific dendrites after eliminating larval dendrites during metamorphosis. Here we show that the microRNA miR-87 is a critical regulator of dendrite regeneration in Drosophila. miR-87 knockout impairs dendrite regeneration after developmentally-programmed pruning, whereas miR-87 overexpression in C4da neurons leads to precocious initiation of dendrite regeneration. Genetic analyses indicate that the transcriptional repressor Tramtrack69 (Ttk69) is a functional target for miR-87-mediated repression as ttk69 expression is increased in miR-87 knockout neurons and reducing ttk69 expression restores dendrite regeneration to mutants lacking miR-87 function. We further show that miR-87 is required for dendrite regeneration after acute injury in the larval stage, providing a mechanistic link between developmentally programmed and injury-induced dendrite regeneration. These findings thus indicate that miR-87 promotes dendrite regrowth during regeneration at least in part through suppressing Ttk69 in Drosophila sensory neurons and suggest that developmental and injury-induced dendrite regeneration share a common intrinsic mechanism to reactivate dendrite growth. Dendrites are the primary sites for synaptic and sensory inputs. To remodel or repair neuronal connectivity, dendrites often exhibit large-scale structural changes that can be triggered by developmental signals, alterations in sensory inputs, or injury. Despite the importance of dendritic remodeling to nervous system function, the molecular basis for this remodeling is largely unknown. Here we used an unbiased genetic screen and in vivo imaging in Drosophila sensory neurons to demonstrate that the microRNA miR-87 is a critical factor required in neurons to reactivate dendritic growth both in developmental remodeling and following injury. Our work supports the model that miR-87 promotes dendrite regeneration by blocking expression of the transcriptional repressor Tramtrack69 in neurons. This study thus establishes a role for miRNAs in temporal control of dendrite regeneration.
Collapse
Affiliation(s)
- Yasuko Kitatani
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Akane Tezuka
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Eri Hasegawa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Satoyoshi Yanagi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Kazuya Togashi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Masato Tsuji
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Shu Kondo
- Genetic Strains Research Center, National Institute of Genetics, Yata, Mishima, Shizuoka, Japan
| | - Jay Z. Parrish
- Department of Biology, University of Washington, Seattle, Washington, United States of America
- * E-mail: (JZP); (KE)
| | - Kazuo Emoto
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- * E-mail: (JZP); (KE)
| |
Collapse
|
36
|
Abo-Al-Ela HG, Burgos-Aceves MA. Exploring the role of microRNAs in axolotl regeneration. J Cell Physiol 2020; 236:839-850. [PMID: 32638401 DOI: 10.1002/jcp.29920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/30/2020] [Accepted: 06/21/2020] [Indexed: 12/13/2022]
Abstract
The axolotl, Ambystoma mexicanum, is used extensively for research in developmental biology, particularly for its ability to regenerate and restore lost organs, including in the nervous system, to full functionality. Regeneration in mammals typically depends on the healing process and scar formation with limited replacement of lost tissue. Other organisms, such as spiny mice (Acomys cahirinus), salamanders, and zebrafish, are able to regenerate some damaged body components. Blastema is a tissue that is formed after tissue injury in such organisms and is composed of progenitor cells or dedifferentiated cells that differentiate into various cell types during regeneration. Thus, identifying the molecules responsible for initiation of blastema formation is an important aspect for understanding regeneration. Introns, a major source of noncoding RNAs (ncRNAs), have characteristic sizes in the axolotl, particularly in genes associated with development. These ncRNAs, particularly microRNAs (miRNAs), exhibit dynamic regulation during regeneration. These miRNAs play an essential role in timing and control of gene expression to order and organize processes necessary for blastema creation. Master keys or molecules that underlie the remarkable regenerative abilities of the axolotl remain to be fully explored and exploited. Further and ongoing research on regeneration promises new knowledge that may allow improved repair and renewal of human tissues.
Collapse
Affiliation(s)
- Haitham G Abo-Al-Ela
- Department of Aquaculture, Faculty of Fish Resources, Suez University, Suez, Egypt
| | - Mario A Burgos-Aceves
- Department of Chemistry and Biology, University of Salerno, Fisciano, Salerno, Italy
| |
Collapse
|
37
|
Danilov CA, Gu Y, Punj V, Wu Z, Steward O, Schönthal AH, Tahara SM, Hofman FM, Chen TC. Intravenous delivery of microRNA-133b along with Argonaute-2 enhances spinal cord recovery following cervical contusion in mice. Spine J 2020; 20:1138-1151. [PMID: 32145360 DOI: 10.1016/j.spinee.2020.02.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Acute spinal cord injury (SCI) is a devastating condition for which spine decompression and stabilization of injury remains the only therapy available in the clinical setup. However, fibrous scar formation during the healing process significantly impairs full recovery. MicroRNAs (miRs) are small noncoding RNAs that regulate gene expression by binding to target mRNA(s) and initiating translational repression or mRNA degradation. It has been reported that microRNA-133b (miR133b) is highly expressed in regenerating neurons following a SCI in zebrafish, and lentiviral delivery of miR133b at the time of SCI in mice resulted in improved functional recovery. PURPOSE The aim of this study was to investigate whether intravenous delivery of miR133b enhances spinal cord recovery when administered 24 hours following a cervical contusion injury in mice. STUDY DESIGN This is an experimental animal study of acute SCI, investigating the effect of miR133b on spinal cord recovery by targeting scar lesion formation. The approach involved setting an acute SCI in mice, which was followed 24 hours later by intravenous co-delivery of miR133b and Argonaute 2 (Ago2), a protein involved in miRNA stabilization. Readouts of the impact of this intervention included analysis of RNA and protein expression at the lesion site, in particular with regard to markers of scar tissue formation, and determination of motor function recovery by the grip strength meter task. METHODS C57BL6 female mice between 6 and 8 weeks of age were tested. The injury model employed was a unilateral moderate contusion at the cervical fifth level. Twenty-four hours following the injury, the authors co-delivered miR133b, or scrambled miRNA as negative control, along with Ago2 for 3 consecutive days, one dose per day via tail-vein injection. They first investigated the level of miR133b in the spinal cord and in spinal cord lesion after a single dose of injection. Next, they determined the efficacy of miR133b and/or Ago2 delivery in regulating gene and protein expression at the lesion site. Finally, they established the role of miR133b and/or Ago2 in enhancing forelimb gripping recovery as assessed by the grip strength meter task for 8 weeks post-SCI. RESULTS Intravenous delivery of miR133b and/or Ago2 targeted the microenvironment at the lesion site and prevented the increased expression of certain extracellular matrix proteins (ECM), in particular collagen type 1 alpha 1 and tenascin N, which are known to have a key role in scar formation. It also reduced microglia and/or macrophage recruitment to the lesion site. Functional recovery in mice treated with miR133b and/or Ago2 started around 2 weeks postinjury and continued to improve over time, whereas mice in the control group displayed significantly poorer recovery. CONCLUSIONS Our data indicate therapeutic activity of intravenous miR133b and/or Ago2 treatment, possibly via decreasing ECM protein expression and macrophage recruitment at the lesion site, thereby minimizing detrimental fibrous scar formation. CLINICAL SIGNIFICANCE There is an urgent medical need for better treatments of SCIs. Based on our findings in a preclinical model, the miR133b and/or Ago2 system specifically targets fibrous scar formation, a barrier in neuronal regrowth, by remodeling ECM molecules at the injury site. Prevention of scar formation is critical to improved outcomes of treatment. Of note, delivery of miR133b and/or Ago2 was initiated 24 hours after traumatic impact, thus indicating a fairly long window of opportunity providing more time and flexibility for therapeutic intervention. Intravenous miR133b may become a beneficial therapeutic strategy to treat patients with acute SCI.
Collapse
Affiliation(s)
- Camelia A Danilov
- Department of Neurological Surgery, University of Southern California, 2011 Zonal Ave, HMR 414, Los Angeles, CA 90033, USA
| | - Yifei Gu
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Rd, Shanghai, China
| | - Vasu Punj
- Department of Medicine, University of Southern California, Health Sciences Campus, NRT G511, Los Angeles, CA 90033, USA
| | - Zhourui Wu
- Division of Spine, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education of the People's Republic of China, Shanghai 200072, China
| | - Oswald Steward
- Department of Anatomy and Neurobiology, University of California, Irvine, 1105 GNRF, Irvine, CA 92697, USA
| | - Axel H Schönthal
- Department of Molecular Microbiology and Immunology, University of Southern California, 2011 Zonal Ave, HMR 405A, Los Angeles, CA 90033, USA
| | - Stanley M Tahara
- Department of Molecular Microbiology and Immunology, University of Southern California, 2011 Zonal Ave, HMR 510A, Los Angeles, CA 90033, USA
| | - Florence M Hofman
- Department of Pathology, University of Southern California, 2011 Zonal Ave, HMR 315, Los Angeles, CA 90033, USA
| | - Thomas C Chen
- Department of Neurological Surgery, University of Southern California, 1520 San Pablo St, Los Angeles, CA 90089, USA.
| |
Collapse
|
38
|
Cigliola V, Becker CJ, Poss KD. Building bridges, not walls: spinal cord regeneration in zebrafish. Dis Model Mech 2020; 13:13/5/dmm044131. [PMID: 32461216 PMCID: PMC7272344 DOI: 10.1242/dmm.044131] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury is a devastating condition in which massive cell death and disruption of neural circuitry lead to long-term chronic functional impairment and paralysis. In mammals, spinal cord tissue has minimal capacity to regenerate after injury. In stark contrast, the regeneration of a completely transected spinal cord and accompanying reversal of paralysis in adult zebrafish is arguably one of the most spectacular biological phenomena in nature. Here, we review reports from the last decade that dissect the mechanisms of spinal cord regeneration in zebrafish. We highlight recent progress as well as areas requiring emphasis in a line of study that has great potential to uncover strategies for human spinal cord repair. Summary: Unlike mammals, teleost fish are capable of efficient, spontaneous recovery after a paralyzing spinal cord injury. Here, we highlight the major events through which laboratory model zebrafish regenerate spinal cord tissue.
Collapse
Affiliation(s)
- Valentina Cigliola
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.,Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Clayton J Becker
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.,Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA .,Regeneration Next, Duke University, Durham, NC 27710, USA
| |
Collapse
|
39
|
Mesenchymal and Induced Pluripotent Stem Cells-Derived Extracellular Vesicles: The New Frontier for Regenerative Medicine? Cells 2020; 9:cells9051163. [PMID: 32397132 PMCID: PMC7290733 DOI: 10.3390/cells9051163] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine aims to repair damaged, tissues or organs for the treatment of various diseases, which have been poorly managed with conventional drugs and medical procedures. To date, multimodal regenerative methods include transplant of healthy organs, tissues, or cells, body stimulation to activate a self-healing response in damaged tissues, as well as the combined use of cells and bio-degradable scaffold to obtain functional tissues. Certainly, stem cells are promising tools in regenerative medicine due to their ability to induce de novo tissue formation and/or promote organ repair and regeneration. Currently, several studies have shown that the beneficial stem cell effects, especially for mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs) in damaged tissue restore are not dependent on their engraftment and differentiation on the injury site, but rather to their paracrine activity. It is now well known that paracrine action of stem cells is due to their ability to release extracellular vesicles (EVs). EVs play a fundamental role in cell-to-cell communication and are directly involved in tissue regeneration. In the present review, we tried to summarize the molecular mechanisms through which MSCs and iPSCs-derived EVs carry out their therapeutic action and their possible application for the treatment of several diseases.
Collapse
|
40
|
Harris MP, Daane JM, Lanni J. Through veiled mirrors: Fish fins giving insight into size regulation. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e381. [PMID: 32323915 DOI: 10.1002/wdev.381] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/13/2020] [Accepted: 03/19/2020] [Indexed: 12/25/2022]
Abstract
Faithful establishment and maintenance of proportion is seen across biological systems and provides a glimpse at fundamental rules of scaling that underlie development and evolution. Dysregulation of proportion is observed in a range of human diseases and growth disorders, indicating that proper scaling is an essential component of normal anatomy and physiology. However, when viewed through an evolutionary lens, shifts in the regulation of relative proportion are one of the most striking sources of morphological diversity among organisms. To date, the mechanisms via which relative proportion is specified and maintained remain unclear. Through the application of powerful experimental, genetic and molecular approaches, the teleost fin has provided an effective model to investigate the regulation of scaling, size, and relative growth in vertebrate organisms. This article is categorized under: Establishment of Spatial and Temporal Patterns > Regulation of Size, Proportion, and Timing Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Comparative Development and Evolution > Regulation of Organ Diversity.
Collapse
Affiliation(s)
- Matthew P Harris
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob M Daane
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
41
|
Mégret L, Nair SS, Dancourt J, Aaronson J, Rosinski J, Neri C. Combining feature selection and shape analysis uncovers precise rules for miRNA regulation in Huntington's disease mice. BMC Bioinformatics 2020; 21:75. [PMID: 32093602 PMCID: PMC7041117 DOI: 10.1186/s12859-020-3418-9] [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: 01/29/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background MicroRNA (miRNA) regulation is associated with several diseases, including neurodegenerative diseases. Several approaches can be used for modeling miRNA regulation. However, their precision may be limited for analyzing multidimensional data. Here, we addressed this question by integrating shape analysis and feature selection into miRAMINT, a methodology that we used for analyzing multidimensional RNA-seq and proteomic data from a knock-in mouse model (Hdh mice) of Huntington’s disease (HD), a disease caused by CAG repeat expansion in huntingtin (htt). This dataset covers 6 CAG repeat alleles and 3 age points in the striatum and cortex of Hdh mice. Results Remarkably, compared to previous analyzes of this multidimensional dataset, the miRAMINT approach retained only 31 explanatory striatal miRNA-mRNA pairs that are precisely associated with the shape of CAG repeat dependence over time, among which 5 pairs with a strong change of target expression levels. Several of these pairs were previously associated with neuronal homeostasis or HD pathogenesis, or both. Such miRNA-mRNA pairs were not detected in cortex. Conclusions These data suggest that miRNA regulation has a limited global role in HD while providing accurately-selected miRNA-target pairs to study how the brain may compute molecular responses to HD over time. These data also provide a methodological framework for researchers to explore how shape analysis can enhance multidimensional data analytics in biology and disease.
Collapse
Affiliation(s)
- Lucile Mégret
- Sorbonne Université, CNRS UMR8256, INSERM ERL U1164, Brain-C Lab, Paris, France.
| | | | - Julia Dancourt
- Sorbonne Université, CNRS UMR8256, INSERM ERL U1164, Brain-C Lab, Paris, France
| | | | | | - Christian Neri
- Sorbonne Université, CNRS UMR8256, INSERM ERL U1164, Brain-C Lab, Paris, France.
| |
Collapse
|
42
|
Li JH, Shi ZJ, Li Y, Pan B, Yuan SY, Shi LL, Hao Y, Cao FJ, Feng SQ. Bioinformatic identification of key candidate genes and pathways in axon regeneration after spinal cord injury in zebrafish. Neural Regen Res 2020; 15:103-111. [PMID: 31535658 PMCID: PMC6862403 DOI: 10.4103/1673-5374.264460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Zebrafish and human genomes are highly homologous; however, despite this genomic similarity, adult zebrafish can achieve neuronal proliferation, regeneration and functional restoration within 6–8 weeks after spinal cord injury, whereas humans cannot. To analyze differentially expressed zebrafish genes between axon-regenerated neurons and axon-non-regenerated neurons after spinal cord injury, and to explore the key genes and pathways of axonal regeneration after spinal cord injury, microarray GSE56842 was analyzed using the online tool, GEO2R, in the Gene Expression Omnibus database. Gene ontology and protein-protein interaction networks were used to analyze the identified differentially expressed genes. Finally, we screened for genes and pathways that may play a role in spinal cord injury repair in zebrafish and mammals. A total of 636 differentially expressed genes were obtained, including 255 up-regulated and 381 down-regulated differentially expressed genes in axon-regenerated neurons. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment results were also obtained. A protein-protein interaction network contained 480 node genes and 1976 node connections. We also obtained the 10 hub genes with the highest correlation and the two modules with the highest score. The results showed that spectrin may promote axonal regeneration after spinal cord injury in zebrafish. Transforming growth factor beta signaling may inhibit repair after spinal cord injury in zebrafish. Focal adhesion or tight junctions may play an important role in the migration and proliferation of some cells, such as Schwann cells or neural progenitor cells, after spinal cord injury in zebrafish. Bioinformatic analysis identified key candidate genes and pathways in axonal regeneration after spinal cord injury in zebrafish, providing targets for treatment of spinal cord injury in mammals.
Collapse
Affiliation(s)
- Jia-He Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhong-Ju Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bin Pan
- Department of Orthopedics, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Shi-Yang Yuan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Lin-Lin Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Hao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fu-Jiang Cao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shi-Qing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
43
|
Zhang BY, Chang PY, Zhu QS, Zhu YH. Decoding epigenetic codes: new frontiers in exploring recovery from spinal cord injury. Neural Regen Res 2020; 15:1613-1622. [PMID: 32209760 PMCID: PMC7437595 DOI: 10.4103/1673-5374.276323] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury that results in severe neurological disability is often incurable. The poor clinical outcome of spinal cord injury is mainly caused by the failure to reconstruct the injured neural circuits. Several intrinsic and extrinsic determinants contribute to this inability to reconnect. Epigenetic regulation acts as the driving force for multiple pathological and physiological processes in the central nervous system by modulating the expression of certain critical genes. Recent studies have demonstrated that post-SCI alteration of epigenetic landmarks is strongly associated with axon regeneration, glial activation and neurogenesis. These findings not only establish a theoretical foundation for further exploration of spinal cord injury, but also provide new avenues for the clinical treatment of spinal cord injury. This review focuses on the epigenetic regulation in axon regeneration and secondary spinal cord injury. Together, these discoveries are a selection of epigenetic-based prognosis biomarkers and attractive therapeutic targets in the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Bo-Yin Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Peng-Yu Chang
- Department of Radiotherapy, The First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qing-San Zhu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yu-Hang Zhu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | -
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
44
|
Almurshidi B, Carver W, Scott G, Ray SK. Roles of miRNAs in spinal cord injury and potential therapeutic interventions. NEUROIMMUNOLOGY AND NEUROINFLAMMATION 2019; 6:11. [PMID: 33869675 PMCID: PMC8052101 DOI: 10.20517/2347-8659.2019.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Spinal cord injury (SCI) affects approximately 200,000 individuals per year worldwide. There are more than 27 million people worldwide living with long-term disability due to SCI. Historically, it was thought that the central nervous system (CNS) had little ability for regeneration; however, more recent studies have demonstrated potential for repair within the CNS. Because of this, there exists a renewed interest in the discovery of novel approaches to promote regeneration in the CNS including the spinal cord. It is important to know the roles of the microRNAs (miRNAs) in modulation of pathogenesis in SCI and the potentials of the miRNA-based clinical interventions for controlling post-injury symptoms and improving functional recovery. The miRNAs, which are non-coding RNAs with an average of 22 nucleotides in length, are post-transcriptional gene regulators that cause degradation of the target mRNAs and thus negatively control their translation. This review article focuses on current research related to miRNAs and their roles in modulating SCI symptoms, asserting that miRNAs contribute to critical post-SCI molecular processes including neuroplasticity, functional recovery, astrogliosis, neuropathic pain, inflammation, and apoptosis. In particular, miR-96 provides a promising therapeutic opportunity to improve the outcomes of clinical interventions, including the way SCI injuries are evaluated and treated.
Collapse
Affiliation(s)
- Badria Almurshidi
- Department of Environmental Health Sciences, Arnold School of Public Health, CENR, University of South Carolina, Columbia, SC 29209, USA
| | - Wayne Carver
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Geoff Scott
- Department of Environmental Health Sciences, Arnold School of Public Health, CENR, University of South Carolina, Columbia, SC 29209, USA
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| |
Collapse
|
45
|
MicroRNA-31 regulating apoptosis by mediating the phosphatidylinositol-3 kinase/protein kinase B signaling pathway in treatment of spinal cord injury. Brain Dev 2019; 41:649-661. [PMID: 31036380 DOI: 10.1016/j.braindev.2019.04.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/01/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022]
Abstract
Apoptosis is a highly conservative energy demand program for non-inflammatory cell death, which is extremely significant in normal physiology and disease. There are many techniques used for studying apoptosis. MicroRNA (miRNA) is closely related to cell apoptosis, and especially microRNA-31 (miR-31) is involved in apoptosis by regulating a large number of target genes and signaling pathways. In many neurological diseases, cell apoptosis or programmed cell death plays an important role in the reduction of cell number, including the reduction of neurons in spinal cord injuries. In recent years, the phosphoinositol 3-kinase/AKT (PI3K/AKT) signal pathway, as a signal pathway involved in a variety of cell functions, has been studied in spinal cord injury diseases. The PI3K/AKT pathway directly or indirectly affects whether apoptosis occurs in a cell, thereby affecting a significant intracellular event sequence. This paper reviewed the interactions of miR-31 target sites in the PI3K/AKT signaling pathway, and explored new ways to prevent and treat spinal cord injury by regulating the effect of miR-31 on apoptosis.
Collapse
|
46
|
Ceci M, Mariano V, Romano N. Zebrafish as a translational regeneration model to study the activation of neural stem cells and role of their environment. Rev Neurosci 2019; 30:45-66. [PMID: 30067512 DOI: 10.1515/revneuro-2018-0020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
The review is an overview of the current knowledge of neuronal regeneration properties in mammals and fish. The ability to regenerate the damaged parts of the nervous tissue has been demonstrated in all vertebrates. Notably, fish and amphibians have the highest capacity for neurogenesis, whereas reptiles and birds are able to only regenerate specific regions of the brain, while mammals have reduced capacity for neurogenesis. Zebrafish (Danio rerio) is a promising model of study because lesions in the brain or complete cross-section of the spinal cord are followed by an effective neuro-regeneration that successfully restores the motor function. In the brain and the spinal cord of zebrafish, stem cell activity is always able to re-activate the molecular programs required for central nervous system regeneration. In mammals, traumatic brain injuries are followed by reduced neurogenesis and poor axonal regeneration, often insufficient to functionally restore the nervous tissue, while spinal injuries are not repaired at all. The environment that surrounds the stem cell niche constituted by connective tissue and stimulating factors, including pro-inflammation molecules, seems to be a determinant in triggering stem cell proliferation and/or the trans-differentiation of connective elements (mainly fibroblasts). Investigating and comparing the neuronal regeneration in zebrafish and mammals may lead to a better understanding of the mechanisms behind neurogenesis, and the failure of the regenerative response in mammals, first of all, the role of inflammation, considered the main inhibitor of the neuronal regeneration.
Collapse
Affiliation(s)
- Marcello Ceci
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| | - Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicla Romano
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| |
Collapse
|
47
|
Abstract
Nerve injury-induced neuropathic pain is difficult to treat. In this study, we used exosomes derived from human umbilical cord mesenchymal stem cell (UCMSC) as a cell-free therapy for nerve injury-induced pain in rats. Isolated UCMSC exosomes range in size from 30 to 160 nm and contain CD63, HSP60, and CD81 exosome markers. After L5/6 spinal nerve ligation surgery, single intrathecal injection of exosomes reversed nerve ligation-induced mechanical and thermal hypersensitivities of right hindpaw of rats at initial and well-developed pain stages. Moreover, continuous intrathecal infusion of exosomes achieved excellent preventive and reversal effects for nerve ligation-induced pain. In immunofluorescent study, lots of Exo-green-labelled exosomes could be found majorly in the ipsilateral L5 spinal dorsal horn, dorsal root ganglion, and peripheral axons, suggesting the homing ability of UCMSC exosomes. They also appeared in the central terminals or cell bodies of IB4, CGRP, and NF200 sensory neurons. In addition, exosome treatment suppressed nerve ligation-induced upregulation of c-Fos, CNPase, GFAP, and Iba1. All these data suggest that the analgesic effects of exosomes may involve their actions on neuron and glial cells. Exosomes also inhibited the level of TNF-α and IL-1β, while enhanced the level of IL-10, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor in the ipsilateral L5/6 dorsal root ganglion of nerve-ligated rats, indicating anti-inflammatory and proneurotrophic abilities. Protein analysis revealed the content of vascular endothelial growth factor C, angiopoietin-2, and fibroblast growth factor-2 in the exosomes. In summary, intrathecal infusion of exosomes from UCMSCs may be considered as a novel therapeutic approach for nerve injury-induced pain.
Collapse
|
48
|
Ghoreishy A, Khosravi A, Ghaemmaghami A. Exosomal microRNA and stroke: A review. J Cell Biochem 2019; 120:16352-16361. [PMID: 31219202 DOI: 10.1002/jcb.29130] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/20/2022]
Abstract
Blood vessels rupture or occlusion in brain results in stroke. Stroke is the major reason for mortality and dysfunction worldwide. Despite several attempts, there are no any approved therapeutic approaches for stroke subjects. The most neuroprotective agents showed the positive effects in preclinical reports, while there are no significant therapeutic impacts in the clinical trials. MicroRNAs (miRNAs) are small noncoding RNAs which involved in the modulation of a variety of cellular and molecular pathways. Given that deregulation of these molecules is related to initiation and progression of stroke. Exosomes are nano-carriers which are able to transfer different cargos such as miRNAs to recipient cells. Increasing evidence revealed that exosomal miRNAs are one of very important factors which are involved in the pathogenesis of stroke. Hence, more understanding about the role of exosomal miRNAs in stroke pathogenesis could contribute in discovering and developing new therapeutic approaches. Moreover, it has been proved the exosomal miRNAs could be used as noninvasive biomarkers in diagnosis and monitoring response to therapy in subjects with stroke. Herein for first time, we summarized different exosomal miRNAs involved in pathogenesis of stroke.
Collapse
Affiliation(s)
- Abdolreza Ghoreishy
- Department of Neurology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Alireza Khosravi
- Department of Neurology, Clinical Immunology Research Center, School of Medicine, Zahedan University of Medical Science, Zahedan, Iran
| | - Amir Ghaemmaghami
- Department of Psychology, Behaviour, Genetics and Neurobiology Program, University of Toronto, Toronto, Canada
| |
Collapse
|
49
|
Tigchelaar S, Gupta R, Shannon CP, Streijger F, Sinha S, Flibotte S, Rizzuto MA, Street J, Paquette S, Ailon T, Charest-Morin R, Dea N, Fisher C, Dvorak MF, Dhall S, Mac-Thiong JM, Parent S, Bailey C, Christie S, Van Keuren-Jensen K, Nislow C, Kwon BK. MicroRNA Biomarkers in Cerebrospinal Fluid and Serum Reflect Injury Severity in Human Acute Traumatic Spinal Cord Injury. J Neurotrauma 2019; 36:2358-2371. [PMID: 30827169 DOI: 10.1089/neu.2018.6256] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition with variability in injury mechanisms and neurologic recovery. Spinal cord impairment after SCI is measured and classified by a widely accepted standard neurological examination. In the very acute stages post-injury, however, this examination is extremely challenging (and often impossible) to conduct and has modest prognostic value in terms of neurological recovery. The lack of objective tools to classify injury severity and predict outcome is a barrier for clinical trials and thwarts development of therapies for those with SCI. Biological markers (biomarkers) represent a promising, complementary approach to these challenges because they represent an unbiased approach to classify injury severity and predict neurological outcome. Identification of a suitable panel of molecular biomarkers would comprise a fundamental shift in how patients with acute SCI are evaluated, stratified, and treated in clinical trials. MicroRNA are attractive biomarker candidates in neurological disorders for several reasons, including their stability in biological fluids, their conservation between humans and model mammals, and their tissue specificity. In this study, we used next-generation sequencing to identify microRNA associated with injury severity within the cerebrospinal fluid (CSF) and serum of human patients with acute SCI. The CSF and serum samples were obtained 1-5 days post-injury from 39 patients with acute SCI (24 American Spinal Injury Association Impairment Scale [AIS] A, 8 AIS B, 7 AIS C) and from five non-SCI controls. We identified a severity-dependent pattern of change in microRNA expression in CSF and identified a set of microRNA that are diagnostic of baseline AIS classification and prognostic of neurological outcome six months post-injury. The data presented here provide a comprehensive description of the CSF and serum microRNA expression changes that occur after acute human SCI. This data set reveals microRNA candidates that warrant further evaluation as biomarkers of injury severity after SCI and as key regulators in other neurological disorders.
Collapse
Affiliation(s)
- Seth Tigchelaar
- 1International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Rishab Gupta
- 1International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Casey P Shannon
- 2Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
| | - Femke Streijger
- 1International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Sunita Sinha
- 3Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephane Flibotte
- 3Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael A Rizzuto
- 1International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - John Street
- 4Department of Orthopedics, Division of Spine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Scott Paquette
- 5Division of Neurosurgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tamir Ailon
- 5Division of Neurosurgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Raphaele Charest-Morin
- 4Department of Orthopedics, Division of Spine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicolas Dea
- 5Division of Neurosurgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Charles Fisher
- 4Department of Orthopedics, Division of Spine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marcel F Dvorak
- 1International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada.,4Department of Orthopedics, Division of Spine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sanjay Dhall
- 6Department of Neurosurgery, University of California San Francisco, San Francisco, California
| | | | - Stefan Parent
- 8Department of Surgery, Chu Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Christopher Bailey
- 9Division of Orthopaedic Surgery, Schulich Medicine & Dentistry, Victoria Hospital, London, Ontario, Canada
| | - Sean Christie
- 10Division of Neurosurgery, Halifax Infirmary, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Corey Nislow
- 3Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian K Kwon
- 1International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada.,4Department of Orthopedics, Division of Spine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
50
|
Extracellular Vesicle-Mediated Cell⁻Cell Communication in the Nervous System: Focus on Neurological Diseases. Int J Mol Sci 2019; 20:ijms20020434. [PMID: 30669512 PMCID: PMC6359416 DOI: 10.3390/ijms20020434] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/12/2019] [Accepted: 01/17/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes, are membranous particles released by cells into the extracellular space. They are involved in cell differentiation, tissue homeostasis, and organ remodelling in virtually all tissues, including the central nervous system (CNS). They are secreted by a range of cell types and via blood reaching other cells whose functioning they can modify because they transport and deliver active molecules, such as proteins of various types and functions, lipids, DNA, and miRNAs. Since they are relatively easy to isolate, exosomes can be characterized, and their composition elucidated and manipulated by bioengineering techniques. Consequently, exosomes appear as promising theranostics elements, applicable to accurately diagnosing pathological conditions, and assessing prognosis and response to treatment in a variety of disorders. Likewise, the characteristics and manageability of exosomes make them potential candidates for delivering selected molecules, e.g., therapeutic drugs, to specific target tissues. All these possible applications are pertinent to research in neurophysiology, as well as to the study of neurological disorders, including CNS tumors, and autoimmune and neurodegenerative diseases. In this brief review, we discuss what is known about the role and potential future applications of exosomes in the nervous system and its diseases, focusing on cell–cell communication in physiology and pathology.
Collapse
|